You are on page 1of 18

Available online at www.sciencedirect.

com

Engineering organs
Anthony Atala1,2

Applications of regenerative medicine technology may offer The kidney was the first entire organ to be replaced in
novel therapies for patients with injuries, end-stage organ a human, in 1955 [2]. However, this transplant was
failure, or other clinical problems. Currently, patients suffering performed between identical twins, and thus it did not
from diseased and injured organs can be treated with address the immune response to transplanted organs. In
transplanted organs. However, there is a severe shortage of the early 1960s, Murray, who later received the Nobel
donor organs that is worsening yearly as the population ages prize for his work, performed a nonrelated kidney trans-
and new cases of organ failure increase. Scientists in the field of plantation from a nongenetically identical patient into
regenerative medicine and tissue engineering are now applying another. This transplant, which overcame the immuno-
the principles of cell transplantation, material science, and logic barrier, marked a new era in medical therapy and
bioengineering to construct biological substitutes that will opened the door for the use of transplantation as a means
restore and maintain normal function in diseased and injured of therapy for different organ systems. However, lack of
tissues. The stem cell field is also advancing rapidly, opening good immune-suppression, the inability to monitor and
new avenues for this type of therapy. For example, therapeutic control rejection, and the worsening donor shortage
cloning and cellular reprogramming may one day provide a spurred physicians and scientists to look for other
potentially limitless source of cells for tissue engineering alternatives.
applications. Although stem cells are still in the research phase,
some therapies arising from tissue engineering endeavors have Synthetic materials were introduced to replace or rebuild
already entered the clinical setting successfully, indicating the diseased tissues or parts in the human body. The advent
promise regenerative medicine holds for the future. of new manmade materials, such as tetrafluoroethylene
Addresses (Teflon) and silicone, opened a new field which included
1
Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC a wide array of devices that could be applied for human
27157, United States use. However, although these devices could provide for
2
Department of Urology, Wake Forest University School of Medicine,
structural replacement, the functional component of the
Medical Center Boulevard, Winston-Salem, NC 27157, United States
original tissue was not achieved.
Corresponding author: Atala, Anthony (aatala@wfubmc.edu)
Meanwhile, new techniques for cell harvesting, culture,
and expansion were developed. Studies of the extracellu-
Current Opinion in Biotechnology 2009, 20:575–592
lar matrix and its interaction with cells, and with growth
This review comes from a themed issue on factors and their ligands, led the way to a further un-
Tissue, cell and pathway engineering derstanding of cell and tissue growth and differentiation.
Edited by Gail Naughton and Chandran Siddharthan The concept of cell transplantation took hold in the
Available online 5th November 2009
research arena, and culminated with the first human bone
marrow cell transplant in the 1970s. At this time, research-
0958-1669/$ – see front matter ers began to combine the devices and materials with cell
# 2009 Elsevier Ltd. All rights reserved. biology concepts, creating a new field called tissue engin-
DOI 10.1016/j.copbio.2009.10.003
eering. As more scientists from different fields came
together with the common goal of tissue replacement,
the field of tissue engineering became more formally
established.
Introduction
Patients suffering from diseased and injured organs may In the early 1980s, mouse embryonic stem cells were
be treated with transplanted organs. The use of one discovered [3]. However, this new field remained rela-
body part for another or the exchange of parts from one tively dormant until the description of human embryonic
person to another was mentioned in the medical litera- stem cells in 1998 [4]. The description of these cells led to
ture even in antiquity and captured the imagination of one of the most contested ethical debates in the field of
many over time. The maintenance of organs in culture medicine. Then, in 1999, the world awoke to the startling
was a major area of inquiry in the early 1900s. Charles media announcement of the creation of the first cloned
Lindbergh, the first pilot to successfully fly across the mammal, a sheep named Dolly [5].
Atlantic in the 1920s, joined forces with Alexis Carrel, a
Nobel Prize Winner in the field of Medicine, to inves- These fields — cell transplantation, tissue engineering,
tigate the potential of keeping organs alive ex vivo long stem cells, and nuclear transfer — had one unifying
term [1]. concept: the regeneration of living tissues and organs.

www.sciencedirect.com Current Opinion in Biotechnology 2009, 20:575–592


576 Tissue, cell and pathway engineering

Thus in 1999, William Haseltine, then the Scientific cells, their clinical application is also limited because they
Founder and Chief Executive Officer of Human Genome represent an allogenic resource and thus have the poten-
Sciences, coined the term ‘regenerative medicine’, in tial to evoke an immune response. New stem cell tech-
effect bringing all these areas under one defining field nologies (such as cloning, or somatic cell nuclear transfer,
[6]. While organ transplantation remains a mainstay of and reprogramming) promise to overcome this limitation.
treatment for patients with severely compromised organ
function, the number of patients in need of treatment far Two types of nuclear cloning, reproductive cloning and
exceeds the organ supply, and this shortfall is expected to therapeutic cloning, have been described, and a better
worsen as the global population ages. However, recent understanding of the differences between the two types
advances in regenerative medicine suggest that it can may help to alleviate some of the controversy that sur-
provide new alternatives to donor organs. In the last two rounds these technologies [20,21]. Banned in most
decades, scientists have attempted to grow native and countries for human applications, reproductive cloning
stem cells, engineer tissues, and design treatment mod- is used to generate an embryo that has the identical
alities using regenerative medicine techniques for vir- genetic material as its cell source. This embryo can then
tually every tissue of the human body. This article be implanted into the uterus of a female to give rise to an
reviews some of the progress that has been achieved in infant that is a clone of the donor. On the other hand,
this field. therapeutic cloning is used to generate early stage
embryos that are explanted in culture to produce embryo-
Cells for use in organ engineering nic stem cell lines whose genetic material is identical to
Stem cells that of its source. These autologous stem cells have the
In 1981 pluripotent cells were found in the inner cell mass potential to become almost any type of cell in the adult
of the human embryo, and the term ‘human embryonic body, and thus would be useful in tissue and organ
stem cell’ was coined [3]. Human embryonic stem cells replacement applications [22]. Therefore, therapeutic
exhibit two remarkable properties: the ability to prolifer- cloning, which has also been called somatic cell nuclear
ate in an undifferentiated but pluripotent state (self- transfer, may provide an alternative source of transplan-
renewal), and the ability to differentiate into many table cells. Figure 1 shows the strategy of combining
specialized cell types [7]. This makes them an attractive therapeutic cloning with tissue engineering to develop
resource for regenerative medicine techniques. They can tissues and organs.
be isolated by aspirating the inner cell mass from the
embryo during the blastocyst stage (five days postfertili- While promising, somatic cell nuclear transfer technology
zation), and are usually grown on feeder layers consisting has certain limitations that require further improvements
of mouse embryonic fibroblasts or human feeder cells [8]. before therapeutic cloning can be applied widely in
More recent reports have shown that these cells can be replacement therapy. Currently, the efficiency of the
grown without the use of a feeder layer [9] and thus avoid overall cloning process is low. To improve cloning effi-
the exposure of these human cells to mouse viruses and ciency, further improvements are required in the multiple
proteins. These cells have demonstrated longevity in complex steps of nuclear transfer, such as enucleation and
culture by maintaining their undifferentiated state for reconstruction, activation of oocytes, and cell cycle syn-
at least 80 passages when grown using current published chronization between donor cells and recipient oocytes
protocols [4,10]. [23].

Human embryonic stem cells have been shown to differ- Recently, exciting reports of the successful transform-
entiate into cells from all three embryonic germ layers in ation of adult cells into pluripotent stem cells through a
vitro. Skin and neurons have been formed, indicating type of genetic ‘reprogramming’ have been published.
ectodermal differentiation [11–14]. Blood, cardiac cells, Reprogramming is a technique that involves de-differen-
cartilage, endothelial cells, and muscle have been formed, tiation of adult somatic cells to produce patient-specific
indicating mesodermal differentiation [15–17]. Pancreatic pluripotent stem cells, without the use of embryos. Cells
cells have been formed, indicating endodermal differen- generated by reprogramming would be genetically iden-
tiation [18]. In addition, as further evidence of their tical to the somatic cells (and thus, the patient who
pluripotency, embryonic stem cells can form embryoid donated these cells) and would not be rejected. Yamanaka
bodies, which are cell aggregations that contain all three was the first to discover that mouse embryonic fibroblasts
embryonic germ layers while in culture, and can form (MEFs) and adult mouse fibroblasts could be repro-
teratomas in vivo [19]. grammed into an ‘induced pluripotent state (iPS)’ [24].
The resultant iPS cells possessed the immortal growth
The political controversy surrounding stem cells began in characteristics of self-renewing ES cells, expressed genes
1998 with the creation of human embryonic stem (hES) specific for ES cells, and generated embryoid bodies in
cells derived from discarded embryos. In addition to the vitro and teratomas in vivo. When iPS cells were injected
ethical dilemma surrounding the use of embryonic stem into mouse blastocysts, they contributed to a variety of

Current Opinion in Biotechnology 2009, 20:575–592 www.sciencedirect.com


Engineering organs Atala 577

Figure 1

Tissue engineering of the urethra using a collagen matrix. (A) Representative case of a patient with a bulbar stricture. (B) During the urethral repair
surgery, strictured tissue is excised, preserving the urethral plate on the left side, and matrix is anastamosed to the urethral plate in an onlay fashion on
the right. The boxes in both photos indicate the area of interest, including the urethra, which appears white in the left photograph. In the left
photograph, the arrow indicates the area of stricture in the urethra. On the right, the arrow indicates the repaired stricture. Note that the engineered
tissue now obscures the native white urethral tissue in an onlay fashion in the right photograph. (C) Urethrogram six months after repair. (D)
Cystoscopic view of urethra before surgery on the left side, and four months after repair on the right side.

cell types. However, although iPS cells selected in this ing each embryonic germ layer, including cells of adipo-
way were pluripotent, they were not identical to ES cells. genic, osteogenic, myogenic, endothelial, neuronal, and
Unlike ES cells, chimeras made from iPS cells did not hepatic lineages. In this respect, they meet a commonly
result in full-term pregnancies. Gene expression profiles accepted criterion for pluripotent stem cells, without
of the iPS cells showed that they possessed a distinct gene implying that they can generate every adult tissue.
expression signature that was different from that of ES Examples of differentiated cells derived from AFS cells
cells. In addition, the epigenetic state of the iPS cells was and displaying specialized functions include neuronal
somewhere between that found in somatic cells and that lineage secreting the neurotransmitter L-glutamate or
found in ES cells, suggesting that the reprogramming was expressing G-protein-gated inwardly rectifying potassium
incomplete. These results were improved significantly by (GIRK) channels, hepatic lineage cells producing urea,
Wernig and Jaenisch in July 2007 [25] and these iPS cells and osteogenic lineage cells forming tissue-engineered
were completely reprogrammed. It has recently been bone. The cells could be obtained either from amniocent-
shown that reprogramming of human cells is possible esis or chorionic villous sampling in the developing fetus,
[26,27]. or from the placenta at the time of birth. The cells could
be preserved for self-use, and used without rejection, or
An alternate source of stem cells is the amniotic fluid and they could be banked. A bank of 100 000 specimens could
placenta. Amniotic fluid and the placenta are known to potentially supply 99% of the US population with a
contain multiple partially differentiated cell types perfect genetic match for transplantation. Such a bank
derived from the developing fetus. We isolated stem cell may be easier to create than with other cell sources, since
populations from these sources, called amniotic fluid and there are approximately 4.5 million births per year in the
placental stem cells (AFPSC) that express embryonic and USA [28].
adult stem cell markers [28]. The undifferentiated stem
cells expand extensively without feeders and double Finally, it may be possible to employ adult stem cells in
every 36 hours. Unlike human embryonic stem cells, the regenerative process. Adult stem cells, especially
the AFPSC do not form tumors in vivo. Lines maintained hematopoietic stem cells, are the best understood cell
for over 250 population doublings retained long telomeres type in stem cell biology [29]. However, adult stem cell
and a normal karyotype. AFS cells are broadly multi- research remains an area of intense study, as their poten-
potent. Clonal human lines verified by retroviral marking tial for therapy may be applicable to a myriad of degen-
can be induced to differentiate into cell types represent- erative disorders. Within the past decade, adult stem cell

www.sciencedirect.com Current Opinion in Biotechnology 2009, 20:575–592


578 Tissue, cell and pathway engineering

populations have been found in many adult tissues other When native cells are used for tissue reconstitution, donor
than the bone marrow and the gastrointestinal tract, tissue is dissociated into individual cells, which are either
including the brain [30,31], skin [32], and muscle implanted directly into the host or expanded in culture,
[33]. Many other types of adult stem cells have been attached to a support matrix, and reimplanted after
identified in organs all over the body and are thought to expansion. The implanted tissue can be heterologous,
serve as the primary repair entities for their correspond- allogenic, or autologous. Ideally, this approach allows lost
ing organs [34]. The discovery of such tissue-specific tissue function to be restored or replaced in toto and with
progenitors has opened up new avenues for research. limited complications [45]. Native cells and tissues are
Research into adult stem cells has, however, progressed usually preferable for reconstruction. In most cases, the
slowly, mainly because investigators have had great replacement of lost or deficient tissues with functionally
difficulty in maintaining adult non-mesenchymal stem equivalent cells and tissues would improve the outcome
cells in culture. for these patients. This goal may be attainable with the
use of regenerative medicine techniques.
Native targeted progenitor cells
One of the limitations of applying cell-based regenera- Biomaterials
tive medicine techniques to organ replacement has been For cell-based tissue engineering, the expanded cells are
the inherent difficulty of growing specific cell types in seeded onto a scaffold synthesized with the appropriate
large quantities. Even when some organs, such as the biomaterial. In tissue engineering, biomaterials replicate
liver, have a high regenerative capacity in vivo, cell the biologic and mechanical function of the native ECM
growth and expansion in vitro may be difficult. By noting found in tissues in the body by serving as an artificial
the location of the progenitor cells, as well as by explor- ECM. Biomaterials provide a three-dimensional space for
ing the conditions that promote differentiation and/or the cells to form into new tissues with appropriate struc-
self-renewal, it has been possible to overcome some of ture and function, and also can allow for the delivery of
the obstacles that limit cell expansion in vitro. One cells and appropriate bioactive factors (e.g. cell-adhesion
example is the urothelial cell. Urothelial cells could peptides and growth factors), to desired sites in the body
be grown in the laboratory setting in the past, but only [64]. As the majority of mammalian cell types are ancho-
with limited success. It was believed that urothelial cells rage-dependent and will die if no cell-adhesion substrate
had a natural senescence that was hard to overcome. is available, biomaterials provide a cell-adhesion substrate
Several protocols have been developed over the last two that can deliver cells to specific sites in the body with high
decades that have improved urothelial growth and loading efficiency. Biomaterials can also provide mech-
expansion [35–38]. A system of urothelial cell harvesting anical support against in vivo forces such that the pre-
was developed that does not use any enzymes or serum defined three-dimensional structure is maintained during
and has a large expansion potential. Using these tissue development. Furthermore, bioactive signals, such
methods of cell culture, it is possible to expand a as cell-adhesion peptides and growth factors, can be
urothelial strain from a single specimen that initially loaded along with cells to help regulate cellular function.
covers a surface area of 1 cm2 to one covering a surface
area of 4202 m2 (the equivalent area of one football field) Generally, three classes of biomaterials have been used
within eight weeks [35]. for engineering tissues and organs: naturally derived
materials, such as collagen and alginate; acellular tissue
An additional advantage in using native cells is that they matrices, such as bladder submucosa and small-intestinal
can be obtained from the specific organ to be regenerated, submucosa (SIS); and synthetic polymers, such as poly-
expanded, and used in the same patient without rejection, glycolic acid (PGA), poly-lactic acid (PLA), and poly(-
in an autologous manner [35,39–55]. Bladder, ureter, and lactic-co-glycolic acid) (PLGA). Naturally derived
renal pelvis cells can equally be harvested, cultured, and materials and acellular tissue matrices have the potential
expanded in a similar fashion. Normal human bladder advantage of biologic recognition, but synthetic polymers
epithelial and muscle cells can be efficiently harvested can be produced reproducibly on a large scale with
from surgical material, extensively expanded in culture, controlled properties of strength, degradation rate, and
and their differentiation characteristics, growth require- microstructure.
ments, and other biologic properties can be studied
[35,37,38,48,49,56–63]. Major advances in cell culture Collagen is the most abundant and ubiquitous structural
techniques have been made within the past decade, protein in the body, and it may be readily purified from
and these techniques make the use of autologous cells both animal and human tissues with an enzyme treatment
possible for clinical application. However, even now, not and salt/acid extraction [65]. Collagen has long been
all human cells can be grown or expanded in vitro. Liver, known to exhibit minimal inflammatory and antigenic
nerve, and pancreas are examples of human tissues where responses [66], and it has been approved by the U.S. Food
the technology is not yet advanced to the point where and Drug Administration (FDA) for many types of
these cells can be grown and expanded. medical applications, including wound dressings and

Current Opinion in Biotechnology 2009, 20:575–592 www.sciencedirect.com


Engineering organs Atala 579

artificial skin [67]. This material can be processed into a More recently, techniques such as electrospinning have
wide variety of structures such as sponges, fibers, and been used to quickly create highly porous scaffolds in
films [68–70]. various conformations [88–91]. However, one drawback
of the synthetic polymers is lack of biologic recognition.
Alginate, a polysaccharide isolated from seaweed, has As an approach toward incorporating cell recognition
been used as an injectable cell delivery vehicle [71] domains into these materials, copolymers with amino
and a cell immobilization matrix (Lim and Sun, 1980) acids have been synthesized [92–94]. Other biodegrad-
owing to its gentle gelling properties in the presence of able synthetic polymers, including poly(anhydrides) and
divalent ions such as calcium. Efforts have been made to poly(ortho-esters), can also be used to fabricate scaffolds
synthesize biodegradable alginate hydrogels with mech- with controlled properties [95].
anical properties that are controllable in a wide range by
intermolecular covalent cross-linking and with cell- Nanotechnology, which exploits the ability to use small
adhesion peptides coupled to their backbones [72]. molecules that have distinct properties in a small scale,
has been used to create smart biomaterials for regenera-
Recently, natural materials such as alginate and collagen tive medicine [96,97]. Nanoscaffolds have been manu-
have been used as ‘bio-inks’ in a newly developed bio- factured specifically for bladder applications [98]. The
printing technique based on inkjet technology [73,74]. manufacturing of biomaterials can also lead to enhanced
Using this technology, these scaffold materials can be cell alignment and tissue formation [89].
‘printed’ into a desired scaffold shape using a modified
inkjet printer. In addition, several groups have shown that Tissue engineering of specific structures
living cells can also be printed using this technology Investigators around the world, including our laboratory,
[75,76]. This exciting technique can be modified so that have been working toward the development of several
a three-dimensional construct containing a precise cell types and tissues and organs for clinical application.
arrangement of cells, growth factors, and extracellular
matrix material can be printed [77–79]. Such constructs Urethra
may eventually be implanted into a host to serve as the Various strategies have been proposed over the years for
backbone for a new tissue or organ. the regeneration of urethral tissue. Woven meshes of
PGA without cells [99,100] or with cells (Atala et al.
Acellular tissue matrices are collagen-rich matrices pre- [39] #208) were used to regenerate urethras in various
pared by removing cellular components from tissues. The animal models. Naturally derived collagen-based
matrices are often prepared by mechanical and chemical materials such bladder-derived acellular submucosa
manipulation of a segment of bladder tissue [51,80–82]. [82], and an acellular urethral submucosa [101], have also
The matrices slowly degrade after implantation and are been tried experimentally in various animal models for
replaced and remodeled by ECM proteins synthesized urethral reconstruction.
and secreted by transplanted or ingrowing cells. Acellular
tissue matrices have been proved to support cell ingrowth The bladder submucosa matrix [82] was proved to be a
and regeneration of genitourinary tissues, including ure- suitable graft for the repair of urethral defects in rabbits.
thra and bladder, with no evidence of immunogenic The neourethras demonstrated a normal urothelial lumi-
rejection [82,83]. Because the structures of the proteins nal lining and organized muscle bundles. These results
(e.g. collagen and elastin) in acellular matrices are well were confirmed clinically in a series of patients with a
conserved and normally arranged, the mechanical proper- history of failed hypospadias reconstruction wherein the
ties of the acellular matrices are not significantly different urethral defects were repaired with human bladder acel-
from those of native bladder submucosa [80]. lular collagen matrices (Figure 1) [44]. The neourethras
were created by anastomosing the matrix in an onlay
Polyesters of naturally occurring a-hydroxy acids, in- fashion to the urethral plate. The size of the created
cluding PGA, PLA, and PLGA, are widely used in regen- neourethra ranged from 5 to 15 cm. After a three-year
erative medicine. These polymers have gained FDA follow-up, three of the four patients had a successful
approval for human use in a variety of applications, outcome in regard to cosmetic appearance and function.
including sutures [84]. The degradation products of One patient who had a 15-cm neourethra developed a
PGA, PLA, and PLGA are nontoxic, natural metabolites subglanular fistula. The acellular collagen-based matrix
that are eventually eliminated from the body in the form eliminated the necessity of performing additional surgical
of carbon dioxide and water [84]. Because these polymers procedures for graft harvesting, and both operative time
are thermoplastics, they can easily be formed into a three- and the potential morbidity from the harvest procedure
dimensional scaffold with a desired microstructure, gross were decreased. Similar results were obtained in pediatric
shape, and dimension by various techniques, including and adult patients with primary urethral stricture disease
molding, extrusion [85], solvent casting [86], phase sep- using the same collagen matrices [102]. Another study in
aration techniques, and gas foaming techniques [87]. 30 patients with recurrent stricture disease showed that a

www.sciencedirect.com Current Opinion in Biotechnology 2009, 20:575–592


580 Tissue, cell and pathway engineering

healthy urethral bed (two or fewer prior urethral 1 cm, which are treated with a matrix alone, without cells,
surgeries) was needed for successful urethral reconstruc- usually have increased collagen deposition, increased
tion using the acellular collage-based grafts [103]. More fibrosis, and scar formation. Cell-seeded matrices
than 200 pediatric and adult patients with urethral disease implanted in wound beds are able to further lengthen
have been successfully treated in an onlay manner with a the distance for normal tissue formation, without initiat-
bladder-derived collagen-based matrix. One of its advan- ing an adverse fibrotic response. Studies in the field of
tages over nongenital tissue grafts used for urethroplasty, regenerative medicine have shown that very large
is that the material is ‘off the shelf.’ This eliminates the defects, greater than 30 cm, can be successfully treated
necessity of additional surgical procedures for graft har- using cell-seeded scaffolds. This explains the described
vesting, which may decrease operative time, as well as the experimental and clinical results noted with urethral
potential morbidity due to the harvest procedure. repair. Nonseeded matrices are able to replace urethral
segments when used in an onlay fashion because of the
The above techniques, using nonseeded acellular short distances required for tissue ingrowth. However, if a
matrices, were applied experimentally and clinically in tubularized urethral repair is needed, the matrices need to
a successful manner for onlay urethral repairs. However, be seeded with autologous cells in order to avoid the risk
when tubularized urethral repairs were attempted exper- of stricture formation and poor tissue development.
imentally, adequate urethral tissue regeneration was not
achieved, and complications ensued, such as graft con- Bladder
tracture and stricture formation [104]. Autologous rabbit Over the last few decades, several bladder wall substitutes
bladder epithelial and smooth muscle cells were grown have been attempted with both synthetic and organic
and seeded onto preconfigured tubular matrices. Entire materials. Synthetic materials that have been tried in
urethra segments were resected and urethroplasties were experimental and clinical settings include polyvinyl
performed with tubularized collagen matrices seeded sponge, Teflon, collagen matrices, Vicryl (PGA) matrices,
either with cells, or without cells. The tubularized col- and silicone. Most of these attempts have failed because of
lagen matrices seeded with autologous cells formed new mechanical, structural, functional, or biocompatibility pro-
tissue which was histologically similar to native urethra. blems. Usually, permanent synthetic materials used for
The tubularized collagen matrices without cells lead to bladder reconstruction succumb to mechanical failure and
poor tissue development, fibrosis, and stricture formation. urinary stone formation, and the use of degradable
These findings were confirmed clinically. A clinical trial materials leads to fibroblast deposition, scarring, graft con-
using tubularized nonseeded SIS for urethral stricture tracture, and a reduced reservoir volume over time
repair was performed in eight evaluable patients. Two [46,110]. Because of this, there has been an increase in
patients with short inflammatory strictures maintained the use of various collagen-based matrices for tissue regen-
urethral patency. Stricture recurrence developed in the eration. Nonseeded allogenic acellular bladder matrices
other six patients within three months of surgery [105]. have served as scaffolds for the ingrowth of host bladder
Other cell types have also been tried experimentally in wall components. The matrices are prepared by mechani-
acellular bladder collagen matrices, including foreskin cally and chemically removing all cellular components
epidermal cells and oral keratinocytes [106,107]. Vascular from bladder tissue [51,81,83,111,112]. The matrices serve
endothelial growth factor gene-modified urothelial cells as vehicles for partial bladder regeneration, and relevant
have also been used experimentally for urethral recon- antigenicity is not evident. However, in multiple studies
struction [108]. using various materials as nonseeded grafts for cystoplasty,
the urothelial layer was able to regenerate normally, but the
The normal wound healing response to injury has been muscle layer, although present, was not fully developed
studied extensively, and this knowledge has been helpful [51,83,111,113–115]. Studies involving acellular matrices
in maximizing success for the engineering of tissues. At that may provide the necessary environment to promote
the time of tissue injury, cell ingrowth is initiated from cell migration, growth, and differentiation are being con-
the wound edges in order to cover the tissue defect. The ducted [116]. With continued bladder research in this area,
cells from the edges of the native tissue are able to these matrices may have a clinical role in bladder replace-
traverse short distances without any detrimental effects. ment in the future.
If the wound is large, more than a few millimeters in
distance or depth, increased collagen deposition, fibrosis, Human urothelial and muscle cells can be expanded in
and scar formation ensue. Matrices implanted in wound vitro, seeded onto polymer scaffolds, and allowed to
beds are able to lengthen the distances that cells can attach and form sheets of cells. The cell–polymer scaffold
traverse without initiating an adverse fibrotic response. can then be implanted in vivo. Histologic analysis indi-
However, these distances are also limited. The maximum cated that viable cells were able to self-assemble back
distance that adjacent cells from the wound edge have to into their respective tissue types, and would retain their
travel to create normal tissue over a biologic matrix is native phenotype [41]. These experiments demonstrated,
approximately 1 cm [109]. Tissue defects greater than for the first time, that composite layered tissue-engineered

Current Opinion in Biotechnology 2009, 20:575–592 www.sciencedirect.com


Engineering organs Atala 581

structures could be created de novo. Before this study, only 46% of preoperative values, respectively. An average
nonlayered structures had been created in the field of bladder capacity of 95% of the original precystectomy
regenerative medicine. volume was achieved in the cell-seeded tissue-engin-
eered bladder replacements. These findings were con-
Cell-seeded allogenic acellular bladder matrices were firmed radiographically. The subtotal cystectomy
used for bladder augmentation in dogs [51]. The regen- reservoirs that were not reconstructed and the polymer-
erated bladder tissues contained a normal cellular organ- only reconstructed bladders showed a marked decrease in
ization consisting of urothelium and smooth muscle and bladder compliance (10% and 42% total compliance). The
exhibited a normal compliance. Biomaterials preloaded compliance of the cell-seeded tissue-engineered bladders
with cells before their implantation showed better tissue showed almost no difference from preoperative values
regeneration compared with biomaterials implanted with that were measured when the native bladder was present
no cells, in which tissue regeneration depended on (106%). Histologically, the nonseeded scaffold bladders
ingrowth of the surrounding tissue. The bladders showed presented a pattern of normal urothelial cells with a
a significant increase (100%) in capacity when augmented thickened fibrotic submucosa and a thin layer of muscle
with scaffolds seeded with cells, compared to scaffolds fibers. The retrieved tissue-engineered bladders showed
without cells (30%). The acellular collagen matrices can a normal cellular organization, consisting of a trilayer of
be enhanced with growth factors to improve bladder urothelium, submucosa, and muscle. Immunocytochemi-
regeneration [117]. cal analyses confirmed the muscle and urothelial pheno-
type. S-100 staining indicated the presence of neural
It has been well established for decades that the bladder structures [54]. These studies, performed with poly-gly-
is able to regenerate generously over free grafts. Urothe- colic acid based scaffolds, have been repeated by other
lium is associated with a high reparative capacity [118]. investigators, showing similar results in large numbers of
Bladder muscle tissue is less likely to regenerate in a animals long term [114,122]. The strategy of using bio-
normal fashion. Both urothelial and muscle ingrowth are degradable scaffolds with cells can be pursued without
believed to be initiated from the edges of the normal concerns for local or systemic toxicity [123]. However, not
bladder toward the region of the free graft [119,120]. all scaffolds perform well if a large portion of the bladder
Usually, however, contracture or resorption of the graft needs replacement. In a study using SIS for subtotal
has been evident. The inflammatory response toward the bladder replacement in dogs, both the unseeded and
matrix may contribute to the resorption of the free graft. It cell-seeded experimental groups showed graft shrinkage
was hypothesized that building the three-dimensional and poor results [124]. The type of scaffold used is critical
structure constructs in vitro, before implantation, would for the success of these technologies. The use of bio-
facilitate the eventual terminal differentiation of the cells reactors, in which mechanical stimulation is started at the
after implantation in vivo and would minimize the inflam- time of organ production, has also been proposed as an
matory response toward the matrix, thus avoiding graft important parameter for success [125].
contracture and shrinkage. The dog study demonstrated a
major difference between matrices used with autologous A clinical experience involving engineered bladder tis-
cells (tissue-engineered matrices) and those used without sue for cystoplasty reconstruction was conducted starting
cells [51]. Matrices implanted with cells for bladder in 1998. A small pilot study of seven patients was
augmentation retained most of their implanted diameter, reported, using a collagen scaffold seeded with cells
as opposed to matrices implanted without cells for blad- either with or without omentum coverage, or a combined
der augmentation, in which graft contraction and shrink- PGA-collagen scaffold seeded with cells and omental
age occurred. The histomorphology demonstrated a coverage (Figure 2). The patients reconstructed with the
marked paucity of muscle cells and a more aggressive engineered bladder tissue created with the PGA–col-
inflammatory reaction in the matrices implanted without lagen cell-seeded scaffolds with omental coverage
cells. Epithelial–mesenchymal signaling is important for showed increased compliance, decreased end-filling
the differentiation of bladder smooth muscle [121]. pressures, increased capacities and longer dry periods
over time (Figure 3) [126]. It is clear from this experience
The results of initial studies showed that the creation of that the engineered bladders continued their improve-
artificial bladders may be achieved in vivo; however, it ment with time, mirroring their continued development.
could not be determined whether the functional Although the experience is promising in terms of show-
parameters noted were caused by the augmented seg- ing that engineered tissues can be implanted safely, it is
ment or by the intact native bladder tissue. To better just a start in terms of accomplishing the goal of engin-
address the functional parameters of tissue-engineered eering fully functional bladders. This was a limited
bladders, an animal model was designed that required a clinical experience, and the technology is not yet ready
subtotal cystectomy with subsequent replacement with a for wide dissemination, as further experimental and
tissue-engineered organ [54]. Cystectomy-only and non- clinical studies are required. FDA Phase 2 studies have
seeded controls maintained average capacities of 22% and now been completed.

www.sciencedirect.com Current Opinion in Biotechnology 2009, 20:575–592


582 Tissue, cell and pathway engineering

Figure 2

Construction of engineered bladder. (A) Scaffold material seeded with cells for use in bladder repair. (B) The seeded scaffold is anastamosed to native
bladder with running 4-0 poly-glycolic sutures. (C) Implant covered with fibrin glue and omentum.

An area of concern in the field of tissue engineering in the formation during the normal process of tissue regeneration
past was the source of cells for regeneration. The concept due to natural turnover, ageing, and tissue injury. It is
of creating engineered constructs involves initially known that genetically normal nonmalignant progenitor
obtaining cells for expansion from the diseased organ. cells, which are the reservoirs for new cell formation, are
However, it was not known until recently whether the cell programmed to give rise to normal tissue, regardless of
population obtained for later autologous implantation was whether the niche resides in either normal or diseased
normal and would lead to normal tissue formation. For tissues [130–132]. Therefore, although the mechanisms for
example, would the cells obtained from a neuropathic tissue self-assembly and regenerative medicine are not
bladder lead to the engineering of another neuropathic fully understood, it is known that the progenitor cells
bladder? Cultured neuropathic bladder smooth muscle are able to ‘reset’ their program for normal cell differen-
cells possess and maintain different characteristics than tiation. The stem cell niche and its role in normal tissue
normal smooth muscle cells in vitro, as demonstrated by regeneration remains a fertile area of ongoing investigation.
growth assays, contractility, adherence tests, and micro-
array analysis [127–129]. However, when neuropathic Genital tissues and organs
smooth muscle cells were cultured in vitro, seeded onto Reconstructive surgery is required for a wide variety of
matrices and implanted in vivo, the tissue-engineered pathologic penile conditions, including penile carcinoma,
constructs showed the same properties as the tissues trauma, severe erectile dysfunction, and congenital con-
engineered with normal cells [130]. The progenitor cells, ditions such as ambiguous genitalia, hypospadias, and
which reside within stem cell niches within each organ, epispadias. One of the major limitations of phallic recon-
are responsible for new cell differentiation and tissue structive surgery is the availability of sufficient autologous

Figure 3

Cystograms and urodynamic studies of a patient before and after implantation of the tissue-engineered bladder. (A) Preoperative results indicate an
irregular-shaped bladder in the cystogram (left) and abnormal bladder pressures as the bladder is filled during urodynamic studies (right). (B)
Postoperatively, findings are significantly improved.

Current Opinion in Biotechnology 2009, 20:575–592 www.sciencedirect.com


Engineering organs Atala 583

tissue. Nongenital autologous tissue sources have been present in all the rabbits with the engineered corpora.
used for decades. Phallic reconstruction was initially The female rabbits mated with the animals implanted
attempted in the late 1930s, with rib cartilage used as a with engineered corpora and they conceived and deliv-
stiffener for patients with traumatic penile loss [133,134], ered healthy pups. These studies demonstrate that penile
but this process produced unsatisfactory functional and corpora cavernosa tissue can be engineered. The engin-
cosmetic results. Silicone rigid prostheses were popular- eered tissue is able to achieve adequate structural and
ized in the 1970s and have been used widely [135,136]. functional parameters sufficient for erection, copulation,
However, biocompatibility issues have been a problem in ejaculation, and conception in rabbits. Further studies
selected patients [137,138]. Tissue transfer techniques will be needed to confirm the long-term functionality of
with flaps from various nongenital sources such as the these organs. In addition, further studies are needed to
groin and forearm have been used for genital reconstruc- show that comparable human structures can also be
tion [139]. However, operative complications such as engineered.
infection, graft failure, and donor site morbidity are not
negligible. Phallic reconstruction with autologous tissue, Testis
derived from the patient’s own cells, may be preferable in Patients with testicular dysfunction require androgen
selected cases. replacement for somatic development. Conventional
treatment for testicular dysfunction consists of periodic
One of the major components of the phallus is corporal intramuscular injections of chemically modified testos-
smooth muscle. Initial experiments have shown that terone or, more recently, skin patch applications. How-
cultured human corporal smooth muscle cells may be ever, long-term nonpulsatile testosterone therapy is not
used in conjunction with biodegradable polymers to optimal and can cause multiple problems, including
create corpus cavernosum tissue de novo [140]. In a excessive erythropoiesis and bone density changes. To
subsequent study, human corporal smooth muscle cells address the problem, a system was designed in which
and endothelial cells seeded on biodegradable polymer Leydig cells, which produce most of the testosterone in
scaffolds were able to form vascularized cavernosal the male, were microencapsulated in an alginate-poly-L-
muscle when implanted in vivo [55]. Later, in order to lysine solution and injected into castrated animals. Serum
minimize any immune reactions that a synthetic bioma- testosterone was measured serially; the animals were able
terial might cause, naturally derived acellular corporal to maintain testosterone levels in the long term [50].
tissue matrix with the same architecture as native corpora These studies suggest that microencapsulated Leydig
was developed. Acellular collagen matrices were derived cells may be able to replace or supplement testosterone
from processed donor rabbit corpora using cell lysis tech- in situations where anorchia or testicular failure is present.
niques. Human corpus cavernosal muscle and endothelial Microencapsulated Leydig cells offer several advantages.
cells were derived from donor penile tissue, and the cells For example, the encapsulation process creates a semi-
were expanded in vitro and seeded on the acellular permeable barrier between the transplanted cells and the
matrices. The matrices were covered with the appropriate host’s immune system, and it allows for the long-term
cell architecture four weeks after implantation [141]. physiologic release of testosterone.

In order to look at the functional parameters of the Other studies have shown that testicular prostheses cre-
engineered corpora, acellular corporal collagen matrices ated with chondrocytes in bioreactors could be loaded
were obtained from donor rabbit penis and autologous with testosterone, and that these prostheses could provide
corpus cavernosal smooth muscle and endothelial cells controlled testosterone release into the bloodstream
were harvested, expanded and seeded on the matrices. An when implanted. The prostheses were implanted in
entire cross-sectional segment of protruding rabbit phal- athymic mice with bilateral anorchia, and testosterone
lus was excised, leaving the urethra intact. Cell-seeded was released long term, maintaining the androgen level at
matrices were interposed into the excised corporal space. a physiologic range [143]. One could envision combining
Functional and structural parameters (cavernosography, the Leydig cell technology described above with engin-
cavernosometry, mating behavior, and sperm ejaculation) eered prostheses for the long-term functional replace-
were followed, and histological, immunocytochemical, ment of androgen levels.
and Western blot analyses were performed up to six
months after implantation. The engineered corpora Female genital and reproductive tissues
cavernosa achieved adequate structural and functional Congenital malformations of the uterus may have pro-
parameters [142]. This technology was further confirmed found implications clinically. Patients with cloacal exstro-
when the entire rabbit corpora was removed and replaced phy and intersex disorders may not have sufficient uterine
with the engineered scaffolds. Most interestingly, mating tissue present for future reproduction. We investigated
activity in the animals with the engineered corpora the possibility of engineering functional uterine tissue
appeared normal by one month after implantation. The using autologous cells [144]. Autologous rabbit uterine
presence of sperm was confirmed during mating, and was smooth muscle and epithelial cells were harvested, then

www.sciencedirect.com Current Opinion in Biotechnology 2009, 20:575–592


584 Tissue, cell and pathway engineering

grown, and expanded in culture. These cells were seeded poietin have been isolated in culture, and these cells
onto preconfigured uterine-shaped biodegradable poly- could eventually be used to treat anemia that results from
mer scaffolds, and the constructs were used for subtotal end-stage renal failure [158]. Other more ambitious
autologous uterine tissue replacement. Six months after approaches tackle the goal of total renal function replace-
implantation, analyses confirmed the presence of uterine ment. To create kidney tissue that would deliver full
tissue components. Biomechanical analyses and organ renal function, a culture containing all of the cell types
bath studies showed that the functional characteristics comprising the functional nephron units should be used.
of the engineered tissues were similar to those of normal Optimal culture conditions to nurture renal cells have
uterine tissue. Breeding studies using these engineered been extensively studied and cells grown under these
uteri are currently being performed. conditions have been reported to maintain their cellular
characteristics [159]. Cells obtained through the initial
Similarly, several pathologic conditions, including con- process of nuclear transfer were retrieved and expanded
genital malformations and malignancy, can adversely from cloned tissue. Moreover, renal cells placed in a
affect normal vaginal development or anatomy. Vaginal three-dimensional culture environment are able to recon-
reconstruction has traditionally been challenging due to stitute into renal structures.
the paucity of available native tissue. Acellular materials
have been used experimentally for vaginal reconstruction Although isolated renal cells are able to retain their
in rats [145]. The feasibility of using cells to engineer phenotypic and functional characteristics in culture,
vaginal replacements was also investigated [146]. Vaginal transplantation of these cells in vivo may not result in
epithelial and smooth muscle cells of female rabbits were structural remodeling. In addition, cell or tissue com-
harvested, grown, and expanded in culture. These cells ponents cannot be implanted in large volumes due to
were seeded onto biodegradable polymer scaffolds, and limited diffusion of oxygen and nutrients [160]. Thus, a
the cell-seeded constructs were then implanted into cell-support matrix, preferably one that encourages angio-
mice. Functional studies in the tissue-engineered con- genesis, is necessary to allow diffusion across the entire
structs showed similar properties to normal vaginal tissue. implant. A variety of synthetic and naturally derived
When these constructs were used for autologous total materials have been examined in order to determine
vaginal replacement in a rabbit model, patent functional the ideal support structures for regeneration
vaginal structures were noted in the tissue-engineered [43,54,102,126,161]. Biodegradable synthetic materials,
specimens, while the non-cell-seeded structures were such as poly-lactic and poly-glycolic acid polymers, have
noted to be stenotic [147]. These studies indicated that been used to provide structural support for cells. Syn-
a regenerative medicine approach to clinical vaginal re- thetic materials can be easily fabricated and configured in
construction would be a realistic possibility. Clinical trials a controlled manner, which make them attractive options
are currently being conducted. for regenerative medicine. However, naturally derived
materials, such as collagen, laminin, and fibronectin, are
Kidney much more biocompatible and provide a similar extra-
Although the kidney was the first organ to be substituted cellular matrix environment to normal tissue. For this
by an artificial device and the first successfully trans- reason, collagen-based scaffolds have been used increas-
planted organ [2], current modalities of treatment are far ingly in many applications [102,104,126,141,162].
from satisfactory. Renal tissue is arguably one of the most
difficult tissues to replicate in the laboratory. The kidney The feasibility of achieving renal cell growth, expansion,
is very complex and the unique structural and cellular and in vivo reconstitution using regenerative medicine
heterogeneity present within it creates many challenges. techniques was investigated [43]. Donor rabbit kidney
The system of nephrons and collecting ducts within the cells, including distal tubules, glomeruli, and proximal
kidney is composed of multiple functionally and morpho- tubules were plated separately in vitro and after expan-
logically distinct segments. For this reason, appropriate sion, were seeded onto biodegradable poly-glycolic acid
conditions must be provided to ensure the long-term scaffolds and implanted subcutaneously into host athymic
survival, differentiation, and growth of many types of mice. This included implants of proximal tubular cells,
cells at once. Efforts in the area of kidney tissue regen- glomeruli, distal tubular cells, and a mixture of all three
eration have focused on the development of a reliable cell cell types. Histologic examination demonstrated pro-
source [148–153]. Moreover, optimal growth conditions gressive formation and organization of the nephron seg-
have been extensively investigated to provide adequate ments within the polymer fibers with time. BrdU
enrichment to achieve stable renal cell expansion systems incorporation into renal cell DNA was confirmed. These
[154,155–157]. results demonstrated that renal specific cells can be
successfully harvested and cultured, and can sub-
Isolation of particular cell types that produce specific sequently attach to artificial biodegradable polymers.
factors may be a good approach for selective cell therapies However, it was unclear whether the tubular structures
for renal failure. For example, cells that produce erythro- reconstituted de novo from dispersed renal elements, or if

Current Opinion in Biotechnology 2009, 20:575–592 www.sciencedirect.com


Engineering organs Atala 585

Figure 4

Combining therapeutic cloning and tissue engineering to produce kidney tissue. (A) Illustration of the tissue-engineered renal unit. (B) Renal unit
seeded with cloned cells, three months after implantation, showing the accumulation of urine-like fluid. (C) Clear unidirectional continuity between the
mature glomeruli, their tubules, and silastic catheter. (D) Elispot analyses of the frequencies of T cells that secrete IFNg after stimulation with allogenic
renal cells, cloned renal cells, or nuclear donor fibroblasts. Cloned renal cells produce fewer IFNg spots than the allogenic cells, indicating that the
rejection response to cloned cells is diminished. The presented wells are single representatives of duplicate wells.

they merely represented fragments of donor tubules cells of the thin ascending loop of Henle, stained the
which survived the original dissociation and culture pro- tubular sections. Immunohistochemical staining for
cesses intact. Further investigation was conducted in alkaline phosphatase stained proximal tubule-like struc-
order to examine the process [163]. Mouse renal cells tures. Uniform staining for fibronectin in the extracellular
were harvested and expanded in culture. Subsequently, matrix of newly formed tubes was observed. The fluid
single isolated cells were seeded on biodegradable poly- collected from the reservoir was yellow and contained
mers and implanted into immune competent syngeneic 66 mg/dl uric acid (as compared to 2 mg/dl in plasma)
hosts. Renal epithelial cells were observed to reconstitute suggesting that these tubules are capable of uni-
into tubular structures in vivo. Sequential analyses of the directional secretion and concentration of uric acid.
retrieved implants over time demonstrated that renal The creatinine assay performed on the collected fluid
epithelial cells first organized into a cord-like structure showed an 8.2-fold increase in concentration, as compared
with a solid center. Subsequent canalization into a hollow to serum. These results demonstrated that single cells
tube could be seen by two weeks. Histologic examination forming multicellular structures can become organized
with nephron segment specific lactins showed successful into functional renal units that are able to excrete high
reconstitution of proximal tubules, distal tubules, loop of levels of solutes through a urine-like fluid [164].
Henle, collecting tubules, and collecting ducts. These
results showed that single suspended cells are capable of To determine whether renal tissue could be formed using
reconstituting into tubular structures, with homogeneous an alternative cell source, nuclear transplantation (thera-
cell types within each tubule. peutic cloning) was performed to generate histocompa-
tible tissues, and the feasibility of engineering syngeneic
In a subsequent study mouse renal cells were harvested, renal tissues in vivo using these cloned cells was inves-
expanded in culture, and seeded onto a tubular device tigated (Figure 4) [159]. Nuclear material from bovine
constructed from polycarbonate [164]. The tubular device dermal fibroblasts was transferred into unfertilized enu-
was connected at one end to a silastic catheter which cleated donor bovine eggs. Renal cells from the cloned
terminated into a reservoir. The device was implanted embryos were harvested, expanded in vitro, and seeded
subcutaneously in athymic mice. Histological examin- onto three-dimensional renal devices. The devices were
ation of the implanted device demonstrated extensive implanted into the back of the same steer from which the
vascularization as well as formation of glomeruli and cells were cloned, and were retrieved 12 weeks later. This
highly organized tubule-like structures. Immunocyto- process produced functioning renal units. Urine pro-
chemical staining with anti-osteopontin antibody, which duction and viability were demonstrated after transplan-
is secreted by proximal and distal tubular cells and the tation back into the nuclear donor animal. Chemical

www.sciencedirect.com Current Opinion in Biotechnology 2009, 20:575–592


586 Tissue, cell and pathway engineering

analysis suggested unidirectional secretion and concen- pulmonary artery that had been previously repaired.
tration of urea nitrogen and creatinine. Microscopic Seven months after implantation, no evidence of graft
analysis revealed the formation of organized glomeruli occlusion or aneurysmal changes was noted in the reci-
and tubular structures. Immunohistochemical and RT- pient.
PCR analysis confirmed the expression of renal mRNA
and proteins. These studies demonstrated that cells Heart
derived from nuclear transfer can be successfully har- In the United States, over five million people currently
vested, expanded in culture, and transplanted in vivo with live with some form of heart disease, and many more are
the use of biodegradable scaffolds on which the single diagnosed each year. While many medications have been
suspended cells can organize into tissue structures that developed to assist the ailing heart, the treatment for end-
are genetically identical to that of the host. These studies stage heart failure still remains transplantation. Unfortu-
were the first demonstration of the use of therapeutic nately, as with other organs, donor hearts are in short
cloning for the regeneration of tissues in vivo. supply, and even when a transplant can be performed, the
patient must endure the side effects created by lifelong
A naturally derived tissue matrix with existing three- immunosuppression. Thus, alternatives are desperately
dimensional kidney architecture would be preferable to needed, and the development of novel methods to regen-
the artificial matrix used in the above experiments, erate or replace damaged heart muscle using tissue engin-
because it would allow for transplantation of a larger eering and regenerative medicine techniques is an
number of cells, resulting in greater renal tissue volumes. attractive option.
Thus, an acellular collagen-based kidney matrix, which is
similar to the native renal architecture, was developed. In Cell therapy for infracted areas of the heart is attractive, as
a subsequent study it was investigated whether these these methods involve a rather simple injection into the
collagen-based matrices could accommodate large damaged area of a patient’s heart, rather than a rigorous
volumes of renal cells and form kidney structures in vivo surgical procedure, to complete. Various types of stem
[165]. cells have been investigated for their potential to regen-
erate damaged or dead heart tissue in this manner.
Acellular collagen matrices, derived from porcine kid- Skeletal muscle cells, bone marrow stem cells (both
neys, were obtained through a multi-step decellulariza- mesenchymal and hematopoietic), amniotic fluid stem
tion process. During this process, serial evaluation of the cells, and embryonic stem cells have been used for this
matrix for cellular remnants was performed using histo- purpose. In this technique, cells are suspended in a
chemistry, scanning electron microscopy (SEM), and RT- biocompatible matrix that can range from simple normal
PCR. Mouse renal cells were harvested, grown, and saline to complex yet biocompatible hydrogels depending
seeded on decellularized collagen matrices. Cell-matrix on the type of injection to be performed. The cells are
constructs grown in vitro and implanted in the subcu- injected either into the damaged area of the heart itself, or
taneous space of 20 athymic mice were analyzed over into the coronary circulation with the hope that they will
time. Renal cells seeded on the matrix adhered to the home to the damaged area, take up residence there, and
inner surface and proliferated to confluency by seven days begin to repair the tissue. However, injectable therapies
after seeding. Renal tubular and glomerulus-like struc- have been shown to be relatively inefficient, and cell loss
tures were observed eight weeks after implantation. is quite substantial. Newer methods of tissue engineering
include the development of engineered ‘patches,’ which
Blood vessels are comprised of cells adhered to a biomaterial, that can
Xenogenic or synthetic materials have been used as theoretically be used to replace the damaged area of the
replacement blood vessels for complex cardiovascular heart. These techniques have promise, but require
lesions. However, these materials typically lack growth further research into the optimal cell types and biomater-
potential, and may place the recipient at risk for compli- ials for this purpose before they can be used extensively
cations such as stenosis, thromboembolization, or infec- in the clinic (see [172] for an excellent review of these
tion [166]. Tissue-engineered vascular grafts have been methods).
constructed using autologous cells and biodegradable
scaffolds and have been applied in dog and lamb models However, the methods described above could be used
(Figure 5) [167–170]. The key advantage of using these only in cases where a relatively small section of heart
autografts is that they degrade in vivo, and thus allow the muscle was damaged. In cases where a large area or even
new tissue to form without the long-term presence of the whole heart has become nonfunctional, a more radical
foreign material [166]. Application of these techniques approach may be required. In these situations, the use of a
from the laboratory to the clinical setting has begun, with bioartificial heart would be ideal, as rejection would be
autologous vascular cells harvested, expanded, and avoided and the problems associated with a mechanical
seeded onto a biodegradable scaffold [171]. The resultant heart (such as thromboembolus formation) would be
autologous construct was used to replace a stenosed abolished. To this end, Ott et al. recently developed a

Current Opinion in Biotechnology 2009, 20:575–592 www.sciencedirect.com


Engineering organs Atala 587

Figure 5

Studies of an engineered blood vessel implanted in a sheep. (A) An arteriogram showing the engineered vessel graft 130 days after implantation (arrows
indicate graft position). (B) A macroscopic view of the engineered vessel, which consists of a decellularized porcine iliac artery reseeded with endothelial
progenitor cells (EPC) derived from the peripheral blood of the sheep to receive the graft. (C) Microscopic view of graft explanted at day 15
postimplantation indicates a poorly organized thrombotic deposit over the acellular graft media. (D) Microscopic view of the graft at 130 days
postimplantation indicates cellular intimal thickening as pannus from adjacent arterial segments, infiltration of cells into the graft, and a confluent, flattened
cell monolayer lining the luminal surface of the graft (arrow). (E) Section of the 130-day graft stained for extracellular matrix shows preservation of anatomic
features, especially elastin (black, indicated with arrow). (F) Cell staining within the intimal layer (indicated by the letter ‘I’ and in the inset) for smooth muscle
a-actin at 130 days. Note that the medial layer, indicated by the letter ‘m’ does not stain for a-actin. (G) Staining for von Willebrand factor (vWF), an
endothelial cell marker. (H) Preimplant graft seeded with endothelial cells labeled with a fluorescent tracer. (I and J) Fifteen-day and 130-day explanted
grafts still contain a layer of seeded endothelial cells, as indicated by the presence of the fluorescent cells along the lumen of the graft. In H, I, and J, the
lumen is oriented to the left of the photomicrograph. Magnifications in B, C, and the F inset, 200, in D and F, 100, in G, 400, and in H, I, and J, 400.

novel heart construct in vitro using decellularized cada- for liver failure is liver transplantation. However, this
veric hearts. By reseeding the tissue scaffold that therapy is limited by the shortage of donors and the need
remained after a specialized decellularization process for lifelong immunosuppressive therapy. Cell transplan-
with various types of cells that make up a heart (cardi- tation has been proposed as a potential solution for liver
omyocytes, smooth muscle cells, endothelial cells, and failure. This is based on the fact that the liver has
fibrocytes) and culturing the resulting construct in a enormous regenerative potential in vivo, which suggests
bioreactor system designed to mimic physiologic con- that in the right environment, it may be possible to
ditions, this group was able to produce a construct that expand liver cells in vitro in sufficient quantities for tissue
could generate pump function on its own [173]. This engineering [174]. Many approaches have been tried,
study suggests that production of bioartificial hearts may including development of specialized media, coculture
one day be possible. with other cell types, identification of growth factors that
have proliferative effects on these cells, and culture on
Liver three-dimensional scaffolds within bioreactors [174].
The liver can sustain a variety of insults, including viral
infection, alcohol abuse, surgical resection of tumors, and Extracorporeal bioartificial liver devices that use porcine
acute drug-induced hepatic failure. The current therapy hepatocytes have been designed and applied. These

www.sciencedirect.com Current Opinion in Biotechnology 2009, 20:575–592


588 Tissue, cell and pathway engineering

devices are designed to filter and purify the patient’s 5. Wilmut I, Schnieke AE, McWhir J, Kind AJ, Campbell KH: Viable
offspring derived from fetal and adult mammalian cells.. [see
blood as would the patient’s own liver, and the blood is comment] Nature 1997, 385:810-813 [Erratum in Nature
returned to the patient in a manner similar to kidney 1997;386(6621):200].
dialysis. Another cell-based approach is the injection of 6. Haseltine W: A brave new medicine. A conversation with
William Haseltine Interview by Joe Flower. Health Forum J 1999,
liver cell suspensions. This has been performed in animal 42:28-30.
models. Intraportal hepatocyte injection has also been
7. Brivanlou AH, Gage FH, Jaenisch R, Jessell T, Melton D,
used in patients with Crigler–Najjar Syndrome Type 1 Rossant J: Stem cells. Setting standards for human embryonic
[175]; however, complications such as portal vein throm- stem cells.. [see comment] Science 2003, 300:913-916.
bosis and pulmonary embolism are major concerns, 8. Richards M, Fong CY, Chan WK, Wong PC, Bongso A: Human
especially when large cell numbers are used [176]. feeders support prolonged undifferentiated growth of human
inner cell masses and embryonic stem cells.. [see comment]
Finally, cells including stem cells, oval progenitor cells, Nat Biotechnol 2002, 20:933-936.
and mature hepatocytes have been seeded onto liver
9. Amit M, Shariki C, Margulets V, Itskovitz-Eldor J: Feeder layer-
shaped biocompatible matrices to engineer artificial, and serum-free culture of human embryonic stem cells. Biol
implantable livers. These have been tested in various Reprod 2004, 70:837-845.
animal models [177,178]; however, the transplantation 10. Reubinoff BE, Pera MF, Fong CY, Trounson A, Bongso A:
efficiency as well as the functionality of these constructs Embryonic stem cell lines from human blastocysts: somatic
differentiation in vitro.. [see comment] Nat Biotechnol 2000,
must be improved substantially before the technology can 18:399-404 [Erratum in Nat Biotechnol 2000;18(5):559].
be moved into the clinic. 11. Reubinoff BE, Itsykson P, Turetsky T, Pera MF, Reinhartz E, Itzik A,
Ben-Hur T: Neural progenitors from human embryonic stem
Summary and conclusions cells.. [see comment] Nat Biotechnol 2001, 19:1134-1140.
Regenerative medicine efforts are currently underway 12. Schuldiner M, Eiges R, Eden A, Yanuka O, Itskovitz-Eldor J,
Goldstein RS, Benvenisty N: Induced neuronal differentiation of
experimentally for virtually every type of tissue and organ human embryonic stem cells. Brain Res 2001, 913:201-205.
within the human body. As regenerative medicine incorp-
13. Schuldiner M, Yanuka O, Itskovitz-Eldor J, Melton DA,
orates the fields of tissue engineering, cell biology, Benvenisty N: Effects of eight growth factors on the
nuclear transfer, and materials science, personnel who differentiation of cells derived from human embryonic stem
cells. Proc Natl Acad Sci U S A 2000, 97:11307-11312.
have mastered the techniques of cell harvest, culture,
expansion, transplantation, as well as polymer design are 14. Zhang SC, Wernig M, Duncan ID, Brustle O, Thomson JA: In vitro
differentiation of transplantable neural precursors from
essential for the successful application of these technol- human embryonic stem cells.. [see comment] Nat Biotechnol
ogies to extend human life. Various tissues are at different 2001, 19:1129-1133.
stages of development, with some already being used 15. Kaufman DS, Hanson ET, Lewis RL, Auerbach R, Thomson JA:
clinically, a few in preclinical trials, and some in the Hematopoietic colony-forming cells derived from human
embryonic stem cells. Proc Natl Acad Sci U S A 2001,
discovery stage. Recent progress suggests that engineered 98:10716-10721.
tissues may have an expanded clinical applicability in the
16. Kehat I, Kenyagin-Karsenti D, Snir M, Segev H, Amit M,
future and may represent a viable therapeutic option for Gepstein A, Livne E, Binah O, Itskovitz-Eldor J, Gepstein L: Human
those who would benefit from the life-extending benefits embryonic stem cells can differentiate into myocytes with
structural and functional properties of cardiomyocytes.. [see
of tissue replacement or repair. comment] J Clin Invest 2001, 108:407-414.
17. Levenberg S, Golub JS, Amit M, Itskovitz-Eldor J, Langer R:
Acknowledgment Endothelial cells derived from human embryonic stem cells.
The author wishes to thank Jennifer L Olson, PhD, for editorial assistance Proc Natl Acad Sci U S A 2002, 99:4391-4396.
with this manuscript.
18. Assady S, Maor G, Amit M, Itskovitz-Eldor J, Skorecki KL,
Tzukerman M: Insulin production by human embryonic stem
References and recommended reading cells. Diabetes 2001, 50:1691-1697.
Papers of particular interest, published within the period of review,
have been highlighted as: 19. Itskovitz-Eldor J, Schuldiner M, Karsenti D, Eden A, Yanuka O,
Amit M, Soreq H, Benvenisty N: Differentiation of human
embryonic stem cells into embryoid bodies compromising the
 of special interest three embryonic germ layers. Mol Med 2000, 6:88-95.
 of outstanding interest
20. Colman A, Kind A: Therapeutic cloning: concepts and
practicalities. Trends Biotechnol 2000, 18:192-196.
1. Carrel A, Lindbergh CA: The culture of whole organs. Science 21. Vogelstein B, Alberts B, Shine K: Genetics. Please don’t call it
1935, 81:621-623. cloning!. [see comment] Science 2002, 295:1237.
2. Guild WR, Harrison JH, Merrill JP, Murray J: Successful 22. Hochedlinger K, Rideout WM, Kyba M, Daley GQ, Blelloch R,
homotransplantation of the kidney in an identical twin. Trans Jaenisch R: Nuclear transplantation, embryonic stem cells and
Am Clin Climatol Assoc 1955, 67:167-173. the potential for cell therapy. Hematol J 2004, 5(Suppl 3):S114-
S117.
3. Martin GR: Isolation of a pluripotent cell line from early mouse
embryos cultured in medium conditioned by teratocarcinoma 23. Dinnyes A, De Sousa P, King T, Wilmut I: Somatic cell nuclear
stem cells. Proc Natl Acad Sci U S A 1981, 78:7634-7638. transfer: recent progress and challenges. Clon Stem Cells 2002,
4:81-90.
4. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA,
Swiergiel JJ, Marshall VS, Jones JM: Embryonic stem cell lines 24. Takahashi K, Yamanaka S: Induction of pluripotent stem cells
derived from human blastocysts.. [see comment] Science 1998, from mouse embryonic and adult fibroblast cultures by
282:1145-1147 [Erratum in Science 1998;282(5395):1827]. defined factors. Cell 2006, 126:663-676.

Current Opinion in Biotechnology 2009, 20:575–592 www.sciencedirect.com


Engineering organs Atala 589

25. Wernig M, Meissner A, Foreman R, Brambrink T, Ku M, 45. Atala A: Tissue engineering in the genitourinary system. In
Hochedlinger K, Bernstein BE, Jaenisch R: In vitro Tissue Engineering. Edited by Atala A, Mooney DJ. Birkhauser
reprogramming of fibroblasts into a pluripotent ES-cell-like Press; 1997:149.
state. Nature 2007, 448:318-324.
46. Atala A: Autologous cell transplantation for urologic
26. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, reconstruction. J Urol 1998, 159:2-3.
Yamanaka S: Induction of pluripotent stem cells from adult
human fibroblasts by defined factors. Cell 2007, 131:861-872. 47. Yoo JJ, Atala A: A novel gene delivery system using urothelial
tissue engineered neo-organs. J Urol 1997, 158:1066-1070.
27. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J,
Frane JL, Tian S, Nie J, Jonsdottir GA, Ruotti V, Stewart R et al.: 48. Fauza DO, Fishman SJ, Mehegan K, Atala A: Videofetoscopically
Induced pluripotent stem cell lines derived from human assisted fetal tissue engineering: skin replacement.
somatic cells. Science 2007, 318:1917-1920. J Pediatr Surg 1998, 33:357-361.

28. De Coppi P, Bartsch G Jr, Siddiqui MM, Xu T, Santos CC, Perin L, 49. Fauza DO, Fishman SJ, Mehegan K, Atala A: Videofetoscopically
Mostoslavsky G, Serre AC, Snyder EY, Yoo JJ et al.: Isolation of assisted fetal tissue engineering: bladder augmentation.
amniotic stem cell lines with potential for therapy.. [see J Pediatr Surg 1998, 33:7-12.
comment] Nat Biotechnol 2007, 25:100-106.
50. Machluf M, Atala A: Emerging concepts for tissue and organ
29. Ballas CB, Zielske SP, Gerson SL: Adult bone marrow stem cells transplantation. Graft 1998, 1:31-37.
for cell and gene therapies: implications for greater use. J Cell
Biochem Suppl 2002, 38:20-28. 51. Yoo JJ, Meng J, Oberpenning F, Atala A: Bladder augmentation
using allogenic bladder submucosa seeded with cells. Urology
30. Jiao J, Chen DF: Induction of neurogenesis in nonconventional 1998, 51:221-225.
neurogenic regions of the adult central nervous system by niche
astrocyte-produced signals. Stem Cells 2008, 26:1221-1230. 52. Amiel GE, Atala A: Current and future modalities for functional
renal replacement. Urol Clin N Am 1999, 26:235-246.
31. Taupin P: Therapeutic potential of adult neural stem cells.
Recent Patents CNS Drug Discov 2006, 1:299-303. 53. Kershen RT, Atala A: New advances in injectable therapies for
the treatment of incontinence and vesicoureteral reflux. Urol
32. Jensen UB, Yan X, Triel C, Woo SH, Christensen R, Owens DM: A Clin N Am 1999, 26:81-94.
distinct population of clonogenic and multipotent murine
follicular keratinocytes residing in the upper isthmus. J Cell Sci 54. Oberpenning F, Meng J, Yoo JJ, Atala A: De novo reconstitution
2008, 121:609-617. of a functional mammalian urinary bladder by tissue
engineering.. [see comment] Nat Biotechnol 1999, 17:149-155.
33. Crisan M, Casteilla L, Lehr L, Carmona M, Paoloni-Giacobino A,
Yap S, Sun B, Leger B, Logar A, Penicaud L et al.: A reservoir of 55. Park HJ, Yoo JJ, Kershen RT, Moreland R, Atala A:
brown adipocyte progenitors in human skeletal muscle. Stem Reconstitution of human corporal smooth muscle and
Cells 2008, 26:2425-2433. endothelial cells in vivo. J Urol 1999, 162:1106-1109.

34. Weiner LP: Definitions and criteria for stem cells. Methods Mol 56. Liebert M, Wedemeyer G, Abruzzo LV, Kunkel SL, Hammerberg C,
Biol 2008, 438:3-8. Cooper KD, Grossman HB: Stimulated urothelial cells produce
cytokines and express an activated cell surface antigenic
35. Cilento BG, Freeman MR, Schneck FX, Retik AB, Atala A: phenotype. Semin Urol 1991, 9:124-130.
Phenotypic and cytogenetic characterization of human
bladder urothelia expanded in vitro. J Urol 1994, 152:665-670. 57. Tobin MS, Freeman MR, Atala A: Maturational response of normal
human urothelial cells in culture is dependent on extracellular
36. Scriven SD, Booth C, Thomas DF, Trejdosiewicz LK, Southgate J: matrix and serum additives. Surg Forum 1994, 45:786.
Reconstitution of human urothelium from monolayer cultures.
J Urol 1997, 158:1147-1152. 58. Harriss DR: Smooth muscle cell culture: a new approach to the
study of human detrusor physiology and pathophysiology.
37. Liebert M, Hubbel A, Chung M, Wedemeyer G, Lomax MI, Br J Urol 1995, 75(Suppl 1):18-26.
Hegeman A, Yuan TY, Brozovich M, Wheelock MJ, Grossman HB:
Expression of mal is associated with urothelial differentiation 59. Freeman MR, Yoo JJ, Raab G, Soker S, Adam RM, Schneck FX,
in vitro: identification by differential display reverse- Renshaw AA, Klagsbrun M, Atala A: Heparin-binding EGF-like
transcriptase polymerase chain reaction. Differentiation 1997, growth factor is an autocrine growth factor for human urothelial
61:177-185. cells and is synthesized by epithelial and smooth muscle cells in
the human bladder. J Clin Invest 1997, 99:1028-1036.
38. Puthenveettil JA, Burger MS, Reznikoff CA: Replicative
senescence in human uroepithelial cells. Adv Exp Med Biol 60. Solomon LZ, Jennings AM, Sharpe P, Cooper AJ, Malone PS:
1999, 462:83-91. Effects of short-chain fatty acids on primary urothelial cells in
culture: implications for intravesical use in
39. Atala A, Vacanti JP, Peters CA, Mandell J, Retik AB, Freeman MR: enterocystoplasties.. [see comment] J Lab Clin Med 1998,
Formation of urothelial structures in vivo from dissociated 132:279-283.
cells attached to biodegradable polymer scaffolds in vitro. J
Urol 1992, 148:658-662. 61. Lobban ED, Smith BA, Hall GD, Harnden P, Roberts P, Selby PJ,
Trejdosiewicz LK, Southgate J: Uroplakin gene expression by
40. Atala A, Cima LG, Kim W, Paige KT, Vacanti JP, Retik AB, normal and neoplastic human urothelium. Am J Pathol 1998,
Vacanti CA: Injectable alginate seeded with chondrocytes as a 153:1957-1967.
potential treatment for vesicoureteral reflux. J Urol 1993,
150:745-747. 62. Nguyen HT, Park JM, Peters CA, Adam RM, Orsola A, Atala A,
Freeman MR: Cell-specific activation of the HB-EGF and ErbB1
41. Atala A, Freeman MR, Vacanti JP, Shepard J, Retik AB: genes by stretch in primary human bladder cells. In Vitro Cell
Implantation in vivo and retrieval of artificial structures Dev Biol Anim 1999, 35:371-375.
consisting of rabbit and human urothelium and human bladder
muscle. J Urol 1993, 150:608-612. 63. Rackley RR, Bandyopadhyay SK, Fazeli-Matin S, Shin MS,
Appell R: Immunoregulatory potential of urothelium:
42. Atala A, Kim W, Paige KT, Vacanti CA, Retik AB: Endoscopic characterization of NF-kappaB signal transduction. J Urol
treatment of vesicoureteral reflux with a chondrocyte-alginate 1999, 162:1812-1816.
suspension. J Urol 1994, 152:641-643 [discussion 644].
64. Kim BS, Mooney DJ: Development of biocompatible synthetic
43. Atala A, Schlussel RN, Retik AB: Renal cell growth in vivo after extracellular matrices for tissue engineering. Trends
attachment to biodegradable polymer scaffolds. J Urol 1995, Biotechnol 1998, 16:224-230.
153.
65. Li ST: Biologic biomaterials: tissue derived biomaterials
44. Atala A, Guzman L, Retik AB: A novel inert collagen matrix for (collagen). In The Biomedical Engineering Handbook. Edited by
hypospadias repair. J Urol 1999, 162:1148-1151. JD B. CRS Press; 1995:627-647.

www.sciencedirect.com Current Opinion in Biotechnology 2009, 20:575–592


590 Tissue, cell and pathway engineering

66. Furthmayr H, Timpl R: Immunochemistry of collagens and 89. Choi JS, Lee SJ, Christ GJ, Atala A, Yoo JJ: The influence of
procollagens. Int Rev Connect Tissue Res 1976, 7:61-99. electrospun aligned poly(epsilon-caprolactone)/collagen
nanofiber meshes on the formation of self-aligned skeletal
67. Cen L, Liu W, Cui L, Zhang W, Cao Y: Collagen tissue muscle myotubes. Biomaterials 2008, 29:2899-2906.
engineering: development of novel biomaterials and
applications. Pediatr Res 2008, 63:492-496. 90. Lee SJ, Liu J, Oh SH, Soker S, Atala A, Yoo JJ: Development of a
composite vascular scaffolding system that withstands
68. Yannas IV, Burke JF, Gordon PL, Huang C, Rubenstein RH: physiological vascular conditions. Biomaterials 2008,
Design of an artificial skin. II. Control of chemical composition. 29:2891-2898.
J Biomed Mater Res 1980, 14:107-132.
91. Lee SJ, Oh SH, Liu J, Soker S, Atala A, Yoo JJ: The use of thermal
69. Yannas IV, Burke JF: Design of an artificial skin. I. Basic design treatments to enhance the mechanical properties of
principles. J Biomed Mater Res 1980, 14:65-81. electrospun poly(epsilon-caprolactone) scaffolds.
Biomaterials 2008, 29:1422-1430.
70. Cavallaro JF, Kemp PD, Kraus KH: Collagen fabrics as
biomaterials. Biotechnol Bioeng 1994, 43:781-791. 92. Barrera DA, Zylstra E, Lansbury PT, Langer R: Synthesis and RGD
peptide modification of a new biodegradable copolymer
71. Smidsrod O, Skjak-Braek G: Alginate as immobilization matrix poly (lactic acid-co-lysine). J Am Chem Soc 1993,
for cells. Trends Biotechnol 1990, 8:71-78. 115:11010-11011.
72. Rowley JA, Madlambayan G, Mooney DJ: Alginate hydrogels as 93. Cook AD, Hrkach JS, Gao NN, Johnson IM, Pajvani UB,
synthetic extracellular matrix materials. Biomaterials 1999, Cannizzaro SM, Langer R: Characterization and development of
20:45-53. RGD-peptide-modified poly(lactic acid-co-lysine) as an
73. Campbell PG, Weiss LE: Tissue engineering with the aid of interactive, resorbable biomaterial. J Biomed Mater Res 1997,
inkjet printers. Expert Opin Biol Ther 2007, 7:1123-1127. 35:513-523.

74. Boland T, Xu T, Damon B, Cui X: Application of inkjet printing to 94. Intveld PJA, Shen ZR, Takens GAJ: Glycine glycolic acid based
tissue engineering. Biotechnol J 2006, 1:910-917. copolymers. J Polym Sci Polym Chem 1994, 32:1063-1069.

75. Nakamura M, Kobayashi A, Takagi F, Watanabe A, Hiruma Y, 95. Peppas NA, Langer R: New challenges in biomaterials.. [see
Ohuchi K, Iwasaki Y, Horie M, Morita I, Takatani S: Biocompatible comment] Science 1994, 263:1715-1720.
inkjet printing technique for designed seeding of individual 96. Boccaccini AR, Blaker JJ: Bioactive composite materials for
living cells. Tissue Eng 2005, 11:1658-1666. tissue engineering scaffolds. Expert Rev Med Devices 2005,
76. Laflamme MA, Gold J, Xu C, Hassanipour M, Rosler E, Police S, 2:303-317.
Muskheli V, Murry CE: Formation of human myocardium in the 97. Harrison BS, Atala A: Carbon nanotube applications for tissue
rat heart from human embryonic stem cells. Am J Pathol 2005, engineering. Biomaterials 2007, 28:344-353.
167:663-671.
98. Harrington DA, Cheng EY, Guler MO, Lee LK, Donovan JL,
77. Xu T, Rohozinski J, Zhao W, Moorefield EC, Atala A, Yoo JJ: Claussen RC, Stupp SI: Branched peptide-amphiphiles as
Inkjet-mediated gene transfection into living cells combined
self-assembling coatings for tissue engineering scaffolds.
with targeted delivery. Tissue Eng Part A 2009, 15:95-101. J Biomed Mater Res A 2006, 78:157-167.
78. Ilkhanizadeh S, Teixeira AI, Hermanson O: Inkjet printing of
99. Bazeed MA, Thuroff JW, Schmidt RA, Tanagho EA: New
macromolecules on hydrogels to steer neural stem cell
treatment for urethral strictures. Urology 1983, 21:53-57.
differentiation. Biomaterials 2007, 28:3936-3943.
100. Olsen L, Bowald S, Busch C, Carlsten J, Eriksson I: Urethral
79. Roth EA, Xu T, Das M, Gregory C, Hickman JJ, Boland T: Inkjet
reconstruction with a new synthetic absorbable device. An
printing for high-throughput cell patterning. Biomaterials 2004,
experimental study. Scand J Urol Nephrol 1992, 26:323-326.
25:3707-3715.
101. Sievert KD, Bakircioglu ME, Nunes L, Tu R, Dahiya R, Tanagho EA:
80. Dahms SE, Piechota HJ, Dahiya R, Lue TF, Tanagho EA:
Homologous acellular matrix graft for urethral reconstruction
Composition and biomechanical properties of the bladder
in the rabbit: histological and functional evaluation. J Urol
acellular matrix graft: comparative analysis in rat, pig and
2000, 163:1958-1965.
human. Br J Urol 1998, 82:411-419.
102. El-Kassaby AW, Retik AB, Yoo JJ, Atala A: Urethral stricture
81. Piechota HJ, Dahms SE, Nunes LS, Dahiya R, Lue TF, Tanagho EA:
repair with an off-the-shelf collagen matrix. J Urol 2003,
In vitro functional properties of the rat bladder regenerated
169:170-173 [discussion 173].
by the bladder acellular matrix graft. J Urol 1998,
159:1717-1724. 103. El-Kassaby A, Aboushwareb T, Atala A: Randomized
comparative study between buccal mucosal and acellular
82. Chen F, Yoo JJ, Atala A: Acellular collagen matrix as a possible
bladder matrix grafts in complex anterior urethral strictures.
‘‘off the shelf’’ biomaterial for urethral repair. Urology 1999,
J Urol 2008, 179:1432-1436.
54:407-410.
104. De Filippo RE, Yoo JJ, Atala A: Urethral replacement using cell
83. Probst M, Dahiya R, Carrier S, Tanagho EA: Reproduction of
seeded tubularized collagen matrices. J Urol 2002, 168:1789-
functional smooth muscle tissue and partial bladder
1792 [discussion 1792–1783].
replacement. Br J Urol 1997, 79:505-515.
105. le Roux PJ: Endoscopic urethroplasty with unseeded small
84. Gilding D: Biodegradable polymers. In Biocompatibility of
intestinal submucosa collagen matrix grafts: a pilot study.
Clinical Implant Materials. Edited by Williams D. CRC Press; 1981:
209-232.
J Urol 2005, 173:140-143.
85. Freed LE, Vunjak-Novakovic G, Biron RJ, Eagles DB, Lesnoy DC,
Barlow SK, Langer R: Biodegradable polymer scaffolds for 106. Fu Q, Deng CL, Liu W, Cao YL: Urethral replacement using
tissue engineering. Biotechnology (NY) 1994, 12:689-693. epidermal cell-seeded tubular acellular bladder collagen
matrix. BJU Int 2007, 99:1162-1165.
86. Mikos AG, Lyman MD, Freed LE, Langer R: Wetting of poly(L-
lactic acid) and poly(DL-lactic-co-glycolic acid) foams for 107. Li C, Xu Y, Song L, Fu Q, Cui L, Yin S: Preliminary experimental
tissue culture. Biomaterials 1994, 15:55-58. study of tissue-engineered urethral reconstruction
using oral keratinocytes seeded on BAMG. Urol Int 2008,
87. Harris LD, Kim BS, Mooney DJ: Open pore biodegradable 81:290-295.
matrices formed with gas foaming. J Biomed Mater Res 1998,
42:396-402. 108. Guan Y, Ou L, Hu G, Wang H, Xu Y, Chen J, Zhang J, Yu Y, Kong D:
Tissue engineering of urethra using human vascular
88. Han D, Gouma PI: Electrospun bioscaffolds that mimic the endothelial growth factor gene-modified bladder urothelial
topology of extracellular matrix. Nanomedicine 2006, 2:37-41. cells. Artif Organs 2008, 32:91-99.

Current Opinion in Biotechnology 2009, 20:575–592 www.sciencedirect.com


Engineering organs Atala 591

109. Dorin RP, Pohl HG, De Filippo RE, Yoo JJ, Atala A: Tubularized 128. Hipp J, Andersson KE, Kwon TG, Kwak EK, Yoo J, Atala A:
urethral replacement with unseeded matrices: what is the Microarray analysis of exstrophic human bladder smooth
maximum distance for normal tissue regeneration? World J muscle. BJU Int 2008, 101:100-105.
Urol 2008, 26:323-326.
129. Dozmorov MG, Kropp BP, Hurst RE, Cheng EY, Lin HK:
110. Atala A: Commentary on the replacement of urologic Differentially expressed gene networks in cultured smooth
associated mucosa. J Urol 1995, 156:338. muscle cells from normal and neuropathic bladder.
J Smooth Muscle Res 2007, 43:55-72.
111. Sutherland RS, Baskin LS, Hayward SW, Cunha GR:
Regeneration of bladder urothelium, smooth muscle, blood 130. Lai JY, Yoon CY, Yoo JJ, Wulf T, Atala A: Phenotypic and
vessels and nerves into an acellular tissue matrix. J Urol 1996, functional characterization of in vivo tissue engineered
156:571-577. smooth muscle from normal and pathological bladders. J Urol
2002, 168:1853-1857 [discussion 1858].
112. Wefer J, Sievert KD, Schlote N, Wefer AE, Nunes L, Dahiya R,
Gleason CA, Tanagho EA: Time dependent smooth muscle 131. Faris RA, Konkin T, Halpert G: Liver stem cells: a
regeneration and maturation in a bladder acellular matrix potential source of hepatocytes for the treatment
graft: histological studies and in vivo functional evaluation. of human liver disease.. [see comment] Artif Organs 2001,
J Urol 2001, 165:1755-1759. 25:513-521.
113. Kropp BP, Sawyer BD, Shannon HE, Rippy MK, Badylak SF, 132. Haller H, de Groot K, Bahlmann F, Elger M, Fliser D: Stem cells
Adams MC, Keating MA, Rink RC, Thor KB: Characterization of and progenitor cells in renal disease. Kidney Int 2005,
small intestinal submucosa regenerated canine detrusor: 68:1932-1936.
assessment of reinnervation, in vitro compliance and
contractility. J Urol 1996, 156:599-607. 133. Frumpkin AP: Reconstruction of male genitalia. Am Rev Sov
Med 1944, 2:14.
114. Jayo MJ, Jain D, Wagner BJ, Bertram TA: Early cellular and
stromal responses in regeneration versus repair of a 134. Goodwin WE, Scott WW, Phalloplasty: J Urol1952, 68:903-908.
mammalian bladder using autologous cell and biodegradable
scaffold technologies.. [see comment] J Urol 2008, 180:392-397.
135. Bretan PN: History of prosthetic treatment of impotence. In
115. Zhang Y: Bladder reconstruction by tissue engineering — with Genitourinary Prostheses. Edited by Montague DK. WB Saunders;
or without cells?. [see comment] J Urol 2008, 180:10-11. 1989:1-5.
116. Chun SY, Lim GJ, Kwon TG, Kwak EK, Kim BW, Atala A, Yoo JJ: 136. Small MP, Carrion HM, Gordon JA: Small-Carrion penile
Identification and characterization of bioactive factors in prosthesis. New implant for management of impotence.
bladder submucosa matrix. Biomaterials 2007, 28:4251-4256. Urology 1975, 5:479-486.
117. Kikuno N, Kawamoto K, Hirata H, Vejdani K, Kawakami K, 137. Thomalla JV, Thompson ST, Rowland RG, Mulcahy JJ: Infectious
Fandel T, Nunes L, Urakami S, Shiina H, Igawa M et al.: Nerve complications of penile prosthetic implants. J Urol 1987,
growth factor combined with vascular endothelial growth 138:65-67.
factor enhances regeneration of bladder acellular matrix graft
in spinal cord injury-induced neurogenic rat bladder. BJU Int 138. Nukui F, Okamoto S, Nagata M, Kurokawa J, Fukui J:
2008, 103:1424-1428. Complications and reimplantation of penile implants. Int J Urol
1997, 4:52-54.
118. de Boer WI, Schuller AG, Vermey M, van der Kwast TH:
Expression of growth factors and receptors during specific 139. Jordan GH: Penile reconstruction, phallic construction, and
phases in regenerating urothelium after acute injury in vivo. urethral reconstruction. Urol Clin N Am 1999, 26:1-13.
Am J Pathol 1994, 145:1199-1207.
140. Kershen RT, Yoo JJ, Moreland RB, Krane RJ, Atala A:
119. Baker R, Kelly T, Tehan T, Putnam C, Beaugard E: Subtotal Reconstitution of human corpus cavernosum smooth muscle
cystectomy and total bladder regeneration in treatment of in vitro and in vivo. Tissue Eng 2002, 8:515-524.
bladder cancer. J Am Med Assoc 1958, 168:1178-1185.
141. Falke G, Yoo JJ, Kwon TG, Moreland R, Atala A: Formation of
120. Gorham SD, French DA, Shivas AA, Scott R: Some observations corporal tissue architecture in vivo using human cavernosal
on the regeneration of smooth muscle in the repaired urinary muscle and endothelial cells seeded on collagen matrices.
bladder of the rabbit. Eur Urol 1989, 16:440-443. Tissue Eng 2003, 9:871-879.
121. Master VA, Wei G, Liu W, Baskin LS: Urothlelium facilitates the 142. Kwon TG, Yoo JJ, Atala A: Autologous penile corpora
recruitment and trans-differentiation of fibroblasts into cavernosa replacement using tissue engineering techniques.
smooth muscle in acellular matrix. J Urol 2003, 170:1628-1632. J Urol 2002, 168:1754-1758.
122. Jayo MJ, Jain D, Ludlow JW, Payne R, Wagner BJ, McLorie G, 143. Raya-Rivera AM, Baez C, Atala A, Yoo JJ: Tissue engineered
Bertram TA: Long-term durability, tissue regeneration and neo- testicular prostheses with prolonged testosterone release.
organ growth during skeletal maturation with a neo-bladder World J Urol 2008, 26:307-314.
augmentation construct. Regener Med 2008, 3:671-682.
144. Wang T, Koh C, Yoo JJ: Creation of an engineered uterus for
123. Kwon TG, Yoo JJ, Atala A: Local and systemic effects of a tissue surgical reconstruction. In American Academy of Pediatrics
engineered neobladder in a canine cystoplasty model. J Urol Section on Urology; 2003.
2008, 179:2035-2041.
145. Wefer J, Sekido N, Sievert KD, Schlote N, Nunes L, Dahiya R,
124. Zhang Y, Frimberger D, Cheng EY, Lin HK, Kropp BP: Challenges Jonas U, Tanagho EA: Homologous acellular matrix graft for
in a larger bladder replacement with cell-seeded and vaginal repair in rats: a pilot study for a new reconstructive
unseeded small intestinal submucosa grafts in a subtotal approach. World J Urol 2002, 20:260-263.
cystectomy model. BJU Int 2006, 98:1100-1105.
146. De Filippo RE, Yoo JJ, Atala A: Engineering of vaginal tissue in
125. Farhat WA, Yeger H: Does mechanical stimulation have any role vivo. Tissue Eng 2003, 9:301-306.
in urinary bladder tissue engineering? World J Urol 2008,
26:301-305. 147. De Filippo RE, Bishop CE, Filho LF, Yoo JJ, Atala A: Tissue
engineering a complete vaginal replacement from a small
126. Atala A, Bauer SB, Soker S, Yoo JJ, Retik AB: Tissue-engineered biopsy of autologous tissue. Transplantation 2008, 86:208-214
autologous bladders for patients needing cystoplasty.. [see [Erratum in Transplantation 2008;86(5):751. Note: De Philippo,
comment] Lancet 2006, 367:1241-1246. Roger E (corrected to De Filippo, Roger E)].
127. Lin HK, Cowan R, Moore P, Zhang Y, Yang Q, Peterson JA Jr, 148. Ikarashi K, Li B, Suwa M, Kawamura K, Morioka T, Yao J, Khan F,
Tomasek JJ, Kropp BP, Cheng EY: Characterization of Uchiyama M, Oite T: Bone marrow cells contribute to
neuropathic bladder smooth muscle cells in culture. J Urol regeneration of damaged glomerular endothelial cells. Kidney
2004, 171:1348-1352. Int 2005, 67:1925-1933.

www.sciencedirect.com Current Opinion in Biotechnology 2009, 20:575–592


592 Tissue, cell and pathway engineering

149. Kale S, Karihaloo A, Clark PR, Kashgarian M, Krause DS, 165. Amiel GE, Yoo JJ, Atala A: Renal tissue engineering using a
Cantley LG: Bone marrow stem cells contribute to repair of the collagen-based kidney matrix. Tissue Eng Suppl 2000, 6:685.
ischemically injured renal tubule. J Clin Invest 2003, 112:42-49.
166. Matsumura G, Miyagawa-Tomita S, Shin’oka T, Ikada Y,
150. Lin F, Cordes K, Li L, Hood L, Couser WG, Shankland SJ, Kurosawa H: First evidence that bone marrow cells contribute
Igarashi P: Hematopoietic stem cells contribute to the to the construction of tissue-engineered vascular autografts
regeneration of renal tubules after renal ischemia–reperfusion in vivo. Circulation 2003, 108:1729-1734.
injury in mice. J Am Soc Nephrol 2003, 14:1188-1199.
167. Watanabe M, Shin’oka T, Tohyama S, Hibino N, Konuma T,
151. Lin F, Moran A, Igarashi P: Intrarenal cells, not bone marrow- Matsumura G, Kosaka Y, Ishida T, Imai Y, Yamakawa M et al.:
derived cells, are the major source for regeneration in Tissue-engineered vascular autograft: inferior vena
postischemic kidney. J Clin Invest 2005, 115:1756-1764. cava replacement in a dog model. Tissue Eng 2001,
7:429-439.
152. Prockop DJ: Marrow stromal cells as stem cells for
nonhematopoietic tissues. Science 1997, 276:71-74. 168. Shinoka T, Breuer CK, Tanel RE, Zund G, Miura T, Ma PX,
153. Yokoo T, Ohashi T, Shen JS, Sakurai K, Miyazaki Y, Utsunomiya Y, Langer R, Vacanti JP, Mayer JE Jr: Tissue engineering heart
Takahashi M, Terada Y, Eto Y, Kawamura T et al.: Human valves: valve leaflet replacement study in a lamb model. Ann
mesenchymal stem cells in rodent whole-embryo culture are Thorac Surg 1995, 60:S513-516.
reprogrammed to contribute to kidney tissues. Proc Natl Acad 169. Shinoka T, Shum-Tim D, Ma PX, Tanel RE, Isogai N, Langer R,
Sci U S A 2005, 102:3296-3300. Vacanti JP, Mayer JE Jr: Creation of viable pulmonary artery
154. Carley WW, Milici AJ, Madri JA: Extracellular matrix specificity autografts through tissue engineering. J Thorac Cardiovasc
for the differentiation of capillary endothelial cells. Exp Cell Res Surg 1998, 115:536-545 [discussion 545–536].
1988, 178:426-434.
170. Shinoka T, Shum-Tim D, Ma PX, Tanel RE, Langer R, Vacanti JP,
155. Humes HD, Cieslinski DA: Interaction between growth factors Mayer JE Jr: Tissue-engineered heart valve leaflets: does cell
and retinoic acid in the induction of kidney tubulogenesis in origin affect outcome? Circulation 1997, 96: II-102-07.
tissue culture. Exp Cell Res 1992, 201:8-15.
171. Shin’oka T, Imai Y, Ikada Y: Transplantation of a tissue-
156. Milici AJ, Furie MB, Carley WW: The formation of fenestrations engineered pulmonary artery. N Engl J Med 2001, 344:532-533.
and channels by capillary endothelium in vitro. Proc Natl Acad
Sci U S A 1985, 82:6181-6185. 172. Jawad H, Ali NN, Lyon AR, Chen QZ, Harding SE, Boccaccini AR:
Myocardial tissue engineering: a review. J Tissue Eng Regener
157. Schena FP: Role of growth factors in acute renal failure. Kidney Med 2007, 1:327-342.
Int Suppl 1998, 66:S11-15.
173. Ott HC, Matthiesen TS, Goh SK, Black LD, Kren SM, Netoff TI,
158. Aboushwareb T, Egydio F, Straker L, Gyabaah K, Atala A, Yoo JJ: Taylor DA: Perfusion-decellularized matrix: using nature’s
Erythropoietin producing cells for potential cell therapy. World platform to engineer a bioartificial heart. Nat Med 2008.
J Urol 2008, 26:295-300.
174. Bhandari RN, Riccalton LA, Lewis AL, Fry JR, Hammond AH,
159. Lanza RP, Chung HY, Yoo JJ, Wettstein PJ, Blackwell C, Tendler SJ, Shakesheff KM: Liver tissue engineering: a role for
Borson N, Hofmeister E, Schuch G, Soker S, Moraes CT et al.: co-culture systems in modifying hepatocyte function and
Generation of histocompatible tissues using nuclear viability. Tissue Eng 2001, 7:345-357.
transplantation. Nat Biotechnol 2002, 20:689-696.
175. Fox IJ, Chowdhury JR, Kaufman SS, Goertzen TC, Chowdhury NR,
160. Folkman J, Hochberg M: Self-regulation of growth in three Warkentin PI, Dorko K, Sauter BV, Strom SC: Treatment
dimensions. J Exp Med 1973, 138:745-753. of the Crigler–Najjar syndrome type I with hepatocyte
161. Tachibana M, Nagamatsu GR, Addonizio JC: Ureteral transplantation.. [see comment] N Engl J Med 1998,
replacement using collagen sponge tube grafts. J Urol 1985, 338:1422-1426.
133:866-869.
176. Nieto JA, Escandon J, Betancor C, Ramos J, Canton T, Cuervas-
162. Murray MM, Forsythe B, Chen F, Lee SJ, Yoo JJ, Atala A, Mons V: Evidence that temporary complete occlusion of
Steinert A: The effect of thrombin on ACL fibroblast splenic vessels prevents massive embolization and sudden
interactions with collagen hydrogels. J Orthop Res 2006, death associated with intrasplenic hepatocellular
24:508-515. transplantation. Transplantation 1989, 47:449-450.

163. Fung LCT, Elenius K, Freeman M, Donovan MJ, Atala A: 177. Kaufmann PM, Kneser U, Fiegel HC, Kluth D, Herbst H, Rogiers X:
Reconstitution of poor EGFr-poor renal epithelial cells into Long-term hepatocyte transplantation using three-
tubular structures on biodegradable polymer scaffold. dimensional matrices. Transplant Proc 1999, 31:1928-1929.
Pediatrics 1996, 98(Suppl):S631.
178. Gilbert JC, Takada T, Stein JE, Langer R, Vacanti JP: Cell
164. Yoo J, Ashkar S, Atala A: Creation of functional kidney transplantation of genetically altered cells on biodegradable
structures with excretion of kidney-like fluid in vivo. Pediatrics polymer scaffolds in syngeneic rats. Transplantation 1993,
1996, 98S:605. 56:423-427.

Current Opinion in Biotechnology 2009, 20:575–592 www.sciencedirect.com

You might also like