BASIC—LIVER, PANCREAS, AND BILIARY TRACT · 2019. 7. 10. · BILIARY TRACT GASTROENTEROLOGY...

14
BASIC—LIVER, PANCREAS, AND BILIARY TRACT Differentiation and Transplantation of Human Embryonic Stem Cell–Derived Hepatocytes HESHAM BASMA,* ALEJANDRO SOTO–GUTIÉRREZ, GOVARDHANA RAO YANNAM,* LIPING LIU,* RYOTARO ITO,* TOSHIYUKI YAMAMOTO,* EWA ELLIS, § STEVEN D. CARSON, SHINTARO SATO, YONG CHEN, DAVID MUIRHEAD, NALU NAVARRO–ÁLVAREZ, RONALD J. WONG, # JAYANTA ROY–CHOWDHURY, ¶, ** JEFFREY L. PLATT, ‡‡,§§ DAVID F. MERCER,* JOHN D. MILLER,* STEPHEN C. STROM, § NAOYA KOBAYASHI, and IRA J. FOX* *Department of Surgery, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska; Department of Surgery, Okayama University Graduate School of Medicine and Dentistry, Shikata-cho, Okayama, Japan; § Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Medicine (Hepatology Division), **Department of Genetics, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, New York; # Department of Pediatrics, Stanford University School of Medicine, Stanford, California; and the ‡‡ Department of Surgery, and the §§ Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan See editorial on page 767. Background & Aims: The ability to obtain unlimited numbers of human hepatocytes would improve the development of cell-based therapies for liver diseases, facilitate the study of liver biology, and improve the early stages of drug discovery. Embryonic stem cells are pluripotent, potentially can differentiate into any cell type, and therefore could be developed as a source of human hepatocytes. Methods: To generate human hepatocytes, human embryonic stem cells were differ- entiated by sequential culture in fibroblast growth factor 2 and human activin-A, hepatocyte growth fac- tor, and dexamethasone. Functional hepatocytes were isolated by sorting for surface asialoglycoprotein-re- ceptor expression. Characterization was performed by real-time polymerase chain reaction, immunohis- tochemistry, immunoblot, functional assays, and transplantation. Results: Embryonic stem cell– de- rived hepatocytes expressed liver-specific genes, but not genes representing other lineages, secreted func- tional human liver–specific proteins similar to those of primary human hepatocytes, and showed human hepatocyte cytochrome P450 metabolic activity. Serum from rodents given injections of embryonic stem cell– derived hepatocytes contained significant amounts of human albumin and 1-antitrypsin. Col- onies of cytokeratin-18 and human albumin– express- ing cells were present in the livers of recipient ani- mals. Conclusions: Human embryonic stem cells can be differentiated into cells with many characteristics of primary human hepatocytes. Hepatocyte-like cells can be enriched and recovered based on asialoglyco- protein-receptor expression and potentially could be used in drug discovery research and developed as therapeutics. T ransplantation of isolated hepatocytes has been shown in the laboratory and in patients to be a minimally invasive intervention that can augment the function of the liver. 1–3 This is an especially exciting alternative to organ transplantation for patients with acute decompensation of the liver or life-threatening liver-based genetic disorders. A shortage of human livers, however, presently limits the use of hepatocytes for trans- plantation. Thus, the availability of an unlimited supply of human hepatocytes would facilitate the development and clinical application of hepatocyte transplantation. An abundant source of human hepatocytes also could be used to repopulate the livers of immune-deficient mice, 4 facilitating the study of human liver disease, hepatitis virus biology, 5 and human drug metabolism. 6,7 Human hepatocyte-like cells have been generated from a variety of putative stem cell sources. 8 –14 However, al- though investigators have generated cells with some characteristics approaching that of human hepatocytes, to date, no approach to generating hepatocytes from stem cells has succeeded in generating a homogeneous population of cells that could provide levels of function comparable with primary liver cells. Here we report a Abbreviations used in this paper: AAT, 1-antitrypsin; AFP, -feto- protein; ASGPR, asialoglycoprotein receptor; CF VII, coagulation factor VII; CYP, cytochrome P450; DMEM/F12, Dulbecco’s modified Eagle medium F-12; EB, embryoid bodies; ES, embryonic stem; hES, human embryonic stem; KSR, Knockout Serum Replacement; RT-PCR, re- verse-transcription polymerase chain reaction. © 2009 by the AGA Institute 0016-5085/09/$36.00 doi:10.1053/j.gastro.2008.10.047 BASIC–LIVER, PANCREAS, AND BILIARY TRACT GASTROENTEROLOGY 2009;136:990 –999

Transcript of BASIC—LIVER, PANCREAS, AND BILIARY TRACT · 2019. 7. 10. · BILIARY TRACT GASTROENTEROLOGY...

Page 1: BASIC—LIVER, PANCREAS, AND BILIARY TRACT · 2019. 7. 10. · BILIARY TRACT GASTROENTEROLOGY 2009;136:990–999. differentiation strategy that produces a relatively homo-geneous

BT

DC

HTND*UPM§

BndfeacohefticbttrntohSsaoimbocp

BA

SIC–LIV

ER,

PA

NCREA

S,A

ND

BILIA

RY

TRA

CT

GASTROENTEROLOGY 2009;136:990–999

ASIC—LIVER, PANCREAS, AND BILIARYRACT

ifferentiation and Transplantation of Human Embryonic Stemell–Derived Hepatocytes

ESHAM BASMA,* ALEJANDRO SOTO–GUTIÉRREZ,‡ GOVARDHANA RAO YANNAM,* LIPING LIU,* RYOTARO ITO,*OSHIYUKI YAMAMOTO,* EWA ELLIS,§ STEVEN D. CARSON,� SHINTARO SATO,‡ YONG CHEN,¶ DAVID MUIRHEAD,�

ALU NAVARRO–ÁLVAREZ,‡ RONALD J. WONG,# JAYANTA ROY–CHOWDHURY,¶,** JEFFREY L. PLATT,‡‡,§§

AVID F. MERCER,* JOHN D. MILLER,* STEPHEN C. STROM,§ NAOYA KOBAYASHI,‡ and IRA J. FOX*Department of Surgery, �Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska; ‡Department of Surgery, Okayamaniversity Graduate School of Medicine and Dentistry, Shikata-cho, Okayama, Japan; §Department of Pathology, University of Pittsburgh School of Medicine,ittsburgh, Pennsylvania; ¶Department of Medicine (Hepatology Division), **Department of Genetics, Marion Bessin Liver Research Center, Albert Einstein College of

# ‡‡

edicine, Bronx, New York; Department of Pediatrics, Stanford University School of Medicine, Stanford, California; and the Department of Surgery, and the§Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan

ut

TmfaalhpoaAufv

atctspc

pVmev

See editorial on page 767.

ackground & Aims: The ability to obtain unlimitedumbers of human hepatocytes would improve theevelopment of cell-based therapies for liver diseases,acilitate the study of liver biology, and improve thearly stages of drug discovery. Embryonic stem cellsre pluripotent, potentially can differentiate into anyell type, and therefore could be developed as a sourcef human hepatocytes. Methods: To generate humanepatocytes, human embryonic stem cells were differ-ntiated by sequential culture in fibroblast growthactor 2 and human activin-A, hepatocyte growth fac-or, and dexamethasone. Functional hepatocytes weresolated by sorting for surface asialoglycoprotein-re-eptor expression. Characterization was performedy real-time polymerase chain reaction, immunohis-ochemistry, immunoblot, functional assays, andransplantation. Results: Embryonic stem cell– de-ived hepatocytes expressed liver-specific genes, butot genes representing other lineages, secreted func-

ional human liver–specific proteins similar to thosef primary human hepatocytes, and showed humanepatocyte cytochrome P450 metabolic activity.erum from rodents given injections of embryonictem cell– derived hepatocytes contained significantmounts of human albumin and �1-antitrypsin. Col-nies of cytokeratin-18 and human albumin– express-

ng cells were present in the livers of recipient ani-als. Conclusions: Human embryonic stem cells can

e differentiated into cells with many characteristicsf primary human hepatocytes. Hepatocyte-like cellsan be enriched and recovered based on asialoglyco-

rotein-receptor expression and potentially could be

sed in drug discovery research and developed asherapeutics.

ransplantation of isolated hepatocytes has beenshown in the laboratory and in patients to be a

inimally invasive intervention that can augment theunction of the liver.1–3 This is an especially excitinglternative to organ transplantation for patients withcute decompensation of the liver or life-threateningiver-based genetic disorders. A shortage of human livers,owever, presently limits the use of hepatocytes for trans-lantation. Thus, the availability of an unlimited supplyf human hepatocytes would facilitate the developmentnd clinical application of hepatocyte transplantation.n abundant source of human hepatocytes also could besed to repopulate the livers of immune-deficient mice,4

acilitating the study of human liver disease, hepatitisirus biology,5 and human drug metabolism.6,7

Human hepatocyte-like cells have been generated fromvariety of putative stem cell sources.8 –14 However, al-

hough investigators have generated cells with someharacteristics approaching that of human hepatocytes,o date, no approach to generating hepatocytes fromtem cells has succeeded in generating a homogeneousopulation of cells that could provide levels of functionomparable with primary liver cells. Here we report a

Abbreviations used in this paper: AAT, �1-antitrypsin; AFP, �-feto-rotein; ASGPR, asialoglycoprotein receptor; CF VII, coagulation factorII; CYP, cytochrome P450; DMEM/F12, Dulbecco’s modified Eagleedium F-12; EB, embryoid bodies; ES, embryonic stem; hES, human

mbryonic stem; KSR, Knockout Serum Replacement; RT-PCR, re-erse-transcription polymerase chain reaction.

© 2009 by the AGA Institute0016-5085/09/$36.00

doi:10.1053/j.gastro.2008.10.047

Page 2: BASIC—LIVER, PANCREAS, AND BILIARY TRACT · 2019. 7. 10. · BILIARY TRACT GASTROENTEROLOGY 2009;136:990–999. differentiation strategy that produces a relatively homo-geneous

dg(p

IamFnmmbplsaptDbbshhce(gasgd

dmbafoMwa

dSmw

wttwfkht(NmTttbti

(Rptu(ppepts

Dl1hmhnl3wbpceFas

BA

SIC–L

IVER

,PA

NCREA

S,A

ND

BIL

IARY

TRA

CT

March 2009 HUMAN ES CELL–DERIVED HEPATOCYTES 991

ifferentiation strategy that produces a relatively homo-eneous population of cells from human embryonic stemhES) cells that show the morphologic and phenotypicroperties of human hepatocytes.

Materials and MethodsDifferentiation ProgramhES cells (H1) were obtained from WiCell Research

nstitute (Madison, WI) and maintained on mitotically in-ctive mouse embryo fibroblast feeder layers in Dulbecco’sodified Eagle medium/Ham’s F-12 medium (DMEM/

12), high (20%) Knockout Serum Replacement (KSR), 4g/mL human recombinant fibroblast growth factor-2, 1mol/L nonessential amino acids, L-glutamine, and 0.1mol/L 2-mercaptoethanol (Invitrogen, Carlsbad, CA). Em-

ryonic bodies (EBs) were generated by plating collagenase-assaged cells at a density of 1–5 � 104 cells per cm2 on

ow-attachment Petri dishes for 48 hours in DMEM/F12upplemented with 15% KSR, 1 mmol/L nonessentialmino acids, and L-glutamine. For differentiation, EBs werelated on 5% Matrigel-Growth Factor Reduced (R&D Sys-ems Inc, Minneapolis, MN), and maintained for 3 days inMEM/F12 media supplemented with 100 ng/mL Recom-

inant activin-A (R&D Systems Inc) and 100 ng/mL fibro-last growth factor-2 (Invitrogen). The concentration oferum (fetal bovine serum or KSR) was 0% for the first 24ours, 0.2% for the second 24 hours, and 2.0% for the last 24ours. Cells then were grown for 8 days in DMEM/F12ontaining 10% fetal bovine serum or KSR, 1 mmol/L non-ssential amino acids, L-glutamine, 1% dimethyl sulfoxideSigma–Aldrich, St. Louis, MO), and 100 ng/mL hepatocyterowth factor (R&D Systems Inc), followed by culture for 3dditional days in DMEM/F12 containing 10% fetal bovineerum or KSR, 1 mmol/L nonessential amino acids, L-lutamine, and 10�7 mol/L dexamethasone (Sigma–Al-rich).

AnimalsMale nonobese diabetic/severe combined immuno-

eficient (NOD/SCID) mice, albumin-urokinase-type plas-inogen activator (Alb-uPA SCID) mice,15 and Nagase anal-

uminemic rats16 were bred in the animal breeding facilityt the University of Nebraska Medical Center and were usedor transplantation procedures. All procedures performedn animals were approved by the University of Nebraskaedical Center Animal Care and Use Committee, and thusere within the guidelines for humane care of laboratorynimals.

TransplantationOne million hES cells, recovered after 18 days of

ifferentiation, were transplanted into the spleens of NOD/CID mice using a 25-gauge needle. Recipient NOD/SCIDice were injected on 2 occasions before transplantation

ith 70 mg/kg retrorsine, separated by 2 weeks, and under- g

ent 50% partial hepatectomy 4 weeks later, at the time ofransplantation.17 Nagase rat recipients similarly werereated twice with 30 mg/kg of retrorsine separated over 2eeks, and underwent a 70% partial hepatectomy just be-

ore transplantation.18 Administration of FK506 at 2 mg/g/day in these animals controls rejection of xenogeneicepatocytes for at least 2 months (unpublished data). Afterransplantation of 1 million asialoglycoprotein receptorASGPR)-sorted differentiated hES cells into the spleen of a

agase analbuminemic rat, human albumin levels wereeasured weekly by enzyme-linked immunosorbent assay.hree-week-old Alb-uPA SCID mice underwent transplan-

ation without treatment with retrorsine or partial hepatec-omy by intrasplenic injection. Graft survival was assessedy serum measurements of human albumin and �1-anti-rypsin (AAT) levels, which were measured by enzyme-linkedmmunosorbent assay.

Polymerase Chain ReactionRNA was prepared using a Qiagen RNeasy Mini Kit

Hilden, Germany). Chromosomal DNA was removed usingNase-free DNase (Qiagen), and complementary DNA wasrepared using the Superscript First-Strand synthesis sys-em (Invitrogen), using 1 �g of total RNA. Specific primerssed for reverse-transcription polymerase chain reaction

RT-PCR) are described in supplementary Table 1 (see sup-lementary material online at www.gastrojournal.org). PCRroducts were resolved on 1% agarose gels and visualized bythidium bromide staining. Human glyceraldehyde-3-phos-hate dehydrogenase and cyclophilin served as internal con-rols. Real-time RT-PCR was performed as previously de-cribed.19

Flow Analysis and Sorting ofASGPR-Expressing ES-Derived HepatocytesES-derived cells were trypsinized and resuspended in

MEM/F12 supplemented with 10% KSR. Cells were al-owed to recover for 30 minutes and then centrifuged at200 rpm. Nonspecific binding was blocked using 100 �Luman Fc for 30 minutes, and then cells were treated for 30inutes with a 100-�L solution containing a goat-anti-

uman ASGPR1 antibody diluted 1:50 (Santa Cruz Biotech-ology Inc, Santa Cruz, CA). A fluorescein isothiocyanate–

abeled mouse anti-goat antibody was added to the cells for0 minutes at a concentration of 0.5 �g/mL. Cells wereashed 3 times with 1 mL fluorescence-activated cell sorteruffer (1% sodium azide and 2% fetal bovine serum inhosphate-buffered saline), resuspended in 500 �L fluores-ence-activated cell sorter buffer, and sorted by flow cytom-try. Cell sorting was performed on a Becton–DickinsonACSVantage DiVa flow cytometer and data acquisitionnd analysis was performed using Becton–Dickinson DiVaoftware (Becton–Dickinson, San Jose, CA).

Electron MicroscopyCells were fixed in 2% paraformaldehyde and 2.5%

lutaraldehyde in Millonig’s phosphate buffer, pH 7.38,

Page 3: BASIC—LIVER, PANCREAS, AND BILIARY TRACT · 2019. 7. 10. · BILIARY TRACT GASTROENTEROLOGY 2009;136:990–999. differentiation strategy that produces a relatively homo-geneous

pth(asba3CoJUaws

sS

FfDcJPmdTv2Tdtmr3cpm[

Ft1efiidte

BA

SIC–LIV

ER,

PA

NCREA

S,A

ND

BILIA

RY

TRA

CT

992 BASMA ET AL GASTROENTEROLOGY Vol. 136, No. 3

ostfixed in buffered 1% osmium tetroxide, washed inriple-distilled water, dehydrated in a graded ethyl alco-ol series, infiltrated and embedded with Epon resin

Electron Microscopy Sciences, Fort Washington, PA),nd polymerized at 60°C for 48 hours. Multiple 1-�mections were stained with 0.5% Tolidine blue and 0.5%orax, and evaluated by light microscopy. Representativereas were selected. Silver sections, 70 nm, mounted on00 mesh Athene copper grids (Ted Pella, Inc, Redding,A), were stained with uranyl acetate followed by Reyn-ld’s lead citrate (Sigma–Aldrich) and examined with aEOL 1230 transmission electron microscope (JEOL,SA, Peabody, MA) at 60 kV. Representative digital im-

ges were taken using a SIS Keenview camera and soft-are (Olympus Soft Imaging Solutions GmbH, Muen-

ter, Germany).

Measurement of Coagulation Factor VIIActivityCoagulation factor VII (CF VII) activity was mea-

ured by monitoring the release of p-nitroanilide frompectrozyme (American Diagnostics, Inc, Stamford, CT)

igure 1. Differentiation of hES cells towards a hepatocyte phenotypeiation of hES cells to hepatocytes. (B) RT-PCR and (C) real-time analys, glucose-6-phosphatase [G6Pase], tyrosine aminotransferase [TAT], anntiated cells (Pou5f1 and Nanog). Lanes: (1) undifferentiated hES cellsactor-2 (FGF-2) and activin-A or at the definitive endoderm (DE) stage, (4n dexamethasone (Diff). Primary human hepatocytes served as the pon supplementary Table 1 (see supplementary material online at www.ifferentiation protocol, after final exposure to dexamethasone (Diff-hESiated hES (Undiff hES), primary human hepatocytes (HH), and HepG2 c

xpressed albumin whereas only 12% co-expressed AFP.

Xa by factor Xa, generated from factor X by tissueactor-bound CF VII.20 A Tmax plate reader (Molecularevices, Sunnyvale, CA) was used to monitor the assays

onducted in 96-well plates (Falcon/BD Biosciences, Sanose, CA). Stocks of purified CF VIIa (Novo Nordisk,rinceton, NJ) and factor X (purified from human plas-a21) and homogenized human brain powder22 were

iluted before use in Tris-buffered saline (0.05 mol/Lris, 0.1 mol/L NaCl, pH 7.6) containing 1 mg/mL bo-ine serum albumin (Sigma). For measurement of CF VII,0 uL of spent medium was combined with 10 uL of theris buffer with bovine serum albumin, 50 uL of 1/100ilution of brain homogenate, 20 uL factor X (concen-ration in the final 160-uL assay volume was approxi-

ately 100 nmol/L), 50 uL of 25 mmol/L calcium chlo-ide, and 10 uL of Spectrozyme FXa (final concentration10 umol/L). A CF VIIa calibration curve was generatedoncurrently using serial dilutions of the pure CF VIIa inlace of the spent medium. The plot of factor Xa [pmol/L]/in vs CF VIIa [pmol/L] was described by y � (1141

�0.73])/([�0.73] � 45.8) � 0.44 in the second assay, for

characterization of hepatocyte-directed cells. (A) Strategy for differen-expression of lineage-specific hepatic markers (albumin [ALB], ASGPRVII), endoderm markers (SOX 17 and AFP), and markers for undiffer-

f), (2) after EB formation (EBs), (3) after treatment with fibroblast growthr culture in HGF and dimethyl sulfoxide (Early Diff), and (5) after culturecontrol. Forward and reverse primers used for these studies are listedojournal.org). (D) Immunohistochemistry of hES cells at day 18 of thealbumin, AFP, and merged albumin and AFP expression. Undifferen-ere used as controls. Scale bar, 50 �m. Approximately 55% of cells

andis ford CF

(Undif) afte

sitivegastr), forells w

Page 4: BASIC—LIVER, PANCREAS, AND BILIARY TRACT · 2019. 7. 10. · BILIARY TRACT GASTROENTEROLOGY 2009;136:990–999. differentiation strategy that produces a relatively homo-geneous

wt

pLlwpowwommtfl5ccattav

(tMb1fmFg

ptcysgmNgNsc

t

pubsdiaatsmcea

FldtTaewwfim

BA

SIC–L

IVER

,PA

NCREA

S,A

ND

BIL

IARY

TRA

CT

March 2009 HUMAN ES CELL–DERIVED HEPATOCYTES 993

hich the CF VIIa dilutions covered the range of activi-ies measured with the spent medium.

Metabolic ActivityES-derived or normal human hepatocytes were

lated in hepatocyte maintenance medium (HMM,onza, Walkersville, MD) media at a density of 1.5 mil-

ion cells per well in collagen-coated, 6-well plates. Cellsere cultured for 72 hours in the presence of 1 mmol/Lhenobarbital or 25 �mol/L �-naphthoflavone as previ-usly described.23,24 After the induction period, cells wereashed 3 times to remove inducers, and media was replacedith fresh media (without inducers). For the measurementf cytochrome P450 (CYP) 1A activity, cells were exposed toedia containing 20 �mol/L 7-ethoxyresorufin and 1.5mol/L salicylamide for 1 hour. The conversion of 7-ethoxy

o 7-hydroxyresorufin in the media was quantified by theuorescence of the 7-hydroxy metabolite measured at35 nm (fluorescence excitation) and 581 nm (fluores-ence emission).24 Salicylamide is used to prevent theonjugation of resorufin formed by de-ethylation. Thenalysis of CYP3A activity was measured by exposing cellso 350 �mol/L testosterone for 1 hour. The conversion ofestosterone to 6�-hydroxytestosterone was measured by

high-pressure liquid chromatographic method as pre-iously described.23

ResultsDifferentiation of hES Toward a HepatocytePhenotypeTo generate ES cell– derived hepatocytes, human

H1) ES cells were plated on low-attachment Petri disheso form embryoid bodies. EBs then were plated on 5%

atrigel for 3 days in recombinant activin-A and fibro-last growth factor-2. Cells then were grown for 8 days in0% serum (or KSR) containing HGF, followed by cultureor 3 additional days in 10% serum or KSR, and 10�7

ol/L dexamethasone (Figure 1A and supplementaryigure 1; see supplementary material online at www.astrojournal.org).

Real-time and RT-PCR analysis was performed on cellreparations at various times during the culture processo determine the time course and degree to which the ESells differentiated toward a hepatocyte phenotype. Anal-sis included markers for endoderm-specific gene expres-ion (sox 17 and �-fetoprotein [AFP]), hepatocyte-specificene expression [albumin, ASGPR 1, and/or CF VII], andarkers for undifferentiated hES cells (Pou5f1 andanog). As shown in Figure 1B and C, endoderm-specific

ene expression increased early whereas Pou5f1 andanog expression gradually decreased and hepatocyte-

pecific gene expression progressively increased over theourse of the differentiation program.

To determine the percentage of cells differentiating

oward a hepatic phenotype, immunohistochemistry was t

erformed for albumin and AFP. As shown in Figure 1D,ndifferentiated H1 hES cells had large nuclei, as showny 4=,6-diamidino-2-phenylindole staining, and expressedome AFP, probably representing low-grade spontaneousifferentiation in culture, but no albumin. After culture

n growth factors, approximately 55% of cells expressedlbumin, only 12% of which co-expressed AFP. There waslso a dramatic change in morphology during the course ofhe differentiation process. As shown in Figure 2, polygonal-haped cells containing multiple nuclei formed and trans-

ission electron microscopy revealed cells containing gly-ogen granules (Figure 2B), mature round nuclei withvenly distributed chromatin, Golgi complexes (Figure 2C),nd well-developed bile canaliculi with apical microvilli and

igure 2. Morphologic analysis of differentiated hES cells. Morpho-ogic analysis of differentiated hES cells in culture indicate that theifferentiation program generated cells morphologically similar to hepa-ocytes, (A) being polygonal in shape with multiple nuclei (arrows).ransmission electron microscopy of differentiated hES showed (B)ccumulation of glycogen rosettes (arrows), (C) round nuclei (Nu) withvenly distributed chromatin, Golgi complexes (G), and liposomes (Li),ith glycogen rosettes identified again by an arrow, and (D and E)ell-developed bile canaliculi (BC) with apical microvilli (Mv) containing

laments (F), and tight junctions (arrows). Scale bar, 50 �m. Originalagnification, 30,000�.

ight junctions (Figure 2D and E).

Page 5: BASIC—LIVER, PANCREAS, AND BILIARY TRACT · 2019. 7. 10. · BILIARY TRACT GASTROENTEROLOGY 2009;136:990–999. differentiation strategy that produces a relatively homo-geneous

mpathrdOitmtUslsS5npa3Ldcec

getemi

etcccfiGutwdhaahe

Fdtttai expre

BA

SIC–LIV

ER,

PA

NCREA

S,A

ND

BILIA

RY

TRA

CT

994 BASMA ET AL GASTROENTEROLOGY Vol. 136, No. 3

Transplant studies then were performed to deter-ine whether differentiated hES cells would, similar to

rimary hepatocytes, home to and engraft in the liverfter injection in the spleen. To allow noninvasiveracking of cells after transplantation, differentiatedES cells were transduced with a recombinant lentivi-us containing the gene encoding firefly luciferase un-er the control of the human albumin promoter.25

ne million cells were transplanted into the spleens ofmmune-deficient NOD/SCID mice. Before transplan-ation, recipient mice were injected twice with 70

g/kg retrorsine, and underwent a 50% partial hepa-ectomy at the time of transplantation (Figure 3A).nder these conditions, the transplanted cells have a

elective growth advantage compared with the nativeiver cells, whose proliferation is blocked by retror-ine.17,18 After transplantation, the serum of NOD/CID mouse recipients of differentiated hES contained00 –1000 ng/mL human albumin and 100 –200g/mL human AAT (Figure 3B). Control mice trans-lanted with primary porcine hepatocytes containedpproximately 500 ng/mL porcine albumin (FigureB). As shown in Figure 3C, whole mouse IVIS (Caliperife Sciences, Hopkinton, MA) imaging performed 21ays after transplantation revealed that differentiatedells containing an active albumin promoter, and thusxpressing luciferase, had traveled through the portal

igure 3. Transplantation of differentiated cells and characterizationifferentiated hES cells were transplanted into retrorsine-treated immunation, (B) human albumin and human �1AT levels were measured byransplanted with primary porcine hepatocytes are shown for comparisransplantation. Before transplantation, differentiated hES cells were tralbumin promoter, and thus expressing luciferase, appear to have trave

n the livers of transplanted mice. There was no evidence of luciferase-

irculation after introduction into the spleen to en- n

raft in the livers of transplanted mice. No luciferase-xpressing ES-derived cells appeared in the location ofhe spleen. At death on posttransplant day 21, how-ver, gross and histologic examination revealed terato-as in the liver, spleen, and peritoneal cavity contain-

ng poorly differentiated cells.

Enrichment for hES-Derived HepatocytesBased on ASGPR ExpressionTo isolate the population of cells with a differ-

ntiated hepatocyte phenotype and reduce or eliminatehe undifferentiated or poorly differentiated cells, orells that might have differentiated toward a differentell lineage, differentiated hES cells were sorted by flowytometry for expression of the ASGPR, a definitiveeature of hepatocytes. As shown in Figure 4A, approx-mately 18%–26% of hES-derived cells expressed AS-

PR. Nucleic acids from the sorted population, fromndifferentiated hES cells, and from hES cells duringhe various stages of the differentiation process thenere assessed by RT-PCR and real-time analysis toetermine the extent to which enrichment toward aepatocyte phenotype resulted from sorting (Figure 4Bnd C). Real-time PCR showed that the sorted cells had

gene expression profile near that of primary adultuman hepatocytes. Cells also expressed the genesncoding bilirubin-UDP glucuronosyl transferase, or-

lls sorted for surface ASGPR expression. (A) One million hepatocyte-ficient NOD/SCID mice after 50% partial hepatectomy. After transplan-yme-linked immunosorbent assay. Pig albumin levels in control mice) Imaging for luciferase-expressing cells was performed 3 weeks after

ced to express firefly luciferase. Differentiated cells containing an activerough the portal circulation after introduction into the spleen to engraftssing cells in the spleen.

of cee-deenz

on. (Cnsduled th

ithine transcarbamylase, and the bile salt export

Page 6: BASIC—LIVER, PANCREAS, AND BILIARY TRACT · 2019. 7. 10. · BILIARY TRACT GASTROENTEROLOGY 2009;136:990–999. differentiation strategy that produces a relatively homo-geneous

pmp(dpeCnisspf

fobp

hVh4hbmt(sts2vmifi

aS

F1ph[NcRechic

BA

SIC–L

IVER

,PA

NCREA

S,A

ND

BIL

IARY

TRA

CT

March 2009 HUMAN ES CELL–DERIVED HEPATOCYTES 995

ump (supplementary Figure 2A; see supplementaryaterial online at www.gastrojournal.org). RT-PCR

erformed for Pdx-1 (a pancreatic marker gene), Nestinectoderm), Brachury (mesendoderm), NKx2.5 (meso-erm), and Pax-6 (ectoderm) showed that the sortingrocess significantly enriched for cells that did notxpress genes representing other lineages (Figure 4D).omplete loss of Pou5f1 and Nanog indicated elimi-ation of less differentiated cells, and loss of Sox17

ndicated enrichment of cells beyond the endodermtage of development. A small amount of AFP expres-ion remained present in sorted cells, indicating theresence of cells that could represent late-gestationetal hepatocytes.

Characterization and Transplantation ofASGPR-Enriched ES-Derived CellsTo determine the capacity of differentiated hES to

unction as primary hepatocytes we examined productionf CF VII, a gene product specific to hepatocytes. Westernlot (Figure 4E) and functional assay of vitamin K–sup-

igure 4. Enrichment of hES differentiated cells by ASGPR express8%–26% of differentiated cells expressed the ASGPR, a definitive featerformed to determine gene expression levels during the various staepatocytes (HH). Expression levels of lineage-specific hepatic marke

G6Pase], CF VII, and ASGPR1), endoderm (Sox17) and the gestationalanog) were examined and messenger RNA expression levels were nyclophilin, and primary human hepatocytes. (D) To assess the lineagT-PCR for Pdx-1 (a pancreatic marker gene), Nestin (ectoderm), Bracntiated hES (lane 1), after EB formation (lane 2), hES differentiated to Dulture in dexamethasone (Diff; lane 5), and Diff stage cells after enepatocytes (lane 7) were used as control cells. (E) Production of CF VII

mmunoblot, and (F) functional CF VII activity in culture supernatants sultured primary human hepatocytes.

lemented cell supernatants from sorted ES-derived d

epatocytes showed production of functionally active CFII on the order of that produced by primary humanepatocytes cultured under the same conditions (FigureF). Cells also were assessed for their ability to generateuman albumin, AAT, and urea in culture. Cells enrichedy sorting for ASGPR expression generated approxi-ately 75% of the albumin, 65% of the AAT, and 65% of

he urea generated by primary human hepatocytesFigure 5A). ES-derived cells also were assessed for expres-ion of human hepatocyte–specific cytochrome P450 me-abolizing genes and P450 metabolic activity. ASGPR-orted differentiated hES cells expressed CYP 1A1, 1A2,B6, 3A4, and 7A1 by real-time PCR (Figure 5B), con-erted testosterone to 6-�-hydroxytestosterone, a specificeasure of CYP 3A4 –mediated metabolism, and showed

nducible hepatic CYP 1A1/1A2–mediated ethoxyresoru-n–O-deethylase activity (Figure 5C).A total of 100,000 –200,000 ASGPR-sorted differenti-

ted cells then were transplanted into each of 5 Alb-uPACID mice. The Alb-uPA SCID mouse is a T- and B-cell–

nd characterization of sorted cells. (A) Flow cytometry showed thatf differentiated hepatocytes. (B) RT-PCR and (C) real-time analysis wasof differentiation and after sorting relative to levels in primary humanbumin [ALB], tyrosine aminotransferase [TAT], glucose-6-phosphatasetocyte marker AFP, and markers for undifferentiated cells (Pou5f1 andlized relative to glyceraldehyde-3-phosphate dehydrogenase (GAPDH),cificity of the differentiation program, mRNA was analyzed further by

(mesendoderm), NKx2.5 (mesoderm), and Pax-6 (ectoderm). Undiffer-e 3), after culture in HGF and dimethyl sulfoxide (Early Diff; lane 4), afterent for ASGPR surface expression (Sorted; lane 6). Primary humanin by unsorted and ASGPR-sorted hES-derived cells was confirmed bymented with vitamin K was shown to be similar to that produced by

ion aure ogesrs (alhepaormae spehuryE (lanrichmproteupple

eficient mouse that carries a tandem array of murine

Page 7: BASIC—LIVER, PANCREAS, AND BILIARY TRACT · 2019. 7. 10. · BILIARY TRACT GASTROENTEROLOGY 2009;136:990–999. differentiation strategy that produces a relatively homo-geneous

umpetmcmirl1dsmtwepN

rwtNtpohpsndr26rgTi

Fd((aEmmwib d celB

ASIC

–LIVER

,PA

NCREA

S,A

ND

BILIA

RY

TRA

CT

996 BASMA ET AL GASTROENTEROLOGY Vol. 136, No. 3

rokinase genes under the control of an albumin pro-oter.15,26 The transgene is toxic to the host liver, and

revents native hepatocytes from responding with regen-ration. At 75 days after transplantation, the serum ofhese animals contained 1000 –2000 ng/mL human albu-

in and AAT (Figure 6A). Serum albumin levels fromontrol animals transplanted with 1,000,000 primary hu-an hepatocytes derived from fresh liver resection spec-

mens (supplementary Table 2; see supplementary mate-ial online at www.gastrojournal.org) or cadaver donorivers not used for transplantation range from less than000 ng/mL to greater than 1 mg/mL depending ononor age and source.27 At death, examination of liverpecimens from transplanted animals revealed small hu-

an cytokeratin-18 staining clusters of engrafted cellshroughout the liver (Figure 6B). No teratomas or tumorsere identified in these animals by gross or histologic

xamination. One million sorted cells also were trans-lanted into the spleen of 1 FK506-immune suppressed

igure 5. Functional analyses of differentiated hES cells enriched toetermined in vitro by primary human hepatocytes (HH) and hES cells

Undiff), hES cells after culture in dexamethasone (Diff), and hES cellssorted). Culture medium used control. (B) Real-time analysis showedt levels similar to that derived from fresh primary human hepatocytesS-derived or normal human hepatocytes were cultured in the preseent of CYP 1A activity, cells were exposed to media containing 2edia was quantified by the fluorescence of the 7-hydroxy metabolitas measured by conversion of testosterone to 6�-hydroxytestos

nducible ethoxyresorufin–O-deethylase (EROD) activity at approximaaseline formation of testosterone by differentiated ASGPR-enriche

agase analbuminemic rat. Before transplantation, the r

ecipient animal was injected with retrorsine and under-ent a 70% partial hepatectomy at the time of transplan-

ation, similar to what had been performed previously inOD-SCID mice, to create an environment in which

here was a selective growth advantage to the trans-lanted cells. At 28 days after transplantation the bloodf this recipient contained 2000 ng/mL (0.002 mg/mL)uman albumin. For comparison, 28 days after trans-lantation with 5,000,000 primary pig hepatocytes,erum porcine albumin levels of approximately 5000g/mL (0.005 mg/mL) were obtained (Figure 6C). At 55ays after transplantation the blood of the animal thateceived ASGPR-sorted differentiated cells contained0,000 ng/mL (0.02 mg/mL) human albumin (FigureC). Examination of liver specimens from this animalevealed large human albumin staining clusters of en-rafted cells present throughout the liver (Figure 6D).hus, ES-derived human hepatocytes were capable of hom-

ng to and engrafting normally in the liver, expanding in

a hepatocyte phenotype. (A) Albumin, urea, and AAT secretion wasg differentiation (n � 5). Analysis involved undifferentiated hES cells

r the Diff stage and after enrichment for ASGPR surface expressionession of cytochrome P450 1A1 (CYP1A1), 1A2, 3A4, 2B6, and 7A1To assess human liver–specific cytochrome P450 metabolic activity,f phenobarbital or 25 �mol/L �-naphthoflavone (BNF). For measure-ol/L 7-ethoxyresorufin and conversion to 7-hydroxyresorufin in theasured at 535 nm (Ex) and 581 nm (Em). Analysis of CYP3A activitye by high-pressure liquid chromatography. Studies showed BNF-25%–30% of that generated by primary human hepatocytes, andls near that produced by cultured primary human hepatocytes.

warddurinafteexpr. (C)nce o0 �me meterontely

esponse to a physiologic proliferation signal, and secreting

Page 8: BASIC—LIVER, PANCREAS, AND BILIARY TRACT · 2019. 7. 10. · BILIARY TRACT GASTROENTEROLOGY 2009;136:990–999. differentiation strategy that produces a relatively homo-geneous

ftpw

tw

dtmbawidfwftn

gettecMgetsoupf

ttAbpAbuTetmccacAwo

Fcmbdabchacpatut(tswto

BA

SIC–L

IVER

,PA

NCREA

S,A

ND

BIL

IARY

TRA

CT

March 2009 HUMAN ES CELL–DERIVED HEPATOCYTES 997

unctional liver-specific proteins after engraftment. Unfor-unately, although there were no intrahepatic tumors, theeritoneum contained tumors histologically consistent

igure 6. Transplantation of ASGPR-enriched hES-derived hepato-ytes in uPA-SCID mice and an immune-suppressed Nagase analbu-inemic rat. A total of 100,000–200,000 hES-derived cells, sortedased on surface ASGPR expression, were transplanted in immune-eficient Alb-uPA SCID mice. (A) After transplantation, human albuminnd human �1AT levels were measured by enzyme-linked immunosor-ent assay. At 75 days after transplantation, the serum of these animalsontained 1000–2000 ng/mL human albumin and AAT. (B) Immuno-istochemistry was performed on liver sections of transplanted animals,nd small clusters of human cytokeratin-18 (CK18)-staining engraftedells, which stain dark brown, with hepatocyte morphology, wereresent throughout the liver. (C) One million ASGPR-sorted differenti-ted hES cells also were transplanted into the spleen of a retrorsine-reated FK506 immune-suppressed Nagase analbuminemic rat thatnderwent 70% partial hepatectomy at the time of transplantation. Afterransplantation, human albumin levels were measured at 20,000 ng/mL0.2 mg/mL) by enzyme-linked immunosorbent assay 55 days afterransplantation. Comparable serum porcine albumin levels were mea-ured in control Alb-uPA SCID mice up to 28 days after transplantationith 4°C 24-hour, University of Wisconsin–preserved primary pig hepa-

ocytes. (D) At that time, immunohistochemistry showed large coloniesf human albumin–expressing cells. ALB, albumin.

ith well-differentiated adenocarcinoma (supplemen- m

ary Figure 2B; see supplementary material online atww.gastrojournal.org).

Discussion

In these studies, we present a simple and repro-ucible method for generating functional human hepa-ocytes from pluripotent ES cells. Although successful

ethods for hepatic differentiation of hES cells haveeen described, none have generated cells with functiondequate for clinical use.8,12,28 –30 We have not determinedhether co-culture with liver nonparenchymal cells might

ncrease the efficiency of hepatic differentiation, as has beenescribed for mouse ES cell differentiation.31 Hepatic dif-erentiation, greater than the 18%–26% described here,ould be desirable, but the simplicity of the protocol may

acilitate clinical application and the eventual scaling uphat will be required to generate ES-derived hepatocytes inumbers that could be used in patients.The protocol we describe incorporates a step involving

enerating EBs. However, based on the work of D’Amourt al,32 we have performed a number of additional real-ime PCR and albumin and urea production experimentshat show essentially identical hepatocyte-specific differ-ntiation whether or not the EB formation step is in-luded in the differentiation program (supplementary

ethods; see supplementary material online at www.astrojournal.org). In fact, there is more extensive andarlier loss of Octamer 3/4, Nanog, Sox7, and AFP whenhe EB step is removed (supplementary Figure 3; seeupplementary material online at www.gastrojournal.rg). In addition, our ability to induce differentiationsing KSR instead of serum allows removal of animalroducts from the process, an important considerationor clinical application.

A critically important component of the differentia-ion protocol relates to enrichment for ES-derived hepa-ocytes. ASGPR expression is unique for liver cells.33

lthough 55% of differentiated cells expressed albuminy immunohistochemistry, significantly fewer cells ex-ressed ASGPR, indicating that enrichment based onSGPR expression may be more selective than sortingased on previously described gene transfer techniquessing reporter genes driven from liver-specific promoters.he present strategy does not depend on transductionfficiency for selecting a relatively homogeneous popula-ion of cells. Although we used flow sorting for enrich-

ent, this technique is relatively time consuming andan lead to significant cell injury. In addition, its efficacyan be affected by sorting speed, which can seriouslyffect the yield and viability of the differentiated ESells recovered and allow recovery of unwanted cells. TheSGPR-based sorting approach outlined, however, alsoould be amenable to enrichment by magnetic sorting34

r panning on antibody-coated plates. Such strategies

ight be more efficient than flow sorting because the
Page 9: BASIC—LIVER, PANCREAS, AND BILIARY TRACT · 2019. 7. 10. · BILIARY TRACT GASTROENTEROLOGY 2009;136:990–999. differentiation strategy that produces a relatively homo-geneous

sarmr

tnipusncAdldlsbEeetsnEccspthstrl

sccscasfsebetEmp

aG1

1

1

1

1

1

1

1

1

1

1

BA

SIC–LIV

ER,

PA

NCREA

S,A

ND

BILIA

RY

TRA

CT

998 BASMA ET AL GASTROENTEROLOGY Vol. 136, No. 3

teps can be performed repeatedly for enhanced selection,nd would result in significantly less damage to theecovered cells. In addition, they might be faster and

ore appropriate for large-scale, high-throughput en-ichment than flow cytometry.

As shown in our studies, tumor risk remains an issuehat must continue to be addressed. It appears thateither the number of cells that can be transplanted into

mmune-deficient mice nor the length of time trans-lanted rodents can be followed up will be adequate tonequivocally determine whether cell preparations areafe for clinical use. Large-scale studies, performed inonhuman primates using frozen stocks of differentiatedells, when possible, may be needed for such an analysis.lthough ES cell– derived hepatocytes may not be imme-iately useful for transplantation therapies, they are

ikely to find early application for the study of humanrug metabolism and drug discovery. Mature human

ivers express important drug-metabolizing enzymes,35

ome of which are inducible after exposure to phenobar-ital or �-naphthoflavone.23,24 Our studies indicate thatS-derived hepatocytes express human CYP genes at lev-ls near those of adult human hepatocytes, and that priorxposure to �-naphthoflavone results in robust induc-ion in the metabolism of ethoxyresorufin, a known sub-trate for CYP 1A1/2, by both ES-derived hepatocytes andormal human liver cells. Our studies also show thatS-derived hepatocytes and normal human hepatocytesonvert testosterone to 6-�-hydroxytestosterone, a spe-ific measure of CYP 3A4 –mediated metabolism, to aimilar degree in culture. Although prior exposure tohenobarbital did not increase the metabolism of testos-erone in ES-derived cells, as occurs with normal humanepatocytes (data not shown), the CYP metabolic activityhown by ES-derived cells is substantial. It is possiblehat further maturation of CYP functional activity mayequire differentiation on a more physiologic extracellu-ar matrix or interaction with other cells.

Although we have examined the use of hES cells in thiseries of experiments, successful generation of hepato-ytes from precursors derived from individual patients36

ould lead to the development of individualized patient-pecific drug regimens and eventually might be used toircumvent the need for life-long immune suppressionfter hepatocyte transplantation. In summary, thesetudies provide a foundation for efficient development ofunctional human hepatocytes from hES cells. Furthertudies will be needed to determine whether the differ-ntiation protocol and enrichment strategy outlined cane scaled for use in patients and can be modified toliminate the risk of contaminating cells and the risk ofumor formation after transplantation. Application ofS-derived hepatocytes for the study of human drugetabolism and drug discovery, however, may soon be

ossible.

Supplementary Data

Note: To access the supplementary materialccompanying this article, visit the online version ofastroenterology at www.gastrojournal.org, and at doi:0.1053/j.gastro.2008.10.047.

References

1. Matas AJ, Sutherland DE, Steffes MW, et al. Hepatocellular trans-plantation for metabolic deficiencies: decrease of plasma biliru-bin in Gunn rats. Science 1976;192:892–894.

2. Groth CG, Arborgh B, Bjorken C, et al. Correction of hyperbiliru-binemia in the glucuronyltransferase-deficient rat by intraportalhepatocyte transplantation. Transplant Proc 1977;9:313–316.

3. Fox IJ, Roy-Chowdhury J. Hepatocyte transplantation. J Hepatol2004;40:878–886.

4. Azuma H, Paulk N, Ranade A, et al. Robust expansion of humanhepatocytes in Fah-/-/Rag2-/-/Il2rg-/- mice. Nat Biotechnol2007;25:903–910.

5. Mercer DF, Schiller DE, Elliott JF, et al. Hepatitis C virus replica-tion in mice with chimeric human livers. Nat Med 2001;7:927–933.

6. Pouton CW, Haynes JM. Embryonic stem cells as a source ofmodels for drug discovery. Nat Rev Drug Discov 2007;6:605–616.

7. Khetani SR, Bhatia SN. Microscale culture of human liver cells fordrug development. Nat Biotechnol 2008;26:120–126.

8. Lavon N, Yanuka O, Benvenisty N. Differentiation and isolation ofhepatic-like cells from human embryonic stem cells. Differentia-tion 2004;72:230–238.

9. Ruhnke M, Ungefroren H, Nussler A, et al. Differentiation of invitro-modified human peripheral blood monocytes into hepato-cyte-like and pancreatic islet-like cells. Gastroenterology 2005;128:1774–1786.

0. Hay DC, Fletcher J, Payne C, et al. Highly efficient differentiationof HESCs to functional hepatic endoderm requires ActivinA andWnt3a signaling. Proc Natl Acad Sci U S A 2008;105:12301–12306.

1. Campard D, Lysy PA, Najimi M, et al. Native umbilical cord matrixstem cells express hepatic markers and differentiate into hepa-tocyte-like cells. Gastroenterology 2008;134:833–848.

2. Cai J, Zhao Y, Liu Y, et al. Directed differentiation of humanembryonic stem cells into functional hepatic cells. Hepatology2007;45:1229–1239.

3. Miki T, Lehmann T, Cai H, et al. Stem cell characteristics ofamniotic epithelial cells. Stem Cells 2005;23:1549–1559.

4. Khurana S, Mukhopadhyay A. In vitro transdifferentiation of adulthematopoietic stem cells: an alternative source of engraftablehepatocytes. J Hepatol 2008;49:998–1007.

5. Rhim JA, Sandgren EP, Palmiter RD, et al. Complete reconstitu-tion of mouse liver with xenogeneic hepatocytes. Proc Natl AcadSci U S A 1995;92:4942–4946.

6. Nagase S, Shimamune K, Shumiya S. Albumin-deficient rat mu-tant. Science 1979;205:590–591.

7. Guo D, Fu T, Nelson JA, et al. Liver repopulation after cell trans-plantation in mice treated with retrorsine and carbon tetrachlo-ride. Transplantation 2002;73:1818–1824.

8. Laconi E, Oren R, Mukhopadhyay DK, et al. Long-term, near-total liver replacement by transplantation of isolated hepato-cytes in rats treated with retrorsine. Am J Pathol 1998;153:319–329.

9. Overbergh L, Giulietti A, Valckx D, et al. The use of real-timereverse transcriptase PCR for the quantification of cytokine gene

expression. J Biomol Tech 2003;14:33–43.
Page 10: BASIC—LIVER, PANCREAS, AND BILIARY TRACT · 2019. 7. 10. · BILIARY TRACT GASTROENTEROLOGY 2009;136:990–999. differentiation strategy that produces a relatively homo-geneous

2

2

2

2

2

2

2

2

2

2

3

3

3

3

3

3

3

R

Se

A

s

C

F

SSS

March 2009 HUMAN ES CELL–DERIVED HEPATOCYTES 999

0. Carson SD. Manifestation of cryptic fibroblast tissue factor oc-curs at detergent concentrations which dissolve the plasmamembrane. Blood Coagul Fibrinolysis 1996;7:303–313.

1. Broze GJ Jr, Majerus PW. Purification and properties of humancoagulation factor VII. J Biol Chem 1980;255:1242–1247.

2. Pitlick FA, Nemerson Y. Purification and characterization of tissuefactor apoprotein. Methods Enzymol 1976;45:37–48.

3. Kostrubsky VE, Ramachandran V, Venkataramanan R, et al. Theuse of human hepatocyte cultures to study the induction ofcytochrome P-450. Drug Metab Dispos 1999;27:887–894.

4. Wen YH, Sahi J, Urda E, et al. Effects of bergamottin on humanand monkey drug-metabolizing enzymes in primary cultured hepa-tocytes. Drug Metab Dispos 2002;30:977–984.

5. Lu Y, Dang H, Middleton B, et al. Bioluminescent monitoring ofislet graft survival after transplantation. Mol Ther 2004;9:428–435.

6. Meuleman P, Libbrecht L, De Vos R, et al. Morphological andbiochemical characterization of a human liver in a uPA-SCIDmouse chimera. Hepatology 2005;41:847–856.

7. Utoh R, Tateno C, Yamasaki C, et al. Susceptibility of chimericmice with livers repopulated by serially subcultured human hepa-tocytes to hepatitis B virus. Hepatology 2008;47:435–446.

8. Schwartz RE, Linehan JL, Painschab MS, et al. Defined conditionsfor development of functional hepatic cells from human embry-onic stem cells. Stem Cells Dev 2005;14:643–655.

9. Ek M, Soderdahl T, Kuppers-Munther B, et al. Expression of drugmetabolizing enzymes in hepatocyte-like cells derived from humanembryonic stem cells. Biochem Pharmacol 2007;74:496–503.

0. Heo J, Factor VM, Uren T, et al. Hepatic precursors derived frommurine embryonic stem cells contribute to regeneration of injuredliver. Hepatology 2006;44:1478–1486.

1. Soto-Gutierrez A, Kobayashi N, Rivas-Carrillo JD, et al. Reversal ofmouse hepatic failure using an implanted liver-assist devicecontaining ES cell-derived hepatocytes. Nat Biotechnol 2006;24:1412–1419.

2. D’Amour KA, Bang AG, Eliazer S, et al. Production of pancreatichormone-expressing endocrine cells from human embryonic stem

cells. Nat Biotechnol 2006;24:1392–1401. a

3. Stockert RJ. The asialoglycoprotein receptor: relationships be-tween structure, function, and expression. Physiol Rev 1995;75:591–609.

4. Handgretinger R, Lang P, Ihm K, et al. Isolation and transplanta-tion of highly purified autologous peripheral CD34(�) progenitorcells: purging efficacy, hematopoietic reconstitution and long-term outcome in children with high-risk neuroblastoma. BoneMarrow Transplant 2002;29:731–736.

5. Strom SC, Pisarov LA, Dorko K, et al. Use of human hepatocytesto study P450 gene induction. Methods Enzymol 1996;272:388–401.

6. Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotentstem cells from adult human fibroblasts by defined factors. Cell2007;131:861–872.

Received August 28, 2008. Accepted October 23, 2008.

eprint requestsAddress requests for reprints to: Ira J. Fox, MD, Department of

urgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213.-mail: [email protected]; fax: (412) 692-6599.

cknowledgementsThe authors thank Melissa Holzapfel for careful preparation of

amples for electron microscopy.

onflict of interestThe authors disclose no conflicts.

undingThe authors disclose the following: Supported by a Grant-in-Aid for

cientific Research (B) of the Japan Society for the Promotion ofcience (to N.K.), National Institutes of Health grants DK-7-0004 (to.C.S), HL52297 (to J.L.P.), DK 067440 (to J.R.–C.), and DK48794

nd AI49472 (to I.J.F.).

BA

SIC–L

IVER

,PA

NCREA

S,A

ND

BIL

IARY

TRA

CT

Page 11: BASIC—LIVER, PANCREAS, AND BILIARY TRACT · 2019. 7. 10. · BILIARY TRACT GASTROENTEROLOGY 2009;136:990–999. differentiation strategy that produces a relatively homo-geneous

S

O

N

S

A

A

C

A

P

N

B

P

N

G

G

999.e1 BASMA ET AL GASTROENTEROLOGY Vol. 136, No. 3

upplementary Table 1. Specific Primers Used for RT-PCR

Genes Primer sequence Product size

ct3/4 5=-TGGGCTCGAGAAGGATGTG-3= 1505=-GCATAGTCGCTGCTTGATCG-3=

anog 5=-TGAACCTCAGCTACAAACAG-3= 3505=-AACTGCATGCAGGACTGCA-3=

ox17 5=-GGCGCAGCAGAATCCAGA-3= 3005=-CCACGACTTGCCCAGCAT-3=

lbumin 5=-ACTTTTATGCCCCGGAACTC-3= 1505=-AGCAGCAGCACGACAGAGTA-3=

FP 5=-ACTGAATCCAGAACACTGCA-3= 1705=-TGCAGTCAATGCATCTTTCA-3=

F VII 5=-GGGAGACTCCCCAAATATCA-3= 5805=-ACGCAGCCTTGGCTTTCTCT-3=

SGR1 5=-GCTCCACGTGAAGCAGTT-3= 6705=-AACTGCAGAAAGCGCCAC-3=

DX1 5=-TGGGCTCGAGAAGGATGTG-3= 3205=-GCATAGTCGCTGCTTGATCG-3=

estin 5=-TGGGCTCGAGAAGGATGTG-3= 3805=-GCATAGTCGCTGCTTGATCG-3=

rachury 5=-TGGGCTCGAGAAGGATGTG-3= 2405=-GCATAGTCGCTGCTTGATCG-3=

ax6 5=-TGGGCTCGAGAAGGATGTG-3= 3305=-GCATAGTCGCTGCTTGATCG-3=

kx 2-5 5=-TGGGCTCGAGAAGGATGTG-3= 1605=-GCATAGTCGCTGCTTGATCG-3=

APDH 5=-GTTGCCATCAATGACCCCTTCATTG-3= 6805=-GCTTCACCACCTTCTTGATGTCATC-3=

APDH, glyceraldehyde-3-phosphate dehydrogenase.

Page 12: BASIC—LIVER, PANCREAS, AND BILIARY TRACT · 2019. 7. 10. · BILIARY TRACT GASTROENTEROLOGY 2009;136:990–999. differentiation strategy that produces a relatively homo-geneous

March 2009 HUMAN ES CELL–DERIVED HEPATOCYTES 999.e2

Supplementary Figure 1. Experimental design. FITC, fluorescein isothiocyanate.

Page 13: BASIC—LIVER, PANCREAS, AND BILIARY TRACT · 2019. 7. 10. · BILIARY TRACT GASTROENTEROLOGY 2009;136:990–999. differentiation strategy that produces a relatively homo-geneous

T

AAA

Nr

Stteiistics of a well-differentiated adenocarcinoma.

999.e3 BASMA ET AL GASTROENTEROLOGY Vol. 136, No. 3

able 2. Transplantation of Control Primary Hepatocytes in uPA-SCID Mice

Maximum human albumin level

Hepatocyte donor group Transplanted animals (n) None 1000–50,000 ng/mL 50,000 ng/mL to 1 mg/mL �1 mg/mL

ge �20 y, fresh 15 3 (20%) 7 (47%) 3 (20%) 2 (13%)ge �20 y, cryopreserved 18 8 (44%) 1 (6%) 5 (28%) 4 (22%)ge �20 y, cryopreserved 37 15 (41%) 10 (27%) 12 (32%) 0 (0%)

OTE. Albumin levels from immune-deficient Alb-uPA SCID mice transplanted with 1,000,000 fresh or cryopreserved primary human hepatocytes

upplementary Figure 2. (A) Real-time analysis showing expression of the genes encoding bilirubin-UDP glucuronosyl transferase, ornithineranscarbamylase (OTC), and bile salt export pump at levels similar to that attained from fresh primary human hepatocytes. Diff, hES cells differentiatedoward a hepatocyte phenotype, but not enriched by sorting for ASGPR; Diff sorted, hES cells differentiated toward a hepatocyte phenotype andnriched by sorting for ASGPR; HH, control fresh primary human hepatocytes. (B) Photomicrograph of H&E-stained tissue section from a peritoneal

mplant obtained from a Nagase rat transplanted with hES-derived hepatocytes enriched based on surface ASGPR expression showing character-

ecovered from liver.

Page 14: BASIC—LIVER, PANCREAS, AND BILIARY TRACT · 2019. 7. 10. · BILIARY TRACT GASTROENTEROLOGY 2009;136:990–999. differentiation strategy that produces a relatively homo-geneous

SoAhe[ccdoitpi

March 2009 HUMAN ES CELL–DERIVED HEPATOCYTES 999.e4

upplementary Figure 3. Differentiation of human ES cells using a program that does not contain an EB formation step. (A) Real-time PCR analysisf Oct 3/4, Nanog, Sox17, Sox7, albumin, and AFP expression indicates that there is more extensive and earlier loss of Oct 3/4, Nanog, Sox7, andFP when the EB step is removed. Analysis of cyclophilin expression was used as the positive control. Undiff, undifferentiated human ES cells; Dir,epatocyte-directed differentiation without the EB formation step; EBs, hepatocyte-directed differentiation containing the EB formation step; HH,xpression levels for control primary human hepatocytes. (B) Real-time PCR analysis for expression of lineage-specific hepatic markers (albumin

ALB], ASGPR 1, glucose-6-phosphatase [G6Pase], and tyrosine aminotransferase [TAT]), an endoderm marker (AFP), and markers for undifferentiatedells (Pou5f1 and Nanog) using a hepatocyte-directed differentiation program that does not involve formation of EBs. Lanes: (1) undifferentiated hESells (Undiff); (2) after treatment with fibroblast growth factor-2 and activin-A, HGF and dimethyl sulfoxide, and then dexamethasone (Dir); and (3) afterifferentiation and the sorting for ASGPR expression (Dir-Sorted). Primary human hepatocytes (HH) served as the positive control. (C) Determinationf albumin and urea secretion in vitro by primary human hepatocytes (HH ) and hES cells after differentiation when the EB formation step is not

ncluded. Protein secretion was assessed in control medium (CM) only; medium from 5 � 105 undifferentiated hES cells (Undiff), hES cells afterreatment with fibroblast growth factor-2 and activin-A, HGF, and dimethyl sulfoxide, and then dexamethasone (Dir); and medium from cultured freshrimary human hepatocytes (HH). Twenty-four hours after culture, the amount of human albumin in the media was measured by enzyme-linked

mmunosorbent assay, and urea was assessed using a urease colorimetric method.