You are on page 1of 9

TISSUE ENGINEERING: Part A

Volume 24, Numbers 7 and 8, 2018


ª Mary Ann Liebert, Inc.
DOI: 10.1089/ten.tea.2017.0161

ORIGINAL ARTICLE

Three-Dimensional Bioprinting of Hepatic Structures


with Directly Converted Hepatocyte-Like Cells
Kyojin Kang, MS,1–3 Yohan Kim, MS,1–3 Hyeryeon Jeon, MS,2,3 Seung Bum Lee, PhD,4 Ji Sook Kim, PhD,5
Su A Park, PhD,6 Wan Doo Kim, PhD,6 Heung Mo Yang, PhD,7 Sung Joo Kim, MD, PhD,7
Jaemin Jeong, PhD,2,3 and Dongho Choi, MD, PhD1–3

Three-dimensional (3D) bioprinting technology is a promising new technology in the field of bioartificial organ
generation with regard to overcoming the limitations of organ supply. The cell source for bioprinting is very
important. Here, we generated 3D hepatic scaffold with mouse-induced hepatocyte-like cells (miHeps), and
investigated whether their function was improved after transplantation in vivo. To generate miHeps, mouse
embryonic fibroblasts (MEFs) were transformed with pMX retroviruses individually expressing hepatic tran-
scription factors Hnf4a and Foxa3. After 8–10 days, MEFs formed rapidly growing hepatocyte-like colonies.
For 3D bioprinting, miHeps were mixed with a 3% alginate hydrogel and extruded by nozzle pressure. After
7 days, they were transplanted into the omentum of Jo2-treated NOD Scid gamma (NSG) mice as a liver
damage model. Real-time polymerase chain reaction and immunofluorescence analyses were conducted to
evaluate hepatic function. The 3D bioprinted hepatic scaffold (25 · 25 mm) expressed Albumin, and ASGR1 and
HNF4a expression gradually increased for 28 days in vitro. When transplanted in vivo, the cells in the hepatic
scaffold grew more and exhibited higher Albumin expression than in vitro scaffold. Therefore, combining 3D
bioprinting with direct conversion technology appears to be an effective option for liver therapy.

Keywords: miHep cells, 3D bioprinting, cell transplantation, direct conversion

Introduction Direct conversion technique has several advantages more


than pluripotent stem cells, such as lower tumori-
genic risk,13 faster conversion rate, and the ability to re-
C ell therapy using stem cells is being explored as
a novel approach that could potentially replace organ
transplantation. Cell therapies for liver diseases can be
pair injured tissues.12
Hepatocytes derived from induced pluripotent stem cells
categorized based on cell source (cell lines, primary hepa- (iPSCs) or embryonic stem cells (ESCs) cannot be main-
tocytes, stem cells, progenitor cells, or nonparenchymal cell tained because they do not multiply.14 Several researchers
types).1,2 Pluripotent stem cells have been used to gener- have suggested methods of long-term culture of hepatocytes,
ate hepatocyte-like cells.3–6 However, the use of pluripotent such as combining them with mesenchymal stem cells
stem cells has led to controversy regarding the danger (MSCs),15 use of collagen sandwiches,16 and the addition of
of tumor formation,7,8 failure of long-term differentiation,9 special media components.17 Although ‘‘sandwich culture,’’
and low differentiation efficiency.10 Therefore, developing an in vitro approach of mimicking three-dimensional (3D)
functional cells that are as effective as primary cells is culture, has been proposed as a solution to long-term culture
crucial. In this article, we introduce directly converted and maintenance of hepatocyte function, it has some limi-
hepatocyte-like cells. Direct conversion technique is con- tations because it cannot be used to construct an actual 3D
sidered to be one of the most effective methods of altering model18; 3D structures are particularly important for gen-
somatic cells without generating pluripotent stem cells.11,12 erating functional hepatocytes in an in vivo-like state.
1
Department of Translational Medicine, Graduate School of Biomedical Science and Engineering, Seongdong-gu, Korea.
2
Department of Surgery, Hanyang University College of Medicine, Seoul, Korea.
3
HY Indang Center of Regenerative Medicine and Stem Cell Research, Hanyang University, Seoul, Korea.
4
Laboratory of Radiation Exposure and Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological
and Medical Science (KIRAMS), Seoul, Korea.
5
Department of Pathology, Hanyang University College of Medicine, Seoul, Korea.
6
Department of Nature-Inspired Nanoconvergence Systems, Korea Institute of Machinery and Materials, Daejeon, Korea.
7
Department of Surgery, Sungkunkwan University College of Medicine, Seoul, Korea.

576
CONSTRUCTION OF MULTILAYERED 3D-PRINTED HEPATIC STRUCTURES 577

3D bioprinting is one of the most promising 3D culture and real-time polymerase chain reaction (PCR) was per-
methods using stem cells, which overcomes many limita- formed using 10 mL of quantitative polymerase chain reaction
tions of earlier cell transplantation methods.19 Nevertheless, (qPCR) PreMix (Dyne bio, Korea), 1 mL of cDNA, and oli-
bioprinting faces some of the same challenges that confront all gonucleotide primers on a CFX Connect Real-Time PCR
tissue engineering and regenerative medicine technologies.20 Detection system (Bio-rad, CA). Reactions were analyzed in
We describe the use of mouse-induced hepatocytes (miHeps) triplicate for each gene; the primers used for mouse Albumin,
in conjunction with 3D bioprinting as a technique that could be ASGPR1, AFP, HNF4a, and 18s rRNA are listed in Supple-
used in cell therapies and drug discovery to eventually facili- mentary Table S1 (Supplementary Data are available online
tate transplantation of bioartificial organs.21,22 at www.liebertpub.com/tea). PCR cycles consisted of 40 cycles
of 95C for 20 s and 60C for 40 s. Melting curves and melting
Materials and Methods peak data were obtained to characterize the PCR products.
Generation of miHeps
Hematoxylin and eosin staining
The production of miHeps has been described previous- and immunohistochemistry
ly.23 In brief, to produce miHeps, 5 · 104 mouse embryonic
fibroblasts (MEFs) were transduced with pMX retroviruses Four-micrometer sections were deparaffinized by washing
expressing hepatic transcription factors Hnf4a and Foxa3 three times with xylene for 5 min each time. The printed mi-
(a gift from Prof. Dong Wook Han of Konkuk University). Heps were rehydrated in a series of 100%, 90%, and 70%
After 48 hrs, the cells were further cultured in miHep medium ethanol for 5 min each, and finally rinsed in tap water. The
on a Type I collagen-coated dish to induce lineage transition sections were stained with hematoxylin (Sigma) for 5 min and
toward miHeps. The cells were cultured in a 100 mm di- counterstained with eosin for 1 min after washing with running
ameter dish (Corning) with DMEM/F-12 (11965; Gibco, water for 5 min. Finally, they were washed in running water
CA) supplemented with 10% fetal bovine serum (Gibco), and dehydrated in 95% ethanol for 5 min. The 3D-printed
10 mM nicotinamide (Sigma-Aldrich, MO), 0.1 mM dexa- mCherry-miHep alginate scaffold was fixed in 4% buffered
methasone (Sigma-Aldrich), 1% insulin-transferrin-selenium formalin and embedded in paraffin. Four-micrometer sections
(ITS) (Gibco), 1% penicillin/streptomycin (Gibco), 20 ng/mL were cut, mounted on silane-coated glass slides, depar-
hepatocyte growth factor (HGF) (Peprotech, NJ), and 20 ng/ affinized, and rehydrated in graded alcohols. After washing,
mL epidermal growth factor (EGF) (Peprotech) at 37C in a endogenous peroxidase was blocked with 3% H2O2 in meth-
CO2 incubator. The medium was changed daily. anol for 15 min at room temperature. Antigen was retrieved by
autoclaving the slides in pH 6.0 citric acid buffer.
Production of mCherry lentivirus For immunohistochemistry, the slides were incubated in a
moist chamber with rabbit anti-mCherry antibody (1:200;
Viruses were produced as previously described.22 mCherry Abcam, Cambridge, United Kingdom), goat antimouse se-
was packaged by cotransfection with psPAX2 lentiviral rum Albumin antibody (1:200; Abcam), mouse antihuman
packaging plasmid and pCMV-VSV-G plasmid in human cytokeratin 18 (1:200; DAKO, CA), cytokeratin 19 (1:100;
embryonic kidney 293T cells. Culture supernatants were Santa Cruz Biotechnology, TX), or Ki67 (1:200; Leica, NCL,
harvested after 48 and 72 hrs and stored at -80C. Lentiviral United Kingdom) overnight at 4C. The next day, the cells
transduction of mCherry was carried out in culture medium were washed twice with staining solution in PBS supple-
supplemented with 8 mg/mL polybrene (Sigma-Aldrich). mented with 1% fetal bovine serum for 5 min, then incubated
with secondary antibody, goat antimouse IgG Alexa Fluor
3D bioprinting 488 (1:250; Molecular probes, OR), goat antirabbit IgG
3D-printed strands of packaged cells were produced with TRITC (1:250; Molecular Probes), or donkey antigoat IgG
a 3D bioprinting system made by the Korea Institute of Ma- Alexa Fluor 488 (1:250; Molecular Probes) for 1 hr 30 min at
chinery and Materials. The system can control the pressure room temperature. Nuclei were counterstained with Hoechst
and velocity needed to fabricate the 3D hydrogel, which was 33342 (1:10000; Molecular Probes) in PBS. After incubation
plotted through the pneumatic dispenser. The material dis- with secondary antibodies, the slides were washed in running
pensed consisted of 3% alginate to form the hydrogel scaffold tap water, dehydrated, cleared, and mounted.
and a calcium chloride (CaCl2) solution to solidify the hy-
drogel. Before 3D printing, the miHep cells were suspended Experimental animal model
in 3% alginate solution. The 3D-printed scaffold measured 25 NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice were purchased
by 25 mm. For cell printing, constant air pressure was applied from the Jackson Laboratory and housed under specific
to the dispenser, and the cell-encapsulating alginate was pathogen-free conditions in accordance with the Principles of
squeezed out onto a 100 mm diameter culture dish. After Laboratory Animal Care and the Guide for the Use of La-
printing, the 3D alginate hydrogel was soaked in 1% CaCl2 boratory Animals of Samsung Biomedical Research Institute
solution to strengthen crosslinking, washed with phosphate- (20170116003). Twenty-four hours after intraperitoneal injec-
buffered saline (PBS), and placed in miHep culture medium. tion of Jo2 antibody/PBS (BD Pharmingen, CA) at 0.1–0.2 mg/
kg body weight, a 3D alginate hydrogel containing miHeps
RNA isolation and quantitative real-time
(*0.5 · 0.5 cm) was implanted into the peritoneal cavity of
polymerase chain reaction
mice. After implantation, the mice received 100 mg/L cipro-
Total RNAs were isolated using Trizol Reagent (Gibco). floxacin (CJ Pharma, Korea) in their drinking water to prevent
Two-microgram RNA samples were reverse transcribed with infection. At termination, mouse tissue and the 3D alginate
a Transcriptor First Strand cDNA Synthesis Kit (Roche, PA), hydrogel scaffold were immediately fixed in 10% formalin.
578 KANG ET AL.

Statistical analysis reprogramming pMX vectors encoding two transcription


Quantitative data are presented as mean – standard devi- factors, Foxa3 and HNF4a (Fig. 1A). After 10–12 days,
ation with inferential statistics ( p-values). Statistical sig- hepatocyte-like cells appeared. To evaluate the hepatic
nificance was evaluated using two-tailed t-tests with characteristics of these miHeps, hepatic marker gene ex-
significance set at *p < 0.05, **p < 0.01, and ***p < 0.001. pression levels were measured by quantitative real-time
polymerase chain reaction (qRT-PCR). Expression levels of
Albumin, AFP, ASGR1, and HNF4a in miHeps were signifi-
Results cantly higher than in MEFs (Fig. 1B). Moreover, miHeps
were positive for hepatic proteins, Albumin, E-cadherin,
Induction of hepatic lineage cells from MEFs by direct
ASGR1, HNF4a, CYP1a2, and CK18 on immunofluores-
conversion methods
cence staining as compared with MEFs (Fig. 1C). Also, Al-
To generate the miHeps, MEFs were prepared from an bumin secretion and urea synthesis were increased on miHeps
E16.5 embryo. The fibroblasts were virally transduced with compared with MEFs (Supplementary Fig. S1). These results

FIG. 1. Generation and


characterization of miHeps.
(A) Scheme of miHep pro-
duction. Epithelial
hepatocyte-like cells appear
10–12 days after infection
with retroviruses carrying
two hepatic transcription
factors. (B) Hepatocyte
marker gene expression
evaluated by qRT-PCR. Ex-
pression of Albumin, AFP,
ASGR1, and HNF4a is up-
regulated during the genera-
tion of miHeps. *p < 0.05,
**p < 0.01, and ***p < 0.001.
MEFs, mouse embryonic fi-
broblasts; miHeps, mouse-
induced hepatocytes; mPHs,
mouseprimary hepatocytes.
(C) Hepatic marker protein
expression evaluated by im-
munofluorescence assays be-
tween MEFs and miHeps.
The miHeps stain for
Albumin, CK18, HNF4a,
E-cadherin, CYP1a2, and
ASGR1. Scale bar, 25 mm.
qRT-PCR, quantitative real-
time polymerase chain
reaction.
CONSTRUCTION OF MULTILAYERED 3D-PRINTED HEPATIC STRUCTURES 579

suggest that direct conversion techniques are effective for minished on day 14 after printing (Fig. 2C). AFP, which is
generating hepatocyte-like cells from fibroblasts. not expressed in normal hepatocytes, gradually decreased.
Overall, these hepatic function data suggest that directly
3D bioprinting of miHeps with alginate, converted miHeps mature during incubation upon 3D-
and improvement of hepatic function printed scaffold.

Before 3D printing, we labeled miHeps with mCherry


Proliferation of functional miHeps
lentivirus to trace miHeps proliferation and migration in
in 3D-printed scaffold
3D scaffold (Fig. 2A). To produce 3D hepatic scaffold,
mCherry-miHeps and alginate mixtures were ejected from We investigated whether 3D-printed hepatic scaffold
a 3D printer (Fig. 2B). Under fluorescence microscopy, we facilitates proliferation of miHeps. As we expected, the mass
confirmed mCherry-miHeps distribution on day 1 (Fig. 2B). of mCherry expressing miHeps increased gradually day by
They were cultured in vitro with miHeps medium for 28 days day (Fig. 3A–E), and proliferation of the cells was confirmed
to form miHeps colonies. To evaluate gene expression in the by fluorescence microscopy (Fig. 3F–J). miHeps appeared to
scaffold, we performed qRT-PCR at several time points. be highly compact by days 14 and 28 after printing, as revealed
Expression of Albumin gradually increased, for up to 28 days, by hematoxylin and eosin (H&E) staining (Fig. 3K–O). Sur-
but ASGR1 and HNF4a increased only on day 28 and di- prisingly, the cells strongly expressed Albumin and CK18,

FIG. 2. Generation of 3D hepatic structures using a 3D bioprinter. (A) mCherry lentivirus infection into miHeps for
tracing proliferation and migration. Scale bar, 100 mm. (B) The 3D bioprinter and the shape of the printed 3D hepatic
structures. Five layers of alginate scaffold each measuring 25 · 25 mm were constructed. And confirming distribution of
mCherry-expressing miHeps under fluorescence microscopy on day 1. Scale bar, 500 mm. (C) Hepatocyte marker gene
expression in the 3D structures evaluated by qRT-PCR after 28 days. Expression of Albumin, ASGR1, and HNF4a increased
over time, whereas AFP expression gradually decreased. **p < 0.01, ***p < 0.001. 3D, three-dimensional.
580 KANG ET AL.

FIG. 3. Proliferation of functional miHeps in 3D hepatic scaffolds in vitro. (A–E) Bright field images of miHeps grown in
3D hepatic structures. Scale bar, 100 mm. (F–J) Fluorescence images of miHeps labeled with mCherry. Scale bar, 100 mm.
(K–O) Hematoxylin and eosin staining. Scale bar, 50 mm. (P–Y) Immunohistochemical analysis of the expression of
Albumin and CK18 using confocal microscopy. Scale bar, 50 mm.

demonstrating that rapidly proliferating miHep cells retained We harvested the scaffold to check the status of the miHeps
typical hepatic characteristics in the scaffold (Fig. 3P–Y). In and confirmed hepatic marker gene expression. As shown in
addition, no cell death was apparent in these structures (Sup- Figure 3, in vivo, proliferating miHeps in the scaffold could
plementary Fig. S2). These data demonstrate that the 3D- be detected after 28 days (Fig. 4C). Surprisingly, the mi-
printed framework facilitates miHep cell proliferation without Heps grew more rapidly and expressed higher levels of
loss of hepatic function. Albumin in vivo than in vitro (Fig. 4C, D). Also, we per-
formed immunocytochemistry to evaluate whether miHeps
differentiated into cholangiocytes in vivo. Interestingly,
Transplantation of a 3D-printed scaffold
some miHeps were stained with CK19 antibody, and they
with miHeps into mice
were colocalized with mCherry expression around the bile
To evaluate miHeps proliferation and hepatic function duct on day 28 (Supplementary Fig. S3). These proliferating
in vivo, 3D hepatic scaffold was transplanted into omentum cells were detected (Supplementary Fig. S4). These results
of Jo2-treated NOD Scid gamma (NSG) mice. 3D-printed confirm that miHeps proliferate and differentiate faster when
hepatic scaffold was stabilized for 7 days in vitro and transplanted on 3D scaffold than in vitro.
transplanted (Fig. 4A). Twenty-eight days after transplan- We further examined the hepatic function of 3D hepatic
tation, miHep scaffold was found in the omentum, and some scaffold around the liver. We were able to detect mCherry
pieces were observed to be also surrounding the liver (Fig. 4B). fluorescence around mouse liver tissue on days 14 and 28
FIG. 4. 3D hepatic scaffolds transplanted into mouse omenta. (A) The experimental schedule. Seven days after 3D
printing, 3D hepatic structures were transplanted in jo2-treated NSG mouse omenta, and harvested on days 14 and 28. (B)
The transplanted 3D hepatic scaffold is well-engrafted around liver tissue at 28 days (red arrow). (C) Proliferation of
miHeps in 3D hepatic scaffolds seen by fluorescence microscopy. The miHeps formed larger colonies in vivo than in vitro
by days 14 and 28. Scale bar, 100 mm. (D) Hepatocyte marker genes (Albumin and AFP) evaluated by qRT-PCR in vitro and
in vivo cultured miHeps. **p < 0.01, ***p < 0.001. NSG, NOD Scid gamma.

FIG. 5. Histological analysis of 3D hepatic scaffolds with mouse liver. (A–D) Fluorescence images of miHeps labeled
with mCherry on days 14 and 28. Scale bar, 250 mm. (E–H) Immunohistochemical analysis of levels of Albumin and CK18
by confocal microscopy on days 14 and 28. Scale bar, 250 mm. White asterisk is mouse liver tissue. (I–L) Hoechst (blue)
labels all nuclei. Red (mCherry) and green (Albumin and CK18) fluorescence colocalize (M–T, enlarged in Q–T).

581
582 KANG ET AL.

(Fig. 5A–D). Albumin (Fig. 5E, G) and CK18 (Fig. 5F, H) Acknowledgment
were also expressed, and expression of both proteins colo- This work was carried out with the support of the ‘‘Co-
calized with mCherry expression (Fig. 5 M–P, enlarged operative Research Program for Agriculture Science and
panel in Q–T). These observations indicate that bioprinted Technology Development (Project No. PJ01100202),’’
3D hepatic scaffold can engraft around liver tissue, and may Rural Development Administration, Republic of Korea.
offer an alternative to liver transplantation.

Disclosure Statement
Discussion
No competing financial interests exist.
Liver transplantation is a vital procedure for patients with
end-stage chronic liver disease.24,25 However, the number of
potential liver transplantation recipients are increasing faster References
than the number of potential donors.21 To overcome this 1. Yu, Y., Fisher, J.E., Lillegard, J.B., Rodysill, B., Amiot, B.,
donor shortage, many researchers have attempted cell ther- and Nyberg, S.L. Cell therapies for liver diseases. Liver
apies using stem cells,26 and are now focusing on generating Transplant 18, 9, 2012.
3D scaffold made up of stem cells within a matrix.27 In this 2. Lee, S.Y., Kim, H.J., and Choi, D. Cell sources, liver
study, we used a direct conversion technique to generate support systems and liver tissue engineering: alternatives to
hepatocyte-like cells as an alternative cell source for cell liver transplantation. Int J Stem Cells 8, 36, 2015.
therapy, together with 3D printing to form 3D scaffold of 3. Nagamoto, Y., Takayama, K., Ohashi, K., Okamoto, R.,
liver organoids. Although hepatocyte-like cells can be de- Sakurai, F., Tachibana, M., Kawabata, K., and Mizuguchi,
rived from human pluripotent stem cells,3–6 there are dis- H. Transplantation of a human iPSC-derived hepatocyte
advantages to this process. The major issues are that their sheet increases survival in mice with acute liver failure. J
differentiation into functional mature hepatocytes is chal- Hepatol 64, 1068, 2016.
lenging to achieve, and sometimes these cells can form 4. Schwartz, R.E., Fleming, H.E., Khetani, S.R., and Bhatia,
tumors in vivo.27 In contrast, directly converted hepatocyte- S.N. Pluripotent stem cell-derived hepatocyte-like cells.
like cells do not need to follow liver development mecha- Biotechnol Adv 32, 504, 2014.
nism during differentiation, and they do not form teratomas 5. Sullivan, G.J., Hay, D.C., Park, I.H., Fletcher, J., Hannoun,
after transplantation.27 Therefore, we transduced HNF4a Z., Payne, C.M., Dalgetty, D., Black, J.R., Ross, J.A.,
and Foxa3 into MEFs, and by 3 or 4 days after infection, Samuel, K., Wang, G., Daley, G.Q., Lee, J.H., Church,
G.M., Forbes, S.J., Iredale, J.P., and Wilmut, I. Generation
the fibroblasts had transformed into polygonal epithelial
of functional human hepatic endoderm from human in-
cells referred to as miHeps.28 This process activates the
duced pluripotent stem cells. Hepatology 51, 329, 2010.
mesenchymal–epithelial transition and hepatocyte markers.29 6. Yi, F., Liu, G.H., and Izpisua Belmonte, J.C. Human in-
Lim et al. have reported that treatment with A83-01, which duced pluripotent stem cells derived hepatocytes: rising
is a transforming growth factor beta receptor inhibitor, promise for disease modeling, drug development and cell
boosts miHeps generation with just one gene. miHeps of therapy. Protein Cell 3, 246, 2012.
generation was first reported with small molecules.29 For 7. Lee, A.S., Tang, C., Rao, M.S., Weissman, I.L., and Wu,
clinical trials of directly converted hepatocyte-like cells to J.C. Tumorigenicity as a clinical hurdle for pluripotent stem
be approved, they must be generated without exogenous cell therapies. Nat Med 19, 998, 2013.
genes or induced pluripotent stem cells.30 Therefore, we 8. Miura, K., Okada, Y., Aoi, T., Okada, A., Takahashi, K.,
will attempt miHeps generation without exogenous genes Okita, K., Nakagawa, M., Koyanagi, M., Tanabe, K., Oh-
such as mRNA or other small molecules in the future. nuki, M., Ogawa, D., Ikeda, E., Okano, H., and Yamanaka,
The geometry of the extracellular matrix that was formed S. Variation in the safety of induced pluripotent stem cell
using collagen sandwiches can maintain hepatocytes long lines. Nat Biotechnol 27, 743, 2009.
term and to expand their function.18 Therefore, we hy- 9. Hannoun, Z., Steichen, C., Dianat, N., Weber, A., and
pothesized that miHeps would also maintain their hepatic Dubart-Kupperschmitt, A. The potential of induced plurip-
function and proliferation on 3D scaffold or 3D structure. otent stem cell derived hepatocytes. J Hepatol 65, 182, 2016.
Although collagen is theoretically the optimal extracellular 10. Song, Z.H., Cai, J., Liu, Y.X., Zhao, D.X., Yong, J., Duo,
matrix material, we used alginate hydrogels as bioink. This S.G., Song, X.J., Guo, Y.S., Zhao, Y., Qin, H., Yin, X.L.,
is because collagen requires certain temperatures and pH Wu, C., Che, J., Lu, S.C., Ding, M.X., and Deng, H.K.
levels to harden,31 and 3D bioprinters are incapable of Efficient generation of hepatocyte-like cells from human
induced pluripotent stem cells. Cell Res 19, 1233, 2009.
handling collagen as an extracellular matrix material.
11. Du, Y., Wang, J., Jia, J., Song, N., Xiang, C., Xu, J., Hou,
Therefore, further development of bioinks and techniques
Z., Su, X., Liu, B., Jiang, T., Zhao, D., Sun, Y., Shu, J.,
for seeding cells under the desired conditions is needed. Guo, Q., Yin, M., Sun, D., Lu, S., Shi, Y., and Deng, H.
In summary, we have shown that miHeps can be used as a Human hepatocytes with drug metabolic function induced
liver cell source in 3D hepatic scaffold and that they in- from fibroblasts by lineage reprogramming. Cell Stem Cell
crease in number and function when seeded upon scaffold 14, 394, 2014.
and incubated. Thus, the transplantation of 3D-printed mi- 12. Sekiya, S., and Suzuki, A. Direct conversion of mouse fi-
Heps suggest that 3D bioprinting techniques will improve broblasts to hepatocyte-like cells by defined factors. Nature
cell therapy techniques and survival rates. Although chal- 475, 390, 2011.
lenges related to 3D bioprinters, bioink, and cell sources 13. Ben-David, U., and Benvenisty, N. The tumorigenicity of
remain, bioprinting is poised to become one of the most human embryonic and induced pluripotent stem cells. Nat
promising methods of creating bioartificial organs. Rev Cancer 11, 268, 2011.
CONSTRUCTION OF MULTILAYERED 3D-PRINTED HEPATIC STRUCTURES 583

14. Jorns, C., Gramignoli, R., Saliem, M., Zemack, H., Mork, 27. Song, W., Lu, Y.C., Frankel, A.S., An, D., Schwartz, R.E.,
L.M., Isaksson, B., Nowak, G., Ericzon, B.G., Strom, S., and Ma, M. Engraftment of human induced pluripotent
and Ellis, E. Strategies for short-term storage of hepato- stem cell-derived hepatocytes in immunocompetent mice
cytes for repeated clinical infusions. Cell Transplant 23, via 3D co-aggregation and encapsulation. Sci Rep 5, 16884,
1009, 2014. 2015.
15. Rebelo, S.P., Costa, R., Silva, M.M., Marcelino, P., Brito, 28. Huang, P., He, Z., Ji, S., Sun, H., Xiang, D., Liu, C., Hu, Y.,
C., and Alves, P.M. Three-dimensional co-culture of hu- Wang, X., and Hui, L. Induction of functional hepatocyte-
man hepatocytes and mesenchymal stem cells: improved like cells from mouse fibroblasts by defined factors. Nature
functionality in long-term bioreactor cultures. J Tissue Eng 475, 386, 2011.
Regen Med 11, 2034–2045, 2017. 29. Lim, K.T., Lee, S.C., Gao, Y., Kim, K.P., Song, G., An,
16. Sattler, C.A., Michalopoulos, G., Sattler, G.L., and Pitot, S.Y., Adachi, K., Jang, Y.J., Kim, J., Oh, K.J., Kwak, T.H.,
H.C. Ultrastructure of adult rat hepatocytes cultured on Hwang, S.I., You, J.S., Ko, K., Koo, S.H., Sharma, A.D.,
floating collagen membranes. Cancer Res 38, 1539, 1978. Kim, J.H., Hui, L., Cantz, T., Scholer, H.R., and Han, D.W.
17. Lee, C.H., and Edwards, A.M. Stimulation of DNA syn- Small molecules facilitate single factor-mediated hepatic
thesis and c-fos mRNA expression in primary rat hepato- reprogramming. Cell Rep 15, 814, 2016.
cytes by estrogens. Carcinogenesis 22, 1473, 2001. 30. Lin, T., and Wu, S. Reprogramming with small molecules
18. Choi, H.J., and Choi, D. Successful mouse hepatocyte instead of exogenous transcription factors. Stem Cells Int
culture with sandwich collagen gel formation. J Korean 2015, 794632, 2015.
Surg Soc 84, 202, 2013. 31. Wang, Y.J., Liu, H.L., Guo, H.T., Wen, H.W., and Liu, J.
19. Stanton, M.M., Samitier, J., and Sanchez, S. Bioprinting of Primary hepatocyte culture in collagen gel mixture and
3D hydrogels. Lab Chip 15, 3111, 2015. collagen sandwich. World J Gastroenterol 10, 699, 2004.
20. Murphy, S.V., and Atala, A. 3D bioprinting of tissues and
organs. Nat Biotechnol 32, 773, 2014.
21. Dutkowski, P., Oberkofler, C.E., Bechir, M., Mullhaupt, B.,
Address correspondence to:
Geier, A., Raptis, D.A., and Clavien, P.A. The model for
Dongho Choi, MD, PhD
end-stage liver disease allocation system for liver trans-
plantation saves lives, but increases morbidity and cost: a Department of Surgery
prospective outcome analysis. Liver Transpl 17, 674, 2011. Hanyang University College of Medicine
22. Jeon, H., Kang, K., Park, S.A., Kim, W.D., Paik, S.S., Lee, Seoul 04763
S.H., Jeong, J., and Choi, D. Generation of multilayered 3D Korea
structures of HepG2 cells using a bio-printing technique.
Gut Liver 11, 121–128, 2016. E-mail: crane87@hanyang.ac.kr
23. Huang, P., Zhang, L., Gao, Y., He, Z., Yao, D., Wu, Z.,
Cen, J., Chen, X., Liu, C., Hu, Y., Lai, D., Hu, Z., Chen, L., Jaemin Jeong, PhD
Zhang, Y., Cheng, X., Ma, X., Pan, G., Wang, X., and Hui, Department of Surgery
L. Direct reprogramming of human fibroblasts to functional Hanyang University College of Medicine
and expandable hepatocytes. Cell Stem Cell 14, 370, 2014. Seoul 04763
24. Lee, H.W., and Suh, K.S. Liver transplantation for ad- Korea
vanced hepatocellular carcinoma. Clin Mol Hepatol 22,
309, 2016. E-mail: jmj1103@gmail.com
25. Florman, S., and Miller, C.M. Live donor liver transplan-
tation. Liver Transpl 12, 499, 2006. Received: April 10, 2017
26. Kwon, Y.J., Lee, K.G., and Choi, D. Clinical implications of Accepted: July 10, 2017
advances in liver regeneration. Clin Mol Hepatol 21, 7, 2015. Online Publication Date: January 9, 2018
Reproduced with permission of copyright owner. Further reproduction
prohibited without permission.

You might also like