Open access peer-reviewed chapter

Optimization of a Decellularization/Recellularization Strategy for Transplantable Bioengineered Liver

Written By

Quanyu Chen, Xiaolin You, Jiejuan Lai, Shifang Jiang, Hongyu Zhang and Lianhua Bai

Submitted: 11 August 2019 Reviewed: 03 September 2019 Published: 27 November 2019

DOI: 10.5772/intechopen.89493

From the Edited Volume

Xenotransplantation - Comprehensive Study

Edited by Shuji Miyagawa

Chapter metrics overview

839 Chapter Downloads

View Full Metrics

Abstract

The liver is a complex organ that requires constant perfusion for the delivery of nutrients and oxygen and the removal of waste in order to survive. Efforts to recreate or mimic the liver microstructure via a ground-up approach are essential for liver tissue engineering. A decellularization/recellularization strategy is one of the approaches aiming at the possibility of producing a fully functional organ with in vitro-developed construction for clinical applications to replace failed livers, such as end-stage liver disease (ESLD). However, the complexity of the liver microarchitecture along with the limited suitable hepatic component, such as the optimization of the extracellular matrix (ECM) of the biomaterials, the selection of the seed cells, and development of the liver-specific three-dimensional (3D) niche settings, pose numerous challenges. In this chapter, we have provided a comprehensive outlook on how the physiological, pathological, and spatiotemporal aspects of these drawbacks can be turned into the current challenges in the field, and put forward a few techniques with the potential to address these challenges, mainly focusing on a decellularization-based liver regeneration strategy. We hypothesize the primary concepts necessary for constructing tissue-engineered liver organs based on either an intact (from a naïve liver) or a partial (from a pretreated liver) structure via simulating the natural development and regenerative processes.

Keywords

  • tissue engineering
  • decellularization
  • recellularization
  • thrombogenicity
  • hemocompatibility
  • partial hepatectomy transplantation

1. Introduction

The liver is the largest internal organ in the human body, accounting for approximately 2–5% of the total body volume [1, 2]. Physiologically, the liver possesses over 500 different functions [3] and any severe damage could be life-threatening, such as that caused by ESLD, including acute liver failure and chronic liver disease.

In modern times, the failure of solid organs, such as ESLD caused by injury or disease, has become a major challenge in clinics [4]. According to the U.S. Centers for Disease Control and Prevention (https://www.cdc.gov/), in 2014, 38,170 people died of ESLD. Currently, orthotopic liver transplant (OLT) is an ideal therapy for ESLD. However, a shortage of liver organ donors severely limits OLT usage. The Department of Health and Human Services in the United States has estimated that (https://optn.transplant.hrsa.gov) 22 people on the National Transplant Waiting List die each day, while one person is added to the waiting list every 10 min. Additionally, people fortunate enough to receive an organ transplantation have to suffer from the lifelong use of immunosuppressants against chronic rejection. Therefore, new technologies are eagerly needed to create a transplantable liver [5]. Tissue engineering is a mixed field that aims to fabricate functional organs in vitro [6]. Over the decades, great progress has been achieved in the laboratory, and even some livers have been used in clinics [7]. Tissue engineering by using a decellularization/recellularization strategy, which maintains the architecture, vascular system, and ECM components, has been shown to be a promising tool for solid organs, such as liver.

Liver tissue engineering by using decellularization/recellularization strategy (Figure 1) involves biomimicking the architecture and physiological features of the native liver. The procedure generally needs three major components: a scaffolding platform, seed cells, and a 3D microenvironment. Despite the numerous advances over the years, it is still an enormous challenge to fabricate a liver organ [8]. Generating liver organ-specific 3D structure scaffold to keep as much as original biochemical, physiochemical, and biomechanical ECM microenvironment is the one of the main hurdles in liver engineering field [9]. Such physiological 3D structure also plays a remarkable role in influencing seeded cell long-term survival and complex liver tissue mass formation [10]. To achieve this, scientists have been working with different scaffolding systems for liver tissue engineering. Studies have shown that a construction strategy based on a combination of a decellularized naïve liver matrix and recellularization with seed cells has led to constructs that match human organs in size and structure. However, the present constructs still only fulfill partial functions of the liver. The preservation of a functional ECM during decellularization, cellular differentiation [11], and a lack of endothelial-lying vascular networks limits the long-term functional integration of constructs after in vivo transplantation. As techniques continue develop, some methods with the potential to overcome these challenges should be explored in the near future, which will further boost the development of a tissue-engineered liver with improved functions. In this chapter, we have tried to focus on the possibility of liver tissue engineering by using a decellularization/recellularization strategy and to describe the current advancements made in the field to address a possible clinical transplantation.

Figure 1.

The decellularization/recellularization strategy in liver tissue engineering. Mammals donor-derived livers undergo a process of decellularization to obtain decellularized liver scaffolds (DLS) (step a–c), and then recellularized seed cells are placed onto the scaffolds (step d–f). Finally, the recellularized scaffolds are placed into 3D culture conditions in a bioreactor to construct liver-like tissues or organs with an overall structure and vasculature (step g).

Advertisement

2. Decellularization-based scaffold biomaterials

The term “biomaterials” traditionally means a nonliving substance used for a medical purpose. As the technology of biomaterials developed, the definition expanded to include substances to control the biological environment of cells and tissues for increased compatibility with a host to allow for colonization, proliferation, and differentiation of cells while maintaining their specific morphologies, configurations and avoiding immunological rejection. Based on the increasing knowledge of ECM biology, scaffold biomaterials can be grouped as synthetic materials, natural materials, or a decellularized matrix [12]. Moreover, modifications have been made to enhance the biologically active signals of scaffolds, leading to improved cell attachment, survival, and tissue formation [13, 14].

Biomaterials with required properties have been well studied from synthetic materials. For instance, a nanofibrous matrix made of poly and poly-embedded growth factors was transplanted into animals and restored cardiac regeneration by promoting vascularization [15]. Zawaneh et al. have reported the design of an injectable synthetic and biodegradable polymeric biomaterial consisting of polyethylene glycol and a polycarbonate of dihydroxyacetone that is easily extruded through narrow-gage needles, biodegrades into inert products, and is well tolerated by soft tissues [16]. Those chemically and biologically modified synthetic materials could result in a better way to mimic and control seed cell responses [17, 18]. Another advantage of synthetic materials is their easier to predict and control the degradation of synthetic scaffolds. However, despite this wealth of knowledge, the ability of synthetic biomaterials to support cell attachment, or induce phenotypic expression is much lower than that of natural biomaterials [19, 20, 21]; thus, natural biomaterials have been extensively studied [22].

Natural biomaterials include collagen, alginate, and chitosan. These types of biomaterials are inherently able to facilitate for seed cell attachment, proliferation, and functional differentiation, thus they hold significant promise for liver tissue engineering [23, 24]. However, traditional natural materials have poor inherent bioactivity, acidic byproducts, etc., and alone cannot rebuild the complex architecture of solid organs like liver. Other limitations include their unpredictable degradation kinetics; generally, weak mechanical strength, and risk of evoking an immune response [25], etc. also need to be considered.

Decellularized scaffolds (matrices) being natural biomaterials, which are deprived of cellular components while maintaining their original architecture and vascular system, have been widely studied and used in more complex tissue engineering [26]. In the case of liver tissue engineering, the use of decellularization/recellularization strategy was inspired by a pioneer study of heart tissue engineering from the Ott group in 2008 [27]. After that, liver tissue engineered by using this approach has been fabricated [28, 29, 30]. Compared to those derived from other synthetic or natural biomaterial scaffolds, the decellularized liver scaffold (DLS) mostly preserves the native complex liver ECM components, spatial microstructure, and perusable vascular architecture [31, 32] as more “biocompatible ways” for seed cells attaching and reorganizing on a complex 3D level [33]. Therefore, the DLS might have more favorable advantages than other scaffolds for clinical application although the biocompatibility signal between ECM of the DLS and seed cells is still unclear. Scientists have recellularized stem cells onto the natural 3D DLS and have found that these culturing cells not only survive better in the scaffold structure than their culturing in 2D environment but also differentiate into functional cells as well [34]. Zhang et al. seeded adult mouse liver hepatic stem/progenitor cells onto the DLS that generated from naïve liver (nDLS) and cultured the complex in bioreactor, which formed a liver-like construction. Importantly, the nDLS/cell construction was able to repair a cirrhotic liver and even replace the failure liver [35].

Although many studies have been performed in the DLS field for liver tissue engineering [36, 37, 38, 39, 40], unfortunately, because of the nDLS being a lack of “active microenvironmental” support in existing ECM components, the optimization of the nDLS biomaterials become an important procedure for improving the skill of liver tissue engineering. Many protocols have been applied to modify the non-bioactive decellularized scaffolds. The application of a variety of growth factors [41] to promote the survival, proliferation, and differentiation of cells, like insulin-like growth factor 1 thought to promote hepatic cell differentiation from bone marrow-derived mesenchymal stem cells, vascular endothelial growth factor applied to enhance the vascularization of tissue-engineered tissues or organs. Additionally, the complex synergistic and antagonistic actions between different kinds of growth factors in vivo, more attention should be paid to the combined and sequential application of different growth factors. Consideration of optimizing the ECM of nDLS foir its behave like “naïve liver regenerative niche” might be a nice way to induce liver-like tissue formation spontaneously both in vitro and in vivo. Based on this, recently, Yang et al. has presented a very interesting experiment: the authors generated an acellular liver scaffold from pretreated naïve liver. They pretreated a naïve liver by performing a 30–55% partial hepatectomy, and the liver was maintained in vivo for 3–5 days until acute liver regeneration occurred, which allowed for the generation of the scaffold from the regenerative liver (rDLS) (Figure 2). These rDLS retain a variety of higher level of supporting growth factors for liver spontaneous regeneration as compared to that of nDLS, including their collagens, growth factors (HGF, TGF-α, IL-6, b-FGF, VEGF), glycosaminoglycans, antithrombotic proteins, and other matrix proteins [42]. Since the novel rDLS possesses a natural liver regenerative microenvironment, so-called “bioactive” ECM, it has shown more efficiency than nDLS in promoting primary hepatocyte survival and antithrombotic activity. Notably, when recellularized the rDLS with intrahepatic stem/progenitor cells and cultured them in 3D environment, a more likely liver organ was formatted as compared to the nDLS recellularized with the same stem/progenitor cells, after transplanted into recipients [42]. This pioneer study demonstrated that “bioactive” scaffolds of the rDLS obtained from a regenerative liver possess an advanced natural “active state niche” as compared to nDLS (“still state niche”) for promoting primary hepatocyte survival, resistance to thrombosis, and liver-like organ construction. Other forms of bioactive factors are also involved in liver tissue engineering, like microRNAs, etc. [43, 44]. Furthermore, it needs to be mentioned that the advantage of highly conserved each specific ECM protein of decellularized scaffold among species of which the ECM are recognizable within and between species largely without immune rejection [45, 46] when properly processed to remove cellular antigens that would induce an immune rejection without damaging the ECM.

Figure 2.

Generation of a porcine decellularized liver scaffold from naïve livers and livers that had undergone partial hepatectomy (PHx). (A) Perfusion procedure for liver organ decellularization. (B) Blood-vessel tree of a decellularized scaffold from a naïve liver (nDLS). (C) Blood vessel tree of a decellularized scaffold from a partial hepatectomy (PHx) liver (rDLS).

Advertisement

3. Seed cells response to the natural three-dimensional-decellularized liver biomaterial scaffold

Cellular components are an integral part of any tissue engineering. In the case of the liver, it is important to find appropriate cells, such as hepatocytes or stem cells and to seed them into biomaterial scaffolds to regenerate liver tissues or organs [47]. Appropriate seed cells contain parenchymal such as hepatocytes, cholangiocytes, and supportive cells like liver sinusoidal endothelial cells, Kupffer cells, hepatic stellate cells, and pit cells [48]. Hepatocytes account for 60–65% of a liver’s cell population [1, 2, 49], which is important for liver tissue engineering. If it is difficult to obtain patient-derived hepatocytes, along with challenging isolation, culture, and the low yields of these cells in vitro [50], stem cells are required for liver tissue engineering [51, 52].

Stem cells are generally grouped as embryonic stem cells (ESCs), somatic stem cells (SSCs), and inducible pluripotent stem cells (iPS) [53]. ESCs have a higher regenerative capacity and can be manipulated to differentiate into other cell types [54, 55]. For liver tissue engineering, ESCs are considered beneficial for the purpose of cell differentiation. For instance, epithelial cells differentiate from ESCs, which could cover the interior of vessels (arteries, veins, and capillaries) of DLS, and the interior of vessels is one of the major players of the angiogenesis process in physiological and pathological conditions involved in thrombus resistant effects. Due to the ethical problems with ESCs, tetratomics and expanded adult human hepatocytes [56], iPS are described as an alternative for adult human hepatocyte differentiation [57, 58, 59, 60, 61]. More studies about iPS are under active investigation at present [62], but dozens of publications regarding iPS-derived hepatic lineages have varied from report to report, making it difficult to compare the relative successes of the various modified protocols in enhancing hepatocyte differentiation [63, 64]. Moreover, cultured human hepatocytes often upregulate inappropriate immature markers, such as alpha-fetal protein (AFP). Consequently, any comparisons made to these altered adult hepatocytes may make the candidate immaturely appear more strongly functional than they truly are. Indeed, an examination of published accounts reveals that many protocols lead to fetal hepatocyte-like cells, but in some cases, the characterization reported is not sufficient to determine the fetal versus mature nature of the resulting differentiated hepatic cells. Given the seemingly fetal nature of iPS-derived hepatic cells produced to date, it is apparent that additional, careful modification of differentiation protocols is still required for further investigation before clinical implementation. Somatic stem cells could overcome the obstacles caused by ECSs, thereby making them more appropriate for liver tissue engineering [65, 66].

SSCs are composed of intrahepatic SSCs and extrahepatic SSCs. Bone marrow-, umbilical-, and fat tissue-derived mesenchymal stem cells are well accepted extrahepatic SSCs [67, 68, 69], while oval cells, especially neuro-glial antigen 2 (Ng2)-expressing cells (Ng2+HSP), are currently identified as intrahepatic stem/progenitor cells. Isolation of the Ng2+HSP should be completed by using a specific protocol [70]. Other sources of SSC behaviors seeded in the DLS have also influenced liver tissue engineering. Several studies have demonstrated that liver-derived mesenchymal stem cell (MSC)-like cells can differentiate into hepatocytes and cholangiocytes in nDLS and that the functional differentiation of MSCs in certain situations could be an alternative approach for an engineered liver organ transplantation in the treatment or replacement of ESLD [35, 71]. Our recently studied animal models have revealed that intrahepatic MSC-like SSCs repaired injured livers better than extrahepatic MSCs [unpublished]. Contrary to past hypotheses, extrahepatic bone marrow-derived MSCs do not seem to directly differentiate themselves into hepatocytes, in particularly in vivo, compared to local (liver) MSC-like cells, such as above mentioned the Ng2+HSP. As the Ng2+HSP has been demonstrated to have a role in tissue repair [70] and failed liver replacement [35] in liver cirrhosis murine model, we recently further demonstrated that the intrahepatic Ng2+HSP cells are capable of more efficiency than extrahepatic BM-MSCs in self-renewal and hepatocyte and cholangiocyte differentiations (unpublished) (Figure 3). Interestingly, by using the Ng2+HSP, Zhang et al. have successfully reconstituted a liver construct in vitro that is very similar to a naïve liver organ [35]. In addition, the immuno-modulatory, anti-inflammatory, antiapoptotic, and angiogenic properties of the intrahepatic MSC-like Ng2+HSP in the liver still need to be further investigated for liver tissue engineering.

Figure 3.

Murine intrahepatic and extrahepatic mesenchymal stem cells (MSCs). (A) Cultured and immunofluorescently stained of intrahepatic neuro-glial antigen 2 (Ng2)-expressing mesenchymal stem cell (MSC)-like stem/progenitor cells (Ng2+HSP). (B) Cultured and immunofluorescently stained identical bone marrow (BM)-derived-MSCs (BM-MSCs), as visualized by optical microscopy, scale bar = 100 μM.

Advertisement

4. Decellularization/recellularization strategy-based liver construction

With the development of decellularization approaches, such as the detergent perfusion technique, whole decellularized scaffolds from liver organs have been produced DLS with an ECM structure and bioactive components being used fabricate bioengineered liver tissues, thus serving as a platform for liver organ bioengineering. Within the past several decades, numerous accomplishments have been driven by the development of these construction strategies. To date, decellularization-based liver construction strategies are constantly advancing such as maintaining complete hepatic vessel networks [72].

Despite the well-conserved macroscopic structure of a liver organ obtained by using decellularization, it is still difficult to avoid some disruption to the ECM composition and ultrastructure through decellularization, which leads to impairment of the natural 3D microenvironment, for example, an impairment of glycosaminoglycans within the ECM by enzymes [73] can cause altered stiffness. Therefore, improved measures for preserving the integrity of the ECM during the decellularization process are required [74, 75]. An functional engineered liver tissue usually uses stem cells or progenitor cells that need to differentiate into multiple kinds of repair cells, which is a challenge to directly seed cells to colonize in relevant sites of DLS to induce their differentiation into specific cell types. Whether an engineering formed liver organ can successfully fulfill its functions depends not only on its physically decellularized scaffold structure but also on an effective recellularization. Therefore, how to populate seed cells like differentiated hepatocytes from different kinds of seed cells or stem cells themselves onto the DLS needs to be carefully considered. In particular, how to manipulate the DLS to enhance the targeted specific colonization of cells to specific areas of DLS such as perfused endothelial cells [76, 77, 78], has drawn much attention. To ensure the long-term survival of an engineered liver by allowing exchanges of oxygen, nutrients, and disposal of metabolic waste [79], a functional vascular network and thrombosis after transplantation also needs to be considered. Despite the conservation of the general vascular structure by DLS, the formation of a functional vascular network remains a challenge for liver organ construction. The mainstream strategy to fabricate an engineered liver organ with a functional vascular network includes also the procedure of prevascularization. The initial approaches have been successfully used in spontaneous lineage of endothelial cells in DLS vascular networks after recellularization with stem cells [80, 81, 82] to challenge thrombosis after transplantation when exposed to blood, thus leading to localized organ failures [83]. There are two nice approaches showed that endothelialization of vasculature and immobilization of heparin on nDLS could reduce its incidence of thrombosis [84]. More recently, from a pretreated naïve liver obtained rDLS, exhibited except for strong promoting primary hepatocyte survival but also antithrombosis more effect [biomaterials 2018]. Notably, after transplantation guiding the rDLS/cells complex forms complex liver-like tissues (geometries) more effective on rDLS than on nDLS (Figure 4), meanwhile combined with better organization of endothelial lineage in rDLS than in nDLS [42]. This suggests that rDLS possesses an advanced “bioactive natural regeneration state niche” relative to the nDLS, which preserves a “still state niche.” Therefore, the spontaneous manipulation of the ECM on DLS is a more promising strategy for decellularization-based liver tissue. In the future, the objective of a decellularization-based liver construction strategy could be based on generating a 3D decellularized biomaterial scaffold with natural “regenerative bioactive niche” for the seed cell attachment, proliferation and differentiation of cells, and developing a transplantable “new” liver in vitro that maintains the structures and functions of a naïve liver.

Figure 4.

Comparison of the murine liver-lobule-like tissue construction formation between rDLS and nDLS after portal-renal arterialized auxiliary heterotopic liver transplantation. (A) Schematic of the procedure. The left green cycle indicates the DLS, and the right green cycle indicates the end-to-end anastomosis of the PV (scaffold)-L-RA (recipient). The green arrows in the panels indicate the right-RA. The right bottom cartoon shows the end-to-side anastomosis of the IVC (scaffold)-IVC (recipient). (Ba–d) Exposure of the right-side kidney (the square indicates the kidney) (a). Nephrectomy of the right-side kidney (the square indicates the lack of kidney) (b). The cell-loaded DLS where the kidney was removed (the bold arrow indicates the PV, thin arrow indicates the IVC, and the green arrow indicates a right renal artery (right-RA)). The left-side renal artery (L-RA) was connected to the PV with cross-clamping of the PV and the IVC of the recellularized scaffold (c). The noncell loaded DLS was connected to the recipient by the same procedure as the cell-loaded DLS where the kidney was removed (d). (Ca–c) DLS seeded with Ng2+HSP cells formed a liver-lobule-like construct in rDLS (a and b) after approximately 20–40 days (a, indicated as a cycle), for two lobes with better blood patency (b), represented with a white arrow; there was no visible blood flow in the nDLS loaded with Ng2+HSP cells for the same time (c). (D) Blood flow velocity (flow, arbitrary unit, AU) was measured in rDLS and nDLS at 45 min within 100 s after the operation by a near-infrared-LDF system, scale bar = 50 μM.

In summary, compared with other strategies that can only fabricate partial structures, a decellularization/recellularization-based liver tissue engineering strategy enables the construction of the liver structures with complete blood vessel network at a clinically relevant scale, thus becoming a more promising approach for liver tissue engineering. However, in order to provide a promising route for developing a functional bioartificial liver with potential applications for humans by such strategy, several questions must be answered: (1) Is the use of a decellularized liver matrix the only possible solution? (2) What kinds of cells need to be chosen for recellularization? Extrahepatic cells? or possibly resident stem/progenitors cells? (3) What is the optimal decellularized liver scaffold (DLS)? (4) What is the length of time for incubation in a bioreactor? (5) Would the technique be applicable to a human liver with its extensively sinusoidal surface?

Advertisement

5. Conclusion and challenges

Clearly, decellularization/recellularization through the development of in vitro and in vivo tissue and organ models for liver bioengineering are advancing strategies. This, combined with multidisciplinary team-workers performing focused, systematic studies to address critical questions, is essential for the success of this strategy. The following critical issues might need to be addressed before clinical applications: (1) preservation and modification of a functional ECM structure to better mimic the regenerative niche; (2) selection of effective seed cell sources for recellularization; (3) modification of blood-vessel networks for “endothelialized DLS”; (4) long-term survival by preventing from thrombosis and functions after transplantation; and (5) immune rejection. In the coming years, many new techniques will be explored, which are expected to have the potential to address these challenges.

Advertisement

Funding information

This work was funded by the National Natural Science Foundation of China (NNSFC), Grant/Award Numbers: 81570573 and 81873586; the Army Medical University, Grant/Award Numbers: 2018XLC2009; and the Army Medical University-affiliated Southwest Hospital, Grant/Award Numbers: SWH2017ZYLX-03.

Advertisement

Abbreviations

ESLDend-stage liver disease
ECMextracellular matrix
3Dthree-dimensional
OLTorthotopic liver transplant
DLSdecellularized liver scaffold
nDLSthe decellularized scaffold that generated from naïve liver
rDLSthe decellularized scaffold that generated from pretreated liver
HGFhepatic growth factor
TGF-αtransforming growth factor-alpha
IL-6interleukin 6
b-FGFfibroblast growth factor-beta
VEGFvascular endothelial growth factor
ESCsembryonic stem cells
SSCssomatic stem cells
iPSinducible pluripotent stem cells
Ng2+HSPneuro-glial antigen 2 (Ng2)-expressing cells
MSCmesenchymal stem cell

References

  1. 1. Tanaka M, Iwakiri Y. Lymphatics in the liver. Current Opinion in Immunology. 2018;53:137-142
  2. 2. Nahmias Y, Berthiaume F, Yarmush ML. Integration of technologies for hepatic tissue engineering. Advances in Biochemical Engineering/Biotechnology. 2007;103:309-329
  3. 3. Eslam M, George J. Targeting IFN-lambda: Therapeutic implications. Expert Opinion on Therapeutic Targets. 2016;20:1425-1432
  4. 4. Shafiee A, Atala A. Tissue engineering: Toward a new era of medicine. Annual Review of Medicine. 2017;68:29-40
  5. 5. Atala A. A new era in stem cells translational medicine. Stem Cells Translational Medicine. 2012;1:1-2
  6. 6. Soto-Gutierrez A, Wertheim JA, Ott HC, Gilbert TW. Perspectives on whole-organ assembly: Moving toward transplantation on demand. The Journal of Clinical Investigation. 2012;122:3817-3823
  7. 7. Berthiaume F, Maguire TJ, Yarmush ML. Tissue engineering and regenerative medicine: History, progress, and challenges. Annual Review of Chemical and Biomolecular Engineering. 2011;2:403-430
  8. 8. 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 on a Chip. 2015;15:3822-3837
  9. 9. Lv T, Liang W, Li L, Cui X, Wei X, Pan H, et al. Novel calcitonin gene-related peptide/chitosan-strontium-calcium phosphate cement: Enhanced proliferation of human umbilical vein endothelial cells in vitro. Journal of Biomedical Materials Research. Part B, Applied Biomaterials. 2019;107:19-28
  10. 10. Brown JH, Das P, DiVito MD, Ivancic D, Tan LP, Wertheim JA. Nanofibrous PLGA electrospun scaffolds modified with type I collagen influence hepatocyte function and support viability in vitro. Acta Biomaterialia. 2018;73:217-227
  11. 11. Tandon V, Zhang B, Radisic M, Murthy SK. Generation of tissue constructs for cardiovascular regenerative medicine: From cell procurement to scaffold design. Biotechnology Advances. 2013;31:722-735
  12. 12. Rowley AT, Nagalla RR, Wang SW, Liu WF. Extracellular matrix-based strategies for immunomodulatory biomaterials engineering. Advanced Healthcare Materials. 2019;8:e1801578
  13. 13. Perez RA, Won JE, Knowles JC, Kim HW. Naturally and synthetic smart composite biomaterials for tissue regeneration. Advanced Drug Delivery Reviews. 2013;65:471-496
  14. 14. Atala A, Kasper FK, Mikos AG. Engineering complex tissues. Science Translational Medicine. 2012;4:112r-160r
  15. 15. Lakshmanan R, Kumaraswamy P, Krishnan UM, Sethuraman S. Engineering a growth factor embedded nanofiber matrix niche to promote vascularization for functional cardiac regeneration. Biomaterials. 2016;97:176-195
  16. 16. Zawaneh PN, Singh SP, Padera RF, Henderson PW, Spector JA, Putnam D. Design of an injectable synthetic and biodegradable surgical biomaterial. Proceedings of the National Academy of Sciences of the United States of America. 2010;107:11014-11019
  17. 17. Huling J, Ko IK, Atala A, Yoo JJ. Fabrication of biomimetic vascular scaffolds for 3D tissue constructs using vascular corrosion casts. Acta Biomaterialia. 2016;32:190-197
  18. 18. Shamay Y, Shah J, Isik M, Mizrachi A, Leibold J, Tschaharganeh DF, et al. Quantitative self-assembly prediction yields targeted nanomedicines. Nature Materials. 2018;17:361-368
  19. 19. Bacakova L, Novotna K, Parizek M. Polysaccharides as cell carriers for tissue engineering: The use of cellulose in vascular wall reconstruction. Physiological Research. 2014;63(Suppl 1):S29-S47
  20. 20. Rustad KC, Sorkin M, Levi B, Longaker MT, Gurtner GC. Strategies for organ level tissue engineering. Organogenesis. 2010;6:151-157
  21. 21. Mano JF, Silva GA, Azevedo HS, Malafaya PB, Sousa RA, Silva SS, et al. Natural origin biodegradable systems in tissue engineering and regenerative medicine: Present status and some moving trends. Journal of the Royal Society Interface. 2007;4:999-1030
  22. 22. Rodríguez-Vázquez M, Vega-Ruiz B, Ramos-Zúñiga R, Saldaña-Koppel DA, Quiñones-Olvera LF. Chitosan and its potential use as a scaffold for tissue engineering in regenerative medicine. BioMed Research International. 2015;2015:821279
  23. 23. Patel NM, Yazdi IK, Tasciotti E, Birla RK. Optimizing cell seeding and retention in a three-dimensional bioengineered cardiac ventricle: The two-stage cellularization model. Biotechnology and Bioengineering. 2016;113:2275-2285
  24. 24. Mogosanu GD, Grumezescu AM. Natural and synthetic polymers for wounds and burns dressing. International Journal of Pharmaceutics. 2014;463:127-136
  25. 25. Lee J, Yoo JJ, Atala A, Lee SJ. Controlled heparin conjugation on electrospun poly(epsilon-caprolactone)/gelatin fibers for morphology-dependent protein delivery and enhanced cellular affinity. Acta Biomaterialia. 2012;8:2549-2558
  26. 26. Arenas-Herrera JE, Ko IK, Atala A, Yoo JJ. Decellularization for whole organ bioengineering. Biomedical Materials. 2013;8:14106
  27. 27. Ott HC, Matthiesen TS, Goh SK, Black LD, Kren SM, Netoff TI, et al. Perfusion-decellularized matrix: Using nature’s platform to engineer a bioartificial heart. Nature Medicine. 2008;14:213-221
  28. 28. Uygun BE, Soto-Gutierrez A, Yagi H, Izamis ML, Guzzardi MA, Shulman C, et al. Organ reengineering through development of a transplantable recellularized liver graft using decellularized liver matrix. Nature Medicine. 2010;16:814-820
  29. 29. Li Q , Uygun BE, Geerts S, Ozer S, Scalf M, Gilpin SE, et al. Proteomic analysis of naturally-sourced biological scaffolds. Biomaterials. 2016;75:37-46
  30. 30. Ciccocioppo R, Dos SC, Baumgart DC, Cangemi GC, Cardinale V, Ciacci C, et al. Proceedings of the signature series event of the international society for cellular therapy: “Advancements in cellular therapies and regenerative medicine in digestive diseases,” London, United Kingdom, May 3, 2017. Cytotherapy. 2018;20:461-476
  31. 31. Mattei G, Magliaro C, Pirone A, Ahluwalia A. Decellularized human liver is too heterogeneous for designing a generic extracellular matrix mimic hepatic scaffold. Artificial Organs. 2017;41:E347-E355
  32. 32. Orlando G, Booth C, Wang Z, Totonelli G, Ross CL, Moran E, et al. Discarded human kidneys as a source of ECM scaffold for kidney regeneration technologies. Biomaterials. 2013;34:5915-5925
  33. 33. Nelson CM, Bissell MJ. Of extracellular matrix, scaffolds, and signaling: Tissue architecture regulates development, homeostasis, and cancer. Annual Review of Cell and Developmental Biology. 2006;22:287-309
  34. 34. Batchelder CA, Martinez ML, Tarantal AF. Natural scaffolds for renal differentiation of human embryonic stem cells for kidney tissue engineering. PLoS One. 2015;10:e143849
  35. 35. Zhang H, Siegel CT, Li J, Lai J, Shuai L, Lai X, et al. Functional liver tissue engineering by an adult mouse liver-derived neuro-glia antigen 2-expressing stem/progenitor population. Journal of Tissue Engineering and Regenerative Medicine. 2018;12:e190-e202
  36. 36. Sellaro TL, Ranade A, Faulk DM, McCabe GP, Dorko K, Badylak SF, et al. Maintenance of human hepatocyte function in vitro by liver-derived extracellular matrix gels. Tissue Engineering. Part A. 2010;16:1075-1082
  37. 37. Mussbach F, Settmacher U, Dirsch O, Xie C, Dahmen U. Bioengineered livers: A new tool for drug testing and a promising solution to meet the growing demand for donor organs. European Surgical Research. 2016;57:224-239
  38. 38. Rana D, Zreiqat H, Benkirane-Jessel N, Ramakrishna S, Ramalingam M. Development of decellularized scaffolds for stem cell-driven tissue engineering. Journal of Tissue Engineering and Regenerative Medicine. 2017;11:942-965
  39. 39. Mussbach F, Dahmen U, Dirsch O, Settmacher U. Liver engineering as a new source of donor organs: A systematic review. Chirurg. 2016;87:504-513
  40. 40. Badylak SF, Taylor D, Uygun K. Whole-organ tissue engineering: Decellularization and recellularization of three-dimensional matrix scaffolds. Annual Review of Biomedical Engineering. 2011;13:27-53
  41. 41. Macri L, Silverstein D, Clark RA. Growth factor binding to the pericellular matrix and its importance in tissue engineering. Advanced Drug Delivery Reviews. 2007;59:1366-1381
  42. 42. Yang W, Chen Q , Xia R, Zhang Y, Shuai L, Lai J, et al. A novel bioscaffold with naturally-occurring extracellular matrix promotes hepatocyte survival and vessel patency in mouse models of heterologous transplantation. Biomaterials. 2018;177:52-66
  43. 43. Dong X, Pan R, Zhang H, Yang C, Shao J, Xiang L. Modification of histone acetylation facilitates hepatic differentiation of human bone marrow mesenchymal stem cells. PLoS One. 2013;8:e63405
  44. 44. Cao B, He G, Yang H, Chang H, Li S, Deng A. Development of a highly sensitive and specific enzyme-linked immunosorbent assay (ELISA) for the detection of phenylethanolamine A in tissue and feed samples and confirmed by liquid chromatography tandem mass spectrometry (LC-MS/MS). Talanta. 2013;115:624-630
  45. 45. Moser PT, Ott HC. Recellularization of organs: What is the future for solid organ transplantation? Current Opinion in Organ Transplantation. 2014;19:603-609
  46. 46. Farre R, Otero J, Almendros I, Navajas D. Bioengineered lungs: A challenge and an opportunity. Archivos de Bronconeumología. 2018;54:31-38
  47. 47. Murphy SV, Atala A. Organ engineering—Combining stem cells, biomaterials, and bioreactors to produce bioengineered organs for transplantation. BioEssays. 2013;35:163-172
  48. 48. Uygun BE, Yarmush ML. Engineered liver for transplantation. Current Opinion in Biotechnology. 2013;24:893-899
  49. 49. Jain E, Damania A, Kumar A. Biomaterials for liver tissue engineering. Hepatology International. 2014;8:185-197
  50. 50. Kaserman JE, Wilson AA. Patient-derived induced pluripotent stem cells for alpha-1 antitrypsin deficiency disease modeling and therapeutic discovery. Chronic Obstructive Pulmonary Disease. 2018;5:258-266
  51. 51. Zhang L, Ye JS, Decot V, Stoltz JF, de Isla N. Research on stem cells as candidates to be differentiated into hepatocytes. Bio-medical Materials and Engineering. 2012;22:105-111
  52. 52. Zhang L, Zhao YH, Guan Z, Ye JS, de Isla N, Stoltz JF. Application potential of mesenchymal stem cells derived from Wharton’s jelly in liver tissue engineering. Bio-medical Materials and Engineering. 2015;25:137-143
  53. 53. Ren X, Ott HC. On the road to bioartificial organs. Pflügers Archiv. 2014;466:1847-1857
  54. 54. Radhakrishnan S, Trentz OA, Martin CA, Reddy MS, Rela M, Chinnarasu M, et al. Effect of passaging on the stemness of infrapatellar fat padderived stem cells and potential role of nucleostemin as a prognostic marker of impaired stemness. Molecular Medicine Reports. 2019;20:813-829
  55. 55. Lorzadeh N, Kazemirad N. Embryonic stem cells and infertility. American Journal of Perinatology. 2018;35:925-930
  56. 56. Yu Y, Fisher JE, Lillegard JB, Rodysill B, Amiot B, Nyberg SL. Cell therapies for liver diseases. Liver Transplantation. 2012;18:9-21
  57. 57. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663-676
  58. 58. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861-872
  59. 59. Touboul T, Hannan NR, Corbineau S, Martinez A, Martinet C, Branchereau S, et al. Generation of functional hepatocytes from human embryonic stem cells under chemically defined conditions that recapitulate liver development. Hepatology. 2010;51:1754-1765
  60. 60. Sharma NS, Wallenstein EJ, Novik E, Maguire T, Schloss R, Yarmush ML. Enrichment of hepatocyte-like cells with upregulated metabolic and differentiated function derived from embryonic stem cells using S-NitrosoAcetylPenicillamine. Tissue Engineering. Part C, Methods. 2009;15:297-306
  61. 61. Moore RN, Dasgupta A, Rajaei N, Yarmush ML, Toner M, Larue L, et al. Enhanced differentiation of embryonic stem cells using co-cultivation with hepatocytes. Biotechnology and Bioengineering. 2008;101:1332-1343
  62. 62. Espejel S, Roll GR, McLaughlin KJ, Lee AY, Zhang JY, Laird DJ, et al. Induced pluripotent stem cell-derived hepatocytes have the functional and proliferative capabilities needed for liver regeneration in mice. The Journal of Clinical Investigation. 2010;120:3120-3126
  63. 63. Hirata M, Yamaoka T. Hepatocytic differentiation of iPS cells on decellularized liver tissue. Journal of Artificial Organs. 2017;20:318-325
  64. 64. Schepers A, Li C, Chhabra A, Seney BT, Bhatia S. Engineering a perfusable 3D human liver platform from iPS cells. Lab on a Chip. 2016;16:2644-2653
  65. 65. Grath A, Dai G. Direct cell reprogramming for tissue engineering and regenerative medicine. Journal of Biological Engineering. 2019;13:14
  66. 66. Kumar SA, Delgado M, Mendez VE, Joddar B. Applications of stem cells and bioprinting for potential treatment of diabetes. World Journal of Stem Cells. 2019;11:13-32
  67. 67. Klimczak A, Kozlowska U. Mesenchymal stromal cells and tissue-specific progenitor cells: Their role in tissue homeostasis. Stem Cells International. 2016;2016:4285215
  68. 68. Verstegen M, Willemse J, van den Hoek S, Kremers GJ, Luider TM, van Huizen NA, et al. Decellularization of whole human liver grafts using controlled perfusion for transplantable organ bioscaffolds. Stem Cells and Development. 2017;26:1304-1315
  69. 69. Coronado RE, Somaraki-Cormier M, Ong JL, Halff GA. Hepatocyte-like cells derived from human amniotic epithelial, bone marrow, and adipose stromal cells display enhanced functionality when cultured on decellularized liver substrate. Stem Cell Research. 2019;38:101471
  70. 70. Zhang H, Siegel CT, Shuai L, Lai J, Zeng L, Zhang Y, et al. Repair of liver mediated by adult mouse liver neuro-glia antigen 2-positive progenitor cell transplantation in a mouse model of cirrhosis. Scientific Reports. 2016;6:21783
  71. 71. Martin C, Olmos É, Collignon M, De Isla N, Blanchard F, Chevalot I, et al. Revisiting MSC expansion from critical quality attributes to critical culture process parameters. Process Biochemistry. 2017;59:231-243
  72. 72. Shirakigawa N, Ijima H. Decellularization of liver and organogenesis in rats. Methods in Molecular Biology. 2018;1577:271-281
  73. 73. He M, Callanan A. Comparison of methods for whole-organ decellularization in tissue engineering of bioartificial organs. Tissue Engineering. Part B, Reviews. 2013;19:194-208
  74. 74. Struecker B, Hillebrandt KH, Voitl R, Butter A, Schmuck RB, Reutzel-Selke A, et al. Porcine liver decellularization under oscillating pressure conditions: A technical refinement to improve the homogeneity of the decellularization process. Tissue Engineering. Part C, Methods. 2015;21:303-313
  75. 75. Manso AM, Okada H, Sakamoto FM, Moreno E, Monkley SJ, Li R, et al. Loss of mouse cardiomyocyte talin-1 and talin-2 leads to beta-1 integrin reduction, costameric instability, and dilated cardiomyopathy. Proceedings of the National Academy of Sciences of the United States of America. 2017;114:E6250-E6259
  76. 76. Shirakigawa N, Ijima H, Takei T. Decellularized liver as a practical scaffold with a vascular network template for liver tissue engineering. Journal of Bioscience and Bioengineering. 2012;114:546-551
  77. 77. Devalliere J, Chen Y, Dooley K, Yarmush ML, Uygun BE. Improving functional re-endothelialization of acellular liver scaffold using REDV cell-binding domain. Acta Biomaterialia. 2018;78:151-164
  78. 78. Agarwal T, Maiti TK, Ghosh SK. Decellularized caprine liver-derived biomimetic and pro-angiogenic scaffolds for liver tissue engineering. Materials Science & Engineering. C, Materials for Biological Applications. 2019;98:939-948
  79. 79. Sun Y, Liu Y, Li S, Liu C, Hu Q. Novel compound-forming technology using bioprinting and electrospinning for patterning a 3D scaffold construct with multiscale channels. Micromachines (Basel). 2016;7:238-256
  80. 80. Unger RE, Sartoris A, Peters K, Motta A, Migliaresi C, Kunkel M, et al. Tissue-like self-assembly in cocultures of endothelial cells and osteoblasts and the formation of microcapillary-like structures on three-dimensional porous biomaterials. Biomaterials. 2007;28:3965-3976
  81. 81. Unger RE, Dohle E, Kirkpatrick CJ. Improving vascularization of engineered bone through the generation of pro-angiogenic effects in co-culture systems. Advanced Drug Delivery Reviews. 2015;94:116-125
  82. 82. Ko IK, Peng L, Peloso A, Smith CJ, Dhal A, Deegan DB, et al. Bioengineered transplantable porcine livers with re-endothelialized vasculature. Biomaterials. 2015;40:72-79
  83. 83. Watanabe M, Yano K, Okawa K, Yamashita T, Tajima K, Sawada K, et al. Construction of sinusoid-scale microvessels in perfusion culture of a decellularized liver. Acta Biomaterialia. 2019;95:307-318
  84. 84. Hussein KH, Park KM, Kang KS, Woo HM. Heparin-gelatin mixture improves vascular reconstruction efficiency and hepatic function in bioengineered livers. Acta Biomaterialia. 2016;38:82-93

Written By

Quanyu Chen, Xiaolin You, Jiejuan Lai, Shifang Jiang, Hongyu Zhang and Lianhua Bai

Submitted: 11 August 2019 Reviewed: 03 September 2019 Published: 27 November 2019