Vous êtes sur la page 1sur 16

5.539.

From Tissue to Organ Engineering


A Atala, Wake Forest University School of Medicine, Winston-Salem, NC, USA
ã 2011 Elsevier Ltd. All rights reserved.

5.539.1. Introduction 548


5.539.2. Biomaterials 548
5.539.2.1. Naturally Derived Materials 549
5.539.2.2. Acellular Tissue Matrices 549
5.539.2.3. Synthetic Polymers 549
5.539.3. Cells for Use in Tissue Engineering 549
5.539.3.1. Embryonic Stem Cells 549
5.539.3.2. Laboratory-Generated Stem Cells 550
5.539.3.3. Amniotic Fluid and Placental Stem Cells 551
5.539.3.4. Adult Stem Cells and Native Progenitor Cells 551
5.539.4. Cell Therapy with Injectable Substances 551
5.539.4.1. Bulking Agents 551
5.539.4.2. Injectable Muscle Cells 552
5.539.4.3. Endocrine Replacement 552
5.539.5. Tissue Engineering of Specific Structures 553
5.539.5.1. Urethra 553
5.539.5.2. Bladder 554
5.539.5.3. Kidney 556
5.539.5.4. Male Reproductive Organs 557
5.539.5.5. Female Reproductive Organs 558
5.539.5.6. Blood Vessels 558
5.539.5.7. Heart 558
5.539.5.8. Liver 559
5.539.5.9. Articular Cartilage and Trachea 559
5.539.6. Summary and Conclusion 560
References 560

Glossary Embryonic stem (ES) cell A broadly multipotent cell type


Acellular matrix Collagen-rich scaffolds prepared found in the inner cell mass of a blastocyst; ES cells can
by removing all cellular components from a donor differentiate into nearly every cell type in the body.
tissue. Induced pluripotent stem (iPS) cell A somatic cell that has
Adult stem cell An undifferentiated cell found in postnatal been dedifferentiated to a stem cell-like phenotype via the
tissues that can self-renew as well as produce progeny that introduction of functional gene products involved in
can differentiate into the various cell types that comprise maintaining pluripotency.
the tissue. Reproductive cloning The use of cloning procedures to
Amniotic fluid stem (AFS) cell A multipotent cell type produce a live offspring that is a genetic copy of an adult
found in the amniotic fluid surrounding an embryo; AFS animal.
cells may be mesenchymal in origin and differentiation to Therapeutic cloning The use of cloning procedures to
neural, hepatic, osteogenic, and chondrogenic lineages, produce a blastocyst in vitro in order to obtain embryonic
among others, has been demonstrated. stem cells for cell therapy or tissue engineering applications.

Abbreviations FDA Food and Drug Administration


AFPSC Amniotic fluid and placental stem cell FLM Fluorescent latex microspheres
AFS Amniotic fluid stem GIRK G-protein-gated inwardly rectifying potassium
CDC Centers for disease control iPS Induced pluripotent state
DNA Deoxyribonucleic acid IFN Interferon
ECM Extracellular matrix MEF Murine embryonic fibroblast
EliSPOT Enzyme-linked immunosorbent spot assay MMP Matrix metalloproteinase
ES Embryonic stem mtDNA Mitochondrial DNA

547
548 Organ Engineering

PGA Polyglycolic acid RNA Ribonucleic acid


PLA Polylactic acid RT-PCR Reverse transcription polymerase chain
PLGA Poly(lactic-co-glycolic) acid reaction
RGD Arginine-glycine-aspartate SCNT Somatic cell nuclear transfer

5.539.1. Introduction Most current strategies for tissue engineering depend upon
a sample of autologous cells from the diseased organ of the host.
Patients suffering from diseased and injured organs may be Aging itself is not a barrier to successful applications of engi-
treated with transplanted organs. However, there is a severe neered tissues, as cells can be expanded from young and old
shortage of donor organs that is worsening yearly. As modern patients alike. However, for many patients with extensive end-
medicine increases the human lifespan, the aging population stage organ failure, a tissue biopsy may not yield enough normal
grows, and the need for organs grows with it, as aging organs cells for expansion and transplantation. In other instances, pri-
are generally more prone to failure. Scientists in the field of mary autologous human cells cannot be expanded from a par-
regenerative medicine and tissue engineering apply the princi- ticular organ, such as the pancreas. In these situations, stem cells
ples of cell transplantation, material science, and bioengineering are envisioned as being an alternative source of cells from which
to construct biological substitutes that can prolong life by reduc- the desired tissue can be derived. Stem cells can be derived from
ing mortality from these diseases. Therapeutic cloning, where the discarded human embryos (human embryonic stem cells), from
nucleus from a donor cell is transferred into an enucleated oocyte fetal tissue, or from adult sources (bone marrow, fat, skin).
in order to extract pluripotent embryonic stem cells, offers a Therapeutic cloning has also played a role in the develop-
potentially limitless source of cells for tissue engineering applica- ment of the field of regenerative medicine. This type of cloning,
tions. The stem cell field is also advancing rapidly, opening new which has also been called nuclear transplantation and nuclear
options for therapy. This chapter reviews recent advances that transfer, involves the introduction of a nucleus from a donor
have occurred in regenerative medicine and describes applica- cell into an enucleated oocyte to generate an embryo with a
tions of these new technologies that offer promise of a longer life. genetic makeup identical to that of the donor. Stem cells can be
The field of regenerative medicine encompasses various derived from this source, which may have the future potential
areas of technology, such as tissue engineering, stem cells, and to be used therapeutically.
cloning. Tissue engineering, one of the major components of
regenerative medicine, follows the principles of cell transplan-
tation, materials science, and engineering toward the develop- 5.539.2. Biomaterials
ment of biological substitutes that can restore and maintain
normal function. Tissue engineering strategies generally fall For cell-based tissue engineering, the expanded cells are seeded
into two categories: the use of acellular matrices, which depend onto a scaffold synthesized with the appropriate biomaterial.
on the body’s natural ability to regenerate for proper orienta- In tissue engineering, biomaterials replicate the biologic and
tion and direction of new tissue growth, and the use of matrices mechanical function of the native ECM found in tissues in the
with cells. Acellular tissue matrices are usually prepared by body by serving as an artificial ECM. Biomaterials provide a
manufacturing artificial scaffolds, or by removing cellular com- three-dimensional space for the cells to form into new tissues
ponents from tissues via mechanical and chemical manipula- with appropriate structure and function, and also can allow
tion to produce collagen-rich matrices.1–4 These matrices tend for the delivery of cells and appropriate bioactive factors (e.g.,
to slowly degrade on implantation and are generally replaced cell-adhesion peptides, growth factors), to desired sites in the
by the extracellular matrix (ECM) proteins that are secreted by body.13 As the majority of mammalian cell types are anchorage-
the ingrowing cells. Cells can also be used for therapy via dependent and will die if no cell-adhesion substrate is available,
injection, either with carriers such as hydrogels, or alone. biomaterials provide a cell-adhesion substrate that can deliver
When cells are used for tissue engineering, a small piece cells to specific sites in the body with high loading efficiency.
of donor tissue is dissociated into individual cells. These cells Biomaterials can also provide mechanical support against in vivo
are either implanted directly into the host, or are expanded in forces such that the predefined three-dimensional structure is
culture, attached to a support matrix, and then reimplanted maintained during tissue development. Furthermore, bioactive
into the host after expansion. The source of donor tissue can be signals, such as cell-adhesion peptides and growth factors, can be
heterologous (such as bovine), allogeneic (same species, dif- loaded along with cells to help regulate cellular function.
ferent individual), or autologous. Ideally, both structural and The ideal biomaterial should be biodegradable and bio-
functional tissue replacement will occur with minimal compli- resorbable to support the replacement of normal tissue
cations. The preferred cells to use are autologous cells, where without inflammation. Incompatible materials are destined
a biopsy of tissue is obtained from the host, the cells are for an inflammatory or foreign-body response that eventually
dissociated and expanded in culture, and the expanded cells leads to rejection and/or necrosis. Degradation products, if
are implanted into the same host.4–12 The use of autologous produced, should be removed from the body via metabolic
cells, although it may cause an inflammatory response, avoids pathways at an adequate rate that keeps the concentration of
rejection, and thus, the deleterious side effects of immunosup- these degradation products in the tissues at a tolerable level.14
pressive medications can be avoided. The biomaterial should also provide an environment in which
From Tissue to Organ Engineering 549

appropriate regulation of cell behavior (adhesion, prolifera- are often prepared by mechanical and chemical manipula-
tion, migration, and differentiation) can occur such that func- tion of a segment of tissue.1–4 The matrices slowly degrade
tional tissue can form. Cell behavior in the newly formed tissue upon implantation, and are replaced and remodeled by
has been shown to be regulated by multiple interactions of ECM proteins synthesized and secreted by transplanted or
the cells with their microenvironment, including interactions ingrowing cells.
with cell-adhesion ligands15 and with soluble growth factors.
Since biomaterials provide temporary mechanical support
5.539.2.3. Synthetic Polymers
while the cells undergo spatial reorganization into tissue, the
properly chosen biomaterial should allow the engineered tis- Polyesters of naturally occurring a-hydroxy acids, including
sue to maintain sufficient mechanical integrity to support itself PGA, PLA, and PLGA, are widely used in tissue engineering.
in early development, while in late development, it should These polymers have gained FDA approval for human use in
have begun degradation such that it does not hinder further a variety of applications, including sutures.24 The ester bonds
tissue growth.13 in these polymers are hydrolytically labile, and these polymers
In general, three classes of biomaterials are used for engi- degrade by nonenzymatic hydrolysis. The degradation pro-
neering tissues: naturally derived materials (e.g., collagen and ducts of PGA, PLA, and PLGA are nontoxic natural metabolites
alginate), acellular tissue matrices (e.g., bladder submucosa and are eventually eliminated from the body in the form
and small intestinal submucosa), and synthetic polymers of carbon dioxide and water.24 The degradation rate of these
such as polyglycolic acid (PGA), polylactic acid (PLA), and polymers can be tailored from several weeks to several years
poly(lactic-co-glycolic acid) PLGA). These classes of biomater- by altering crystallinity, initial molecular weight, and the
ials have been tested with respect to their biocompatibility.16,17 copolymer ratio of lactic to glycolic acid. Since these polymers
Naturally derived materials and acellular tissue matrices have are thermoplastics, they can be easily formed into a three-
the potential advantage of biological recognition. However, dimensional scaffold with a desired microstructure, gross
synthetic polymers can be produced reproducibly on a large shape, and dimension by various techniques, including mold-
scale with controlled properties such as strength, degradation ing, extrusion, solvent casting,25 phase separation techniques,
rate, and microstructure. and gas foaming techniques.26 Many applications in tissue
engineering often require a scaffold with high porosity and
ratio of surface area to volume. Other biodegradable synthetic
5.539.2.1. Naturally Derived Materials
polymers, including poly(anhydrides) and poly(ortho-esters),
Collagen is the most abundant and ubiquitous structural protein can also be used to fabricate scaffolds for tissue engineering
in the body, and may be readily purified from both animal and with controlled properties.27
human tissues with an enzyme treatment and salt/acid extrac-
tion.18 Collagen implants, under normal conditions, degrade
through a process involving phagocytosis of collagen fibrils by 5.539.3. Cells for Use in Tissue Engineering
fibroblasts.19 This is followed by sequential attack by lysosomal
5.539.3.1. Embryonic Stem Cells
enzymes including cathepsins B1 and D. Under inflammatory
conditions, the implants can be rapidly degraded largely by Human embryonic stem cells exhibit two remarkable pro-
matrix metalloproteinases (MMPs) and collagenases.19 How- perties: the ability to proliferate in an undifferentiated but
ever, the in vivo resorption rate of a collagen implant can be pluripotent state (self-renewal), and the ability to differentiate
regulated by controlling the density of the implant and the extent into many specialized cell types.28 They can be isolated by
of intermolecular cross-linking. The lower the density, the greater aspirating the inner cell mass from the embryo during the
the space between collagen fibers and the larger the pores for cell blastocyst stage (5 days postfertilization), and are usually
infiltration, leading to a higher rate of implant degradation. grown on feeder layers consisting of mouse embryonic fibro-
Collagen contains cell-adhesion domain sequences (e.g., RGD) blasts (MEFs) or human feeder cells.29 More recent reports
that may assist in retaining the phenotype and activity of many have shown that these cells can be grown without the use of
types of cells, including fibroblasts20 and chondrocytes.21 a feeder layer30 and can thus avoid the exposure of these
Alginate, a polysaccharide isolated from seaweed, has been human cells to mouse viruses and proteins. These cells have
used as an injectable cell delivery vehicle22 and a cell immobi- demonstrated longevity in culture by maintaining their undif-
lization matrix23 owing to its gentle gelling properties in the ferentiated state for at least 80 passages when grown using
presence of divalent ions, such as calcium. Alginate is relatively current published protocols.31,32
biocompatible and approved by the Food and Drug Adminis- Human embryonic stem cells have been shown to differen-
tration (FDA) for human use as wound dressing material. tiate into cells from all three embryonic germ layers in vitro.
Alginate is a family of copolymers of D-mannuronate and Skin and neurons have been formed, indicating ectodermal
L-guluronate. The physical and mechanical properties of algi- differentiation.33–36 Blood, cardiac cells, cartilage, endothelial
nate gel are strongly correlated with the proportion and length cells, and muscle have been formed, indicating mesodermal
of polygluronic block in the alginate chains.22 differentiation.37–39 Pancreatic cells have been formed, indicat-
ing endodermal differentiation.40 In addition, as further evi-
dence of their pluripotency, embryonic stem cells can form
5.539.2.2. Acellular Tissue Matrices
embryoid bodies, which are cell aggregations that contain all
Acellular tissue matrices are collagen-rich matrices prepared three embryonic germ layers while in culture, and can form
by removing cellular components from tissues. The matrices teratomas in vivo.41
550 Organ Engineering

5.539.3.2. Laboratory-Generated Stem Cells because of immunologic incompatibility, and immunosup-


pressive drugs are usually required.55 The use of transplantable
In addition, stem cells for tissue engineering can be generated
tissue and organs derived from therapeutic cloning may poten-
through cloning procedures. There has been tremendous inter-
tially lead to the avoidance of immune responses that typically
est in the field of nuclear cloning since the birth of Dolly in
are associated with transplantation of nonautologous tissues.56
1997, but frogs were the first successfully cloned vertebrates
Although it looks promising, somatic cell nuclear trans-
derived from nuclear transfer42–45 but the nuclei were derived
fer technology has certain limitations that require further
from nonadult sources. In the past 15 years, tremendous
improvements before therapeutic cloning can be applied widely
advances in nuclear cloning technology have been reported,
in replacement therapy. Currently, the efficiency of the overall
indicating the relative immaturity of the field. Dolly was not
cloning process is low. The majority of embryos derived from
the first cloned mammal to be produced via nuclear transfer;
animal cloning do not survive after implantation.58–60 To
in fact, live lambs were produced in 1996 using nuclear transfer
improve cloning efficiency, further improvements are required
and differentiated epithelial cells derived from embryonic
in the multiple complex steps of nuclear transfer, such as enu-
discs.46 The significance of Dolly was that she was the first
cleation and reconstruction, activation of oocytes, and cell cycle
mammal to be derived from an adult somatic cell using nuclear
synchronization between donor cells and recipient oocytes.61
transfer.47 Since then, animals from several species have been
Recently, exciting reports of the successful transformation
grown using nuclear transfer technology, including cattle,48
of adult cells into pluripotent stem cells through a type of
goats,49 mice,50 and pigs.51,52
genetic “reprogramming” have been published. Reprogram-
Two types of nuclear cloning, reproductive cloning and
ming is a technique that involves dedifferentiation of adult
therapeutic cloning, have been described, and a better under-
somatic cells to produce patient-specific pluripotent stem
standing of the differences between the two types may help
cells, without the use of embryos. Cells generated by repro-
to alleviate some of the controversy that surrounds these
gramming would be genetically identical to the somatic cells
technologies.53,54 Banned in most countries for human appli-
(and thus, the patient who donated these cells) and would
cations, reproductive cloning is used to generate an embryo
not be rejected. Yamanaka was the first to discover that MEFs
that has the identical genetic material as its cell source. This
and adult mouse fibroblasts could be reprogrammed into
embryo can then be implanted into the uterus of a female
an ‘induced pluripotent state (iPS)’.62 This group used MEFs
to give rise to an infant that is a clone of the donor. On the
engineered to express a neomycin resistance gene from the
other hand, therapeutic cloning is used to generate early stage
Fbx15 locus, a gene expressed only in ES cells. They examined
embryos that are explanted in culture to produce embryonic
24 genes that were thought to be important for embryonic
stem cell lines whose genetic material is identical to that of its
stem cells and identified four key genes that, when introduced
source. These autologous stem cells have the potential to
into the reporter fibroblasts, resulted in drug-resistant cells.
become almost any type of cell in the adult body, and thus
These were Oct3/4, Sox2, c-MYC, and Klf4. This experiment
would be useful in tissue and organ replacement applica-
indicated that expression of the four genes in these transgenic
tions.55 Therefore, therapeutic cloning, which has also been
MEFs led to expression of a gene specific for ES cells. MEFs and
called somatic cell nuclear transfer, may provide an alternative
adult fibroblasts were cotransduced with retroviral vectors,
source of transplantable cells. Figure 1 shows the strategy
each carrying one of the four genes, and transduced cells were
of combining therapeutic cloning with tissue engineering to
selected via drug resistance. The resultant iPS cells possessed
develop tissues and organs. According to data from the Centers
the immortal growth characteristics of self-renewing ES cells,
for Disease Control (CDC), an estimated 3000 Americans
expressed genes specific for ES cells, and generated embryoid
die every day of diseases that could have been treated with
bodies in vitro and teratomas in vivo. When iPS cells were
stem cell-derived tissues56,57 With current allogeneic tissue
injected into mouse blastocysts, they contributed to a variety
transplantation protocols, rejection is a frequent complication
of cell types. However, although iPS cells selected in this way
were pluripotent, they were not identical to ES cells. Unlike ES
cells, chimeras made from iPS cells did not result in full-term
pregnancies. Gene expression profiles of the iPS cells showed
Therapeutic Cloning Strategies
that they possessed a distinct gene expression signature that
was different from that of ES cells. In addition, the epigenetic
Somatic
cell biopsy
Enucleated state of the iPS cells was somewhere between that found in
oocyte
somatic cells and that found in ES cells, suggesting that the
Patient Nuclear transfer reprogramming was incomplete.
These results were improved significantly by Wernig and
Transplantation Embryonic stem Jaenisch in July 2007.63 Fibroblasts were infected with retroviral
cells
vectors and selected for the activation of endogenous Oct4 or
Hematopoietic Nanog genes. Results from this study showed that DNA methyl-
cells Genetically matched tissue
Neurons ation, gene expression profiles, and the chromatin state of the
reprogrammed cells were similar to those of ES cells. Teratomas
Pancreatic
islet cells Hepatocytes induced by these cells contained differentiated cell types repre-
Cardiomyocytes senting all three embryonic germ layers. Most importantly, the
Renal cells
reprogrammed cells from this experiment were able to form
Figure 1 Strategy for therapeutic cloning and tissue engineering. viable chimeras and contribute to the germline-like ES cells,
From Tissue to Organ Engineering 551

suggesting that these iPS cells were completely reprogrammed. The undifferentiated stem cells expand extensively without fee-
Wernig et al. observed that the number of reprogrammed colo- ders and double every 36 h. Unlike human embryonic stem
nies increased when drug selection was initiated later (day 20 cells, the AFPSC do not form tumors in vivo. Lines maintained
rather than day 3 posttransduction). This suggests that repro- for over 250 population doublings retained long telomeres and
gramming is a slow and gradual process and may explain why a normal karyotype. AFS cells are broadly multipotent. Clonal
previous attempts resulted in incomplete reprogramming. human lines verified by retroviral marking can be induced to
It has recently been shown that reprogramming of human differentiate into cell types representing each embryonic germ
cells is possible.64,65 Yamanaka showed that retrovirus-mediated layer, including cells of adipogenic, osteogenic, myogenic, endo-
transfection of OCT3/4, SOX2, KLF4, and c-MYC generates thelial, neuronal, and hepatic lineages. In this respect, they meet
human iPS cells that are similar to hES cells in terms of mor- a commonly accepted criterion for pluripotent stem cells, with-
phology, proliferation, gene expression, surface markers, and out implying that they can generate every adult tissue. Examples
teratoma formation. Thompson’s group showed that retroviral of differentiated cells derived from AFS cells and displaying
transduction of OCT4, SOX2, NANOG, and LIN28 could gener- specialized functions include neuronal lineage secreting the
ate pluripotent stem cells without introducing any oncogenes neurotransmitter L-glutamate or expressing G-protein-gated
(c-MYC). Both studies showed that human iPS were similar but inwardly rectifying potassium (GIRK) channels, hepatic lineage
not identical to hES cells. cells producing urea, and osteogenic lineage cells forming
Another concern is that these iPS cells contain three tissue-engineered bone. The cells could be obtained either
to six retroviral integrations (one for each factor), which from amniocentesis or chorionic villous sampling in the devel-
may increase the risk of tumorigenesis. Yamanaka et al. oping fetus, or from the placenta at the time of birth. The cells
studied the tumor formation in chimeric mice generated could be preserved for self-use, and used without rejection, or
from Nanog-iPS cells and found 20% of the offspring devel- they could be banked. A bank of 100,000 specimens could
oped tumors due to the retroviral expression of c-MYC.66 An potentially supply 99% of the US population with a perfect
alternative approach would be to use a transient expression genetic match for transplantation. Such a bank may be easier
method, such as adenovirus-mediated system, since both to create than with other cell sources, since there are approxi-
Jaenisch and Yamanaka showed strong silencing of the mately 4.5 million births per year in the United States.69
viral-controlled transcripts in iPS cells.66,67 This indicates that
these viral genes are only required for the induction, not the
5.539.3.4. Adult Stem Cells and Native Progenitor Cells
maintenance, of pluripotency.
Another concern is the use of transgenic donor cells for One of the limitations of applying cell-based regenerative
reprogrammed cells in the mouse studies. Both studies used medicine techniques to organ replacement has been the inher-
donor cells from transgenic mice harboring a drug resistance ent difficulty of growing specific cell types in large quantities.
gene driven by Fbx15, Oct3/4, or Nanog promoters so that, if Even when some organs, such as the liver, have a high regener-
these ES cell-specific genes were activated, the resulting cells ative capacity in vivo, cell growth and expansion in vitro may be
could be easily selected using neomycin. However, the use of difficult. By studying the privileged sites for committed precur-
genetically modified donors hinders clinical applicability for sor cells in specific organs, as well as exploring the conditions
humans. To assess whether iPS cells can be derived from non- that promote differentiation, one may be able to overcome
transgenic donor cells, wild-type MEF and adult skin cells were the obstacles that limit cell expansion in vitro. For example,
retrovirally transduced with Oct3/4, Sox2, c-MYC, and Klf4 urothelial cells could be grown in the laboratory setting in
and ES-like colonies were isolated by morphology alone, with- the past, but only with limited expansion. Several protocols
out the use of drug selection for Oct4 or Nanog.67 iPS cells from were developed over the past two decades that identified the
wild-type donor cells formed teratomas and generated live undifferentiated cells, and kept them undifferentiated during
chimeras. This study suggests that transgenic donor cells are their growth phase.10,70–73 Using these methods of cell culture,
not necessary to generate iPS cells. it is now possible to expand a urothelial strain from a single
Although this is an exciting phenomenon, it is unclear why specimen that initially covered a surface area of 1 cm2 to
reprogramming adult fibroblasts and mesenchymal stromal one covering a surface area of 4202 m2 (the equivalent of one
cells have similar efficiencies.62 It would seem that cells that football field) within 8 weeks.10 These studies indicated that
are already multipotent could be reprogrammed with greater it should be possible to collect autologous bladder cells
efficiency, since the more undifferentiated donor nucleus the from human patients, expand them in culture, and return
better SCNT performs.68 This further emphasizes our limited them to the donor in sufficient quantities for reconstructive
understanding of the mechanism of reprogramming, yet the purposes.10,71–76 Major advances have been achieved within
potential for this area of study is exciting. the past decade on the possible expansion of a variety of primary
human cells, with specific techniques that make the use of
autologous cells possible for clinical application.
5.539.3.3. Amniotic Fluid and Placental Stem Cells
An alternate source of stem cells is the amniotic fluid and
placenta. Amniotic fluid and the placenta are known to con- 5.539.4. Cell Therapy with Injectable Substances
tain multiple partially differentiated cell types derived from the
5.539.4.1. Bulking Agents
developing fetus. We isolated stem cell populations from these
sources, called amniotic fluid and placental stem cells (AFPSC) Injectable bulking agents can be endoscopically used in the
that express embryonic and adult stem cell markers.69 treatment of both urinary incontinence and vesicoureteral
552 Organ Engineering

reflux. The advantages in treating urinary incontinence and Myoblasts were labeled with fluorescent latex microspheres
vesicoureteral reflux with this minimally invasive approach (FLM) in order to track them after injection. Labeled myoblasts
include the simplicity of this quick outpatient procedure and were directly injected into the proximal urethra and lateral
the low morbidity associated with it. Several investigators are bladder walls of nude mice with a microsyringe in an open
seeking alternative implant materials that would be safe for surgical procedure. Tissue harvested up to 35 days postinjec-
human use.77 tion contained the labeled myoblasts, as well as evidence
The ideal substance for the endoscopic treatment of reflux of differentiation of the labeled myoblasts into regenerative
and incontinence should be injectable, nonantigenic, nonmi- myofibers. The authors reported that a significant portion of
gratory, volume stable, and safe for human use. Toward this the injected myoblast population persisted in vivo. Similar
goal long-term studies were conducted to determine the effect techniques of sphincteric derived muscle cells have been used
of injectable chondrocytes in vivo.78 It was initially determined for the treatment of urinary incontinence in a pig model.84 The
that alginate, a liquid solution of gluronic and mannuronic fact that myoblasts can be labeled and survive after injection
acid, embedded with chondrocytes, could serve as a synthetic and begin the process of myogenic differentiation further sup-
substrate for the injectable delivery and maintenance of carti- ports the feasibility of using cultured cells of muscular origin as
lage architecture in vivo. Alginate undergoes hydrolytic biodeg- an injectable bioimplant.
radation and its degradation time can be varied depending on The use of injectable muscle precursor cells has also been
the concentration of each of the polysaccharides. The use of investigated for use in the treatment of urinary incontinence
autologous cartilage for the treatment of vesicoureteral reflux due to irreversible urethral sphincter injury or maldevelop-
in humans would satisfy all the requirements for an ideal ment. Muscle precursor cells are the quiescent satellite cells
injectable substance. found in each myofiber that proliferate to form myoblasts
Chondrocytes derived from an ear biopsy can be readily and eventually myotubes and new muscle tissue. Intrinsic
grown and expanded in culture. Neocartilage formation can be muscle precursor cells have previously been shown to play an
achieved in vitro and in vivo using chondrocytes cultured on active role in the regeneration of injured striated urethral
synthetic biodegradable polymers. In these experiments, the sphincter.85 In a subsequent study, autologous muscle precur-
cartilage matrix replaced the alginate as the polysaccharide sor cells were injected into a rat model of urethral sphincter
polymer underwent biodegradation. This system was adapted injury, and both replacement of mature myotubes and restora-
for the treatment of vesicoureteral reflux in a porcine model.79 tion of functional motor units were noted in the regenerating
These studies showed that chondrocytes can be easily harvested sphincteric muscle tissue.86 This is the first demonstration of
and combined with alginate in vitro, the suspension can be the replacement of both sphincter muscle tissue and its inner-
easily injected cystoscopically, and the elastic cartilage tissue vation by the injection of muscle precursor cells. As a result,
formed is able to correct vesicoureteral reflux without any muscle precursor cells may be a minimally invasive solution
evidence of obstruction. for urinary incontinence in patients with irreversible urinary
Two multicenter clinical trials were conducted using this sphincter muscle insufficiency.
engineered chondrocyte technology. Patients with vesicoureteral
reflux were treated at ten centers throughout the US. The
5.539.4.3. Endocrine Replacement
patients had a similar success rate as with other injectable sub-
stances in terms of cure (Figure 5). Chondrocyte formation was Patients with testicular dysfunction and hypogonadal disor-
not noted in patients who had treatment failure. It is supposed ders are dependent on androgen replacement therapy to
that the patients who were cured have a biocompatible region of restore and maintain physiological levels of serum testosterone
engineered autologous tissue present, rather than a foreign and its metabolites, dihydrotestosterone, and estradiol.87,88
material.80 Patients with urinary incontinence were also treated Currently available androgen replacement modalities, such as
endoscopically with injected chondrocytes at three different testosterone tablets and capsules, Depo-Provera injections, and
medical centers. Phase 1 trials showed an approximate success skin patches may be associated with fluctuating serum levels
rate of 80% at follow-up 3 and 12 months postoperatively.81 and complications such as fluid and nitrogen retention, eryth-
Several of the clinical trials involving bioengineered products ropoiesis, hypertension, and bone density changes.89 Since
have been placed on hold because of the costs involved with the Leydig cells of the testes are the major source of testosterone
specific technology. With a bioengineered product, costs are in men, implantation of heterologous Leydig cells or gonadal
usually high because of the biological nature of the therapies tissue fragments have previously been proposed as a method
involved. As with any therapy, the cost that the medical health for chronic testosterone replacement.90,91 These approaches,
care system can allow for a specific technology is limited. There- however, were limited by the failure of the tissues and cells to
fore, the costs of bioengineered products have to be lowered for produce testosterone.
them to have an impact clinically. This is currently being Encapsulation of cells in biocompatible and semiperme-
addressed for multiple tissue-engineered technologies. As the able polymeric membranes has been an effective method to
technologies advance overtime, and the volume of the applica- protect against a host immune response as well as to maintain
tion is considered, costs will naturally decrease. viability of the cells while allowing the secretion of desired
therapeutic agents.92,93 Alginate–poly-L-lysine-encapsulated
Leydig cell microspheres were used as a novel method for
5.539.4.2. Injectable Muscle Cells
testosterone delivery in vivo.88 Elevated stable serum testos-
The potential use of injectable cultured myoblasts for the treat- terone levels were noted in castrated adult rats over the course
ment of stress urinary incontinence has been investigated.82,83 of the study, suggesting that microencapsulated Leydig cells
From Tissue to Organ Engineering 553

may be a potential therapeutic modality for testosterone of performing additional surgical procedures for graft harvest-
supplementation. ing, and both operative time and the potential morbidity from
the harvest procedure were decreased. Similar results were
obtained in pediatric and adult patients with primary urethral
5.539.5. Tissue Engineering of Specific Structures stricture disease using the same collagen matrices.99 Another
study in 30 patients with recurrent stricture disease showed
Investigators around the world, including those in our labora- that a healthy urethral bed (two or fewer prior urethral sur-
tory, have been working toward the development of several cell geries) was needed for successful urethral reconstruction using
types and tissues and organs for clinical application. the acellular collage-based grafts.100 More than 200 pediatric
and adult patients with urethral disease have been successfully
treated in an onlay manner with a bladder-derived collagen-
5.539.5.1. Urethra
based matrix. One of its advantages over nongenital tissue grafts
Various strategies have been proposed over the years for the used for urethroplasty is that the material is “off the shelf.” This
regeneration of urethral tissue. Woven meshes of PGA without eliminates the necessity of additional surgical procedures for
cells94,95 or with cells96 were used to regenerate urethras in graft harvesting, which may decrease operative time, as well as
various animal models. Naturally derived collagen-based mate- the potential morbidity due to the harvest procedure.
rials such bladder-derived acellular submucosa,1 and an acellu- The above techniques, using nonseeded acellular matrices,
lar urethral submucosa,97 have also been tried experimentally in were applied experimentally and clinically in a successful man-
various animal models for urethral reconstruction. ner for onlay urethral repairs. However, when tubularized
The bladder submucosa matrix1 proved to be a suitable urethral repairs were attempted experimentally, adequate ure-
graft for repair of urethral defects in rabbits. The neourethras thral tissue regeneration was not achieved, and complications
demonstrated a normal urothelial luminal lining and organized ensued, such as graft contracture and stricture formation.101
muscle bundles. These results were confirmed clinically in a Autologous rabbit bladder epithelial and smooth muscle cells
series of patients with a history of failed hypospadias recon- were grown and seeded onto preconfigured tubular matrices.
struction wherein the urethral defects were repaired with Entire urethra segments were resected and urethroplasties were
human bladder acellular collagen matrices (Figure 2).98 The performed with tubularized collagen matrices either seeded
neourethras were created by anastomosing the matrix in an with cells, or without cells. The tubularized collagen matrices
onlay fashion to the urethral plate. The size of the created seeded with autologous cells formed new tissue that was histo-
neourethra ranged from 5 to 15 cm. After a 3-year follow-up, logically similar to native urethra. The tubularized collagen
three of the four patients had a successful outcome with regard matrices without cells lead to poor tissue development, fibro-
to cosmetic appearance and function. One patient who had a sis, and stricture formation. These findings were confirmed
15-cm neourethra created developed a subglandular fistula. clinically. A clinical trial using tubularized nonseeded SIS
The acellular collagen-based matrix eliminated the necessity for urethral stricture repair was performed in eight evaluable
patients. Two patients with short inflammatory strictures main-
tained urethral patency. Stricture recurrence developed in the
other six patients within 3 months of surgery.102 Other cell
types have also been tried experimentally in acellular bladder
collagen matrices, including foreskin epidermal cells and oral
keratinocytes.103,104 Vascular endothelial growth factor gene-
modified urothelial cells have also been used experimentally
(a) for urethral reconstruction.105
The normal wound healing response to injury has been
(b) studied extensively, and this knowledge has been helpful in
maximizing success for the engineering of tissues. At the time
of tissue injury, cell ingrowth is initiated from the wound edges
in order to cover the tissue defect. The cells from the edges of
(c) (d)
the native tissue are able to traverse short distances without any
detrimental effects. If the wound is large, more than a few milli-
Figure 2 Tissue engineering of the urethra using a collagen matrix. meters in distance or depth, increased collagen deposition, fibro-
(a) Representative case of a patient with a bulbar stricture. (b) During the sis, and scar formation ensue. Matrices implanted in wound beds
urethral repair surgery, strictured tissue is excised, preserving the are able to lengthen the distances that cells can traverse without
urethral plate on the left side, and matrix is anastamosed to the urethral initiating an adverse fibrotic response. However, these distances
plate in an onlay fashion on the right. The boxes in both photos indicate are also limited. The maximum distance that adjacent cells from
the area of interest, including the urethra, which appears white in the left the wound edge have to travel to create normal tissue over a
photograph. In the left photograph, the arrow indicates the area of
biologic matrix is approximately 1 cm.106 Tissue defects greater
stricture in the urethra. On the right, the arrow indicates the repaired
stricture. Note that the engineered tissue now obscures the native
than 1 cm, that are treated with a matrix alone, without cells,
white urethral tissue in an onlay fashion in the right photograph. usually have increased collagen deposition, increased fibrosis,
(c) Urethrogram 6 months after repair. (d) Cystoscopic view of urethra and scar formation. Cell-seeded matrices implanted in wound
before surgery on the left side, and 4 months after repair on the beds are able to further lengthen the distance for normal tissue
right side. formation, without initiating an adverse fibrotic response.
554 Organ Engineering

Studies in the field of regenerative medicine have shown that It has been well established for decades that the bladder is
very large defects, greater than 30 cm, can be successfully treated able to regenerate generously over free grafts. Urothelium is
using cell-seeded scaffolds. This explains the described experi- associated with a high reparative capacity.116 Bladder muscle
mental and clinical results noted with urethral repair. Non- tissue is less likely to regenerate in a normal fashion. Both
seeded matrices are able to replace urethral segments when urothelial and muscle ingrowth are believed to be initiated
used in an onlay fashion because of the short distances required from the edges of the normal bladder toward the region
for tissue ingrowth. However, if a tubularized urethral repair is of the free graft.117,118 Usually, however, contracture or resorp-
needed, the matrices need to be seeded with autologous cells in tion of the graft has been evident. The inflammatory response
order to avoid the risk of stricture formation and poor tissue toward the matrix may contribute to the resorption of the free
development. graft. It was hypothesized that building the three-dimensional
structure constructs in vitro, before implantation, would facili-
tate the eventual terminal differentiation of the cells after
5.539.5.2. Bladder
implantation in vivo and would minimize the inflammatory
Currently, gastrointestinal segments are commonly used as response toward the matrix, thus avoiding graft contracture
tissues for bladder replacement or repair. However, gastroin- and shrinkage. The dog study demonstrated a major difference
testinal tissues are designed to absorb specific solutes, whereas between matrices used with autologous cells (tissue-engineered
bladder tissue is designed for the excretion of solutes. Due to matrices) and those used without cells.4 Matrices implanted
the problems encountered with the use of gastrointestinal with cells for bladder augmentation retained most of their
segments, numerous investigators have attempted alternative implanted diameter, as opposed to matrices implanted without
materials and tissues for bladder replacement or repair. cells for bladder augmentation, in which graft contraction
Over the last few decades, several bladder wall substitutes and shrinkage occurred. The histomorphology demonstrated a
have been attempted with both synthetic and organic materi- marked paucity of muscle cells and a more aggressive inflamma-
als. Synthetic materials that have been tried in experimental tory reaction in the matrices implanted without cells. Epithelial-
and clinical settings include polyvinyl sponge, Teflon, collagen mesenchymal signaling is important for the differentiation of
matrices, Vicryl (PGA) matrices, and silicone. Most of these bladder smooth muscle.119
attempts have failed because of mechanical, structural, func- The results of initial studies showed that the creation of
tional, or biocompatibility problems. Usually, permanent syn- artificial bladders may be achieved in vivo; however, it could
thetic materials used for bladder reconstruction succumb to not be determined whether the functional parameters noted
mechanical failure and urinary stone formation, and use of were caused by the augmented segment or by the intact native
degradable materials lead to fibroblast deposition, scarring, bladder tissue. To better address the functional parameters of
graft contracture, and a reduced reservoir volume overtime.7,107 tissue-engineered bladders, an animal model was designed that
Because of this, there has been an increase in the use of various required a subtotal cystectomy with subsequent replacement
collagen-based matrices for tissue regeneration. Nonseeded with a tissue-engineered organ.11 Cystectomy-only and non-
allogeneic acellular bladder matrices have served as scaffolds seeded controls maintained average capacities of 22% and 46%
for the ingrowth of host bladder wall components. The matri- of preoperative values, respectively. An average bladder capac-
ces are prepared by mechanically and chemically removing all ity of 95% of the original precystectomy volume was achieved
cellular components from bladder tissue.3,4,108–110 The matri- in the cell-seeded tissue-engineered bladder replacements.
ces serve as vehicles for partial bladder regeneration, and rele- These findings were confirmed radiographically (Figure 3).
vant antigenicity is not evident. However, in multiple studies The subtotal cystectomy reservoirs that were not reconstructed
using various materials as nonseeded grafts for cystoplasty, the and the polymer-only reconstructed bladders showed a marked
urothelial layer was able to regenerate normally, but the muscle decrease in bladder compliance (10% and 42% total compli-
layer, although present, was not fully developed.4,108,109,111–113 ance). The compliance of the cell-seeded tissue-engineered
Studies involving acellular matrices that may provide the nec- bladders showed almost no difference from preoperative
essary environment to promote cell migration, growth, and values that were measured when the native bladder was present
differentiation are being conducted.114 With continued blad- (106%). Histologically, the nonseeded scaffold bladders pre-
der research in this area, these matrices may have a clinical role sented a pattern of normal urothelial cells with a thickened
in bladder replacement in the future. fibrotic submucosa and a thin layer of muscle fibers. The
Cell-seeded allogeneic acellular bladder matrices were used retrieved tissue-engineered bladders showed a normal cellular
for bladder augmentation in dogs.4 The regenerated bladder organization, consisting of a trilayer of urothelium, submu-
tissues contained a normal cellular organization consisting cosa, and muscle. Immunocytochemical analyses confirmed
of urothelium and smooth muscle and exhibited a normal the muscle and urothelial phenotype. S-100 staining indicated
compliance. Biomaterials preloaded with cells before their the presence of neural structures.11 These studies, performed
implantation showed better tissue regeneration compared with polyglocolic acid based-scaffolds, have been repeated by
with biomaterials implanted with no cells, in which tissue other investigators, showing similar long-term results in a large
regeneration depended on ingrowth of the surrounding tissue. number of animals.111,120 The strategy of using biodegradable
The bladders showed a significant increase (100%) in capacity scaffolds with cells can be pursued without concerns for local
when augmented with scaffolds seeded with cells, compared or systemic toxicity.121 However, not all scaffolds perform well
to scaffolds without cells (30%). The acellular collagen matri- if a large portion of the bladder needs replacement. In a study
ces can be enhanced with growth factors to improve bladder using SIS for subtotal bladder replacement in dogs, both the
regeneration.115 unseeded and cell-seeded experimental groups showed graft
From Tissue to Organ Engineering 555

shrinkage and poor results.122 The type of scaffold used is


Subtotal Polymer only Tissue-engineered critical for the success of these technologies. The use of bior-
cystectomy only implants neobladder
eactors, in which mechanical stimulation is started at the time
of organ production, has also been proposed as an important
parameter for success.123
A clinical experience involving engineered bladder tissue for
cystoplasty reconstruction was conducted starting in 1998.
A small pilot study of seven patients was reported, using a
collagen scaffold seeded with cells either with or without omen-
tum coverage, or a combined PGA-collagen scaffold seeded
with cells and omental coverage (Figure 4). The patients
reconstructed with the engineered bladder tissue created with
the PGA-collagen cell-seeded scaffolds with omental coverage
11M 11M 11M showed increased compliance, decreased end-filling pres-
sures, increased capacities, and longer dry periods overtime
Figure 3 Gross specimens and cystograms at 11 months of the
(Figure 5).124 It is clear from this experience that the engineered
cystectomy-only, nonseeded controls, and cell-seeded tissue-engineered
bladder replacements in dogs. The cystectomy-only bladder had a bladders continued their improvement with time, mirroring
capacity of 22% of the preoperative value and a decrease in bladder their continued development. Although the experience is
compliance to 10% of the preoperative value. The nonseeded controls promising in terms of showing that engineered tissues can be
showed significant scarring with a capacity of 46% of the preoperative implanted safely, it is just a start in terms of accomplishing the
value and a decrease in bladder compliance to 42% of the preoperative goal of engineering fully functional bladders. This was a limited
value. An average bladder capacity of 95% of the original precystectomy clinical experience, and the technology is not yet ready for wide
volume was achieved in the cell-seeded tissue-engineered bladder dissemination, as further experimental and clinical studies are
replacements and the compliance showed almost no difference from required. FDA Phase 2 studies have now been completed.
preoperative values that were measured when the native bladder was
An area of concern in the field of tissue engineering in the
present (106%).
past was the source of cells for regeneration. The concept of
creating engineered constructs involves initially obtaining cells
for expansion from the diseased organ. However, it was not
known until recently whether the cell population obtained
for later autologous implantation was normal and would
lead to normal tissue formation. For example, would the
cells obtained from a neuropathic bladder lead to the engineer-
ing of another neuropathic bladder? Cultured neuropathic
(a) (b) (c)
bladder smooth muscle cells possess and maintain different
characteristics than normal smooth muscle cells in vitro, as
Figure 4 Construction of engineered bladder. (a) Scaffold material demonstrated by growth assays, contractility, adherence tests,
seeded with cells for use in bladder repair. (b) The seeded scaffold is and microarray analysis.125–127 However, when neuropathic
anastamosed to native bladder with running 4–0 polyglycolic sutures. smooth muscle cells were cultured in vitro, seeded onto matri-
(c) Implant covered with fibrin glue and omentum. ces and implanted in vivo, the tissue-engineered constructs

Pves
30 cnH20/Div

3 min/div
t1 t3 t4 t6 t8 t9 11 13
t2 t5 t7 10 12

Pves
30 cnH20/Div

5 min/div
t1 t2 t4 t5
t3

Figure 5 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.
556 Organ Engineering

showed the same properties as the tissues engineered with of proximal tubules, distal tubules, loops of Henle, collecting
normal cells.128 The progenitor cells, which reside within tubules, and collecting ducts. These results clearly showed that
stem cell niches within each organ, are responsible for new single cell suspensions grown in vitro are capable of reconsti-
cell differentiation and tissue formation during the normal tuting tubule structures. The tubules contained homogenous
process of tissue regeneration due to natural turnover, aging, cell types within each tubule.143
and tissue injury. It is known that genetically normal nonma- Further investigations by our group have demonstrated that
lignant progenitor cells, which are the reservoirs for new cell renal tubular development from digested renal units is possible
formation, are programmed to give rise to normal tissue in the murine model as well. Joraku et al. developed an in vitro
regardless of whether the niche resides in either normal or method for cultivation of renal cells that allowed for develop-
diseased tissues.128–130 Therefore, although the mechanisms ment of tubular structures. This technique involves digestion of
for tissue self-assembly and regenerative medicine are not the entire murine kidney followed by cultivation of the result-
fully understood, it is known that the progenitor cells are ing cells in a specific renal media. Immunohistochemistry
able to “reset” their program for normal cell differentiation. revealed cells expressing proximal and distal tubule markers
The stem cell niche and its role in normal tissue regeneration as well as markers for glomerular and endothelial cells. When
remains a fertile area of ongoing investigation. these cells were allowed to grow on rat-tail type 1 collagen, cells
from the thick ascending loop of Henle stained positive for
Tamm–Horsfall protein in an architectural pattern reminiscent
5.539.5.3. Kidney
of natural tubules.144
The kidney is a complex organ with multiple cell types, derived Yoo et al. evaluated whether murine renal cells grown
from combining anlagen and a complex functional anatomy in vitro could produce functional results in vivo. They harvested
that renders it one of the most difficult to reconstruct.5,131 the cells, expanded them in culture and seeded them onto
While the metanephros is responsible for the development of a tubular device constructed from polycarbonate. At one
the proximal section of the nephrons, the ureteric bud forms end of the tubular device was a silastic catheter, which termi-
the collecting ducts and distal structures. The large vessels nated into a reservoir. The device was subcutaneously
of the kidney are induced from extrarenal tissues. Divergent implanted into athymic mice. The implanted device demon-
embryologic origin converges to produce at least 26 distinct strated extensive vascularization in addition to glomerular
functional cells in the kidney.132 Previous efforts in tissue formation and highly organized tubular architecture. Immu-
engineering of the kidney have been directed toward the devel- nohistochemistry for alkaline phosphatase showed positivity
opment of extracorporeal renal support systems made of in the proximal tubule-like structures. Osteopontin, which is
biological and synthetic components133–141 and these ex vivo secreted by the proximal and distal tubular cells and the cells of
renal replacement devices are known to be life-sustaining. the thin loop of Henle, was found on immunocytochemical
However, there would be obvious benefits for patients with staining of the tubular sections. In addition, the ECM of the
end-stage kidney disease if these devices could be implanted newly formed tubules stained uniformly positive for fibronec-
long term without the need for an extracorporeal perfusion tin. Importantly, fluid collected in the reservoir of the device.
circuit or immunosuppressive drugs. This fluid was yellow and the uric acid concentration was
We have explored the possibility of seeding an implantable 66 mg dl 1 (as compared to 2 mg dl 1 in plasma). The creati-
biomaterial with a heterogeneous population of renal cells to nine concentration of the fluid (27.91  7.56 mg dl 1) was
evaluate function and viability. Atala et al. plated donor rabbit 8.2 times higher than that found in the serum (4.49  0.08 mg
kidney cells, including distal tubules, glomeruli, and proximal dl 1).145 These studies demonstrate that single cells can form
tubules in vitro and after expansion, these were seeded onto multicellular structures, become organized into functional
biodegradeable PGA scaffolds and implanted subcutaneously renal units, and are capable of unidirectional excretion of
into athymic mice. The implants consisted of individual cell solutes through production of a urine-like fluid.
types and a mixture of all three. When examined histologically, Next, we applied the principles of both tissue engineering
progressive formation and organization of the nephron seg- and therapeutic cloning in an effort to produce genetically
ments within the polymer fibers was noted. Additionally, BrdU identical renal tissue in a large animal model, the cow (Bos
incorporation into the renal cell DNA was confirmed.142 It was taurus).146 Bovine skin fibroblasts from adult Holstein steers
unclear if the tubular structures found on the scaffolds were obtained by ear notch, and single donor cells were isolated
occurred de novo from the implanted cells or if they merely and microinjected into the perivitelline space of donor enu-
represented fragments of donor tubules that had survived cleated oocytes (nuclear transfer). The resulting blastocysts were
intact the original dissociation and culture process. To further implanted into progestin-synchronized recipients to allow for
investigate this question, mouse renal cells were harvested and further in vivo growth. After 12 weeks, cloned renal cells were
expanded in culture. Subsequently, a single cell suspension was harvested, expanded in vitro, then seeded onto biodegradable
created from the isolated cells, and these were seeded on bio- scaffolds consisting of polycarbonate membranes and three
degradable polymers and implanted into immunocompetent collagen-coated cylindrical silastic catheters (Figure 6(a)). The
synergic hosts. In this experiment, renal epithelial cells recon- ends of the three membranes of each scaffold were connected to
stituted tubular structures in vivo. The analyses of the retrieved catheters that terminated into a collecting reservoir. This created
implants indicated that the renal epithelial cells first organized a renal neoorgan with a mechanism for collecting the excreted
into a structure with a solid center. Next, canalization into a urinary fluid (Figure 6(b)). These devices were transplanted
hollow tube could be seen at 2 weeks. Histologic examination subcutaneously into the same steer from which the genetic mate-
with nephron-specific lactins revealed successful reconstitution rial originated, and then retrieved 12 weeks after implantation.
From Tissue to Organ Engineering 557

Porous membrane These studies demonstrated that cells derived from nuclear
Silastic Coated transfer can be successfully harvested, expanded in culture, and
catheter collagen Glomerulus
Renal
transplanted in vivo with the use of biodegradable scaffolds on
units which the single suspended cells can organize into tissue struc-
tures that are genetically identical to that of the host. These
Tubule studies were the first demonstration of the use of therapeutic
Reservoir
cloning for regeneration of tissues in vivo.
(a) (b) Investigations into the possibility of using other cell types to
produce renal tissue have also been promising. Perin et al. have
been able to induce AFPSC to differentiate into renal tissues.
Membrane
They labeled human AFPSC with a green fluorescent protein
Allogeneic Cloned Autologous (c) and microinjected the cells into developing murine embryonic
(d) kidneys. They found that the labeled cells assisted in forming
both C- and S- shaped bodies, and the cells expressed RNA for
Figure 6 Combining therapeutic cloning and tissue engineering to
early markers of renal development.153 This study demon-
produce kidney tissue. (a) Illustration of the tissue-engineered renal unit.
strates that AFPSC can differentiate into renal lineages when
(b) Renal unit seeded with cloned cells, 3 months after implantation,
showing the accumulation of urine-like fluid. (c) Clear unidirectional cultured in vitro with renal precursors. Although it looks
continuity between the mature glomeruli, their tubules, and silastic promising, further in vivo studies and functional assays are
catheter. (d) Elispot analyses of the frequencies of T cells that secrete IFN required prior to clinical applications with AFPSC. For more
FX after stimulation with allogeneic renal cells, cloned renal cells, or information on renal tissue engineering strategies, please see
nuclear donor fibroblasts. Cloned renal cells produce fewer IFNg spots Chapter 5.540, Kidney Tissue Engineering in this book.
than the allogeneic cells, indicating that the rejection response to cloned
cells is diminished. The presented wells are single representatives of
duplicate wells. 5.539.5.4. Male Reproductive Organs
Reconstructive surgery is required for a wide variety of patho-
Chemical analysis of the collected urine-like fluid, includ- logic penile conditions, including penile carcinoma, trauma,
ing urea nitrogen and creatinine levels, electrolyte levels, severe erectile dysfunction, and congenital conditions such as
specific gravity, and glucose concentration, revealed that ambiguous genitalia, hypospadias, and epispadias. One of the
the implanted renal cells possessed filtration, reabsorption, major limitations of phallic reconstructive surgery is the avail-
and secretory capabilities. Histological examination of the ability of sufficient autologous tissue. Nongenital autologous
retrieved implants revealed extensive vascularization and tissue sources have been used for decades. Phallic reconstruc-
self-organization of the cells into glomeruli and tubule-like tion was initially attempted in the late 1930s, with rib cartilage
structures. A clear continuity between the glomeruli, the used as a stiffener for patients with traumatic penile loss,154,155
tubules, and the silastic catheter was noted (Figure 6(c)). but this process produced unsatisfactory functional and cos-
Immunohistochemical analysis with renal-specific antibo- metic results. Silicone rigid prostheses were popularized in the
dies revealed the presence of renal proteins, RT-PCR analysis 1970s and have been used widely.156,157 However, biocompat-
confirmed the transcription of renal-specific RNA in the ibility issues have been a problem in selected patients.158,159
cloned specimens, and Western blot analysis confirmed the Tissue transfer techniques with flaps from various nongenital
presence of elevated renal-specific protein levels. sources such as the groin and forearm have been used for genital
Since previous studies have shown that bovine clones harbor reconstruction.160 However, operative complications such as
oocyte mitochondrial DNA,147–149 the donor egg’s mitochon- infection, graft failure, and donor site morbidity are not negligi-
drial DNA (mtDNA) was thought to be a potential source of ble. Phallic reconstruction with autologous tissue, derived from
immunologic incompatibility. Differences in mtDNA-encoded the patient’s own cells, may be preferable in selected cases.
proteins expressed by cloned cells could stimulate a T cell One of the major components of the phallus is corporal
response specific for mtDNA-encoded minor histocompatibility smooth muscle. Initial experiments have shown that cultured
antigens when the cloned cells are implanted back into the human corporal smooth muscle cells may be used in conjunc-
original nuclear donor.150 Maternally transmitted minor histo- tion with biodegradable polymers to create corpus cavernosum
compatibility antigens in mice have been shown to stimulate tissue de novo.161 In a subsequent study, human corporal
both skin allograft rejection in vivo and cytotoxic T lympho- smooth muscle cells and endothelial cells seeded on biode-
cytes expansion in vitro150 that could prevent the use of these gradable polymer scaffolds were able to form vascularized
cloned constructs in patients with chronic rejection of major cavernosal muscle when implanted in vivo.162 Later, in order
histocompatibility matched human renal transplants.151,152 to minimize any immune reactions that a synthetic biomaterial
We tested for a possible T cell response to the cloned renal might cause, naturally derived acellular corporal tissue matrix
devices using delayed-type hypersensitivity testing in vivo and with the same architecture as native corpora was developed.
EliSPOT analysis of interferon-g secreting T cells in vitro. Acellular collagen matrices were derived from processed donor
Both analyses revealed no evidence of a T cell response, indi- rabbit corpora using cell lysis techniques. Human corpus caver-
cating that rejection of cloned renal cells will not necessarily nosal muscle and endothelial cells were derived from donor
occur in the presence of oocyte-derived mtDNA (Figure 6(d)). penile tissue, and the cells were expanded in vitro and seeded
This finding may represent a step forward in overcoming the on the acellular matrices. The matrices were covered with the
histocompatibility problem of stem cell therapy.152 appropriate cell architecture 4 weeks after implantation.163
558 Organ Engineering

In order to look at the functional parameters of the 5.539.5.5. Female Reproductive Organs
engineered corpora, acellular corporal collagen matrices were
Congenital malformations of the uterus may have profound
obtained from donor rabbit penis and autologous corpus
implications clinically. Patients with cloacal exstrophy and
cavernosal smooth muscle and endothelial cells were har-
intersex disorders may not have sufficient uterine tissue pres-
vested, expanded, and seeded on the matrices. An entire
ent for future reproduction. We investigated the possibility
cross-sectional segment of protruding rabbit phallus was
of engineering functional uterine tissue using autologous
excised, leaving the urethra intact. Cell-seeded matrices were
cells.167 Autologous rabbit uterine smooth muscle and epithe-
interposed into the excised corporal space. Functional and
lial cells were harvested, then grown, and expanded in culture.
structural parameters (cavernosography, cavernosometry, mat-
These cells were seeded onto preconfigured uterine-shaped
ing behavior, and sperm ejaculation) were followed, and
biodegradable polymer scaffolds, which were then used for
histological, immunocytochemical, and Western blot analyses
subtotal uterine tissue replacement in the corresponding autol-
were performed up to 6 months after implantation. The engi-
ogous animals. Upon retrieval 6 months after implantation,
neered corpora cavernosa achieved adequate structural and
histological, immunocytochemical, and Western blot analyses
functional parameters.164 This technology was further con-
confirmed the presence of normal uterine tissue components.
firmed when the entire rabbit corpora was removed and
Biomechanical analyses and organ bath studies showed that
replaced with the engineered scaffolds. Most interestingly, mat-
the functional characteristics of these tissues were similar to
ing activity in the animals with the engineered corpora
those of normal uterine tissue. Breeding studies using these
appeared normal by 1 month after implantation. The presence
engineered uteri are currently being performed.
of sperm was confirmed during mating, and was present
Similarly, several pathologic conditions, including congeni-
in all the rabbits with the engineered corpora. The female
tal malformations and malignancy, can adversely affect normal
rabbits mated with the animals implanted with engineered
vaginal development or anatomy. Vaginal reconstruction has
corpora and they conceived and delivered healthy pups.
traditionally been challenging due to the paucity of available
These studies demonstrate that penile corpora cavernosa tissue
native tissue. The feasibility of engineering vaginal tissue in vivo
can be engineered. The engineered tissue is able to achieve
was investigated.168 Vaginal epithelial and smooth muscle cells
adequate structural and functional parameters sufficient for
of female rabbits were harvested, grown, and expanded in cul-
erection, copulation, ejaculation, and conception in rabbits.
ture. These cells were seeded onto biodegradable polymer scaf-
Further studies will be needed to confirm the long-term func-
folds, and the cell-seeded constructs were then implanted into
tionality of these organs. In addition, further studies are
nude mice for up to 6 weeks. Immunocytochemical, histologi-
needed to show that comparable human structures can also
cal, and Western blot analyses confirmed the presence of vaginal
be engineered.
tissue phenotypes. Electrical field stimulation studies in the
Patients with testicular dysfunction require androgen
tissue-engineered constructs showed similar functional proper-
replacement for somatic development. Conventional treat-
ties to those of normal vaginal tissue. When these constructs
ment for testicular dysfunction consists of periodic intramus-
were used for autologous total vaginal replacement, patent vagi-
cular injections of chemically modified testosterone or, more
nal structures were noted in the tissue-engineered specimens,
recently, skin patch applications. However, long-term non-
while the noncell-seeded structures were noted to be stenotic.168
pulsatile testosterone therapy is not optimal and can cause
multiple problems, including excessive erythropoiesis and
bone density changes. To address the problem, a system was 5.539.5.6. Blood Vessels
designed in which Leydig cells, which produce most of Xenogenic or synthetic materials have been used as replace-
the testosterone in the male, were microencapsulated in an ment blood vessels for complex cardiovascular lesions. How-
alginate–poly-L-lysine solution and injected into castrated ani- ever, these materials typically lack growth potential, and may
mals. Serum testosterone was measured serially; the animals place the recipient at risk for complications such as stenosis,
were able to maintain testosterone levels in the long term.165 thromboembolization, or infection.169 Tissue-engineered vas-
These studies suggest that microencapsulated Leydig cells may cular grafts have been constructed using autologous cells and
be able to replace or supplement testosterone in situations biodegradable scaffolds and have been applied in dog and
where anorchia or testicular failure is present. Microencapsu- lamb models.170–173 The key advantage of using these auto-
lated Leydig cells offer several advantages. For example, the grafts is that they degrade in vivo, and thus allow the new tissue
encapsulation process creates a semipermeable barrier between to form without the long-term presence of foreign material.169
the transplanted cells and the host’s immune system, and it Application of these techniques from the laboratory to
allows for the long-term physiologic release of testosterone. the clinical setting has begun, with autologous vascular cells
Other studies have shown that testicular prostheses created harvested, expanded, and seeded onto a biodegradable scaf-
with chondrocytes in bioreactors could be loaded with testos- fold.174 The resultant autologous construct was used to replace
terone, and that these prostheses could provide controlled a stenosed pulmonary artery that had been previously repaired.
testosterone release into the bloodstream when implanted. Seven months after implantation, no evidence of graft occlu-
The prostheses were implanted in athymic mice with bilateral sion or aneurysmal changes was noted in the recipient.
anorchia, and testosterone was released long term, maintain-
ing the androgen level at a physiologic range.166 One could
5.539.5.7. Heart
envision combining the Leydig cell technology described
above with engineered prostheses for the long-term functional In the United States, over 5 million people currently live with
replacement of androgen levels. some form of heart disease, and many more are diagnosed each
From Tissue to Organ Engineering 559

year. While many medications have been developed to assist the therapy. Cell transplantation has been proposed as a potential
ailing heart, the treatment for end-stage heart failure still remains solution for liver failure. The fact that the liver has enormous
transplantation. Unfortunately, as with other organs, donor regenerative potential in vivo suggests that in the right envi-
hearts are in short supply, and even when a transplant can be ronment, it may be possible to expand liver cells in vitro in
performed, the patient must endure the side effects created by sufficient quantities for tissue engineering.177 Many approaches
lifelong immunosuppression. Thus, alternatives are desperately have been tried, including development of specialized media,
needed, and the development of novel methods to regenerate or coculture with other cell types, identification of growth factors
replace damaged heart muscle using tissue engineering and that have proliferative effects on these cells, and culture on
regenerative medicine techniques is an attractive option. three-dimensional scaffolds within bioreactors.177
Cell therapy for infarcted areas of the heart is attractive, as Extracorporeal bioartificial liver devices that use porcine
these methods involve a rather simple injection into the dam- hepatocytes have been designed and applied. These devices
aged area of a patient’s heart, rather than a rigorous surgical are designed to filter and purify the patient’s blood as would
procedure, to complete. Various types of stem cells have been the patient’s own liver, and the blood is returned to the patient
investigated for their potential to regenerate damaged or dead in a manner similar to kidney dialysis. Another cell-based
heart tissue in this manner. Skeletal muscle cells, bone marrow approach is the injection of liver cell suspensions. This has
stem cells (both mesenchymal and hematopoietic), amniotic been performed in animal models. Intraportal hepatocyte
fluid stem cells, and embryonic stem cells have been used for injection has also been used in patients with Crigler–Najjar
this purpose. In this technique, cells are suspended in a bio- Syndrome Type 1178; however, complications such as portal
compatible matrix that can range from simple normal saline to vein thrombosis and pulmonary embolism are major con-
complex yet biocompatible hydrogels depending on the type cerns, especially when large cell numbers are used.179 Finally,
of injection to be performed. The cells are either injected into cells including stem cells, oval progenitor cells, and mature
the damaged area of the heart itself, or they are injected hepatocytes have been seeded onto liver-shaped biocompati-
into the coronary circulation with the hope that they will ble matrices to engineer artificial, implantable livers. These
home to the damaged area, take up residence there, and have been tested in various animal models;180,181 however,
begin to repair the tissue. However, injectable therapies have the transplantation efficiency as well as the functionality of
been shown to be relatively inefficient, and cell loss is quite these constructs must be improved substantially before the
substantial. Newer methods of tissue engineering include the technology can be moved into the clinic.
development of engineered “patches,” which comprise cells
adhered to a biomaterial that can theoretically be used to
5.539.5.9. Articular Cartilage and Trachea
replace the damaged area of the heart. These techniques hold
promise, but require further research into the optimal cell types Full-thickness articular cartilage lesions have limited healing
and biomaterials for this purpose before they can be used capacity and thus represent a difficult management issue for
extensively in the clinic (see Jawad et al.175 for an excellent the clinicians who treat adult patients with damaged articular
review of these methods). cartilage.182,183 Large defects can be associated with mechani-
However, the methods described above could only be used cal instability and may lead to degenerative joint disease if left
in cases where a relatively small section of heart muscle was untreated.184,185 Chondrocytes were expanded and cultured
damaged. In cases where a large area or even the whole heart onto biodegradable scaffolds to create engineered cartilage
has become nonfunctional, a more radical approach may be for use in large osteochondral defects in rabbits.186 When
required. In these situations, the use of a bioartificial heart sutured to a subchondral support, the engineered cartilage
would be ideal, as rejection would be avoided and the pro- was able to withstand physiologic loading and underwent
blems associated with a mechanical heart (such as throm- orderly remodeling of the large osteochondral defects in
boembolus formation) would be abolished. To this end, Ott adult rabbits, providing a biomechanically functional tem-
et al. recently developed a novel heart construct in vitro using plate that was able to undergo orderly remodeling when sub-
decellularized cadaveric hearts. By reseeding the tissue scaffold jected to quantitative structural and functional analyses.
that remained after a specialized decellularization process Few treatment options are currently available for patients
with various types of cells that make up a heart (cardiomyo- who suffer from severe congenital tracheal pathology, such as
cytes, smooth muscle cells, endothelial cells, and fibrocytes) stenosis, atresia, and agenesis, due to the limited availability of
and culturing the resulting construct in a bioreactor system autologous transplantable tissue in the neonatal period. Tissue
designed to mimic physiologic conditions, this group was engineering in the fetal period may be a viable alternative for
able to produce a construct that could generate pump function the surgical treatment of these prenatally diagnosed congential
on its own.176 This study suggests that production of bioartifi- anomalies, as cells could be harvested and grown into trans-
cial hearts may one day be possible. plantable tissue in parallel with the remainder of gestation.
Chondrocytes from both elastic and hyaline cartilage speci-
mens have been harvested from fetal lambs, expanded
5.539.5.8. Liver
in vitro, then dynamically seeded onto biodegradable scaf-
The liver can sustain a variety of insults, including viral infection, folds.187 The constructs were then implanted as replacement
alcohol abuse, surgical resection of tumors, and acute drug- tracheal tissue in fetal lambs. The resultant tissue-engineered
induced hepatic failure. The current therapy for liver failure is cartilage was noted to undergo engraftment and epithelializa-
liver transplantation. However, this therapy is limited by the tion, while maintaining its structural support and patency.
shortage of donors and the need for lifelong immunosuppressive Furthermore, if native tracheal tissue is unavailable, engineered
560 Organ Engineering

cartilage may be derived from bone marrow-derived mesen- 28. Brivanlou, A. H.; Gage, F. H.; Jaenisch, R.; Jessell, T.; Melton, D.; Rossant, J.
chymal progenitor cells as well.188 Science 2003, 300, 913–916.
29. Richards, M.; Fong, C. Y.; Chan, W. K.; Wong, P. C.; Bongso, A. Nat. Biotechnol.
2002, 20, 933–936.
5.539.6. Summary and Conclusion 30. Amit, M.; Shariki, C.; Margulets, V.; Itskovitz-Eldor, J. Biol. Reprod. 2004, 70,
837–845.
31. Reubinoff, B. E.; Pera, M. F.; Fong, C. Y.; Trounson, A.; Bongso, A. Nat.
Regenerative medicine efforts are currently underway experi- Biotechnol. 2000, 18, 399–404; [see comment; erratum in Nat. Biotechnol.
mentally for virtually every type of tissue and organ within the 2000, 18(5), 559].
human body. As regenerative medicine incorporates the fields of 32. Thomson, J. A.; Itskovitz-Eldor, J.; Shapiro, S. S.; et al. Science 1998, 282,
1145–1147; [see comment; erratum in Science 1998, 282(5395), 1827].
tissue engineering, cell biology, nuclear transfer, and materials
33. Reubinoff, B. E.; Itsykson, P.; Turetsky, T.; et al. Nat. Biotechnol. 2001, 19,
science, personnel who have mastered the techniques of cell 1134–1140.
harvest, culture, expansion, transplantation, as well as polymer 34. Schuldiner, M.; Eiges, R.; Eden, A.; et al. Brain Res. 2001, 913, 201–205.
design are essential for the successful application of these tech- 35. Schuldiner, M.; Yanuka, O.; Itskovitz-Eldor, J.; Melton, D. A.; Benvenisty, N. Proc.
nologies to extend human life. Various tissues are at different Natl. Acad. Sci. USA 2000, 97, 11307–11312.
36. Zhang, S. C.; Wernig, M.; Duncan, I. D.; Brustle, O.; Thomson, J. A. Nat.
stages of development, with some already being used clinically, Biotechnol. 2001, 19, 1129–1133.
a few in preclinical trials, and some in the discovery stage. 37. Kaufman, D. S.; Hanson, E. T.; Lewis, R. L.; Auerbach, R.; Thomson, J. A. Proc.
Recent progress suggests that engineered tissues may have an Natl. Acad. Sci. USA 2001, 98, 10716–10721.
expanded clinical applicability in the future and may represent a 38. Kehat, I.; Kenyagin-Karsenti, D.; Snir, M.; et al. J. Clin. Investig. 2001, 108,
407–414.
viable therapeutic option for those who would benefit from the
39. Levenberg, S.; Golub, J. S.; Amit, M.; Itskovitz-Eldor, J.; Langer, R. Proc. Natl.
life-extending benefits of tissue replacement or repair. Acad. Sci. USA 2002, 99, 4391–4396.
40. Assady, S.; Maor, G.; Amit, M.; Itskovitz-Eldor, J.; Skorecki, K. L.; Tzukerman, M.
Diabetes 2001, 50, 1691–1697.
Acknowledgment 41. Itskovitz-Eldor, J.; Schuldiner, M.; Karsenti, D.; et al. Mol. Med. 2000, 6, 88–95.
42. Gurdon, J. B. J. Embryol. Exp. Morphol. 1962, 10, 622–640.
43. Gurdon, J. B. J. Hered. 1962, 53, 5–9.
The author wishes to thank Jennifer L. Olson, Ph.D., for edito- 44. Gurdon, J. B. Dev. Biol. 1962, 5, 68–83.
rial assistance with this manuscript. 45. Gurdon, J. B.; Elsdale, T. R.; Fischberg, M. Nature 1958, 182, 64–65.
46. Campbell, K. H.; Mcwhir, J.; Ritchie, W. A.; Wilmut, I. Nature 1996, 380, 64–66.
47. Wilmut, I.; Schnieke, A. E.; Mcwhir, J.; Kind, A. J.; Campbell, K. H. Nature 1997,
385, 810–813; [see comment; erratum in Nature 1997, 386(6621), 200].
References 48. Cibelli, J. B.; Stice, S. L.; Golueke, P. J.; et al. Science 1998, 280, 1256–1258.
49. Baguisi, A.; Behboodi, E.; Melican, D. T.; et al. Nat. Biotechnol. 1999, 17,
1. Chen, F.; Yoo, J. J.; Atala, A. Urology 1999, 54, 407–410. 456–461.
2. Dahms, S. E.; Piechota, H. J.; Dahiya, R.; Lue, T. F.; Tanagho, E. A. Br. J. Urol. 50. Wakayama, T.; Perry, A. C.; Zuccotti, M.; Johnson, K. R.; Yanagimachi, R. Nature
1998, 82, 411–419. 1998, 394, 369–374.
3. Piechota, H. J.; Dahms, S. E.; Nunes, L. S.; Dahiya, R.; Lue, T. F.; Tanagho, E. A. 51. Betthauser, J.; Forsberg, E.; Augenstein, M.; et al. Nat. Biotechnol. 2000, 18,
J. Urol. 1998, 159, 1717–1724. 1055–1059.
4. Yoo, J. J.; Meng, J.; Oberpenning, F.; Atala, A. Urology 1998, 51, 221–225. 52. De Sousa, P. A.; Dobrinsky, J. R.; Zhu, J.; et al. Biol. Reprod. 2002, 66, 642–650.
5. Amiel, G. E.; Atala, A. Urol. Clin. North Am. 1999, 26, 235–246. 53. Colman, A.; Kind, A. Trends Biotechnol. 2000, 18, 192–196.
6. Amiel, G. E.; Komura, M.; Shapira, O.; et al. Tissue Eng. 2006, 12, 54. Vogelstein, B.; Alberts, B.; Shine, K. Science 2002, 295, 1237.
2355–2365. 55. Hochedlinger, K.; Rideout, W. M.; Kyba, M.; Daley, G. Q.; Blelloch, R.;
7. Atala, A. J. Urol. 1998, 159, 2–3. Jaenisch, R. Hematol. J. 2004, 5(Suppl. 3), S114–S117.
8. Atala, A. Adv. Exp. Med. Biol. 1999, 462, 31–42. 56. Lanza, R. P.; Cibelli, J. B.; West, M. D. Nat. Biotechnol. 1999, 17, 1171–1174.
9. Atala, A. BJU Int. 2001, 88, 765–770. 57. Lanza, R. P.; Cibelli, J. B.; West, M. D.; Dorff, E.; Tauer, C.; Green, R. M. Science
10. Cilento, B. G.; Freeman, M. R.; Schneck, F. X.; Retik, A. B.; Atala, A. J. Urol. 1994, 2001, 292, 1299.
152, 665–670. 58. Hochedlinger, K.; Jaenisch, R. Curr. Opin. Cell Biol. 2002, 14, 741–748.
11. Oberpenning, F.; Meng, J.; Yoo, J. J.; Atala, A. Nat. Biotechnol. 1999, 17, 59. Rideout, W. M. 3rd.; Eggan, K.; Jaenisch, R. Science 2001, 293, 1093–1098.
149–155. 60. Solter, D. Nat. Rev. Genet. 2000, 1, 199–207.
12. Yoo, J. J.; Park, H. J.; Lee, I.; Atala, A. J. Urol. 1999, 162, 1119–1121. 61. Dinnyes, A.; De Sousa, P.; King, T.; Wilmut, I. Cloning Stem Cells 2002, 4,
13. Kim, B. S.; Mooney, D. J. Trends Biotechnol. 1998, 16, 224–230. 81–90.
14. Bergsma, J. E.; Rozema, F. R.; Bos, R. R.; Boering, G.; De Bruijn, W. C.; 62. Takahashi, K.; Yamanaka, S. Cell 2006, 126, 663–676.
Pennings, A. J. Biomaterials 1995, 16, 267–274. 63. Wernig, M.; Meissner, A.; Foreman, R.; et al. Nature 2007, 448, 318–324.
15. Hynes, R. O. Cell 1992, 69, 11–25. 64. Takahashi, K.; Tanabe, K.; Ohnuki, M.; et al. Cell 2007, 131, 861–872.
16. Pariente, J. L.; Kim, B. S.; Atala, A. J. Biomed. Mater. Res. 2001, 55, 33–39. 65. Yu, J.; Vodyanik, M. A.; Smuga-Otto, K.; et al. Science 2007, 318, 1917–1920;
17. Pariente, J. L.; Kim, B. S.; Atala, A. J. Urol. 2002, 167, 1867–1871. 1151526.
18. Li, S. T. In The Biomedical Engineering Handbook; Bronzino, J. D., Ed.; 66. Okita, K.; Ichisaka, T.; Yamanaka, S. Nature 2007, 448, 313–317.
CRC Press: Boca Raton, FL, 1995; pp 627–647. 67. Meissner, A.; Wernig, M.; Jaenisch, R. Nat. Biotechnol. 2007, 25, 1177–1181.
19. Arora, P. D.; Manolson, M. F.; Downey, G. P.; Sodek, J.; McCulloch, C. A. G. 68. Blelloch, R.; Wang, Z.; Meissner, A.; Pollard, S.; Smith, A.; Jaenisch, R. Stem
J. Biol. Chem. 2000, 275, 35432–35441. Cells 2006, 24, 2007–2013.
20. Silver, F. H.; Pins, G. J. Long Term Eff. Med. Implants 1992, 2, 67–80. 69. De Coppi, P.; Bartsch, G., Jr.; Siddiqui, M. M.; et al. Nat. Biotechnol. 2007, 25,
21. Sams, A. E.; Nixon, A. J. Osteoarthr. Cartil. 1995, 3, 47–59. 100–106.
22. Smidsrod, O.; Skjak-Braek, G. Trends Biotechnol. 1990, 8, 71–78. 70. Scriven, S. D.; Booth, C.; Thomas, D. F.; Trejdosiewicz, L. K.; Southgate, J.
23. Lim, F.; Sun, A. M. Science 1980, 210, 908–910. J. Urol. 1997, 158, 1147–1152.
24. Gilding, D. In Biocompatibility of Clinical Implant Materials; Williams, D., Ed.; 71. Liebert, M.; Hubbel, A.; Chung, M.; et al. Differentiation 1997, 61, 177–185.
CRC Press: Boca Raton, FL, 1981; pp 209–232. 72. Liebert, M.; Wedemeyer, G.; Abruzzo, L. V.; et al. Semin. Urol. 1991, 9, 124–130.
25. Mikos, A. G.; Lyman, M. D.; Freed, L. E.; Langer, R. Biomaterials 1994, 15, 55–58. 73. Puthenveettil, J. A.; Burger, M. S.; Reznikoff, C. A. Adv. Exp. Med. Biol. 1999,
26. Harris, L. D.; Kim, B. S.; Mooney, D. J. J. Biomed. Mater. Res. 1998, 42, 462, 83–91.
396–402. 74. Freeman, M. R.; Yoo, J. J.; Raab, G.; et al. J. Clin. Investig. 1997, 99,
27. Peppas, N. A.; Langer, R. Science 1994, 263, 1715–1720. 1028–1036.
From Tissue to Organ Engineering 561

75. Harriss, D. R. Br. J. Urol. 1995, 75(Suppl. 1), 18–26. 126. Hipp, J.; Andersson, K. E.; Kwon, T. G.; Kwak, E. K.; Yoo, J.; Atala, A. BJU Int.
76. Nguyen, H. T.; Park, J. M.; Peters, C. A.; et al. In Vitro Cell. Dev. Biol. Anim. 2008, 101, 100–105.
1999, 35, 371–375. 127. Lin, H. K.; Cowan, R.; Moore, P.; et al. J. Urol. 2004, 171, 1348–1352.
77. Kershen, R. T.; Atala, A. Urol. Clin. North Am. 1999, 26, 81–94. 128. Lai, J. Y.; Yoon, C. Y.; Yoo, J. J.; Wulf, T.; Atala, A. J. Urol. 2002, 168,
78. Atala, A.; Cima, L. G.; Kim, W.; et al. J. Urol. 1993, 150, 745–747. 1853–1857; discussion 1858.
79. Atala, A.; Kim, W.; Paige, K. T.; Vacanti, C. A.; Retik, A. B. J. Urol. 1994, 152, 129. Faris, R. A.; Konkin, T.; Halpert, G. Artif. Organs 2001, 25, 513–521.
641–643; discussion 644. 130. Haller, H.; De Groot, K.; Bahlmann, F.; Elger, M.; Fliser, D. Kidney Int. 2005, 68,
80. Diamond, D. A.; Caldamone, A. A. J. Urol. 1999, 162, 1185–1188. 1932–1936.
81. Bent, A. E.; Tutrone, R. T.; Mclennan, M. T.; Lloyd, L. K.; Kennelly, M. J.; 131. Auchincloss, H.; Bonventre, J. V. Nat. Biotechnol. 2002, 20, 665–666.
Badlani, G. Neurourol. Urodyn. 2001, 20, 157–165. 132. Al-Awqati, Q.; Oliver, J. A. Kidney Int. 2002, 61, 387–395.
82. Chancellor, M. B.; Yokoyama, T.; Tirney, S.; et al. Neurourol. Urodyn. 2000, 19, 133. Aebischer, P.; Ip, T. K.; Miracoli, L.; Galletti, P. M. ASAIO Trans. 1987, 33,
279–287. 96–102.
83. Yokoyama, T.; Huard, J.; Chancellor, M. B. World J. Urol. 2000, 18, 56–61. 134. Aebischer, P.; Ip, T. K.; Panol, G.; Galletti, P. M. Life Support Syst. 1987, 5,
84. Strasser, H.; Berjukow, S.; Marksteiner, R.; et al. Exp. Gerontol. 2004, 39, 159–168.
1259–1265. 135. Amiel, G. E.; Yoo, J. J.; Atala, A. World J. Urol. 2000, 18, 71–79.
85. Yiou, R.; Lefaucheur, J. P.; Atala, A. Anat. Embryol. 2003, 206, 429–435. 136. Humes, H. D.; Buffington, D. A.; MacKay, S. M.; Funke, A. J.; Weitzel, W. F. Nat.
86. Yiou, R.; Yoo, J. J.; Atala, A. Transplantation 2003, 76, 1053–1060. Biotechnol. 1999, 17, 451–455.
87. Machluf, M.; Orsola, A.; Atala, A. World J. Urol. 2000, 18, 80–83. 137. Humes, H. D.; MacKay, S. M.; Funke, A. J.; Buffington, D. A. Kidney Int. 1999, 55,
88. Machluf, M.; Orsola, A.; Boorjian, S.; Kershen, R.; Atala, A. Endocrinology 2003, 2502–2514.
144, 4975–4979. 138. Ip, T. K.; Aebischer, P.; Galletti, P. M. ASAIO Trans. 1988, 34, 351–355.
89. Santen, R.; Swerdloff, R. Clinical aspects of androgen therapy. In Workshop 139. Joki, T.; Machluf, M.; Atala, A.; et al. Nat. Biotechnol. 2001, 19, 35–39.
Conference on Androgen Therapy: Biologic and Clinical Consequences, 140. Lanza, R. P.; Hayes, J. L.; Chick, W. L. Nat. Biotechnol. 1996, 14, 1107–1111.
Pennsylvania State: Philadelphia, PA, 1990. 141. MacKay, S. M.; Funke, A. J.; Buffington, D. A.; Humes, H. D. ASAIO J. 1998, 44,
90. Tai, J.; Johnson, H. W.; Tze, W. J. Transplantation 1989, 47, 1087–1089. 179–183.
91. Van Dam, J. H.; Teerds, K. J.; Rommerts, F. F. Arch. Androl. 1989, 22, 123–129. 142. Atala, A.; Schlussel, R. N.; Retik, A. B. J. Urol. 1995, 153.
92. De Vos, P.; De Haan, B.; Van Schilfgaarde, R. Biomaterials 1997, 18, 273–278. 143. Fung, L. C. T.; Elenius, K.; Freeman, M.; Donovan, M. J.; Atala, A. Pediatrics
93. Tai, I. T.; Sun, A. M. FASEB J. 1993, 7, 1061–1069. 1996, 98(Suppl.), S631.
94. Bazeed, M. A.; Thuroff, J. W.; Schmidt, R. A.; Tanagho, E. A. Urology 1983, 21, 144. Joraku, A.; Stern, K. A.; Atala, A.; Yoo, J. J. Methods 2009, 47, 129–133.
53–57. 145. Yoo, J.; Ashkar, S.; Atala, A. Pediatrics 1996, 98S, 605.
95. Olsen, L.; Bowald, S.; Busch, C.; Carlsten, J.; Eriksson, I. Scand. J. Urol. Nephrol. 146. Lanza, R. P.; Chung, H. Y.; Yoo, J. J.; et al. Nat. Biotechnol. 2002, 20, 689–696.
1992, 26, 323–326. 147. Evans, M. J.; Gurer, C.; Loike, J. D.; Wilmut, I.; Schnieke, A. E.; Schon, E. A.
96. Atala, A.; Vacanti, J. P.; Peters, C. A.; Mandell, J.; Retik, A. B.; Freeman, M. R. Nat. Genet. 1999, 23, 90–93.
J. Urol. 1992, 148, 658–662. 148. Hiendleder, S.; Schmutz, S. M.; Erhardt, G.; Green, R. D.; Plante, Y. Mol. Reprod.
97. Sievert, K. D.; Bakircioglu, M. E.; Nunes, L.; Tu, R.; Dahiya, R.; Tanagho, E. A. Dev. 1999, 54, 24–31.
J. Urol. 2000, 163, 1958–1965. 149. Steinborn, R.; Schinogl, P.; Zakhartchenko, V.; et al. Nat. Genet. 2000, 25,
98. Atala, A.; Guzman, L.; Retik, A. B. J. Urol. 1999, 162, 1148–1151. 255–257.
99. El-Kassaby, A. W.; Retik, A. B.; Yoo, J. J.; Atala, A. J. Urol. 2003, 169, 170–173; 150. Fischer Lindahl, K.; Hermel, E.; Loveland, B. E.; Wang, C. R. Annu. Rev. Immunol.
discussion 173. 1991, 9, 351–372.
100. El Kassaby, A.; Aboushwareb, T.; Atala, A. J. Urol. 2008, 179, 1432–1436. 151. Hadley, G. A.; Linders, B.; Mohanakumar, T. Transplantation 1992, 54,
101. De Filippo, R. E.; Yoo, J. J.; Atala, A. J. Urol. 2002, 168, 1789–1792; discussion 537–542.
1792–1793. 152. Yard, B. A.; Kooymans-Couthino, M.; Reterink, T.; et al. Kidney Int. 1993, 39
102. Le Roux, P. J. J. Urol. 2005, 173, 140–143. (Suppl.), S133–S138.
103. Fu, Q.; Deng, C. L.; Liu, W.; Cao, Y. L. BJU Int. 2007, 99, 1162–1165. 153. Perin, L.; Giuliani, S.; Jin, D.; et al. Cell Prolif. 2007, 40, 936–948.
104. Li, C.; Xu, Y.; Song, L.; Fu, Q.; Cui, L.; Yin, S. Urol. Int. 2008, 81, 290–295. 154. Frumpkin, A. P. Am. Rev. Sov. Med. 1944, 2, 14.
105. Guan, Y.; Ou, L.; Hu, G.; et al. Artif. Organs 2008, 32, 91–99. 155. Goodwin, W. E.; Scott, W. W. J. Urol. 1952, 68, 903–908.
106. Dorin, R. P.; Pohl, H. G.; De Filippo, R. E.; Yoo, J. J.; Atala, A. World J. Urol. 156. Bretan, P. N. In Genitourinary Prostheses; Montague, D. K., Ed.; WB Saunders:
2008, 26, 323–326. Philadelphia, PA, 1989; pp 1–5.
107. Atala, A. J. Urol. 1995, 156, 338. 157. Small, M. P.; Carrion, H. M.; Gordon, J. A. Urology 1975, 5, 479–486.
108. Probst, M.; Dahiya, R.; Carrier, S.; Tanagho, E. A. Br. J. Urol. 1997, 79, 505–515. 158. Nukui, F.; Okamoto, S.; Nagata, M.; Kurokawa, J.; Fukui, J. Int. J. Urol. 1997, 4,
109. Sutherland, R. S.; Baskin, L. S.; Hayward, S. W.; Cunha, G. R. J. Urol. 1996, 156, 52–54.
571–577. 159. Thomalla, J. V.; Thompson, S. T.; Rowland, R. G.; Mulcahy, J. J. J. Urol. 1987,
110. Wefer, J.; Sievert, K. D.; Schlote, N.; et al. J. Urol. 2001, 165, 1755–1759. 138, 65–67.
111. Jayo, M. J.; Jain, D.; Wagner, B. J.; Bertram, T. A. J. Urol. 2008, 180, 392–397. 160. Jordan, G. H. Urol. Clin. North Am. 1999, 26, 1–13.
112. Kropp, B. P.; Sawyer, B. D.; Shannon, H. E.; et al. J. Urol. 1996, 156, 599–607. 161. Kershen, R. T.; Yoo, J. J.; Moreland, R. B.; Krane, R. J.; Atala, A. Tissue Eng.
113. Zhang, Y. J. Urol. 2008, 180, 10–11. 2002, 8, 515–524.
114. Chun, S. Y.; Lim, G. J.; Kwon, T. G.; et al. Biomaterials 2007, 28, 4251–4256. 162. Park, H. J.; Yoo, J. J.; Kershen, R. T.; Moreland, R.; Atala, A. J. Urol. 1999, 162,
115. Kikuno, N.; Kawamoto, K.; Hirata, H.; et al. BJU Int. 2009, 103, 1424–1428. 1106–1109.
116. De Boer, W. I.; Schuller, A. G.; Vermey, M.; Van Der Kwast, T. H. Am. J. Pathol. 163. Falke, G.; Yoo, J. J.; Kwon, T. G.; Moreland, R.; Atala, A. Tissue Eng. 2003, 9,
1994, 145, 1199–1207. 871–879.
117. Baker, R.; Kelly, T.; Tehan, T.; Putnam, C.; Beaugard, E. J. Am. Med. Assoc. 1958, 164. Kwon, T. G.; Yoo, J. J.; Atala, A. J. Urol. 2002, 168, 1754–1758.
168, 1178–1185. 165. Machluf, M.; Atala, A. Graft 1998, 1, 31–37.
118. Gorham, S. D.; French, D. A.; Shivas, A. A.; Scott, R. Eur. Urol. 1989, 16, 166. Raya-Rivera, A. M.; Baez, C.; Atala, A.; Yoo, J. J. World J. Urol. 2008, 26,
440–443. 307–314.
119. Master, V. A.; Wei, G.; Liu, W.; Baskin, L. S. J. Urol. 2003, 170, 1628–1632. 167. Wang, T.; Koh, C.; Yoo, J. J. Creation of an Engineered Uterus for Surgical
120. Jayo, M. J.; Jain, D.; Ludlow, J. W.; et al. Regen. Med. 2008, 3, 671–682. Reconstruction. American Academy of Pediatrics Section on Urology:
121. Kwon, T. G.; Yoo, J. J.; Atala, A. J. Urol. 2008, 179, 2035–2041. New Orleans, LA, 2003.
122. Zhang, Y.; Frimberger, D.; Cheng, E. Y.; Lin, H. K.; Kropp, B. P. BJU Int. 2006, 168. De Filippo, R. E.; Yoo, J. J.; Atala, A. Tissue Eng. 2003, 9, 301–306.
98, 1100–1105. 169. Matsumura, G.; Miyagawa-Tomita, S.; Shin’oka, T.; Ikada, Y.; Kurosawa, H.
123. Farhat, W. A.; Yeger, H. World J. Urol. 2008, 26, 301–305. Circulation 2003, 108, 1729–1734.
124. Atala, A.; Bauer, S. B.; Soker, S.; Yoo, J. J.; Retik, A. B. Lancet 2006, 367, 170. Shinoka, T.; Breuer, C. K.; Tanel, R. E.; et al. Ann. Thorac. Surg. 1995, 60,
1241–1246. S513–S516.
125. Dozmorov, M. G.; Kropp, B. P.; Hurst, R. E.; Cheng, E. Y.; Lin, H. K. J. Smooth 171. Shinoka, T.; Shum-Tim, D.; Ma, P. X.; et al. J. Thorac. Cardiovasc. Surg. 1998,
Muscle Res. 2007, 43, 55–72. 115, 536–545; discussion 545–546.
562 Organ Engineering

172. Shinoka, T.; Shum-Tim, D.; Ma, P. X.; et al. Circulation 1997, 96(II), 180. Gilbert, J. C.; Takada, T.; Stein, J. E.; Langer, R.; Vacanti, J. P. Transplantation
102–107. 1993, 56, 423–427.
173. Watanabe, M.; Shin’oka, T.; Tohyama, S.; et al. Tissue Eng. 2001, 7, 429–439. 181. Kaufmann, P. M.; Kneser, U.; Fiegel, H. C.; Kluth, D.; Herbst, H.; Rogiers, X.
174. Shin’oka, T.; Imai, Y.; Ikada, Y. N Engl J. Med. 2001, 344, 532–533. Transplant. Proc. 1999, 31, 1928–1929.
175. Jawad, H.; Ali, N. N.; Lyon, A. R.; Chen, Q. Z.; Harding, S. E.; Boccaccini, A. R. 182. Hunter, W. Clin. Orthop. Relat. Res. 1995, 317, 3–6.
J. Tissue Eng. Regen. Med. 2007, 1, 327–342. 183. O’Driscoll, S. W. J. Bone Joint Surg. Am. 1998, 80, 1795–1812.
176. Ott, H. C.; Matthiesen, T. S.; Goh, S. K.; et al. Nat. Med. 2008, 14(2), 184. Buckwalter, J. A.; Lohmander, S. J. Bone Joint Surg. Am. 1994, 76, 1405–1418.
213–221. 185. Buckwalter, J. A.; Mankin, H. J. Arthritis Rheum. 1998, 41, 1331–1342.
177. Bhandari, R. N.; Riccalton, L. A.; Lewis, A. L.; et al. Tissue Eng. 2001, 7, 186. Schaefer, D.; Martin, I.; Jundt, G.; et al. Arthritis Rheum. 2002, 46,
345–357. 2524–2534.
178. Fox, I. J.; Chowdhury, J. R.; Kaufman, S. S.; et al. N Engl J. Med. 1998, 338, 187. Fuchs, J. R.; Terada, S.; Ochoa, E. R.; Vacanti, J. P.; Fauza, D. O. J. Pediatr. Surg.
1422–1426. 2002, 37, 1000–1006; discussion 1000–1006.
179. Nieto, J. A.; Escandon, J.; Betancor, C.; Ramos, J.; Canton, T.; Cuervas-Mons, V. 188. Fuchs, J. R.; Hannouche, D.; Terada, S.; Vacanti, J. P.; Fauza, D. O. J. Pediatr.
Transplantation 1989, 47, 449–450. Surg. 2003, 38, 984–987.

Vous aimerez peut-être aussi