Human hepatocytes loaded in 3D bioprinting generate mini-liver
 
Cheng Zhong, Hai-Yang Xie, Lin Zhou, Xiao Xu and Shu-Sen Zheng
Hangzhou, China
 
 
Author Affiliations: Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China (Zhong C, Xie HY, Zhou L, Xu X and Zheng SS)
Corresponding Author: Shu-Sen Zheng, MD, PhD, FACS, Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China (Tel: +86-571-87236352; Email: shusenzheng@zju.edu.cn)
 
© 2016, Hepatobiliary Pancreat Dis Int. All rights reserved.
doi: 10.1016/S1499-3872(16)60119-4
Published online August 4, 2016.
 
 
Contributors: ZSS proposed the study. ZC, XHY, ZL and XX performed the research and wrote the first draft. All authors contributed to the design and interpretation of the study and to further drafts. ZSS is the guarantor.
Funding: This study was supported by grants from the National Natural Science Foundation of Major Research and Development Plan of China (91542205) and 151 Talents Project of Zhejiang Province (12-1-058).
Ethical approval: This study was approved by the Ethics Committee of the First Affiliated Hospital of Zhejiang University School of Medicine.
Competing interest: No benefits in any form have been received or will be received from a commercial party related directly or indirectly to the subject of this article.
 
 
BACKGROUND: Because of an increasing discrepancy between the number of potential liver graft recipients and the number of organs available, scientists are trying to create artificial liver to mimic normal liver function and therefore, to support the patient’s liver when in dysfunction. 3D printing technique meets this purpose. The present study was to test the feasibility of 3D hydrogel scaffolds for liver engineering.
 
METHODS: We fabricated 3D hydrogel scaffolds with a bioprinter. The biocompatibility of 3D hydrogel scaffolds was tested. Sixty nude mice were randomly divided into four groups, with 15 mice in each group: control, hydrogel, hydrogel with L02 (cell line HL-7702), and hydrogel with hepatocyte growth factor (HGF). Cells were cultured and deposited in scaffolds which were subsequently engrafted into livers after partial hepatectomy and radiation-induced liver damage (RILD). The engrafted tissues were examined after two weeks. The levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), albumin, total bilirubin, CYP1A2, CYP2C9, glutathione S-transferase (a-GST), and UDP-glucuronosyl transferase (UGT-2) were compared among the groups. Hematoxylin-eosin (HE) staining and immunohistochemistry of cKit and cytokeratin 18 (CK18) of engrafted tissues were evaluated. The survival time of the mice was also compared among the four groups.
 
RESULTS: 3D hydrogel scaffolds did not impact the viability of cells. The levels of ALT, AST, albumin, total bilirubin, CYP1A2, CYP2C9, a-GST and UGT-2 were significantly improved in mice engrafted with 3D scaffold loaded with L02 compared with those in control and scaffold only (P<0.05). HE staining showed clear liver tissue and immunohistochemistry of cKit and CK18 were positive in the engrafted tissue. Mice treated with 3D scaffold+L02 cells had longer survival time compared with those in control and scaffold only (P<0.05).
 
CONCLUSION: 3D scaffold has the potential of recreating liver tissue and partial liver functions and can be used in the reconstruction of liver tissues.
 
(Hepatobiliary Pancreat Dis Int 2016;15:512-518)
 
KEY WORDS: 3D printing; hepatocyte; liver; tissue engineering
 
 
Introduction
Liver transplantation is the gold standard of care for patients with end-stage liver disease and those with tumors of liver origin in the setting of liver dysfunction.[1] Today, one of the greatest hurdles that liver transplantation faces is the growing discrepancy between the demand and availability of donor livers, although this gap has been slightly reduced since the model for end-stage liver disease allocation system was adopted.[2] Different strategies have evolved to expand the donor pool.[3, 4] Despite efforts to expand this resource, solutions to expand the donor pool will require the use of more borderline organs. Further research is needed on optimal organ sources, including the use of organ regeneration in the laboratory.[5, 6]
 
In recent years, a significant amount of attention has been focused on the concept of tissue engineering. The principle of tissue engineering assumes that a variety of cells can be coaxed into synthesizing new tissue when they are seeded onto an appropriate scaffold in an appropriate growth and differentiation environment.[7] Bioprinting is defined as the use of printing technology to deposit living cells, an extracellular matrix, biochemical factors, drugs, and biomaterials on a receiving solid or gel substrate or liquid reservoir.[8] Progress has been made in cell biology, material sciences, and tissue engineering that has enabled researchers to develop cutting-edge technology that has led to the creation of non-modular tissue constructs such as skin,[9] bladder,[10] and vessels.[11] Therefore, the use of 3D printing with other classes of nano-scale functional building blocks, including semiconductor, magnetic, plasmonic, and ferroelectric nanoparticles could expand the opportunities for engineering of bionic tissues and organs.[12] Techniques such as seeding cells into non-adhesive molds or self-folding scaffolds have been used to fabricate 3D tissue constructs with complex geometries.[8, 13]
 
The grid pattern design was adopted as a simple model of liver lobules. As a kind of transplantation, the function of printed tissues was more important than the shape. Difference from previous reports,[14] our study adopted the new design of printed scaffold with pores. Compared with the previous in vivo researches which did not emphasize the morphology and functions,[15-17] our in vivo study evaluated the morphology and function changes in 3D scaffold loaded with hepatocyte in detail. This model may be highly applicable for future studies on the regeneration of hepatocyte tissues.
 
 
Methods
Cell culture
L02 cells (HL-7702, a cell line of human hepatocytes) were provided by the Cell Bank of Shanghai, Chinese Academy Science. As a type of potential seed cell in liver tissue engineering,[18, 19] L02 cells were maintained in DMEM-LG medium (Hyclone Laboratories Inc., Logan, UT, USA) with 10% fetal bovine serum (Gibco, Life Technologies Corp., Grand Island, NY, USA). Trypsin was purchased from Amresco (Solon, OH, USA). The ELISA kits for alanine aminotransferase (ALT), aspartate aminotransferase (AST), albumin and total bilirubin were obtained from Cloud Clone Corp. (Houston, TX, USA). cKit and cytokeratin 18 (CK18) antibodies were manufactured by Abcam (London, UK). An Olympus CKX31-A12PHP inverted microscope was used for observation of cells. A Hitachi SU1510 scanning electron microscope was used for cell ultrastructure observation.
 
Cellular proliferation
To evaluate the effect of hydrogels on L02 cellular proliferation, an methylthiotetrazole (MTT) assay was performed at day 1, 3, 5, and 7 post-culture. Accordingly, 3000 cells/well were placed in a 96-well dish. A total of 20 µL of the MTT solution (0.5%) was added to each well, and cells were allowed to incubate with the MTT solution for 4 hours. The supernatant was subsequently discarded, and 150 µL of dimethyl sulfoxide were added to each well. Low-speed oscillation for 10 minutes was used to fully dissolve the crystals. The optical density (OD) values for each well were measured at an absorbance of 570 nm, with a blank well serving as the control.
 
Preparation of hydrogel scaffolds
A 1% collagen type I solution (BD Biosciences) and a 1.5% chitosan (w/v) HCl solution (0.2 mol/L), both buffered with a 0.5 mol/L morpholinoethanesulfonic acid solution (MES) and NaOH (1 mol/L), were mixed in a 10:1 ratio (w/w) with a pH of 7.2.[20] 1-ethyl-3-(3-dimethyl aminopropyl) carbodiimide (EDC) and N-hydroxysuccinimide (NHS), both at 10% (w/v) in MES, were mixed with the collagen-chitosan solution. The solution was allowed to cross-link for 5 minutes using MES and 1 mol/L NaOH, and then glycine was added. After the incubation of gelation for 30 minutes at 37 ��, complete endothelial basal medium (EBM, Clonetics) was added and the gels were returned to the incubator for 18-24 hours. The biocompatibility of the hydrogels was tested with MTT. The design of the 3D printing scaffold is presented in Fig. 1B. The 3D printing scaffold morphology by eye, under microscope and electron microscope are presented in Fig. 1C-F. The scaffold has 50 layers of cell-hydrogel.
 
3D printing
Regenovo Biotechnology Co., Ltd. (Hangzhou, China) provided the 3D bioprinter (Fig. 1A). The 3D scaffold was fabricated with hydrogel and there were pores in the scaffold according to a grid pattern design. Fig. 1B is the picture of 3D scaffold. This design was different from a previous model.[15]
 
The microscale injection systems, comprising a nozzle with a 210 µm internal diameter, were attached to reservoirs for the bio-ink. The solution comprising hydrogel and L02 cells was about 2×106 cells/mL. The software “SolidWorks” was used to design the 3D model. The software “3D-Bioprint” was used to load the 3D model. The microscale injection systems and bio-ink reservoirs were mounted onto the tool that could build the micrometer-resolution 3-axis XY-Z stage. After cross-linking, a solution of CaCl2 (10%, 50 mL) was used to maintain the 3D pattern. The maximum molding speed was 170 mm/s, the position accuracy was ±0.01 mm, and the size of the molding was 15×15×2 mm.
 
The 3D scaffold was examined by scanning electron microscopy (JSM-6701F, J EOL, ROKYO, Japan) after platinum coating and inverted microscopy (Olympus, IX83, Japan).
 
In vivo experiments
The experiments were performed using nude mice with initial weights of 16-20 g. Sixty mice (30 males and 30 females) were classified to four groups randomly, with 15 mice in each group. The mice were divided into four groups: group 1: control, radiation-induced liver damage (RILD) combined with 2/3 hepatectomy; group 2: group 1+hydrogels; group 3: group 2 with L02 deposition (3D printing); group 4: group 2+hepatocyte growth factor (HGF). The protocol of RILD (50 Gy) combined with 2/3 partial hepatectomy was adopted from previous reports.[21, 22] The possibility of the proliferation of host hepatocytes was excluded because RILD suppresses the proliferation of host hepatocytes. To detect the effects of 3D printing scaffold with different loading in the body, each of the 3D scaffolds were subsequently engrafted into the liver after partial hepatectomy. Five mice of each group were sacrificed after 2 weeks and the rest 10 mice of each group were used in survival analysis. the engrafted scaffolds were collected, sliced, stained with hematoxylin-eosin (HE) and examined using immunohistochemical cKit and CK18 analyses. As the biomarkers of the hepatocyte surface, cKit and CK18 were the specific antigens in hepatocytes.
 
Serum ALT, AST, albumin, total bilirubin, CYP1A2, CYP2C9, a-GST and UGT-2 were tested with ELISA kits. The animal survivals were recorded in all groups. The ethics committee of Zhejiang University approved the animal experiments.
 
Statistical analysis
The differences among groups were evaluated with the one-way ANOVA and Chi-squared test using SPSS software version 12.0 SPSS Inc. (Chicago, IL, USA). P values less than 0.05 were considered significant. The experiments were repeated three times.
 
 
Results
Biocompatibility
The MTT assay of the cells on the 1st, 3rd, 5th, and 7th days of the culture were not significantly different between the cells cultured regularly and those cultured with hydrogel at any time point (P>0.05; Fig. 2).
 
Morphology
The morphology of the engrafted scaffold in different groups was showed in Fig. 3. HE staining showed hepatocyte-like cells and cKit and CK18 were positive in mice from group 3. In the control group, tissue regeneration and cKit+/CK18+ hybridization were not detected. Some nonspecific cells assembled around the mice liver. In the hydrogel group, tissue regeneration and cKit+/CK18+ hybridization were not detectable as well. Some nonspecific cells assembled around the mice liver. Compared with the engrafted tissues in group 3, the tissues showed immature behavior in group 4 and were chaotic and the morphology was random. Only the engrafted tissues in group 3 showed significant liver function.
 
Liver function
At day 5 and 10, the ALT, AST, and total bilirubin levels in group 3 were significantly decreased (P<0.05; Fig. 4) compared with groups 1 and 2. The albumin level was significantly higher at day 5 and 10 (P<0.05; Fig. 4) in group 3 compared with groups 1 and 2. The levels of CYP1A2, CYP2C9, a-GST and UGT-2 were significantly higher at day 1, 5 and 10 (P<0.05; Fig. 5) in group 3 compared with groups 1 and 2. These findings indicated that the liver function in group 3 had rapidly recovered. There was no significant difference between groups 3 and 4 in these variables (P>0.05).
 
Survival analysis
The median survival time of the mice was 8.5, 8, 14 and 11 days in groups 1, 2, 3 and 4, respectively (Fig. 6). The log-rank (Mantel-Cox) test between groups 1 and 3 showed: χ2=8.108; df=3; P<0.05; between groups 2 and 3 were: χ2=5.535; df=1; P<0.05.
 
 
Discussion
One major obstacle that precludes the large regeneration of liver tissue was the lack of an accompanying microvasculature.[23, 24] Without a functional circulatory system, the potential size of the engineered liver tissue is limited to a few hundred pores. Invading microvessels limit the penetration depth and, therefore, impede the successful incorporation of larger constructs.[25] In addition, these vessels, while migrating and developing within the implanted liver tissue, may distort or destroy the construction of the engineered tissue.[26] Therefore, from the standpoint of design, it would be more beneficial to print a preinstalled tissue with biomaterials that preferentially directed vessel ingrowth, or ideally, built a network with the microvasculature in place prior to implanting.[9, 27]
 
The success of liver tissue engineering largely depends on the capacity to construct an organ using cells that are viewed three-dimensionally as well as the successful incorporation into a target organ.[16, 28] Prior to this study, our laboratory has addressed the issue of 3D-cell patterning and functionalization. Relevant methodologies were combined to initiate the present study and to realize a potentially workable platform to generate tissue constructs for liver disease therapy.[29]
 
Here, we introduced a conceptually novel approach that addressed the aforementioned challenges via 3D printing of hydrogel materials and viable cell-seeded hydrogels in the anatomic geometries of the human liver. Because the metabolic function of hepatocyte-like cells is the core of liver tissue engineering, the architecture of the printed hydrogels enables the growth of the engineering organs that exhibit enhanced functionalities. Importantly, we verified that the grid pattern printing process did not affect the vitality of hepatocytes. Our data is consistent with Faulkner-Jones’s report.[15] Our 3D printing tissues were close to natural liver. The pores in the grid hydrogels were suitable for medium exchange. Although the hydrogels had tiny expansion,[30] the morphology of regenerated tissues were closed to the liver tissue after engrafting. This phenomenon was not reported previously. Two studies from the same laboratory[31, 32] detailed the principles and methods that undergirded the direct cell writing biofabrication process and adapted the microfluidic devices for the creation of an organ screening model, establishing a novel organ metabolism study platform. Our study is consistent with Chang and colleagues. 3D printing technology provides a suitable environment for cell growth and metabolism and therefore, the cells in printed scaffold were alive. As more persuasive results, the in vivo results in our research revealed that the grid pattern printing was a novel platform for the study of liver function.
 
Our in vivo data were similar to those of previous cell transplantation reports.[21, 33] The serum albumin concentrations were significantly higher in the transplant group compared with that of the controls. The survival rate of patients with transplanted tissues improved significantly compared with those of controls. The median survival time of our mice with RILD plus a partial hepatectomy was 8.5 days, while this time was expanded to 14 days with the application of our 3D scaffold device, significantly longer than controls. Our data indicated that 3D scaffold device is applicable to improve the liver function and prolong the survival time in our mice model.
 
L02 cells were used in this research. According to previous reports,[18, 19] L02 cells have potential application in liver tissue engineering. Other authors found that L02 cell line possesses a feature of cancer.[4, 5] Although our short-term observation did not show tumor like proliferation, the safety and tumor formation cannot be ruled out according to the present data. Other cells, including stem cells, will be studied in in vivo experiments in the next stage of experiment. The comprehensive evaluation of types of seed cells will achieve a conclusion as to which cell will be better survived and proliferated in 3D printed device.
 
Our approach offers the ability to create spatially heterogeneous constructs by extruding an organized structure of materials in a layer-by-layer process until the final stereo-lithographic geometry is complete. This concept of 3D printed device with living cells together with hydrogel materials and growing these into functional organs without scaffold degradation represents a new direction in biological systems.[14, 34] Indeed, such 3D printed organs are distinct from engineered tissues and they offer a way of attaining a three dimensional merger of scaffolds without degradation. Overall, the 3D printing methods described in the present study are highly suitable for cell placement and scaffold fabrication according to a predefined design;[35] however, the technology applied in tissue engineering is still in its infancy stage and requires further improvement. Especially in terms of vascularization, repeatability, and large-scale production of 3D printed organs, additional research still needs to be conducted.[36]
 
In conclusion, the 3D printed hepatocytes behave the same as natural hepatocytes, 3D printing tissues can be used in liver tissue engineering and liver disease model research in vitro.
 
 
References
1 Zheng SS, Xu X, Wu J, Chen J, Wang WL, Zhang M, et al. Liver transplantation for hepatocellular carcinoma: Hangzhou experiences. Transplantation 2008;85:1726-1732. PMID: 18580463
2 Wertheim JA, Petrowsky H, Saab S, Kupiec-Weglinski JW, Busuttil RW. Major challenges limiting liver transplantation in the United States. Am J Transplant 2011;11:1773-1784. PMID: 21672146
3 Chang HL, Qu K, Liu C, Lv Y, Yu L, Liu XM, et al. Liver transplantation using DCD donors: the current strategy to expand the organ donor pool in China. Am J Transplant 2013;13:1939- 1940. PMID: 23731262
4 Sutaria R, Adams DH. Efforts to expand the donor pool for liver transplantation. F1000 Med Rep 2010;2. PMID: 20948842
5 Nelson B. 3-Dimensional bioprinting makes its mark: new tissue and organ printing methods are yielding critical new tools for the laboratory and clinic. Cancer Cytopathol 2015;123:203- 204. PMID: 25873242
6 Annual Data Report of the US Organ Procurement and Transplantation Network. Preface. Am J Transplant 2014;14:5-7. PMID: 23635097
7 Mannoor MS, Jiang Z, James T, Kong YL, Malatesta KA, Soboyejo WO, et al. 3D printed bionic ears. Nano Lett 2013;13:2634-2639. PMID: 23260439
8 Tasoglu S, Demirci U. Bioprinting for stem cell research. Trends Biotechnol 2013;31:10-19. PMID: 24373164
9 Gledhill K, Guo Z, Umegaki-Arao N, Higgins CA, Itoh M, Christiano AM. Melanin transfer in human 3D skin equivalents generated exclusively from induced pluripotent stem cells. PLoS One 2015;10:e0136713. PMID: 26308443
10 Thoma C. Regenerative medicine: Rapid 3D bladder model engineering. Nat Rev Urol 2015;12:178. PMID: 25800400
11 He M, Zhao Y, Wang B, Xi Q, Zhou J, Liang Z. 3D printing fabrication of amorphous thermoelectric materials with ultralow thermal conductivity. Small 2015;11:5889-5894. PMID: 26448629
12 Palakkan AA, Hay DC, Anil Kumar PR, Kumary TV, Ross JA. Liver tissue engineering and cell sources: issues and challenges. Liver Int 2013;33:666-676. PMID: 23490085
13 Loo Y, Goktas M, Tekinay AB, Guler MO, Hauser CA, Mitraki A. Self-assembled proteins and peptides as scaffolds for tissue regeneration. Adv Healthc Mater 2015;4:2557-2586. PMID: 26461979
14 Zocca A, Elsayed H, Bernardo E, Gomes CM, Lopez-Heredia MA, Knabe C, et al. 3D-printed silicate porous bioceramics using a non-sacrificial preceramic polymer binder. Biofabrication 2015;7:025008. PMID: 26000907
15 Faulkner-Jones A, Fyfe C, Cornelissen DJ, Gardner J, King J, Courtney A, et al. Bioprinting of human pluripotent stem cells and their directed differentiation into hepatocyte-like cells for the generation of mini-livers in 3D. Biofabrication 2015;7:044102. PMID: 26486521
16 Jang M, Neuzil P, Volk T, Manz A, Kleber A. On-chip three-dimensional cell culture in phaseguides improves hepatocyte functions in vitro. Biomicrofluidics 2015;9:034113. PMID: 26180570
17 Powers MJ, Domansky K, Kaazempur-Mofrad MR, Kalezi A, Capitano A, Upadhyaya A, et al. A microfabricated array bioreactor for perfused 3D liver culture. Biotechnol Bioeng 2002;78:257-269. PMID: 11920442
18 Zhao L, Chang J, Zhai W. Preparation and HL-7702 cell functionality of titania/chitosan composite scaffolds. J Mater Sci Mater Med 2009;20:949-957. PMID: 19034620
19 Wu XB, Peng CH, Huang F, Kuang J, Yu SL, Dong YD, et al. Preparation and characterization of chitosan porous microcarriers for hepatocyte culture. Hepatobiliary Pancreat Dis Int 2011;10:509-515. PMID: 21947725
20 McBane JE, Vulesevic B, Padavan DT, McEwan KA, Korbutt GS, Suuronen EJ. Evaluation of a collagen-chitosan hydrogel for potential use as a pro-angiogenic site for islet transplantation. PLoS One 2013;8:e77538. PMID: 24204863
21 Baimakhanov Z, Yamanouchi K, Sakai Y, Koike M, Soyama A, Hidaka M, et al. Efficacy of multilayered hepatocyte sheet transplantation for radiation-induced liver damage and partial hepatectomy in a rat model. Cell Transplant 2016;25: 549-558. PMID: 26224253
22 Landis CS, Zhou H, Liu L, Hetherington HP, Guha C. Liver regeneration and energetic changes in rats following hepatic radiation therapy and hepatocyte transplantation by 31P MRSI. Liver Int 2015;35:1145-1151. PMID: 25775097
23 Dias Garcia RI, Tsuji DH, Imamura R, Mauad T, Ferraz da Silva LF. Effects of hepatocyte growth factor injection and reinjection on healing in the rabbit vocal fold. J Voice 2012;26:667.e7-12. PMID: 22296997
24 Lokmic Z, Mitchell GM. Engineering the microcirculation. Tissue Eng Part B Rev 2008;14:87-103. PMID: 18454636
25 Chang CC, Boland ED, Williams SK, Hoying JB. Direct-write bioprinting three-dimensional biohybrid systems for future regenerative therapies. J Biomed Mater Res B Appl Biomater 2011;98:160-170. PMID: 21504055
26 Dew L, MacNeil S, Chong CK. Vascularization strategies for tissue engineers. Regen Med 2015;10:211-224. PMID: 25835483
27 Wolf S, Arend O, Jung F, Toonen H, Bertram B, Reim M. The microvasculature and microcirculation of the peri-macular capillary network. New diagnostic possibilities with the scanning laser ophthalmoscope. Ophthalmologe 1992;89:45-49. PMID: 1581692
28 Yoon No D, Lee KH, Lee J, Lee SH. 3D liver models on a microplatform: well-defined culture, engineering of liver tissue and liver-on-a-chip. Lab Chip 2015;15:3822-3837. PMID: 26279012
29 Fox IJ, Duncan SA. Engineering liver tissue from induced pluripotent stem cells: a first step in generating new organs for transplantation? Hepatology 2013;58:2198-2201. PMID: 24114924
30 Ali W, Gebert B, Hennecke T, Graf K, Ulbricht M, Gutmann JS. Design of thermally responsive polymeric hydrogels for brackish water desalination: effect of architecture on swelling, deswelling, and salt rejection. ACS Appl Mater Interfaces 2015;7:15696-15706. PMID: 26090770
31 Chang R, Emami K, Wu H, Sun W. Biofabrication of a three-dimensional liver micro-organ as an in vitro drug metabolism model. Biofabrication 2010;2:045004. PMID: 21079286
32 Snyder JE, Hamid Q, Wang C, Chang R, Emami K, Wu H, et al. Bioprinting cell-laden matrigel for radioprotection study of liver by pro-drug conversion in a dual-tissue microfluidic chip. Biofabrication 2011;3:034112. PMID: 21881168
33 Park KM, Hussein KH, Ghim JH, Ahn C, Cha SH, Lee GS, et al. Hepatic differentiation of porcine embryonic stem cells for translational research of hepatocyte transplantation. Transplant Proc 2015;47:775-779. PMID: 25891729
34 Du C, Narayanan K, Leong MF, Wan AC. Induced pluripotent stem cell-derived hepatocytes and endothelial cells in multi-component hydrogel fibers for liver tissue engineering. Biomaterials 2014;35:6006-6014. PMID: 24780169
35 Sudo R. Multiscale tissue engineering for liver reconstruction. Organogenesis 2014;10:216-224. PMID: 24500493
36 Ohashi K, Okano T. Functional tissue engineering of the liver and islets. Anat Rec (Hoboken) 2014;297:73-82. PMID: 24343912
 
Received October 9, 2015
Accepted after revision May 26, 2016