Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Mar 1.
Published in final edited form as: Biotechnol Adv. 2014 Jan 16;32(2):504–513. doi: 10.1016/j.biotechadv.2014.01.003

Pluripotent Stem Cell-Derived Hepatocyte-Like Cells

R E Schwartz 1,3,5, HE Fleming 1,3, S N Bhatia 1,2,3,4,5,+
PMCID: PMC4043206  NIHMSID: NIHMS572864  PMID: 24440487

Abstract

Liver disease is an important clinical problem, impacting over 30 million Americans and over 600 million people worldwide. It is the 12th leading cause of death in the United States and the 16th worldwide. Due to a paucity of donor organs, several thousand Americans die yearly while waiting for liver transplantation. Unfortunately, alternative tissue sources such as fetal hepatocytes and hepatic cell lines are unreliable, difficult to reproduce, and do not fully recapitulate hepatocyte phenotype and function. As a consequence, alternative cell sources that do not have these limitations have been sought. Human embryonic stem (hES) cell- and induced pluripotent stem (iPS) cell-derived hepatocyte-like cells may enable cell based therapeutics, the study of the mechanisms of human disease and human development, and provide a platform for pharmacology and toxicology drug screening. iPS cells can be differentiated in a stepwise fashion with high efficiency and reproducibility into hepatocyte-like cells that exhibit morphologic and phenotypic characteristics of hepatocytes. In addition, iPS-derived hepatocyte-like cells possess some functional hepatic activity as they secrete urea, alpha-1-antitrypsin, and albumin. However, the combined phenotypic and functional traits exhibited by iPS-derived hepatocyte-like cells resemble a relatively immature hepatic phenotype that more closely resembles that of fetal hepatocytes rather than adult hepatocytes. Specifically, iPS-derived hepatocyte-like cells express fetal markers such as alpha fetoprotein and lack key mature hepatocyte functions, as reflected by drastically reduced activity (0.1%) of many detoxification enzymes (i.e. CYP2A6, CYP3A4). These key differences between iPS-derived hepatocyte-like cells and adult hepatocytes have limited the use of stem cells as a renewable source of functional adult human hepatocytes for in vitro and in vivo applications. Unfortunately, the developmental pathways that control hepatocyte maturation from a fetal into an adult hepatocyte are poorly understood, which has hampered the field in its efforts to induce further maturation of iPS-derived hepatic lineage cells. This review analyzes recent developments in the derivation of hepatocyte-like cells, and proposes important points to consider and assays to perform during their characterization. In the future, we envision that iPS-derived hepatocyte-like cells will be used as in vitro models of human disease, and in the longer term, provide an alternative cell source for drug testing and clinical therapy.

INTRODUCTION

Chronic liver disease is a significant cause of morbidity and mortality, impacting over 600 million people worldwide [1]. As a result, the number of people living with end stage liver disease is increasing, and over 1 million people die each year from acute and chronic liver disease across the globe [1]. Liver transplantation is currently the only definitive and curative treatment for acute and chronic liver failure [2]. First accomplished in 1967 by Thomas Starzl, liver transplantation has been an unquestioned clinical success; however, the demand for liver transplantation has significantly outstripped the supply of donor organs [2-4]. As a consequence, multiple attempts to expand the availability of donor organs have been employed: opt-out organ donation programs, the use of suboptimal donor organs (deceased cardiac donors or steatotic (fatty) livers), split donor transplantation, and living donor liver transplantation [4].

The search for alternatives to whole organ transplantation has been focused on expanding the availability of replacement liver tissue, such as developing cell-based therapies that include hepatocyte transplantation, engineered hepatic tissue constructs, and the bioartificial liver [5-9]. In particular, hepatocyte transplantation has been performed clinically for more than 15 years, primarily in the setting of acute liver failure and inherited liver metabolic disorders. A general problem facing hepatocyte transplantation is the limited repopulation capacity of engrafted cells, although in the case of some metabolic disorders, replacement of just 2–5% of the liver parenchyma with normal hepatocytes may be sufficient to improve liver function significantly. For example, Fox et al reported the successful treatment of a 10-year-old with one such metabolic disorder, termed Crigler–Najjar disease, who was experiencing recurrent episodes of brain injury resulting from elevated bilirubin. The patient was shown to respond well to infusion of 7.5×109 hepatocytes, based on an improvement in metabolic function and reduced need for phototherapy [7]. However, hepatocyte transplantation has not been widely adopted, due to a variety of technical reasons including the inability to monitor graft health and frequent signs of rejection [8]. Moreover, these clinical treatments require human liver tissue as a cell source of the transplanted hepatocytes which, as mentioned, is in very short supply.

Based on the apparent success of hepatocyte transplantation combined with the challenges in sourcing appropriate donor cells, a strong focus has been placed on developing a safe and reliable method to expand the small number of available human hepatocytes. Indeed, the liver has been known for its capacity to regenerate since antiquity, as encapsulated in the story of Prometheus. Modern studies have shown that in vivo, human hepatocytes are capable of cellular proliferation based on the observed replacement of damaged hepatocytes following injury, or even during the daily turnover of the liver [10]. However, in vitro, researchers have been unable to induce and/or support the cellular proliferation of human hepatocytes; rather, attempts to culture human hepatocytes have led to the loss of differentiated function rather than any increase in cell number [11-12]. Consequently, attempts to expand adult human hepatocytes have been unsuccessful as a target approach to achieving cellular therapy of the liver, and alternatives are under active investigation. Other approaches include utilizing cell lines derived from hepatocellular carcinoma, or generated through SV40 or Big T antigen transformation, both of which have enabled the expansion and creation of in vitro model systems [13]. Regrettably, these cell lines poorly recapitulate hepatocyte function, as cultured cells lose most of their differentiated function and acquire genetic abnormalities.

Several alternative sources have been proposed as options to circumvent the limited supply of human hepatocytes, including using human fetal tissue or even xenogeneic material, but both paths have been sidelined due to a variety of ethical, sourcing, and safety issues [14]. While still prone to some ethical and safety challenges, pluripotent stem cell-based therapies overcome many of the drawbacks that challenge other cell lines and fetal tissue, and thus are considered by many as an ideal alternative source of human hepatocytes [15-16]. Human pluripotent stem cells include embryonic stem (hES) cells, first isolated from human blastocysts by James Thomson and colleagues [17], as well as the more recently described induced pluripotent stem (iPS) cells first generated by Yamanaka and colleagues following the forced expression of a panel of transcription factors in adult-derived cells [18-19]. These cell lines are defined as pluripotent in that they can self-renew in culture, maintain genetic stability, and differentiate into cell lineages of all three germ layers including endodermal hepatocyte-like cells (HLCs) [17-19]. Importantly, iPS cells can be derived from adult tissue in a reliable manner and have been shown to differentiate efficiently into hepatocyte-like cells [18-21]. However, unlike relatively simple in vitro models designed to reproduce embryonic differentiation, in vivo development advances through a much more complex, structured and highly organized series of patterning and differentiation events in which cell-extracellular matrix and cell-cell interactions are tightly controlled and play an important role [22-25]. Consequently, the hepatocyte-like cells generated from pluripotent stem cells in culture exhibit many morphologic and phenotypic characteristics of human hepatocytes. However, the examination of their functional traits has been more limited, and many signs suggest that only partial differentiation has been attained, as discussed below.

Despite the challenges inherent in performing developmental studies in an in vitro setting, and the roadblocks that remain regarding the current capacity to treat patients with human hepatocytes from any available derivation source, the importance of being able to develop experimental models to study human disease states cannot be overstated. To date, many genome-wide association screens (GWAS) have identified a variety of genetic variants associated with human liver disease [26]. However, many of these variants represent novel loci whose contribution to liver disease is entirely unknown. Linking GWAS findings to biologic mechanisms has been an ongoing challenge in the genetics community. In most studies, mouse models have been employed; however, the usefulness of mouse models is unclear given its low-throughput nature and the physiologic and metabolic differences between humans and rodents [27-28]. Similarly, zebrafish models have been employed as a higher-throughput system to examine numerous genetic variants but given the significant developmental, structural, physiologic, and metabolic differences between species, the value of these models is less clear [29-30]. Consequently, cell culture systems have been employed, despite the observation that although no single cell type tested to date fully recapitulates hepatocyte morphology, phenotype, and function [31-33]. In contrast, iPS cells offer the potential to establish patient-specific cell types such as iHLCs, thus facilitating in vitro modeling of rare diseases, and may one day enable personalized medicine [18-19]. When combined with the capacity to engineer genetic changes in established iPS lines [34], patient-specific iPS cells and iHLCs can be utilized to study genetic variants identified in GWAS studies, as well as a host of other monogenic alterations to assay their impact on hepatocyte differentiation, phenotype and function [35]. For example, recent papers have demonstrated that iHLCs can recapitulate the disease phenotype of alpha-1-antitrypsin disease, familial hyperlipidemia, and Wilson’s disease, although the capacity to apply this approach to study polygenic disorders remains to be seen [35-37]. And yet, in order to maximize the potential use of iPS-derived liver cells either for studies of disease models and treatment, or for eventual cell transplantation therapies, it is essential that efficient and reproducible iHLC differentiation protocols are established.

Significant progress has been made over the past few years in the derivation of pluripotent stem cell derived hepatocyte-like cells and consequently, a multitude of differing protocols have been developed to generate iHLCs [20-21, 38-40]. Most of these protocols share general themes regarding their approach to differentiation induction, however, specific differences are apparent upon close examination of the individual methodologies.

In this review, we overview some of the most established and cited iHLC methods. We stress the importance of detailed characterization of derived cell types, and that multiple phenotypic and functional read outs are required in this effort. Based on an analysis of the available literature, we provide a summary of assays that can be applied during the analysis of iHLC populations, towards the goal of promoting a unified field and achieving a robust, mature source of this very important and clinically relevant lineage.

RESULTS AND DISCUSSION

For decades, researchers have been attempting to promote the in vitro differentiation of primitive, and now pluripotent, cells towards specific lineages of all three germ layers. A typical approach to designing such protocols has been to mimic the patterns and stages observed during embryologic development, in order to recreate the necessary molecular and cellular cues. In the case of pluripotent cells, most protocols apply either one of several cellular aggregation strategies or promote differentiation in a monolayer culture. Pluripotent stem cells can be aggregated in suspension or using specialized plates, which results in the formation of three-dimensional structures called embryoid bodies (EBs) that may serve to replicate some of the cell-cell and cell-matrix signals that are experienced during development in vivo [41]. By culturing EBs in specific cytokine cocktails or on different extracellular matrices - again, designed in an effort to mimic signals observed during embryonic liver development - it is possible to improve the efficiency of pluripotent stem cell-derived HLC generation [42]. However, all existing protocols still suffer from relatively low differentiation efficiency, and tend to lead to the production of a variety of alternate cell lineages, likely because EBs develop regional differentiation over time in what appears to be a stochastic and spontaneous process. As a result, cultures that utilize an embryoid body step typically introduce an unpredictable degree of variability between differentiation attempts. To address this problem, several groups have either switched to a monolayer-style culture (often co-culture), and/or use hepatocyte reporter constructs that are only turned on at specific stages of development, or sort desired populations based on the expression of cell surface markers that are upregulated during the course of differentiation [43-44]. Using these protocols, relatively pure subpopulations of cells can be isolated, and which have been tested functionally in a variety of hepatocyte transplantation assays. Unfortunately, the outcome of these experiments has varied widely, and to date, no papers have demonstrated the ability to re-plate and use these cells in vitro [43].

More recently, several groups have optimized the differentiation procedure and eliminated the use of poorly defined components such as serum, fibroblast feeder cells, embryoid bodies, and other undefined culture medium components, and have optimized their protocols based on a growing understanding of mouse hepatic development. These next generation protocols typically start with pluripotent stem cells and in a stepwise manner, expose the cultured cells to a series of defined factors in order to obtain iHLCs [18-21]. According to these methods, iHLCs are obtained with a much higher efficiency than earlier aggregation protocols, although scaleability remains a challenge in light of the numbers of cells that may be required for future applications. The key to the success of these protocols was the ability to promote the pluripotent cells to adopt a definitive endoderm fate at high efficiency. First described in 2006 using embryonic stem cells, this class of protocols identified the role that activin A and WNT3 signaling play during the establishment of the early primitive streak, and that ultimately lead to endoderm specification [45-46]. These insights opened the door to further refinements in the methodology, and enabled the derivation of visceral, endodermal-derived tissues. These updated protocols yield hepatic lineage cells that can be considered hepatic-like based on morphologic and some phenotypic analyses [20-21, 38]. However, functional and other specific phenotypic analyses demonstrate that the resulting iHLCs exhibit an immature hepatic phenotype, in that they resemble fetal hepatocytes more than adult hepatocytes. Notably, iHLCs persistently express fetal markers like alpha fetoprotein (AFP) and lack key mature hepatocyte functions, as reflected by drastically reduced activity (0.1%) of many detoxification enzymes (e.g. CYP2A6, CYP3A4) [20-21, 38]. These subtle but important differences between iHLCs and adult hepatocytes have limited the use of stem cells as a renewable source of functional human hepatocytes.

Despite the development of multiple differentiation protocols it is unclear as to which protocol leads to the production of the most terminally differentiated hepatocyte-like cells. This challenge in comparison with adult-derived cells typically stems from the inclusion of only minimally detailed functional analysis. Therefore, it is important to review the various approaches that numerous labs use to validate the identity of the lineages and precursors that result from hepatic differentiation methodologies. Indeed, in order to best optimize a protocol designed to yield efficient, robust progeny of a desired lineage, it is essential that appropriate functional tests are performed, and that reasonable control and ‘benchmark’ comparisons are made as part of the refining process. As an example, we have summarized three differentiation protocols established by Si-Tayyeb et al, Song et al, and Touboul et al [20-21, 38]. All protocols share a step-wise process to generate definitive endoderm, followed by hepatic-specified endoderm, followed by hepatoblasts, and finally yield hepatocytes, although the precise culture conditions, growth factor combinations, kinetics, efficiency, and overall protocol complexity vary in each case (Figure 1). In terms of the reported characterization of the cells generated during the course of the procedures, Si-Tayyeb et al completed immunofluorescence analysis, periodic acid Schiff staining for glycogen production, LDL uptake, albumin secretion, and in vivo transplantation. Song et al completed immunofluorescence analysis, periodic acid Schiff staining for glycogen production albumin production assay, urea metabolism assay, and CYP2B analysis. Touboul et al completed immunofluorescence analysis, flow cytometry analysis, indocyanine green assay, CYP3A5/CYP3A7 (fetal cytochrome P450) analysis and in vivo transplantation. All three of these examples highlight that, particularly in the case of the hepatocyte lineage, no single assay can conclusively confirm a hepatic identity in isolation. However, as illustrated by these three publications, there are a multitude of morphologic, phenotypic, and functional tests which can be combined to demonstrate with confidence that pluripotent stem cell-derived HLCs are similar in quality and function to primary human hepatocytes. For the purposes of this review, we will describe a panel of these assays below.

Figure 1. Generation of hepatocyte-like cells from iPS cells via three stepwise protocols.

Figure 1

Schematic outlining the differentiation kinetics and growth factors utilized in three different but commonly used protocols established by Si-Tayyeb et al, Song et al, and Touboul et al [20-21, 38].

Cellular morphology

Pluripotent stem cell derived hepatocyte-like cells should be cuboidal or polygonal in appearance and have enlarged nuclei with an increased cytoplasm to nucleus ratio. In electron microscopy studies, abundant mitochondria, perioxisomes, lipid bodies, intact golgi apparati, and rough endoplasmic reticulum should be present. In addition, abundant microvilli and vesicles should be visible, and junctional complexes should be present on either side of nearby hepatocytes, consistent with bile canalicular network formation.

Gene and protein expression

In addition to the overall morphology of the population, confirming lineage identity depends on demonstrating the presence or absence of expression of particular genes and proteins. Based on developmental studies in vivo, numerous expected molecular phenotypes have been defined throughout the differentiation process as pluripotent cells transition to an adult hepatocyte state.Table 1 outlines expected expression patterns for various stages along the differentiation cascade. In terms of RNA expression analysis, the preferred methodology is to use quantitiative real-time PCR (qRT-PCR) analysis of pluripotent stem cell-derived HLCs compared to a high-quality human hepatocyte reference (see below). At each differentiation step it is important to characterize and demonstrate the commitment or specification to the specified lineage. Protein immunofluorescence or immunohistochemistry staining should be performed to confirm qRT-PCR data as well as to help determine marker co-expression. This per-cell assay also allows for an assessment of overall differentiation efficiency within a given culture well or plate. Cell polarization, an essential feature of functional hepatocytes, can also be examined in this manner, in that several proteins (BSEP, MRP-2, G6P, and several others) should only be expressed on apical or basolateral membranes. Finally, at least a selection of the imaging stains should be confirmed by Western blot of bulk populations, to decrease possibility of off-target staining results.

Table 1.

Marker expression throughout hepatocyte differentiation

Gene iPS Definitive
Endoderm
Hepatic
Specified
Hepatoblast
specification
Fetal
Hepatocyte
Adult
Hepatocyte
Endoderm
OCT3/4 + NT NT +
Nanog + NT NT
SSEA4 + NT NT
TRA1-60 + NT NT
FOXA2 + +,lower + + +
GATA-4 + +,lower + + +
Cereberus +
FGF17 +
Goosecoid +
HNF4α * + + + +
hHex + +,very low
MixL1 +
Sox7 -*
Sox17 + +,lower +,lower +,lower +,very low
APOA1 + + + +
APOB + + + +
BMP6 +
DUSP6 +
TBX3 + + + +
AFP + +
Decorin + + +
HNF1α + + +
GSTA1 + + +
KRT19 + +/−
TTR + + +
α1AT + +
Albumin +/− + +
ASGPR1 +
CPS1 +
CK8 + +
CK18 + +
CYP1A2 +
CYP2A6 +
CYP2B6 +
CYP2C9 +
CYP2C19 +
CYP2D6 +
CYP3A4 +
CYP3A7 + very low
MAOA/B very low +
UGT1A1 +
MRP2 very low +
BSEP very low +
*

Expression of Sox7 or HNF4α at the definitive endoderm stage indicates production of embryonic rather than definitive endoderm

Functional characterization

Pluripotent stem cell-derived HLCs should exhibit functional characteristics of hepatocytes, in addition to their morphologic and phenotypic traits. Hepatocytes are notable for their wide variety of metabolic and other functional capacities, spanning over 500 classes of functions such as energy metabolism, bile production, and synthetic or detoxification functions, and thus to conclusively validate their identity, a series of in vitro assays can be performed to demonstrate activity in a variety of organized functional categories (Tables 2-3).

Table 2.

Characteristic Functional Traits in both Fetal and Adult Hepatocytes

Fetal Hepatocyte Adult Hepatocyte

Synthetic Function
 AFP Production [71] +
 Albumin Production [42] + +
 Alpha-1-Antitrypsin Production [9] + +

Energy Metabolism
Lipid Metabolism
 LDL-R Expression [35] + +
 LDL Uptake [35] + +
Glucose Metabolism
 Glucose Synthesis [72] + +
 Glycogen Production [20] + +
 Glucose-6-Phosphatase Activity [72] + +
Protein Metabolism + +
 Ammonia Uptake/Urea Production [9] + +

Bile Production and Metabolism
 Production and Secretion of bilirubin
 monoglucuronide and diglucuronide [73]
NT +
 Uptake of bile acids and secretion into hepatic
 biliary canalicular networks [74]
+ +

Detoxification
Cytochrome P450 Function
 CYP1A2 [52] +
 CYP2A6 [52] +
 CYP3A4 [52] +
 CYP3A4/CYP3A7 + + (same as CYP3A4)
Drug Metabolism Assays
 Buproprion [52] NT +
 Testosterone [52] NT +
Drug Induction Assays
 Omeprazole (CYP1A2, CYP2B6, CYP3A4) [52] +
 Rifampin (CYP2A6, CYP2B6, CYP3A4) [52] +
 Β-Naphthloflavone (CYP1A2, CYP2B6) [52] NT +
 Phenobarbital (CYP2A6, CYP2B6, CYP3A4) [52] NT +
Drug Metabolism Assays
 Buproprion [52] NT +
 Testosterone [52] NT +

Table 3.

Example functional assays with expected outcomes using adult hepatocytes

Assay name Assay format Specific examples
Cytochrome
P450 induction
Pretreatment of cells with a variety
of known cytochrome P450 inducers
  • β-naphtholflavone (CYP1A, CYP2A6, CYP2B6)

  • omeprazole (CYP1A, CYP2A6, CYP2B6, CYP3A4)

  • phenobarbital (CYP2A6, CYP2B6, CYP3A4)

  • rifampin (CYP2A6, CYP2B6, CYP3A4)

Drug toxicity Known hepatotoxins lead to
measurable TC50 via MTT or
mitochondrial activity assays
  • chlorporamazine

  • troglitazone

  • tolcapone

  • rifampin

Drug inhibition Evaluate drugs that lead to dose-
dependent inhibition of cytochrome
P450 function
  • methoxsalen (CYP2A6)

  • sulfaphenazole (CYP2C9)

  • probenacid (glucoronidation)

Drug-Drug
interaction
Evaluate for drug-mediated
modulation of cytochrome P450
expression that result in altered
toxicity of combined drug
  • phenobarbitol or rifampin pretreatment followed by acetaminophen dosing leads to increased toxicity

In vitro cell culture may enable phenotypic and functional iHLC characterization across a variety of individual parameters, however, a higher bar – and to some, a necessary bar to reach - is to achieve in vivo engraftment and function, and in some cases, functional rescue. In conducting these assays, there are three essential sets of decisions that must be considered: choice of recipient model and route/dose of administered cells, evaluation methods used to determine the kinetics and extent of donor cell engraftment, and specific functional outcomes examined to assess both local and systemic functional output of the engrafted cells. A variety of models are available, each with relative strengths and weaknesses. Two immunodeficient, metabolic mutation mouse models have often been utilized to assay for robust engraftment and repopulation of primary human hepatocytes include Fah//Rag2//IL2rγ/ or alb-uPA severe combined immunodeficient mice [47-48]. These models offer benefits in that endogenous liver cells are compromised, and thus minimal donor cell engraftment may lead to a measureable functional read out, and yet – as with most animal models – there remain some questions as to the absolute relevance of the outcome, and whether engraftment in this setting will directly correlate with function in a human. Nonetheless, although no iHLC transplant studies to date have observed significant repopulation with functional activity, small islands of stem cell derived hepatocyte-like cells have been detected using these compromised hosts. Highlighting the importance of selecting an appropriate host model for functional engraftment studies, a recent report described very high levels of albumin-staining following the transplantation of iHLCs in a mouse model that has been traditionally utilized for toxicity testing of candidate drugs [49]. In this case the stem cell-derived donor cells did not carry any genetic or functional advantage over the surviving host cells, but did appear to be selected to repopulate the damaged liver. Unfortunately, the degree to which the local human albumin-staining cells contributed to circulating human markers of functional hepatocyte integration was extremely and surprisingly low, but this observation went largely undiscussed in the original publication. Overall, the unusually high levels of engraftment are encouraging for the field, but clearly additional work remains to be done to demonstrate fully functional engraftment of iHLCs in an in vivo setting. Given these observations, some researchers have begun to explore alternate animal models, including rodents. Indeed, one group has presented data in oral presentations showing that radiation preconditioning of host rats can enhance the engraftment of human hepatocytes and iHLCs. Consequently, it seems likely that identifying the most amenable, but appropriate, host model is both a central challenge and an active area of investigation in this field.

Finally, in all of the assays utilized to characterize candidate iHLC populations, particularly their functional traits, comparison with an appropriate reference cell type is critical. Hepatocyte reference controls such as cell lines or immortalized hepatocytes have been used extensively, and offer a reproducible benchmark population, however, these lines do not exhibit appropriate, physiological levels of most hepatocyte-specific functions. Freshly isolated or uncultured, cryopreserved primary human hepatocytes are generally considered the ideal reference controls for this purpose. However, many functional tests require that cells are cultured for periods of ours to days, and once maintained in traditional culture systems the morphology, phenotype, and function of primary human hepatocytes all rapidly decline. Several updated in vitro platforms have been developed to preserve hepatic morphology, phenotype, and many functions of primary adult hepatocytes (ie. matrigel overlay, collagen gel sandwich, micropatterned cocultured hepatocytes, and three-dimensional aggregates) [50-54]. Of these culture systems, micropatterned cocultured human hepatocytes have been shown to most faithfully recapitulate human hepatocyte function with maintenance of hepatic morphology, phenotype and function for up to four weeks [52]. Other alternatives include human liver tissue homeogenates, human liver microsome isolation, and tissue sections that have been shown to faithfully recapitulate gene expression, cytochrome P450 activity and hepatocyte phenotype, although these systems cannot be used for dynamic studies. For many characterization assays that do not require periods of culture, such as gene expression, one of these alternatives or uncultured primary human hepatocytes offer the ideal ‘benchmark’. However, for other methods that require sample collection or observation over time in culture, we encourage the use of platforms such as the MPCC, or related assays, for benchmarking purposes.

Unfortunately, while dozens of published papers demonstrate HLC production from a variety of cell types, including pluripotent stem cells, the criteria used to identify the resulting hepatocyte-like cells has varied from report to report. Due to this lack of standardization of what defines an HLC, it has been difficult to compare the relative success of various protocols, and thus it is close to impossible to identify candidate method modifications that may enhance iHLC production. As mentioned above, many groups have benchmarked their iHLC populations against cultured primary human hepatocytes, which contribute to the challenge in comparing results across platforms due to the variability in most human hepatocyte culture systems. Furthermore, multiple publications have established that unless specific culture model systems are employed, such as the micropatterned coculture system, cultured hepatocytes rapidly lose their phenotype and the majority of typical functions. Moreover, cultured human hepatocytes upregulate inappropriate and often immature markers such as AFP. Consequently, any comparisons made to these altered adult hepatocytes may make the candidate iHLCs appear more strongly functional than they truly are. Indeed, examination of published accounts reveals that many protocols lead to fetal hepatocyte-like cells, although in some cases the characterization reported is not sufficient to determine the fetal versus mature nature of the resulting HLCs. Given the seemingly fetal nature of iHLCs produced to date, it is apparent that additional, careful modification of differentiation protocols will be required before the potential of pluripotent stem cell-derived hepatocyte-like cells can be realized.

Even after reaching a suitably characterized iHLC state, a remaining issue is the question of the stability of their phenotype and function in long-term culture, similar to the alterations observed in cultured adult hepatocytes [55]. Consequently a variety of strategies have been undertaken to improve the hepatocyte-specific functions and survival of primary hepatocytes in vitro. Numerous studies have focused on strategies that aim to recapitulate the normal liver microenvironment and provide missing microenvironmental cues including soluble factors [55-56], cell-matrix interactions [57-59], and heterotypic cell-cell interactions with nonparenchymal cells [60-61]. Microfabrication approaches (semiconductor-driven microtechnology tools which enable micrometer-scale control over cell adhesion, shape and multi-cellular interactions) have been utilized to control tissue microarchitecture in order to define the best cell-matrix interactions and to achieve an optimal balance of homotypic and heterotypic cellular interactions to promote hepatocyte function [62-63]. These approaches have culminated in photolithographic cell patterning techniques and robust hepatocyte culture model systems which has been used extensively for drug development and pathogen modeling. These approaches will be critical to improving the development and culture of iHLCs and will not only improve the robustness of the iHLC system but will help untangle the role that soluble factors, cell-matrix interactions and homotypic and heterotypic cell-cell interactions play in hepatic development. Notably, this area of study has also raised the possibility that the observed heterogeneity of cell types produced using current iHLC differentiation protocols may actually required for robust iHLC differentiation. That is, during the course of iHLC generation, differentiating progenitors may require signals produced by other cell types developed in parallel. Thus, it may not be possible to achieve a pure population of functional iHLCs, unless replacement signals and/or factors can be defined and provided with appropriate kinetics. All of the above underscore the need for live cell reporters that allow differentiation state to be monitored, and permit subsequent isolation of desired populations.

The differentiation protocols currently used to coax pluripotent stem cells to generate hepatocyte-like cells rely on the addition of exogenous growth factors identified in developmental studies. Subsequent work has established the key transcription factors that are activated during the course of the hepatic differentiation process. Other groups have demonstrated that direct reprogramming strategies which bypass the pluripotent stage can be applied to convert fibroblasts into cardiomyocytes and neurons by overexpressing key lineage-specific transcriptional regulators. Motivated by these findings, two groups have recently shown that mouse fibroblasts can be reprogrammed into hepatocyte-like cells via the overexpression of key transcription factors (ie. Foxa2/3, HNF1α and GATA4 or Foxa1/2/3 and HNF4α) [64-65]. In both examples, induced hepatocytes exhibited only minimal hepatocyte-specific functions including very low albumin secretion, triglyceride synthesis, and cytochrome P450 function. Transplantation of induced hepatocytes into FAH deficient mice (a genetic model of hereditary tyrosinemia that develops liver failure without NTBC drug treatment) led to liver repopulation and survival of less than half of the hosts. This partial success stands in contrast to reported outcomes using primary mouse hepatocytes which repopulate the complete liver with rare failure to rescue the mice from death. Whether these differences in repopulation and in vitro hepatic function reflect variable reprogramming, dysregulated function or immature cells is still unclear.

iPS and iHLCs offer a dizzying array of opportunities including hepatocyte-like cell generation for possible cell replacement therapy. However, this noble goal is likely still far on the horizon, and thus their greatest strength, or at least the most near-term potential of iHLCs, may lie in applying them to serve as a platform for disease modeling, or for mechanistic toxicity studies in idiosyncratic responses. The cost of drug development is heavily influenced by the attrition rate of tested compounds; for every drug that reaches the marketplace, 5,000 to 10,000 molecules were tested preclinically [66]. Utilizing iHLCs for this sort of application will require an experimental platform that is robust and scaleable, and that involves multiple essential steps. First, a target disease that exhibits a recognizable in vitro phenotype must be selected, and candidate tissue donors with the target disease need to be identified, as well as healthy control subjects. Second, iPS cells need to be derived, characterized, and grown in an easily scaleable platform. Third, differentiation of iPS cells into iHLCs needs to be efficient, complete, consistent, and conducted in a format appropriate for large-scale small molecule testing (ie. at least 96, 384, or 1024 wells). While, as discussed above, current best-available iHLCs remain incompletely differentiated, reports of the successful application of this process have been described for patients with alpha-1-antitrypsin disease and familial hyperlipidemia [35-36]. However, in the case of polygenic, and thus genetically complex diseases such as nonalcoholic fatty liver disease, screening for a specific phenotypic or functional outcome may not be possible, even after new methods have been developed to achieve more complete and robust stem cell-derived hepatocyte production. Consequently, no matter what disease is being interrogated, a differentiation platform designed for screening or modeling must produce pure populations of fully differentiated cells with minimal heterogeneity and with no stochasticity. Certainly, further improvements currently under examination by the stem cell and engineering communities such as efforts to identify microenvironmental signals to increase the purity, efficiency and maturation of desired cell types are likely to help tackle and solve these various problems.

iHLCs can also enable studies of pathogens that exclusively target human hepatocytes, notably those with profound global health implications such as hepatitis B virus (HBV), hepatitis C virus (HCV) and malaria. For example, current HCV model systems utilize the Huh7 hepatoma carcinoma cell line to examine the HCV viral life cycle in vitro which is limited by the poor hepatic function of the line [67-68]. Consequently, recent approaches using micropatterning techniques have enabled HCV infection in primary human hepatocytes [69]. More recently, we demonstrated that iHLCs express all known entry factors of HCV, support the complete HCV viral cycle, and exhibit a robust anti-viral immune reponse [70]. iHLCs can thus serve as a platform to explore the role that host genetics plays in viral pathogenesis and elucidate the role that these host factors plays in disease pathogenesis.

CONCLUSIONS

In summary, pluripotent stem cell derived hepatocyte-like cells can be generated from iPS cells and hESC in a reproducible and efficient manner. Review of several available methods reveal that there are multiple paths that lead from pluripotency to at least an immature hepatic phenotype that more closely resembles fetal rather than adult hepatocytes. This apparent incomplete differentiation state likely results from our poor understanding of the mechanisms underlying the developmental shift from fetal to adult liver. Moreover, the existing lack of standardization of morphologic, phenotypic, and functional characterization of iHLCs has made comparisons between published papers challenging, if not impossible. In this review, we have illustrated the importance of extensive phenotypic and functional characterization and we encourage the community to apply various standards during hepatocyte-like cell characterization. In addition, the use of well-documented and functional hepatocyte reference controls is key to the future improvement of iHLC generation. This advance will lead to the rapid adoption of this key population and their use in a variety of applications including the study of the mechanisms of human disease and development, and, perhaps in the longer term, as a platform for pharmacology and toxicology drug screening and cell based therapeutics.

ACKNOWLEDGMENTS

We thank Stephen Duncan for insightful discussions. This work was supported by the NIH Roadmap for Medical Research Grant 1 R01 DK085713-01 and Howard Hughes Medical Institute. Robert Schwartz was supported by the AGA Research Scholar Award.

Footnotes

Publisher's Disclaimer: 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 citable 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.

REFERENCES

  • 1.Gonzalez SA, Keeffe EB. Chronic viral hepatitis: epidemiology, molecular biology, and antiviral therapy. Front Biosci. 2011;16:225–50. doi: 10.2741/3685. [DOI] [PubMed] [Google Scholar]
  • 2.Starzl TE, Fung JJ. Themes of liver transplantation. Hepatology. 2010;51(6):1869–84. doi: 10.1002/hep.23595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Starzl TE, et al. Orthotopic homotransplantation of the human liver. Ann Surg. 1968;168(3):392–415. doi: 10.1097/00000658-196809000-00009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Perera MT, Mirza DF, Elias E. Liver transplantation: Issues for the next 20 years. J Gastroenterol Hepatol. 2009;24(Suppl 3):S124–31. doi: 10.1111/j.1440-1746.2009.06081.x. [DOI] [PubMed] [Google Scholar]
  • 5.Nyberg SL, et al. Evaluation of a hepatocyte-entrapment hollow fiber bioreactor: a potential bioartificial liver. Biotechnol Bioeng. 1993;41(2):194–203. doi: 10.1002/bit.260410205. [DOI] [PubMed] [Google Scholar]
  • 6.Fox IJ, Roy-Chowdhury J. Hepatocyte transplantation. J Hepatol. 2004;40(6):878–86. doi: 10.1016/j.jhep.2004.04.009. [DOI] [PubMed] [Google Scholar]
  • 7.Fox IJ, et al. Treatment of the Crigler-Najjar syndrome type I with hepatocyte transplantation. N Engl J Med. 1998;338(20):1422–6. doi: 10.1056/NEJM199805143382004. [DOI] [PubMed] [Google Scholar]
  • 8.Dhawan A, et al. Human hepatocyte transplantation. Methods Mol Biol. 2010;640:525–34. doi: 10.1007/978-1-60761-688-7_29. [DOI] [PubMed] [Google Scholar]
  • 9.Chen AA, et al. Humanized mice with ectopic artificial liver tissues. Proc Natl Acad Sci U S A. 2011;108(29):11842–7. doi: 10.1073/pnas.1101791108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Michalopoulos GK. Liver regeneration. J Cell Physiol. 2007;213(2):286–300. doi: 10.1002/jcp.21172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Kobayashi N, et al. Prevention of acute liver failure in rats with reversibly immortalized human hepatocytes. Science. 2000;287(5456):1258–62. doi: 10.1126/science.287.5456.1258. [DOI] [PubMed] [Google Scholar]
  • 12.Castell JV, et al. Hepatocyte cell lines: their use, scope and limitations in drug metabolism studies. Expert Opin Drug Metab Toxicol. 2006;2(2):183–212. doi: 10.1517/17425255.2.2.183. [DOI] [PubMed] [Google Scholar]
  • 13.Ito M, et al. Review of hepatocyte transplantation. J Hepatobiliary Pancreat Surg. 2009;16(2):97–100. doi: 10.1007/s00534-008-0023-0. [DOI] [PubMed] [Google Scholar]
  • 14.Yu Y, et al. Cell therapies for liver diseases. Liver Transpl. 2012;18(1):9–21. doi: 10.1002/lt.22467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Dalgetty DM, et al. Progress and future challenges in stem cell-derived liver technologies. Am J Physiol Gastrointest Liver Physiol. 2009;297(2):G241–8. doi: 10.1152/ajpgi.00138.2009. [DOI] [PubMed] [Google Scholar]
  • 16.Espejel S, et al. Induced pluripotent stem cell-derived hepatocytes have the functional and proliferative capabilities needed for liver regeneration in mice. J Clin Invest. 2010;120(9):3120–6. doi: 10.1172/JCI43267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Thomson JA, et al. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282(5391):1145–7. doi: 10.1126/science.282.5391.1145. [DOI] [PubMed] [Google Scholar]
  • 18.Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–76. doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
  • 19.Takahashi K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131(5):861–72. doi: 10.1016/j.cell.2007.11.019. [DOI] [PubMed] [Google Scholar]
  • 20.Si-Tayeb K. Highly efficient generation of human hepatocyte-like cells from induced pluripotent stem cells. Hepatology. 2010;51(1):297–305. doi: 10.1002/hep.23354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Touboul T, et al. Generation of functional hepatocytes from human embryonic stem cells under chemically defined conditions that recapitulate liver development. Hepatology. 2010;51(5):1754–65. doi: 10.1002/hep.23506. [DOI] [PubMed] [Google Scholar]
  • 22.Wandzioch E, Zaret KS. Dynamic signaling network for the specification of embryonic pancreas and liver progenitors. Science. 2009;324(5935):1707–10. doi: 10.1126/science.1174497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Zaret KS, Grompe M. Generation and regeneration of cells of the liver and pancreas. Science. 2008;322(5907):1490–4. doi: 10.1126/science.1161431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Matsumoto K, et al. Liver organogenesis promoted by endothelial cells prior to vascular function. Science. 2001;294(5542):559–63. doi: 10.1126/science.1063889. [DOI] [PubMed] [Google Scholar]
  • 25.Deutsch G, et al. A bipotential precursor population for pancreas and liver within the embryonic endoderm. Development. 2001;128(6):871–81. doi: 10.1242/dev.128.6.871. [DOI] [PubMed] [Google Scholar]
  • 26.Ott J, Kamatani Y, Lathrop M. Family-based designs for genome-wide association studies. Nat Rev Genet. 2011;12(7):465–74. doi: 10.1038/nrg2989. [DOI] [PubMed] [Google Scholar]
  • 27.Schmouth JF, et al. Modelling human regulatory variation in mouse: finding the function in genome-wide association studies and whole-genome sequencing. PLoS Genet. 2012;8(3):e1002544. doi: 10.1371/journal.pgen.1002544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Schofield PN, Hoehndorf R, Gkoutos GV. Mouse genetic and phenotypic resources for human genetics. Hum Mutat. 2012;33(5):826–36. doi: 10.1002/humu.22077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Daly AK. Using genome-wide association studies to identify genes important in serious adverse drug reactions. Annu Rev Pharmacol Toxicol. 2012;52:21–35. doi: 10.1146/annurev-pharmtox-010611-134743. [DOI] [PubMed] [Google Scholar]
  • 30.Lieschke GJ, Currie PD. Animal models of human disease: zebrafish swim into view. Nat Rev Genet. 2007;8(5):353–67. doi: 10.1038/nrg2091. [DOI] [PubMed] [Google Scholar]
  • 31.Deo RC, MacRae CA. The zebrafish: scalable in vivo modeling for systems biology. Wiley Interdiscip Rev Syst Biol Med. 2011;3(3):335–46. doi: 10.1002/wsbm.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Pattaro C, et al. Genome-wide association and functional follow-up reveals new loci for kidney function. PLoS Genet. 2012;8(3):e1002584. doi: 10.1371/journal.pgen.1002584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Norton N, et al. Genome-wide studies of copy number variation and exome sequencing identify rare variants in BAG3 as a cause of dilated cardiomyopathy. Am J Hum Genet. 2011;88(3):273–82. doi: 10.1016/j.ajhg.2011.01.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Soldner F, et al. Generation of isogenic pluripotent stem cells differing exclusively at two early onset Parkinson point mutations. Cell. 2011;146(2):318–31. doi: 10.1016/j.cell.2011.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Cayo MA, et al. ‘JD’ iPS cell-derived hepatocytes faithfully recapitulate the pathophysiology of familial hypercholesterolemia. Hepatology. 2012 doi: 10.1002/hep.25871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Rashid ST, et al. Modeling inherited metabolic disorders of the liver using human induced pluripotent stem cells. J Clin Invest. 2010;120(9):3127–36. doi: 10.1172/JCI43122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Zhang S, et al. Rescue of ATP7B function in hepatocyte-like cells from Wilson’s disease induced pluripotent stem cells using gene therapy or the chaperone drug curcumin. Hum Mol Genet. 2011;20(16):3176–87. doi: 10.1093/hmg/ddr223. [DOI] [PubMed] [Google Scholar]
  • 38.Song Z, et al. Efficient generation of hepatocyte-like cells from human induced pluripotent stem cells. Cell Res. 2009;19(11):1233–42. doi: 10.1038/cr.2009.107. [DOI] [PubMed] [Google Scholar]
  • 39.Sharma NS, et al. Enrichment of hepatocyte-like cells with upregulated metabolic and differentiated function derived from embryonic stem cells using S-NitrosoAcetylPenicillamine. Tissue Eng Part C Methods. 2009;15(2):297–306. doi: 10.1089/ten.tec.2008.0303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Moore RN, et al. Enhanced differentiation of embryonic stem cells using co-cultivation with hepatocytes. Biotechnol Bioeng. 2008;101(6):1332–43. doi: 10.1002/bit.21987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ungrin MD, et al. Reproducible, ultra high-throughput formation of multicellular organization from single cell suspension-derived human embryonic stem cell aggregates. PLoS One. 2008;3(2):e1565. doi: 10.1371/journal.pone.0001565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Schwartz RE, et al. Defined conditions for development of functional hepatic cells from human embryonic stem cells. Stem Cells Dev. 2005;14(6):643–55. doi: 10.1089/scd.2005.14.643. [DOI] [PubMed] [Google Scholar]
  • 43.Basma H, et al. Differentiation and transplantation of human embryonic stem cell-derived hepatocytes. Gastroenterology. 2009;136(3):990–9. doi: 10.1053/j.gastro.2008.10.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Lavon N. Generation of hepatocytes from human embryonic stem cells. Methods Mol Biol. 2010;640:237–46. doi: 10.1007/978-1-60761-688-7_11. [DOI] [PubMed] [Google Scholar]
  • 45.D’Amour KA, et al. Efficient differentiation of human embryonic stem cells to definitive endoderm. Nat Biotechnol. 2005;23(12):1534–41. doi: 10.1038/nbt1163. [DOI] [PubMed] [Google Scholar]
  • 46.Hay DC, et al. Highly efficient differentiation of hESCs to functional hepatic endoderm requires ActivinA and Wnt3a signaling. Proc Natl Acad Sci U S A. 2008;105(34):12301–6. doi: 10.1073/pnas.0806522105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Azuma H, et al. Robust expansion of human hepatocytes in Fah-/-/Rag2-/-/Il2rg-/- mice. Nat Biotechnol. 2007;25(8):903–10. doi: 10.1038/nbt1326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Dandri M, et al. Repopulation of mouse liver with human hepatocytes and in vivo infection with hepatitis B virus. Hepatology. 2001;33(4):981–8. doi: 10.1053/jhep.2001.23314. [DOI] [PubMed] [Google Scholar]
  • 49.Liu H, et al. In vivo liver regeneration potential of human induced pluripotent stem cells from diverse origins. Sci Transl Med. 2011;3(82):82ra39. doi: 10.1126/scitranslmed.3002376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.LeCluyse EL, Audus KL, Hochman JH. Formation of extensive canalicular networks by rat hepatocytes cultured in collagen-sandwich configuration. Am J Physiol. 1994;266(6 Pt 1):C1764–74. doi: 10.1152/ajpcell.1994.266.6.C1764. [DOI] [PubMed] [Google Scholar]
  • 51.Dunn JC, et al. Hepatocyte function and extracellular matrix geometry: long-term culture in a sandwich configuration. FASEB J. 1989;3(2):174–7. doi: 10.1096/fasebj.3.2.2914628. [DOI] [PubMed] [Google Scholar]
  • 52.Khetani SR, Bhatia SN. Microscale culture of human liver cells for drug development. Nat Biotechnol. 2008;26(1):120–6. doi: 10.1038/nbt1361. [DOI] [PubMed] [Google Scholar]
  • 53.Wu FJ, et al. Enhanced cytochrome P450 IA1 activity of self-assembled rat hepatocyte spheroids. Cell Transplant. 1999;8(3):233–46. doi: 10.1177/096368979900800304. [DOI] [PubMed] [Google Scholar]
  • 54.Wu FJ, et al. Efficient assembly of rat hepatocyte spheroids for tissue engineering applications. Biotechnol Bioeng. 1996;50(4):404–15. doi: 10.1002/(SICI)1097-0290(19960520)50:4<404::AID-BIT7>3.0.CO;2-P. [DOI] [PubMed] [Google Scholar]
  • 55.Guillouzo A. Liver cell models in in vitro toxicology. Environ Health Perspect. 1998;106(Suppl 2):511–32. doi: 10.1289/ehp.98106511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Jindal R, et al. Amino acid-mediated heterotypic interaction governs performance of a hepatic tissue model. FASEB J. 2009;23(7):2288–98. doi: 10.1096/fj.08-114934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Lin P, et al. Assessing porcine liver-derived biomatrix for hepatic tissue engineering. Tissue Eng. 2004;10(7):1046–53. doi: 10.1089/ten.2004.10.1046. [DOI] [PubMed] [Google Scholar]
  • 58.Flaim CJ, Chien S, Bhatia SN. An extracellular matrix microarray for probing cellular differentiation. Nat Methods. 2005;2(2):119–25. doi: 10.1038/nmeth736. [DOI] [PubMed] [Google Scholar]
  • 59.Bissell DM, et al. Support of cultured hepatocytes by a laminin-rich gel. Evidence for a functionally significant subendothelial matrix in normal rat liver. J Clin Invest. 1987;79(3):801–12. doi: 10.1172/JCI112887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Bhatia SN, et al. Effect of cell-cell interactions in preservation of cellular phenotype: cocultivation of hepatocytes and nonparenchymal cells. FASEB J. 1999;13(14):1883–900. doi: 10.1096/fasebj.13.14.1883. [DOI] [PubMed] [Google Scholar]
  • 61.March S, et al. Microenvironmental regulation of the sinusoidal endothelial cell phenotype in vitro. Hepatology. 2009;50(3):920–8. doi: 10.1002/hep.23085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Folch A, Toner M. Microengineering of cellular interactions. Annu Rev Biomed Eng. 2000;2:227–56. doi: 10.1146/annurev.bioeng.2.1.227. [DOI] [PubMed] [Google Scholar]
  • 63.Fukuda J, Sakai Y, Nakazawa K. Novel hepatocyte culture system developed using microfabrication and collagen/polyethylene glycol microcontact printing. Biomaterials. 2006;27(7):1061–70. doi: 10.1016/j.biomaterials.2005.07.031. [DOI] [PubMed] [Google Scholar]
  • 64.Huang P, et al. Induction of functional hepatocyte-like cells from mouse fibroblasts by defined factors. Nature. 2011;475(7356):386–9. doi: 10.1038/nature10116. [DOI] [PubMed] [Google Scholar]
  • 65.Sekiya S, Suzuki A. Direct conversion of mouse fibroblasts to hepatocyte-like cells by defined factors. Nature. 2011;475(7356):390–3. doi: 10.1038/nature10263. [DOI] [PubMed] [Google Scholar]
  • 66.Kola I, Landis J. Can the pharmaceutical industry reduce attrition rates? Nat Rev Drug Discov. 2004;3(8):711–5. doi: 10.1038/nrd1470. [DOI] [PubMed] [Google Scholar]
  • 67.Lindenbach BD, et al. Complete replication of hepatitis C virus in cell culture. Science. 2005;309(5734):623–6. doi: 10.1126/science.1114016. [DOI] [PubMed] [Google Scholar]
  • 68.Wakita T, et al. Production of infectious hepatitis C virus in tissue culture from a cloned viral genome. Nat Med. 2005;11(7):791–6. doi: 10.1038/nm1268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Ploss A, et al. Persistent hepatitis C virus infection in microscale primary human hepatocyte cultures. Proc Natl Acad Sci U S A. 2010;107(7):3141–5. doi: 10.1073/pnas.0915130107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Schwartz RE, et al. Modeling hepatitis C virus infection using human induced pluripotent stem cells. Proc Natl Acad Sci U S A. 2012;109(7):2544–8. doi: 10.1073/pnas.1121400109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Chiao E, et al. Isolation and transcriptional profiling of purified hepatic cells derived from human embryonic stem cells. Stem Cells. 2008;26(8):2032–41. doi: 10.1634/stemcells.2007-0964. [DOI] [PubMed] [Google Scholar]
  • 72.Khuu DN, et al. In vitro differentiated adult human liver progenitor cells display mature hepatic metabolic functions: a potential tool for in vitro pharmacotoxicological testing. Cell Transplant. 2011;20(2):287–302. doi: 10.3727/096368910X516655. [DOI] [PubMed] [Google Scholar]
  • 73.Tada K, et al. Long-term amelioration of bilirubin glucuronidation defect in Gunn rats by transplanting genetically modified immortalized autologous hepatocytes. Cell Transplant. 1998;7(6):607–16. doi: 10.1177/096368979800700611. [DOI] [PubMed] [Google Scholar]
  • 74.Murray JW, et al. Heterogeneous accumulation of fluorescent bile acids in primary rat hepatocytes does not correlate with their homogenous expression of ntcp. Am J Physiol Gastrointest Liver Physiol. 2011;301(1):G60–8. doi: 10.1152/ajpgi.00035.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES