3,701
Views
24
CrossRef citations to date
0
Altmetric
Review

Bioengineering for Organ Transplantation: Progress and Challenges

, , &
Pages 257-261 | Received 01 Jul 2015, Accepted 03 Aug 2015, Published online: 26 Aug 2015

Abstract

Organ transplantation can offer a curative option for patients with end stage organ failure. Unfortunately the treatment is severely limited by the availability of donor organs. Organ bioengineering could provide a solution to the worldwide critical organ shortage. The majority of protocols to date have employed the use of decellularization-recellularization technology of naturally occurring tissues and organs with promising results in heart, lung, liver, pancreas, intestine and kidney engineering. Successful decellularization has provided researchers with suitable scaffolds to attempt cell reseeding. Future work will need to focus on the optimization of organ specific recellularization techniques before organ bioengineering can become clinically translatable. This review will examine the current progress in organ bioengineering and highlight future challenges in the field.

Abbreviations

ACL=

anterior cruciate ligament

CYP 450=

cytochrome P 450

DRT=

decellularization-recellularization technology

ECM=

extracellular matrix

ESC=

embryonic stem cell

ESRD=

end stage renal disease

HUVEC=

human umbilical vein endothelial cell

iHIOs=

induced human intestinal organoids

iMPC=

induced multipotent progenitor cell

iPSC=

induced pluripotent stem cell

MEMS=

microelectromechanical systems

MSC=

mesenchymal stem cell

RAD=

renal assist device

SIS=

small intestine submucosa

SNM=

silicon nanopore membranes

VCA=

vascularized composite allografts.

Introduction

As of May 2015, over 123,000 people were on the waiting list for an organ in the USA alone.Citation1 As a result of this critical organ shortage, 6,885 patients died in the USA while awaiting an organ transplant in 2014.Citation1 Bioengineered organs could provide an inexhaustible organ source and carry the potential benefit of requiring an immunosuppression-free state.Citation2,3 Successful outcomes have been reported with simple, hollow organs including their production and implantation.Citation4-6 The more complex modular organs have proved a greater challenge. There have been considerable advances in this new field but a clinically relevant model still remains elusive. This article explores the achievements of bioengineering organs to date and identifies future challenges.

Decellularization-Recellularization Technology (DRT)

Decellularization

Whole organ bioengineering first requires a scaffold to allow cells to develop and function.Citation2,7 The extracellular matrix (ECM) performs this task in vivo. Current approaches to achieving a bioengineered organ construct have employed the use of a supporting structure generated by decellularization of naturally occurring human or animal tissues and organs.Citation8 In doing so, allogeneic and xenogeneic cellular antigens are removed allowing the probable elimination of immunogenicity.Citation8 Ott et al. (2008) have demonstrated a successful protocol for decellularization of a rat heart.Citation9 In doing so, the ability to generate a biocompatible organ construct entered the realms of possibility. The technique produced a “complex, biocompatible cardiac ECM scaffold with a perfusable vascular tree, patent valves and a 4-chamber-geometry template for biomimetic tissue engineering”.Citation9 Further protocols were subsequently developed but none were considered ideal for generating intact scaffolds with products either containing residual cell debris or resulting in destruction of ECM proteins.Citation10 Effective decellularization protocols have since become established and in general, include the use of “physical, ionic, chemical and enzymatic methods,” with the organ's own vascular network being used for detergent delivery.Citation11,12 The result is an intact acellular organ scaffold of ECM that retains the structure of the original organ that supports cell attachment, proliferation and integration.Citation13

Recellularization

Once a satisfactory organ scaffold has been produced, recellularization is required to achieve a functional organ product for implantation. The principles of recellularization include obtaining a renewable source of cells, cell seeding onto the scaffold and finally organ specific culture.Citation14 Significant work has already been carried out to establish effective and reproducible protocols that create functional organ products.

The first important goal is to identify an ethically approved and renewable source of cells for scaffold seeding. Fetal and adult cells, embryonic (ESC), mesenchymal (MSC) and induced pluripotent stem cells (iPSC) have all been used.Citation14 MSCs have shown promise, being easily and ethically obtained from bone marrow stroma or adipose tissue.Citation15 In addition, they have shown a good degree of differentiation, attachment and persistence on organ scaffolds.Citation16,17 iPSCs, in particular, have also shown potential. They are created via the reprogramming of human somatic cells and exhibit characteristics that resemble embryonic stem cells.Citation18 iPSCs have been efficiently differentiated into alveolar epithelial cells and myocytes but their adhesive potential to organ scaffolds has been inferior to that of MSCs.Citation19-21 Interestingly, although immature iPSCs prompt a detrimental immune response, self tolerance has been achieved with terminally differentiated iPSCs, further paving the way for their use in organ bioengineering.Citation22

Cell seeding techniques are largely organ specific.Citation14 Perfusion seeding via the vasculature is common to all organs but non-vascular routes (e.g. airway and ureter, in lung or kidney respectively) have also shown good results.Citation14 The use of multiple perfusion routes, optimization of scaffold coating, mechanical environment and rate of cell perfusion have shown promise in improving cell concentration and diffusion over the scaffold.Citation14

Successful seeding of the scaffold vasculature has been essential to the survival of a bioengineered organ.Citation14 The majority of recellularization protocols to date have used endothelial cells but more recent work has shown potential benefit with iPSCs.Citation9,20 Interestingly, the success of vasculature seeding techniques has previously been shown to be largely dependent on perfusion flow rates.Citation23

Cell culture is required after sufficient cell seeding onto an organ scaffold. Delivery of nutrients to the entire 3-dimensional organ construct requires a bioreactor for perfusion and to supply an environment that encourages cell growth in an organ specific manner.Citation14

Organ Specific Progress and Challenges

Heart

The accomplishment of attaining a viable decellularized heart scaffold was a major turning point in the pursuit of achieving a bioengineered organ. In 2008, Ott and colleagues constructed a suitable heart scaffold from a rat heart, before reseeding and maturing the matrix to develop a structure capable of contracting and responding to drugs.Citation9 This was the first protocol that produced a recellularized, functional organ scaffold.Citation9 Further studies have demonstrated the potential of decellularized heart ECM to direct differentiation of progenitor cells, notably iPSCs, into those of cardiac lineage.Citation20,24 Seeding techniques have employed the use of perfusion via the aorta and coronary arteries. However, there has been universal difficulty in establishing adequate cellularization of the ventricular wall. Direct injection attempts have resulted in dense cellularity at the injection site with poor distribution throughout the remainder.Citation9,25 The first evidence of the potential of a scaled-up model was recently reported using porcine heart scaffolds.Citation25 Successfully decellularized porcine hearts were reseeded with murine cardiomyocytes and human umbilical vein endothelial cells (HUVECs) resulting in a construct that exhibited intrinsic electrical activity.Citation25 Further work is still required to achieve a fully functional bioartificial heart of appropriate clinical scale with focus on suitability of cardiac cell sources (including the use of conspecific cells), enhancing reseeding techniques to promote sufficient dispersion, and optimization of in vitro organ culture.Citation25

Lung

Challenges in developing a biocompatible lung scaffold initially revolved around the production of a highly elastic matrix to facilitate reproduction of the complex lung architecture required to support gas exchange.Citation26 The development of a suitable scaffold followed by successful reseeding with endothelial cells and epithelial cells, was reported in 2 initial small animal studies.Citation27,28 Bioreactor systems were used to mimic the mechanical stretch that normal fetal breathing movements and circulation would subject onto the developing lung.Citation27,28 The transplanted bioengineered products were able to exchange oxygen and carbon dioxide in both studies. However, after several hours there was evidence of either significant pulmonary secretions or bleeding into the airways.Citation27,28 These protocols have subsequently been scaled up toward achieving a clinically viable product with porcine and human lungs.Citation13,29 Findings have been encouraging: both porcine and human lungs can be decellularized to produce biocompatible whole-organ scaffolds.Citation13,29 The crucial next step is to achieve successful recellularization of these clinically relevant models. iPSCs have shown particular promise as a cell source, particularly as they would facilitate patient-specific lung bioengineering.Citation30 Expression of Nkx2.1 (the earliest marker of lung-specified endoderm) has already been achieved with iPSCs, indicating the potential for differentiation into all downstream lung epithelial lineages.Citation30 Secondly, more advanced bioreactor systems will be required to allow for prolonged organ culture ex vivo.Citation30 Finally, consideration must be given to developing sufficient but non-destructive sterilization protocols before implantation in vivo.Citation30

Liver

Optimization of protocols for the perfusion decellularization of other organs led to the generation of a 3 dimensional (3D), vasculature-intact liver scaffold from small animal and porcine models.Citation31,32 The ECM composition and vasculature network was successfully retained while removing the cellular components, enabling adequate reseeding.Citation31,32 HUVECs, human fetal liver cells or primary rat hepatocytes were perfused via the portal vein and/or vena cava resulting in the development of engrafted and functional cells typical of the native liver (e.g., CYP 450 and α-fetoprotein expression, production of urea and albumin).Citation31,32 Liver bioengineering has been hindered by the struggle to achieve adequate hepatocyte proliferation once transplanted. Investigation of the suitability of iPSC derived adult hepatocytes have been disappointing in their ability to repopulate the liver.Citation33 Human fibroblasts have since been reprogrammed to form an induced multipotent progenitor cell (iMPC) and have shown promise in their ability to proliferate and successfully repopulate after transplantation.Citation33 Further investigation is required to identify whether these iMPCs can successfully repopulate a decellularized liver matrix on a clinical scale and perform sufficient native hepatocyte function.

Pancreas

Attempts at bioengineering pancreatic tissue can be classified into 3 main areas: islet encapsulation, biomaterial carriers and whole-organ bioengineering.Citation34 Islet encapsulation aims to isolate implanted islet cells from the recipient immune response by hiding the non-self antigens. Unfortunately, difficulties with biocompatibility of capsule materials, inadequate immunoisolation and poor vascularization have limited successful clinical translation.Citation34 Biomaterial carriers involve the use of bioartificial scaffolds that mimic native ECM to allow seeding of pluripotent cells and have yielded promising initial results.Citation34 The decellularization-recellularization approach has since been applied to the pancreas. Successful decellularization and recellularization of mouse pancreata with both endocrine and exocrine cells has been achieved, with cell types displaying appropriate functionality (C-peptide and amylase secretion respectively) and differentiating in their appropriate locations.Citation35 It was noted that insulin gene expression was greater in cells on the 3D scaffolds compared with 2D giving further evidence for possible benefits of whole organ bioengineering.Citation35 Future directions will need to establish DRT protocols on larger scaffolds (porcine or human) and investigate methods to protect the bioengineered organ in vivo from the recipient autoimmune response in type 1 diabetic patients.

Intestine

Intestinal bioengineering remains in the very early stages of development. ECM scaffolds constructed from small intestine submucosa (SIS) have previously been implanted into dogs and have demonstrated development of tissue architecture comparable to native bowel.Citation36 These tissues failed to demonstrate architectural organization or develop any enteric neurons thereby significantly limiting their function in vivo.Citation36 The development of induced human intestinal organoids (iHIOs) has become a promising avenue for intestinal bioengineering as they contain enterocytes, enteroendocrine cells, goblet cells, Paneth and mesenchymal cells.Citation37 These organoid units have been successfully incorporated into synthetic scaffolds of both small intestine and colon, and implanted into small animal models after resection with basic function intact.Citation38,39 In addition, intestinal smooth muscle has been effectively engineered via fibroblast seeding of scaffolds and the use of growth factor.Citation40 More recently, researchers have successfully decellularized small intestine segments and performed recellularization with bone marrow derived stem cells.Citation41 Their work resulted in an intact mucosa, villi, crypts, blood vessels and abundant smooth muscle cells in the muscularis.Citation41 Future work will need to address the requirement for a neuromuscular layer in order that the bioengineered graft can function in vivo.Citation41

Kidney

Several strategies have been employed in the development of a bioartificial kidney. Similarly to the other organs previously discussed, recellularization of decellularized scaffolds has shown promise. Decellularization of kidney tissue slices and whole organs has been documented with maintenance of ECM properties and characteristics.Citation42,43 Song et al. (2013) decellularized rat, porcine and human kidneys creating intact vascular, glomerular and tubular compartments.Citation44 They went onto achieve scaffold repopulation with cells that were capable of producing urine in vitro and in vivo.Citation44 Interestingly, they observed site-specific adhesion and development of polarity during cell seeding.Citation44 Glomerular filtration, glucose and electrolyte absorption, and macromolecular sieving was noted but found to be functionally immature when compared to cadaveric kidneys.Citation44 To facilitate translation toward clinical use, further work is necessary to optimize seeding regimens and organ culture in human-sized scaffolds.Citation44

Other approaches have investigated the possibility of creating an ex vivo bioartificial device. Current haemodialysis techniques in ESRD focus on solute clearance but fail to address the metabolic, endocrine and transport functions physiologically provided by a normal kidney. The Renal Assist Device (RAD) was therefore developed by Humes et al. to address these shortcomings via cell seeding onto the inner surface of the hollow fibers used for microfiltration in a hemofilter.Citation45 It was noted that the technique provided metabolic, endocrine and active transport functions in addition to solute transport and was associated with a significant mortality benefit at 28 d compared with standard haemofiltration in humans.Citation46 Further efforts have investigated the feasibility of producing a device on a scale suitable for implantation by engaging the use of microelectromechanical systems (MEMS) technology and silicon nanopore membranes (SNM).Citation47 Similarly to a natural kidney, this technology can be thought of as having 2 main functional units. Firstly, the filtration unit, containing SNM that is able to function like a physiological glomerulus by filtering substances depending on their molecular weight.Citation47 Secondly, the reabsorption unit, where renal epithelial cells are seeded on a SNM scaffold and facilitate the reabsorption of water and solutes in addition to providing the metabolic and endocrine renal functions.Citation47 Despite the progress, significant optimization of ex vivo bioartificial systems are required before the development of a clinically viable device.

Composite tissue bioengineering

The bioengineering of composite tissue grafts has become an avenue of interest, specifically to orthopaedic and plastic surgeons. Trauma to the hard and soft tissues often results in damage to multiple different tissue types. The construction of interfaces between musculoskeletal tissues and the integration of sufficient vascularity are complex tasks that hold the key to achieving functional integration.Citation48 Similarly to whole organ bioengineering, work has focused on the production of scaffolds in order to provide structural support and proliferation cues to seeded cells by mimicking the role of the natural ECM.Citation48

The development of a tri-phasic scaffold by Spalazzi et al. has shown promise in the development of a ligament-bone interface (anterior cruciate ligament (ACL) to bone).Citation49 The scaffold consists of 3 different, but continuous phases that promote the formation of the distinct tissue regions required: fibroblast and soft tissue, fibrochondrocyte, and bone formation.Citation50 These stratified scaffolds have demonstrated the ability to promote formation of the necessary fibrocartilage interfaces in ACL reconstruction grafts.Citation50 They have also been successfully used to create both cartilage and bone on a single scaffold with subsequent steps made toward achieving an osteochondral interface.Citation50 Tendon-bone interfaces have employed the use of biomimetic ECM nanofiber scaffolds and have shown potential, partly secondary to the ability to modulate cell development by adjusting nanofiber alignment.Citation50

The recent success of clinical vascularized composite allografts (VCAs), including face, upper extremity, lower extremity and abdominal wall transplants have generated an interest in the development of bioengineered structures with multiple tissue types.Citation51 The difficulty in the engineering of such constructs lies in achieving the integration and cooperation of different tissue types in vivo, while maintaining a viable blood supply.Citation51

Conclusion

The bioengineering of whole organs is an exciting area of transplantation research. There have been considerable advances that have led to the development of a suitable decellularized scaffold in small animal, porcine and human models. Further work is now required to optimize organ-specific recellularization techniques before clinical translation becomes a possibility. Should it become possible to create patient-specific organs, essential work is still required to ensure decellularized scaffolds are not recognized by the host immune system. There is also a requirement for a cost-analysis study to be performed in order to compare current techniques with bioengineered technology to ensure feasibility, especially when considering potential availabilities within government health services.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

References

  • UNetsm. [database online]. The Organ Procurement and Transplantation Network. Available at http://optn.transplant.hrsa.gov. Accessed 08 May, 2015
  • Orlando G, Soker S, Stratta RJ, Atala A. Will regenerative medicine replace transplantation? Cold Spring Harb Perspect Med 2013; 3(8):a015693; PMID:23906883; http://dx.doi.org/10.1101/cshperspect.a015693
  • Orlando G, Wood KJ, Soker S, Stratta RJ. How regenerative medicine may contribute to the achievement of an immunosuppression-free state. Transplantation 2011; 92:e36-8; author reply e39; PMID:21989270; http://dx.doi.org/10.1097/TP.0b013e31822f59d8
  • Macchiarini P, Walles T, Biancosino C, Mertsching H. First human transplantation of a bioengineered airway tissue. J Thorac Cardiovasc Surg 2004; 128:638-41; PMID:15457176; http://dx.doi.org/10.1016/j.jtcvs.2004.02.042
  • L'Heureux N, Dusserre N, Konig G, Victor B, Keire P, Wight TN, Chronos NA, Kyles AE, Gregory CR, Hoyt G, et al. Human tissue-engineered blood vessels for adult arterial revascularization. Nat Med 2006; 12:361-5; PMID:16491087; http://dx.doi.org/10.1038/nm1364
  • Atala A, Bauer SB, Soker S, Yoo JJ, Retik AB. Tissue-engineered autologous bladders for patients needing cystoplasty. Lancet 2006; 367:1241-6; PMID:16631879; http://dx.doi.org/10.1016/S0140-6736(06)68438-9
  • Vacanti JP, Morse MA, Saltzman WM, Domb AJ, Perez-Atayde A, Langer R. Selective cell transplantation using bioabsorbable artificial polymers as matrices. J Pediatr Surg 1988; 23:3-9; PMID:2895175; http://dx.doi.org/10.1016/S0022-3468(88)80529-3
  • Gilbert TW, Sellaro TL, Badylak SF. Decellularization of tissues and organs. Biomaterials 2006; 27:3675-83; PMID:16519932
  • Ott HC, Matthiesen TS, Goh SK, Black LD, Kren SM, Netoff TI, Taylor DA. Perfusion-decellularized matrix: Using nature's platform to engineer a bioartificial heart. Nat Med 2008; 14:213-21; PMID:18193059; http://dx.doi.org/10.1038/nm1684
  • Akhyari P, Aubin H, Gwanmesia P, Barth M, Hoffmann S, Huelsmann J, Preuss K, Lichtenberg A. The quest for an optimized protocol for whole-heart decellularization: A comparison of three popular and a novel decellularization technique and their diverse effects on crucial extracellular matrix qualities. Tissue Eng Part C Methods 2011; 17:915-26; PMID:21548726; http://dx.doi.org/10.1089/ten.tec.2011.0210
  • Guyette JP, Gilpin SE, Charest JM, Tapias LF, Ren X, Ott HC. Perfusion decellularization of whole organs. Nat Protoc 2014; 9:1451-68; PMID:24874812; http://dx.doi.org/10.1038/nprot.2014.097
  • Faulk DM, Wildemann JD, Badylak SF. Decellularization and cell seeding of whole liver biologic scaffolds composed of extracellular matrix. J Clin Exp Hepatol 2015; 5:69-80; PMID:25941434; http://dx.doi.org/10.1016/j.jceh.2014.03.043
  • Gilpin SE, Guyette JP, Gonzalez G, Ren X, Asara JM, Mathisen DJ, Vacanti JP, Ott HC. Perfusion decellularization of human and porcine lungs: Bringing the matrix to clinical scale. J Heart Lung Transplant 2014; 33:298-308; PMID:24365767; http://dx.doi.org/10.1016/j.healun.2013.10.030
  • Scarritt ME, Pashos NC, Bunnell BA. A review of cellularization strategies for tissue engineering of whole organs. Front Bioeng Biotechnol 2015; 3:43; PMID:25870857; http://dx.doi.org/10.3389/fbioe.2015.00043
  • Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P, Hedrick MH. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell 2002; 13:4279-95; PMID:12475952; http://dx.doi.org/10.1091/mbc.E02-02-0105
  • Jiang WC, Cheng YH, Yen MH, Chang Y, Yang VW, Lee OK. Cryo-chemical decellularization of the whole liver for mesenchymal stem cells-based functional hepatic tissue engineering. Biomaterials 2014; 35:3607-17; PMID:24462361; http://dx.doi.org/10.1016/j.biomaterials.2014.01.024
  • Daly AB, Wallis JM, Borg ZD, Bonvillain RW, Deng B, Ballif BA, Jaworski DM, Allen GB, Weiss DJ. Initial binding and recellularization of decellularized mouse lung scaffolds with bone marrow-derived mesenchymal stromal cells. Tissue Eng Part A 2012; 18:1-16; PMID:21756220; http://dx.doi.org/10.1089/ten.tea.2011.0301
  • Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, Nie J, Jonsdottir GA, Ruotti V, Stewart R, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 2007; 318:1917-20; PMID:18029452; http://dx.doi.org/10.1126/science.1151526
  • Ghaedi M, Calle EA, Mendez JJ, Gard AL, Balestrini J, Booth A, Bove PF, Gui L, White ES, Niklason LE. Human iPS cell-derived alveolar epithelium repopulates lung extracellular matrix. J Clin Invest 2013; 123:4950-62; PMID:24135142; http://dx.doi.org/10.1172/JCI68793
  • Lu TY, Lin B, Kim J, Sullivan M, Tobita K, Salama G, Yang L. Repopulation of decellularized mouse heart with human induced pluripotent stem cell-derived cardiovascular progenitor cells. Nat Commun 2013; 4:2307; PMID:23942048
  • Oberwallner B, Brodarac A, Choi YH, Saric T, Anic P, Morawietz L, Stamm C. Preparation of cardiac extracellular matrix scaffolds by decellularization of human myocardium. J Biomed Mater Res A 2014; 102:3263-72; PMID:24142588; http://dx.doi.org/10.1002/jbm.a.35000
  • de Almeida PE, Meyer EH, Kooreman NG, Diecke S, Dey D, Sanchez-Freire V, Hu S, Ebert A, Odegaard J, Mordwinkin NM, et al. Transplanted terminally differentiated induced pluripotent stem cells are accepted by immune mechanisms similar to self-tolerance. Nat Commun 2014; 5:3903; PMID: 24875164
  • Lichtenberg A, Cebotari S, Tudorache I, Sturz G, Winterhalter M, Hilfiker A, Haverich A. Flow-dependent re-endothelialization of tissue-engineered heart valves. J Heart Valve Dis 2006; 15:287, 93; discussion 293-4; PMID:16607913
  • Ng SL, Narayanan K, Gao S, Wan AC. Lineage restricted progenitors for the repopulation of decellularized heart. Biomaterials 2011; 32:7571-80; PMID:21783251; http://dx.doi.org/10.1016/j.biomaterials.2011.06.065
  • Weymann A, Patil NP, Sabashnikov A, Jungebluth P, Korkmaz S, Li S, Veres G, Soos P, Ishtok R, Chaimow N, et al. Bioartificial heart: A human-sized porcine model–the way ahead. PLoS One 2014; 9:e111591; PMID:25365554; http://dx.doi.org/10.1371/journal.pone.0111591
  • Nichols JE, Niles JA, Cortiella J. Design and development of tissue engineered lung: Progress and challenges. Organogenesis 2009; 5:57-61; PMID:19794900; http://dx.doi.org/10.4161/org.5.2.8564
  • Ott HC, Clippinger B, Conrad C, Schuetz C, Pomerantseva I, Ikonomou L, Kotton D, Vacanti JP. Regeneration and orthotopic transplantation of a bioartificial lung. Nat Med 2010; 16:927-33; PMID:20628374; ; http://dx.doi.org/10.1038/nm.2193
  • Petersen TH, Calle EA, Zhao L, Lee EJ, Gui L, Raredon MB, Gavrilov K, Yi T, Zhuang ZW, Breuer C, et al. Tissue-engineered lungs for in vivo implantation. Science 2010; 329:538-41; PMID:20576850; http://dx.doi.org/10.1126/science.1189345
  • O'Neill JD, Anfang R, Anandappa A, Costa J, Javidfar J, Wobma HM, Singh G, Freytes DO, Bacchetta MD, Sonett JR, et al. Decellularization of human and porcine lung tissues for pulmonary tissue engineering. Ann Thorac Surg 2013; 96:1046-55; discussion 1055-6; PMID:23870827; http://dx.doi.org/10.1016/j.athoracsur.2013.04.022
  • Gilpin SE, Ott HC. Using nature's platform to engineer bio-artificial lungs. Ann Am Thorac Soc 2015; 12(Suppl 1):S45-9; PMID:25830835; http://dx.doi.org/10.1513/AnnalsATS.201408-366MG
  • Uygun BE, Soto-Gutierrez A, Yagi H, Izamis ML, Guzzardi MA, Shulman C, Milwid J, Kobayashi N, Tilles A, Berthiaume F, et al. Organ reengineering through development of a transplantable recellularized liver graft using decellularized liver matrix. Nat Med 2010; 16:814-20; PMID:20543851; http://dx.doi.org/10.1038/nm.2170
  • Baptista PM, Siddiqui MM, Lozier G, Rodriguez SR, Atala A, Soker S. The use of whole organ decellularization for the generation of a vascularized liver organoid. Hepatology 2011; 53:604-17; PMID:21274881; http://dx.doi.org/10.1002/hep.24067
  • Zhu S, Rezvani M, Harbell J, Mattis AN, Wolfe AR, Benet LZ, Willenbring H, Ding S. Mouse liver repopulation with hepatocytes generated from human fibroblasts. Nature 2014; 508:93-7; PMID:24572354; http://dx.doi.org/10.1038/nature13020
  • Salvatori M, Katari R, Patel T, Peloso A, Mugweru J, Owusu K, Orlando G. Extracellular matrix scaffold technology for bioartificial pancreas engineering: State of the art and future challenges. J Diabetes Sci Technol 2014; 8:159-69; PMID:24876552; http://dx.doi.org/10.1177/1932296813519558
  • Goh SK, Bertera S, Olsen P, Candiello JE, Halfter W, Uechi G, Balasubramani M, Johnson SA, Sicari BM, Kollar E, et al. Perfusion-decellularized pancreas as a natural 3D scaffold for pancreatic tissue and whole organ engineering. Biomaterials 2013; 34:6760-72; PMID:23787110; http://dx.doi.org/10.1016/j.biomaterials.2013.05.066
  • Chen MK, Badylak SF. Small bowel tissue engineering using small intestinal submucosa as a scaffold. J Surg Res 2001; 99:352-8; PMID:11469910; http://dx.doi.org/10.1006/jsre.2001.6199
  • Ross CL, Booth C, Sanders B, Babbar P, Bergman C, Soker T, Sittadjody S, Salvatori M, Al-Shraideh Y, Stratta RJ, et al. Regeneration and bioengineering of transplantable abdominal organs: Current status and future challenges. Expert Opin Biol Ther 2013; 13:103-13; PMID:23110384; http://dx.doi.org/10.1517/14712598.2013.732063
  • Grikscheit TC, Siddique A, Ochoa ER, Srinivasan A, Alsberg E, Hodin RA, Vacanti JP. Tissue-engineered small intestine improves recovery after massive small bowel resection. Ann Surg 2004; 240:748-54; PMID:15492554; http://dx.doi.org/10.1097/01.sla.0000143246.07277.73
  • Grikscheit TC, Ochoa ER, Ramsanahie A, Alsberg E, Mooney D, Whang EE, Vacanti JP. Tissue-engineered large intestine resembles native colon with appropriate in vitro physiology and architecture. Ann Surg 2003; 238:35-41; PMID:12832963.
  • Lee M, Wu BM, Stelzner M, Reichardt HM, Dunn JC. Intestinal smooth muscle cell maintenance by basic fibroblast growth factor. Tissue Eng Part A 2008; 14:1395-402; PMID:18680389; http://dx.doi.org/10.1089/ten.tea.2007.0232
  • Patil PB, Chougule PB, Kumar VK, Almstrom S, Backdahl H, Banerjee D, Herlenius G, Olausson M, Sumitran-Holgersson S. Recellularization of acellular human small intestine using bone marrow stem cells. Stem Cells Transl Med 2013; 2:307-15; PMID:23486834; http://dx.doi.org/10.5966/sctm.2012-0108
  • Nakayama KH, Batchelder CA, Lee CI, Tarantal AF. Decellularized rhesus monkey kidney as a three-dimensional scaffold for renal tissue engineering. Tissue Eng Part A 2010; 16:2207-16; PMID:20156112; http://dx.doi.org/10.1089/ten.tea.2009.0602
  • Ross EA, Williams MJ, Hamazaki T, Terada N, Clapp WL, Adin C, Ellison GW, Jorgensen M, Batich CD. Embryonic stem cells proliferate and differentiate when seeded into kidney scaffolds. J Am Soc Nephrol 2009; 20:2338-47; PMID:19729441; http://dx.doi.org/10.1681/ASN.2008111196
  • Song JJ, Guyette JP, Gilpin SE, Gonzalez G, Vacanti JP, Ott HC. Regeneration and experimental orthotopic transplantation of a bioengineered kidney. Nat Med 2013; 19:646-51; PMID:23584091; http://dx.doi.org/10.1038/nm.3154
  • Humes HD, Buffington DA, MacKay SM, Funke AJ, Weitzel WF. Replacement of renal function in uremic animals with a tissue-engineered kidney. Nat Biotechnol 1999; 17:451-5; PMID:10331803; http://dx.doi.org/10.1038/8626
  • Tumlin J, Wali R, Williams W, Murray P, Tolwani AJ, Vinnikova AK, Szerlip HM, Ye J, Paganini EP, Dworkin L, et al. Efficacy and safety of renal tubule cell therapy for acute renal failure. J Am Soc Nephrol 2008; 19:1034-40; PMID:18272842; http://dx.doi.org/10.1681/ASN.2007080895
  • Kim S, Fissell WH, Humes DH, Roy S. Current strategies and challenges in engineering a bioartificial kidney. Front Biosci (Elite Ed) 2015; 7:215-28; PMID:25553375
  • Atala A, Kasper FK, Mikos AG. Engineering complex tissues. Sci Transl Med 2012; 4:160rv12; PMID:23152327
  • Spalazzi JP, Dagher E, Doty SB, Guo XE, Rodeo SA, Lu HH. In vivo evaluation of a tri-phasic composite scaffold for anterior cruciate ligament-to-bone integration. Conf Proc IEEE Eng Med Biol Soc 2006; 1:525-8; PMID:17946839; http://dx.doi.org/10.1109/IEMBS.2006.259296
  • Lu HH, Subramony SD, Boushell MK, Zhang X. Tissue engineering strategies for the regeneration of orthopedic interfaces. Ann Biomed Eng 2010; 38:2142-54; PMID:20422291; http://dx.doi.org/10.1007/s10439-010-0046-y
  • Bueno EM, Diaz-Siso JR, Sisk GC, Chandawarkar A, Kiwanuka H, Lamparello B, Caterson EJ, Pomahac B. Vascularized composite allotransplantation and tissue engineering. J Craniofac Surg 2013; 24:256-63; PMID:23348296; http://dx.doi.org/10.1097/SCS.0b013e318275f173

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.