Development of Liver Decellularized Extracellular Matrix Bioink for

Mar 9, 2017 - The liver is an important organ and plays major roles in the human body. Because of the lack of liver donors after liver failure and dru...
3 downloads 12 Views 2MB Size
Subscriber access provided by University of Newcastle, Australia

Article

Development of liver decellularized extracellular matrix bioink for 3D cell printing-based liver tissue engineering Hyungseok Lee, Wonil Han, Hyeonji Kim, Dong-Heon Ha, Jinah Jang, Byoung Soo Kim, and Dong-Woo Cho Biomacromolecules, Just Accepted Manuscript • DOI: 10.1021/acs.biomac.6b01908 • Publication Date (Web): 09 Mar 2017 Downloaded from http://pubs.acs.org on March 10, 2017

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Biomacromolecules is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 28

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

1

Development of liver decellularized extracellular matrix

2

bioink for 3D cell printing-based liver tissue engineering

3 4

Hyungseok Lee1,*, Wonil Han2,*, Hyeonji Kim1,

5

Dong-Heon Ha1, Jinah Jang3, Byoung Soo Kim1, and Dong-Woo Cho1, †

6 7 8

1

9

San 31, Hyoja-dong, Nam-gu, Pohang, Kyungbuk 790-784, South Korea

Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH),

10

2

11

y (POSTECH), 77 Cheongam ro, Nam-gu, Pohang, Kyungbuk 790-784, South Korea.

12

3

13

San 31, Hyoja-dong, Nam-gu, Pohang, Kyungbuk 790-784, South Korea

Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technolog

Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH),

14 15

* Hyungseok Lee and Wonil Han contributed equally to this article.

16 17 18



19

Dong-Woo Cho, Ph.D.

20

Department of Mechanical Engineering, POSTECH

21

San 31 Hyoja-dong, Nam-gu, Pohang, Kyungbuk 790-784, South Korea

22

Tel.: +82 54 279 2171; Fax: +82 54 279 5419; E-mail address: [email protected]

Corresponding author

23 1

ACS Paragon Plus Environment

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

24

Abstract

25

The liver is an important organ and plays major roles in the human body. Due to the lack of

26

liver donors after liver failure and drug-induced liver injury, much research has focused on

27

developing liver alternatives and liver in vitro models for transplantation and drug screening.

28

Although numerous studies have been conducted, these systems cannot faithfully mimic the

29

complexity of the liver. Recently, three-dimensional (3D) cell printing technology has

30

emerged as one of a number of innovative technologies that may help to overcome this

31

limitation. However, great scope remains in developing biomaterials optimized for 3D cell

32

printing-based liver tissue engineering. Therefore, in this research, we developed a liver

33

decellularized extracellular matrix (dECM) bioink for 3D cell printing applications and

34

evaluated its characteristics. The liver dECM bioink retained the major ECM components of

35

the liver while cellular components were effectively removed, and further exhibited suitable

36

and adjustable properties for 3D cell printing. We further studied printing parameters with the

37

liver dECM bioink to verify the versatility and fidelity of the printing process. Stem cell

38

differentiation and HepG2 cell functions in the liver dECM bioink in comparison to

39

commercial collagen bioink were also evaluated, and the liver dECM bioink was found to

40

induce stem cell differentiation and enhance HepG2 cell function. Consequently, the results

41

demonstrate that the proposed liver dECM bioink is a promising bioink candidate for 3D cell

42

printing-based liver tissue engineering.

43 44

Keywords: 3D cell printing technology, liver, tissue engineering, decellularized extracellular

45

matrix (dECM), bioink

2

ACS Paragon Plus Environment

Page 2 of 28

Page 3 of 28

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

46

1. Introduction

47

The liver is a vital organ which plays major roles in balancing biochemical environments in

48

the human body, including blood protein synthesis, glucose metabolism, and detoxification of

49

metabolites or other chemicals.1 Liver transplantation is widely used in the case of liver

50

failure; however, transplantation is limited by a lack of liver donors.2, 3 Furthermore, many

51

pharmaceutical products have been banned or not approved because of drug-induced liver

52

injury.4 Numerous studies have focused on developing liver alternatives and liver in vitro

53

models. Takabe et al. have developed a vascularized functional human liver using induced

54

pluripotent stem cells,5 and a recellularized liver graft was developed from a decellularized

55

liver matrix by Uygun et al.6 Additionally, liver spheroids have been studied as a liver in vitro

56

model for drug screening.7-9 Although various studies have been conducted to develop liver

57

alternatives and liver in vitro models, the aforementioned studies could not control the

58

positions of the multiple cell types and of the extracellular matrix (ECM) of the liver.

59

Furthermore, it is nearly impossible to prepare patient-specific liver constructs due to a lack

60

of fabrication methods.

61

Three-dimensional (3D) cell printing is an emerging technology in bioengineering due to its

62

suitability for the fabrication of complex 3D biological constructs.10, 11 The most promising

63

advance in 3D cell printing is that various biomaterials and multiple cell types can be printed

64

simultaneously as intended to fabricate complex biological structures.12, 13 Taking advantage

65

of these advances, Kang et al. have applied 3D cell printing to produce human-scale tissues,

66

and they have also shown that it is possible to achieve patient specificity.14 Further to

67

advances in 3D cell printing, the development of bioink, which encapsulates the cells during

68

the printing process, is another key means of improving the functionality of cell printed 3

ACS Paragon Plus Environment

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

69

constructs. Several studies have focused on liver bioink development. Mixtures, such as

70

gelatin-chitosan, agarose-collagen, and agarose-chitosan, have been employed, for example.15,

71

16

72

remains to be explored. As per another example, when the liver-derived material was used as

73

a liver bioink, the printing uniformity of the bioink had to be improved using poly(ethylene

74

glycol) diacrylate (PEGDA) and an ultraviolet (UV) crosslinking process.17 Occasionally, UV

75

light or radicals associated with the photoinitiator affect cell behavior unexpectedly. As these

76

examples show, much effort has gone into the development of the liver bioink; however, no

77

study has successfully produced a liver-derived bioink that is non-toxic and offers adjustable

78

stiffness and high printability.

79

In this study, we developed and investigated the characteristics and performance of a liver

80

decellularized extracellular matrix (dECM) bioink for 3D cell printing. Initially, we evaluated

81

the biochemical characteristics of the liver dECM bioink after the decellularization process.

82

Then, its rheological properties, printing parameters, and cytotoxicity as well as printed

83

constructs were assessed. Furthermore, stem cell differentiation and the functionality of

84

HepG2 cells in the liver dECM bioink were evaluated and compared with those in

85

commercial collagen bioink. The liver dECM bioink developed here improved the

86

functionality of printed structures.

However, these bioinks did not contain liver-specific biochemical components, which

87 88

2. Materials and Methods

89

2.1. Liver decellularization

90

Porcine liver tissue was sliced roughly into 0.5–1-mm-thick slices and washed with distilled 4

ACS Paragon Plus Environment

Page 4 of 28

Page 5 of 28

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

91

water for 2 hours. Next, the slices were treated with 0.5% Triton X-100 (Sigma-Aldrich, USA)

92

in 1 M NaCl (Samchun Pure Chemicals, Korea) for 9 hours. Any remaining cellular

93

components were washed out with 1% SDS (Affymetrix, USA) for 3 hours, and the sample

94

was sterilized by means of a 1-hour treatment with 0.1% peracetic acid (Sigma-Aldrich, USA)

95

in phosphate-buffered saline (PBS). Between decellularization steps, the tissues were washed

96

with distilled water for 2-5 hours to remove remaining detergent and cell debris.

97

Decellularized tissues were freeze-dried for 48 hours with Free Zone 2.5 (Labconco, USA)

98

and then used for biochemical characterization and liver dECM bioink preparation.

99

2.2. Biochemical characterization of liver dECM

100

To quantify double-stranded DNA content, a DNA quantification kit (Sigma-Aldrich, USA)

101

was purchased and assays were conducted. Deoxyribonucleic acid sodium salt (Sigma-

102

Aldrich, USA) was used for the standard sample of double-stranded DNA. Fluorescence

103

intensity was measured with a SpectraMax Gemini XPS (Molecular Devices, excitation

104

wavelength: 360nm, emission wavelength: 450nm). For the immunohistochemical analysis,

105

native liver and decellularized tissues were fixed in 4% formalin, embedded in paraffin, and

106

sectioned with a microtome. Sectioned samples were stained with hematoxylin and eosin

107

(H&E), Masson's trichrome, anti-fibronectin, and alcian blue, and then the stained samples

108

were examined with a light microscope.

109

2.3. Preparation of biomaterials

110

To prepare the liver dECM bioink for the 3D cell printing system, the liver dECM was

111

lyophilized for 24 hours and ground into powder with a grinder. The liver dECM powder was

112

dissolved in 0.5M acetic acid (Duksan, Korea) with 10% tissue weight of pepsin (Sigma-

113

Aldrich, USA) using a magnetic stirrer for 4 days. The liver dECM solution was centrifuged 5

ACS Paragon Plus Environment

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

114

at 3500 rpm for 10 min to remove large particles. Finally, to prepare the liver dECM bioink,

115

the pH of the liver dECM solution was adjusted to 7.4 with 10 M sodium hydroxide. Porcine

116

skin-derived collagen type I (Dalim Tissen, Korea) was dissolved in 0.5 M acetic acid for

117

four days and neutralized with 10 M sodium hydroxide for the collagen bioink. The liver

118

dECM and collagen bioink were prepared and maintained at below 4 °C. Finally,

119

polycaprolactone (PCL) was also purchased from Polysciences (USA).

120

2.4. Rheological characterization

121

The rheological characteristics of the 1.5% (15 mg/ml) and 3% (30 mg/ml) liver dECM

122

bioinks and collagen bioinks in the same concentrations were evaluated with the Advanced

123

Rheometric Expansion System (TA Instruments, USA). A steady shear sweep analysis of the

124

liver dECM collagen bioinks was performed at 15 °C to evaluate its viscosity. A dynamic

125

frequency sweep analysis was conducted to measure the frequency-dependent storage (G’)

126

and loss (G”) modulus of the liver dECM and collagen bioinks.

127

2.5. Cell preparation and encapsulation in bioinks

128

Human hepatocellular carcinoma (HepG2) cell lines and human bone marrow-derived

129

mesenchymal stem cells (BMMSCs) were purchased from ATCC (USA) and Cefobio (Seoul,

130

Korea) respectively. Both cell types were cultured in Dulbecco’s modified Eagle’s medium

131

(DMEM, Gibco BRL, NY, USA) with 10% (v/v) fetal bovine serum (FBS, Gibco BRL, NY,

132

USA) and 1% (v/v) penicillin/streptomycin (P/S, Sigma-Aldrich, St. Louis, MO, USA) at

133

37 °C in a humidified 5% CO2 atmosphere. All cells used were from passage 2 to 4.

134

BMMSCs and HepG2 cells were encapsulated in bioink separately, at a concentration of 5 x

135

106 cells/ml.

6

ACS Paragon Plus Environment

Page 6 of 28

Page 7 of 28

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

136

2.6. Application to 3D cell printing technology

137

To study the 3D cell printing process with the liver dECM bioink, the printing parameters of

138

pneumatic pressure, printing speed, and nozzle diameter were altered. PCL material was

139

printed with liver dECM bioink to make the hybrid constructs, and PCL was printed at a

140

temperature of 60 °C and a pneumatic pressure of 500 kPa. After the printing process, the

141

printed structures were incubated for 1 hour for gelation. All printing procedures were carried

142

out with an in-house-developed 3D cell-printing system designed to print polymers and

143

bioinks using pneumatic pressure and heat induced by a heating controller.18 The heating

144

temperature and pneumatic pressure can respectively be raised to 150 °C and 650 kPa.

145

2.7. Culture of cell printed constructs

146

The BMMSC and HepG2 cell-printed constructs of 2D patterns were cultured in Dulbecco’s

147

modified Eagle’s medium (DMEM, Gibco BRL, NY, USA) with 10% (v/v) fetal bovine

148

serum (FBS, Gibco BRL, NY, USA) and 1% (v/v) penicillin/streptomycin (P/S, Sigma-

149

Aldrich, St. Louis, MO, USA) at 37 °C in a humidified 5% CO2 atmosphere. No supplements

150

were treated to analyze the biochemical functionalities of liver dECM bioink alone compared

151

to the collagen bioink, which was used as a negative control. The culture media was

152

exchanged every two to three days, and samples were properly harvested in accordance with

153

the analysis.

154

2.8. Evaluation of cell viability

155

To assess the viability of the HepG2 and BMMSCs cells after the 3D cell printing process,

156

samples were stained with live/dead kits (Life Technologies, Germany). 4mM calcein AM for

157

the live cell assay and 2mM ethidium homodimer solution for the dead cell assay were mixed 7

ACS Paragon Plus Environment

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

158

in a ratio of 1 to 4 in PBS and incubated with printed samples at 37 °C for 30 min. This was

159

followed by PBS washing to remove any remainders of the solutions. Live and dead cells

160

were visualized with a Zeiss LSM 510 Meta confocal microscope (Zeiss, Jena, Germany).

161

2.9. Gene expression analysis

162

Samples of printed structure were harvested, and total RNA from each sample was isolated

163

using the RNeasy Isolation Kit (QIAGEN) according to the manufacturer’s instructions.

164

Complementary DNA was synthesized using a Maxima First Strand cDNA Synthesis Kit

165

(Thermo Scientific, Waltham, MA, USA). Gene expression was analyzed with SYBR green

166

PCR Master Mix using real-time PCR (Applied Biosystems, Foster City, CA, USA).

167

The mRNA sequence of target genes was obtained from the National Center for

168

Biotechnology Information (NCBI) gene sequence database, and all primers (Table 1) were

169

designed using Primer Express 3.0 software (Applied Biosystems, Foster City, CA, USA).

170

Forward and reverse primers for glyceraldehyde 3-phosphate dehydrogenase (GAPDH),

171

albumin (ALB), alpha-fetoprotein (AFP), transthyretin (TTR), cytokeratin 18 (CK18),

172

hepatocyte nuclear factor 1 alpha (HNF1A), hepatocyte nuclear factor 3 beta (HNF3B),

173

hepatocyte nuclear factor 4 alpha (HNF4A), and hepatocyte nuclear factor 6 (HNF6) were

174

purchased from Pioneer (Korea). Gene expression levels were all normalized relative to

175

GAPDH.

176

Table 1. Primer sequences for quantitative real-time PCR analysis. Gene GAPDH HNF1α HNF3β

Sense Anti-sense Sense Anti-sense Sense Anti-sense

Sequence ATGGAAATCCCATCACCATCTT CGCCCCACTTGATTTTGG GAGGAGCGAGAGACGCTAGTG CACCCCTCTCTGGATGCATT CTGAAGCCGGAACACCACTAC CGAGGACATGAGGTTGTTGATG 8

ACS Paragon Plus Environment

Page 8 of 28

Page 9 of 28

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

HNF4α HNF6 ALB AFP CK18 TTR

Sense Anti-sense Sense Anti-sense Sense Anti-sense Sense Anti-sense Sense Anti-sense Sense Anti-sense

CGCTACTGCAGGCTCAAGAAA TCTGGACGGCTTCCTTCTTC TGCGCAACCCCAAACC TCCACATCCTCCGGAAGGT TTGCATGAGAAAACGCCAGTA AGCATGGTCGCCTGTTCAC TCGGACACTTATGTATCAGACATGAA GCCTCCTGTTGGCATATGAAG GCCTTGGACAGCAGCAACTC ACCACTTTGCCATCCACTATCC ACCAAATCTTACTGGAAGGCACTT GAGTCGTTGGCTGTGAATACCA

177 178

2.10. Immunofluorescence staining

179

The 3D cell printed liver constructs were fixed with 4% paraformaldehyde. The samples were

180

permeabilized with 0.1% Triton X-100 and were treated with 3% bovine serum albumin

181

(Affimetrix, USA) in PBS for 1 hour to block the nonspecific binding. The samples were

182

washed with PBS three times for 15 min. Anti-human HNF4A rabbit antibody (Abcam, UK)

183

and anti-human AFP mouse antibody (Abcam, UK) were used as primary antibodies and

184

treated overnight at 4 °C. After washing with PBS, the samples were treated with Alexa Fluor

185

594 goat anti-rabbit antibody (Invitrogen, CA, USA) and Alexa Fluor 488 goat anti-mouse

186

antibody (Invitrogen, CA, USA) for 1 hour at 37 °C and counterstained with 4',6-diamidino-

187

2-phenylindole (DAPI). Stained images were obtained with a FV1000 Olympus confocal

188

microscope (Olympus, Tokyo).

189

2.11. Liver function test

190

Constructs fabricated with collagen and the liver dECM bioink with the same concentration

191

of 1.5% were cultured for seven days to conduct a liver function test. Medium samples were

192

collected for analysis for secreted albumin/urea and refreshed every three days. Secreted

193

albumin was quantified with an enzyme-linked immunosorbent assay (ELISA) (DuoSet 9

ACS Paragon Plus Environment

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

194

ELISA development system, Genzyme). Secreted urea was quantified with a urea assay kit

195

(BioVision, USA). Quantification was performed with a microplate reader (Epoch 2

196

Microplate Spectrophotometer, BioTek, USA).

197

2.12. Statistical analysis

198

All variables for liver differentiation and liver-specific functions were expressed as means ±

199

standard deviations (SDs). Evaluation of the difference between the mean values of each

200

group was performed by Student’s t-test, in which a p-value