Elsevier

Biomaterials

Volume 265, January 2021, 120417
Biomaterials

Development of highly functional bioengineered human liver with perfusable vasculature

https://doi.org/10.1016/j.biomaterials.2020.120417Get rights and content

Abstract

Liver tissue engineering offers a promising strategy for liver failure patients. Since transplantation rejection resulting in vessel thrombosis is regarded as a major hurdle, vascular reconstruction is one of indispensable requirements of whole organ engineering. Here we demonstrated a novel strategy for reconstruction of a vascularized bioengineered human liver (VBHL) using decellularized liver scaffolds in an efficient manner. First we achieved fully functional endothelial coverage of scaffolds by adopting the anti-CD31 aptamer as a potent coating agent for re-endothelialization. Through an ex vivo human blood perfusion that recapitulates the blood coagulation response in humans, we demonstrated significantly reduced platelet aggregation in anti-CD31 aptamer coated scaffolds. We then produced VBHL constructs using liver parenchymal cells and nonparenchymal cells, properly organized into liver-like structures with an aligned vasculature. Interestingly, VBHL constructs displayed prominently enhanced long-term liver-specific functions that are affected by vascular functionality. The VBHL constructs formed perfusable vessel networks in vivo as evidenced by the direct vascular connection between the VBHL constructs and the renal circulation. Furthermore, heterotopic transplantation of VBHL constructs supported liver functions in a rat model of liver fibrosis. Overall, we proposed a new strategy to generate transplantable bioengineered livers characterized by highly functional vascular reconstruction.

Introduction

End-stage liver diseases, including chronic liver disease and cirrhosis, cause approximately 2 million of deaths worldwide each year [1]. Liver transplantation is a well-established therapeutic strategy for patients who suffer from hepatic failure. As the demand for organs has grown worldwide, a severe shortage of donor organs had necessitated the development of substitutes for human organs. In this regard, bioengineered liver constructs have currently been suggested as an alternative.

Decellularized extracellular matrix scaffolds, which are obtained by the removal of cells from donor tissue, are as a promising substrate for tissue engineering [[2], [3], [4], [5]]. Owing to preservation of their own complex microstructures and biological signals, the cells seeded into decellularized scaffolds can migrate to the appropriate microenvironment. Thus, decellularized scaffolds have been employed as compatible sources for the fabrication of bioengineered organs. For instance, Song et al. generated functional transplantable kidney grafts by engineering decellularized rat and human kidney matrix [6]. Subsequent work by the same group also demonstrated that decellularized lung scaffolds were repopulated with human cells and then displayed vascular maturation [7].

Likewise, in recent years, a number of studies have been conducted on establishing artificial liver using decellularized liver scaffolds [2,3,[8], [9], [10]]. Given that the liver is a vascular organ with intricate vessels, complete re-endothelialization of the scaffolds is the key to successful reconstruction of the bioengineered liver. To date, various coating agents, such as heparin-gelatin and REDV peptide, have been explored to improve the delivery efficiency of endothelial cells (ECs) [8,11,12]. However, since these chemicals do not specifically interact with ECs, to create a bioengineered liver composed of multiple cell types, there is a need to develop coating agents that interact specifically with ECs so that only ECs attach to the luminal surface of the vasculature within the scaffolds. Although the anti-CD31 antibody coating can selectively capture ECs to ensure better attachment [13], the limitations are that mass production of antibodies requires complicated manufacturing procedures, and the cost of production is very high. Moreover, antibodies derived from other species are possibly recognized as foreign bodies by the human immune system and induce an immune response, leading to undesirable outcomes [14]. These limitations, which obstruct the clinical applications of antibodies led us to develop a new method for efficient re-endothelialization of the liver scaffolds.

Nucleic acid aptamers are composed of synthesized single-stranded oligonucleotides with short sequences. Aptamers are considered alternatives to antibodies due to their low immunogenicity and strong binding affinity for specific proteins [15,16]. It is also beneficial that aptamers can be easily modified and produced cost-effectively in large quantities without much variability or contamination, compared to antibodies [17]. In general, various aptamers have been utilized for medical diagnosis and as therapeutic agents for diseases, such as cancer [18,19] and viral infections [20].

In this study, we aimed to efficiently produce bioengineered human livers with well-organized vascular structures that maintain functional integrity. Hence, we first utilized an anti-CD31 aptamer as a coating agent for the purposes of the re-endothelialization of decellularized liver scaffolds and identified the influence of anti-CD31 aptamers on ECs. To predict vascular thrombosis associated with the rejection in humans, we evaluated the thrombogenicity of the re-endothelialized constructs after continuous human blood perfusion. With these strategies, we fabricated vascularized bioengineered human liver (VBHL) constructs that retained the structural and functional properties of the liver. We also reported the in vivo functionality of VBHL constructs, including graft patency following blood transfusion using the renal circulatory system in rats and the ability to support liver functions by heterotopic implantation into the cirrhotic livers of rats. Eventually, we established a highly functional bioengineered liver with a perfusable vasculature that could feasibly be clinically applied as a substitute for liver transplantation.

Section snippets

Aptamer preparation and analysis

Anti-CD31 single-stranded DNA aptamer (76mer, 2196-18-01) was synthesized and characterized by Aptamer Sciences Inc. (Republic of Korea). After dissolving anti-CD31 aptamer in sterile Ultrapure DEPC-treated water (Invitrogen, USA), it was stored in −20 °C. Prior to conducting the experiments, aptamer was boiled at 95 °C for 10 min and cooled at room temperature to induce proper folding. To confirm that anti-CD31 aptamer specifically targets CD31+ ECs, immunofluorescence staining and flow

Anti-CD31 aptamer specifically binds to ECs

The anti-CD31 aptamer was readily obtained from chemical synthesis and subsequent selection via systematic evolution of ligands by exponential enrichment (SELEX) [24]. As it was designed to target the CD31 protein (Fig. 1A), we attempted to clarify its binding characteristics. First, we observed that the anti-CD31 aptamer bound to HUVECs in a dose-dependent manner (Fig. 1B–C). Of note, the binding affinity of the anti-CD31 aptamer was saturated at 600 nM by up to 99%. Thus, 600 nM was selected

Discussion

Liver tissue engineering has been considered an alternative to the use of donor organs for liver transplantation. DLM has become a promising strategy for the fabrication of artificial livers because it provides an essential physical scaffold for cell engraftment by retaining the necessary biochemical and biomechanical cues. Meanwhile, revascularization of the scaffold is indispensable because endothelial denudation likely causes activation of the extrinsic coagulation pathway and causes

Conclusions

In this study, an efficient strategy is developed to reconstruct vascularized bioengineered liver constructs by using anti-CD31 aptamer. Anti-CD31 aptamer coating of the vascular lumen in the decellularized liver scaffolds promotes the efficient re-endothelialization, thereby leading to formation of perfusable vascular networks with enhanced viability and functionality via activation of integrin-Akt signaling. Furthermore, properly organized liver constructs with highly functioning vasculature

Author contributions

D-H.K. designed and performed the study. D-H.K., M-S.K., H.K.K., N-G.K. and M.G.K. performed the experiments. J.A. fabricated microfluidic devices and helped to analyze the data. S.W.C., N.L.J., and H-M.W. contributed discussion. K–S.K. supervised the study and contributed to writing.

Data availability

All data associated with this study are present in the paper or the Supplementary Data.

CRediT authorship contribution statement

Da-Hyun Kim: Conceptualization, Data curation, Writing - original draft, Writing - review & editing, Investigation. Jungho Ahn: Methodology, Data curation. Hyun Kyoung Kang: Methodology, Visualization. Min-Soo Kim: Validation. Nam-Gyo Kim: Writing - review & editing. Myung Geun Kook: Validation, Formal analysis. Soon Won Choi: Methodology, Formal analysis. Noo Li Jeon: Software, Resources. Heung-Myong Woo: Funding acquisition. Kyung-Sun Kang: Supervision, Project administration.

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgements

We are grateful to Dr James J. Yoo and Dr In Kap Ko at Wake Forest Institute for Regenerative Medicine for helpful discussions. We also thank Dr Jae-Jun Kim at University of California San Francisco and Dr Kyung-Mee Park at Chungbuk National University for experimental advices. This work was supported by the grant from Rural Development Administration (Project No. PJ01100201) and National Research Foundation of Korea (Project No. 2018R1A2B3008483), Republic of Korea.

References (52)

  • M.L. Wong et al.

    Immunogenicity in xenogeneic scaffold generation: antigen removal vs. decellularization

    Acta Biomater.

    (2014)
  • E.C. Stahl et al.

    Evaluation of the host immune response to decellularized lung scaffolds derived from alpha-Gal knockout pigs in a non-human primate model

    Biomaterials

    (2018)
  • J.S. Uzarski et al.

    Essential design considerations for the resazurin reduction assay to noninvasively quantify cell expansion within perfused extracellular matrix scaffolds

    Biomaterials

    (2017)
  • S. Hemmann et al.

    Expression of MMPs and TIMPs in liver fibrosis - a systematic review with special emphasis on anti-fibrotic strategies

    J. Hepatol.

    (2007)
  • A.M. Moon et al.

    Contemporary epidemiology of chronic liver disease and cirrhosis

    Clin. Gastroenterol. Hepatol.

    (2019)
  • B.E. Uygun et al.

    Organ reengineering through development of a transplantable recellularized liver graft using decellularized liver matrix

    Nat. Med.

    (2010)
  • P.M. Baptista et al.

    The use of whole organ decellularization for the generation of a vascularized liver organoid

    Hepatology

    (2011)
  • H.C. Ott et al.

    Perfusion-decellularized matrix: using nature's platform to engineer a bioartificial heart

    Nat. Med.

    (2008)
  • T.Y. Lu et al.

    Repopulation of decellularized mouse heart with human induced pluripotent stem cell-derived cardiovascular progenitor cells

    Nat. Commun.

    (2013)
  • J.J. Song et al.

    Regeneration and experimental orthotopic transplantration of a bioengineered kidney

    Nat. Med.

    (2013)
  • X. Ren et al.

    Engineering pulmonary vasculature in decellularized rat and human lungs

    Nat. Biotechnol.

    (2015)
  • J.E. Nichols et al.

    Production and transplantation of bioengineered lung into a large-animal model

    Sci. Transl. Med.

    (2018)
  • S. Ogiso et al.

    Efficient recellularisation of decellularised whole-liver grafts using biliary tree and foetal hepatocytes

    Sci. Rep.

    (2016)
  • E. Doevendans et al.

    Immunogenicity of innovative and biosimilar monoclonal antibodies

    Antibodies

    (2019)
  • K.M. Song et al.

    Aptamers and their biological applications

    Sensors

    (2012)
  • J. Zhou et al.

    Aptamers as targeted therapeutics: current potential and challenges

    Nat. Rev. Drug Discov.

    (2017)
  • Cited by (22)

    • Collagen-based biomaterials for tissue engineering applications

      2023, Natural Biopolymers in Drug Delivery and Tissue Engineering
    • Improving hemocompatibility of decellularized liver scaffold using Custodiol solution

      2022, Biomaterials Advances
      Citation Excerpt :

      Furthermore, a denude and native tridimensional vascular network is also preserved into acellular scaffolds after whole organ decellularization. Although several studies have been transplanting decellularized liver as auxiliary support through end-to-end or end-to-side anastomosis, problems involving thrombus formation are frequent and promote a lack of oxygen and nutrient diffusion resulting in graft failure after transplantation [44]. In this regard, we have used Custodiol (HTK solution), a leading organ preservation and storage solution, to improve the hemocompatibility of decellularized liver scaffolds.

    • Translational tissue engineering

      2022, Tissue Engineering: Current Status and Challenges
    • Xenoextracellular matrix-rosiglitazone complex-mediated immune evasion promotes xenogenic bioengineered root regeneration by altering M1/M2 macrophage polarization

      2021, Biomaterials
      Citation Excerpt :

      The ECM has been widely recommended as a substitute biomaterial in complex organs reconstruction because of its immune privilege and great potential for clinical application [1,2].

    View all citing articles on Scopus
    View full text