Download as pdf or txt
Download as pdf or txt
You are on page 1of 283

Bup Wan Kim

Editor

Clinical Regenerative
Medicine in Urology

123
Clinical Regenerative Medicine
in Urology
Bup Wan Kim
Editor

Clinical Regenerative
Medicine in Urology
Editor
Bup Wan Kim
Department of Urology
Kyungpook National University Hospital
Daegu
South Korea

ISBN 978-981-10-2722-2    ISBN 978-981-10-2723-9 (eBook)


DOI 10.1007/978-981-10-2723-9

Library of Congress Control Number: 2017954376

© Springer Nature Singapore Pte Ltd. 2018


This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or
part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way,
and transmission or information storage and retrieval, electronic adaptation, computer software,
or by similar or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this
publication does not imply, even in the absence of a specific statement, that such names are
exempt from the relevant protective laws and regulations and therefore free for general use.
The publisher, the authors and the editors are safe to assume that the advice and information in
this book are believed to be true and accurate at the date of publication. Neither the publisher nor
the authors or the editors give a warranty, express or implied, with respect to the material
contained herein or for any errors or omissions that may have been made. The publisher remains
neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Printed on acid-free paper

This Springer imprint is published by Springer Nature


The registered company is Springer Nature Singapore Pte Ltd.
The registered company address is: 152 Beach Road, #21-01/04 Gateway East, Singapore 189721,
Singapore
Preface

Regenerative medicine has emerged as an innovative scientific field that


focuses on developing new approaches to repairing cells, tissues, and organs.
Over the years, many therapeutic strategies have been developed to address
clinical shortcomings by enhancing cellular functions and building viable tis-
sues and organs in the field of medicine. This book gathers recent research
and clinical efforts on regenerative medicine applications in urology.
Specifically, this multidisciplinary book contains reviews on the development
of clinical approaches, which combine stem/progenitor cells with biomateri-
als and scaffolds and growth factors and other bioactive agents in the field of
urology. Clinical Regenerative Medicine in Urology aims to provide clini-
cians and researchers a broad perspective on the development of regenerative
medicine technologies with the intention of disseminating regenerative medi-
cine principles and various therapeutic approaches that target clinical applica-
tions in urology. These include upper and lower urinary tract dysfunctions,
urinary incontinence, neurogenic bladder, and erectile dysfunction.
I would like to thank the authors who contributed to the making of this
book as well as the publishing team at Springer. I hope that this book will
encourage urologists to investigate the new paradigms in regenerative medi-
cine research and explore a means to apply the translational concepts to their
clinical practices.

Daegu, South Korea Bup Wan Kim

v
Contents

Part I  Introduction

1 Current Developments and Future Perspectives of Tissue


Engineering and Regenerative Medicine ����������������������������������������  3
Ji Hyun Kim and James J. Yoo

Part II  Fundamentals of Regenerative Medicine

2 Biomaterials and Tissue Engineering��������������������������������������������  17


Sang Jin Lee, James J. Yoo, and Anthony Atala
3 Cell Technologies������������������������������������������������������������������������������  53
So Young Chun
4 Bioreactors for Regenerative Medicine in Urology����������������������  87
In Kap Ko, Anthony Atala, and James J. Yoo
5 3D Bioprinting for Tissue Engineering������������������������������������������ 105
Sujin Noh, Noehyun Myung, Myeongji Park, Seulgi Kim,
Sung-Uk Zhang, and Hyun-Wook Kang
6 Decellularization����������������������������������������������������������������������������  125
Taekmin Kwon and Kyung Hyun Moon
Part III  Cell and Tissue Applications
7 Kidney ��������������������������������������������������������������������������������������������  145
Bum Soo Kim and Hyun Tae Kim
8 Bladder��������������������������������������������������������������������������������������������  191
Yun-Sok Ha and Tae Gyun Kwon
9 Urethra��������������������������������������������������������������������������������������������  215
Yun-Sok Ha and Tae-Hwan Kim
10 Urethral Sphincter: Stress Urinary Incontinence����������������������  237
Eun Sang Yoo and Jun Nyung Lee
11 Penis and  Testis������������������������������������������������������������������������������  275
Seok Cho and Jae Hyun Bae

vii
Part I
Introduction
Current Developments and Future
Perspectives of Tissue Engineering 1
and Regenerative Medicine

Ji Hyun Kim and James J. Yoo

1.1 Introduction 1.2 Current Developments


in TERM
Tissue engineering and regenerative medicine
(TERM) is an interdisciplinary field encompass- Aligned with the goals of TERM, research activi-
ing many disciplines, including engineering, ties have advanced in many disciplines toward
medicine, and science. The field has gained an developing new therapies that overcome the cur-
enormous attention due to its potential to replace rent limitations associated with conventional
damaged human tissues and organs and restore medical and surgical therapies. Over the years,
normal function [1–3]. Although the early con- numerous strategies have been applied for regen-
cept of tissue engineering was developed based erating or replacing damaged tissues and organs,
on cell culture techniques, recent advances in the which led to the development of new therapies
field combine multiple innovative technologies to and products for patients [4, 6–9]. Accordingly,
accelerate the translation of clinical therapies. In several urological tissue technologies have been
fact, a number of TERM technologies have developed for clinical use, including bladder,
advanced to human clinical trials and commer- urethra, and ureter [9–11]. In this section, we
cialized [4, 5]. In this chapter, we review the cur- review the current status of TERM therapies and
rent developments and recent progresses made in discuss new and innovative technologies that
the field of TERM and discuss the most relevant hold promise in the future.
challenges and future perspectives in the transla-
tion of TERM research.
1.2.1 Cell Source

Cells are one of the main basic components of


TERM. Cells can be injected or infused into
damaged tissues alone or by combining with
cell carrier materials. Cells can also be used to
J.H. Kim (*) • J.J. Yoo engineer tissue constructs by attaching on a
Wake Forest Institute for Regenerative Medicine,
scaffolding system fabricated with biomaterials.
Wake Forest University School of Medicine,
Winston-Salem, NC, USA These cells can mature into tissues and organs
e-mail: jihkim@wakehealth.edu; jyoo@wakehealth.edu by direct proliferation and differentiation into

© Springer Nature Singapore Pte Ltd. 2018 3


B.W. Kim (ed.), Clinical Regenerative Medicine in Urology, DOI 10.1007/978-981-10-2723-9_1
4 J.H. Kim and J.J. Yoo

tissue-­specific cells. Implanted cells can also skin, and urine [18, 23]. Although adult stem cells
stimulate tissue formations indirectly by releas- have lower self-renewal and differentiation capac-
ing biological factors. In TERM therapies, allo- ity than embryonic stem cells, they are relatively
genic and autologous cells have been used free from ethical and safety issues and are easily
frequently. Autologous cells are preferred over translated to patients. Adult mesenchymal stem
allogenic cells because these cells are isolated cells, which are derived from bone marrow or adi-
from the host and therefore minimize rejection pose tissues, have been used in numerous studies
and eliminate side effects caused by immuno- [24]. Bone marrow-derived stem/stromal cells can
suppressive medications [4]. Several types of differentiate into various cell types, including
autologous cells, such as smooth muscle cells, osteocytes, chondrocytes, and myocytes, and can
urothelial cells, and muscle-derived cells, have develop into many tissue types including the bone,
been used in the preclinical and clinical settings cartilage, muscle, vessels, and liver. Since Zuk and
with successful outcomes in the field of urology colleagues have first reported on how to obtain
[12–14]. Autologous cells are usually isolated multipotent stem cells from human adipose tissues
from diseased or damaged tissues of the host; in 2001 [25], the use of adipose-derived stem cells
however, obtaining sufficient amount of normal has been enormously increased. Adipose-derived
cell population may be difficult in selected stem cells are multipotent cells, like bone marrow-­
cases. Additionally, fully differentiated cells derived stem cells. However, these cells are easier
may show limited proliferative capacity [15]. to obtain with high yields than bone marrow-
Therefore, alterative cell sources may be derived stem cells [26, 27]. With these favorable
needed to obtain sufficient numbers for clinical characteristics, adult mesenchymal stem cells have
therapies. been frequently used in bladder and urethral
Stem cells are an alternative cell source that reconstructive applications [9, 28].
can overcome the limitations of autologous Another attractive source of stem cells in
somatic cells [16–20]. Stem cells have the ability TERM is amniotic fluid and placental stem cells.
of self-renewal and can be differentiated into one These cells are a mixture of multipotent stem
or more specialized cell types. Stem cells are tra- cells that express embryonic and adult stem cell
ditionally classified according to their origin: markers [29]. They can differentiate into a vari-
embryonic stem cells, fetal stem cells, or adult ety of cell types, including chondrogenic, osteo-
stem cells. Stem cells can also be classified by genic, hepatic, myogenic, neuronal, and hepatic
their plasticity: totipotent, pluripotent, or multi- lineages. They have over 250 population dou-
potent. Embryonic stem cells have a strong blings and do not form tumors in vivo [29–31].
capacity for self-renewal with the potential to dif- With these advantages, amniotic fluid stem cells
ferentiate into any type of cells, so they are an have shown therapeutic effects in various tissue
effective stem cell source for regenerating tissues regeneration studies. For instance, the injection
and organs. However, the use of these cells is of a combination of early differentiated amniotic
very controversial at the present time. Embryonic fluid stem cells into damaged urethral sphincter
stem cells are obtained from the inner cell mass in mice showed new muscle fiber formation and
of embryos. During the isolation process, the improved urodynamic function [32]. Thus, amni-
embryo is destroyed, and somatic nuclear trans- otic fluid and placental stem cells are considered
fer and cloning processes are required [21, 22]. a promising cell source for cell-based TERM
Aside from the ethical issues, embryonic stem research. Their applications are expected to
cells are known to form teratomas in vivo [22]. increase in preclinical and clinical settings [31].
Thus, many countries prohibit the use of embry- Recently, induced pluripotent stem cells
onic stem cells in research. (iPSC) have gained an enormous attention as
Adult stem cells can be obtained in most tissues novel cell sources for TERM applications. These
and organs in the body, including the bone mar- cells are generated by dedifferentiation of adult
row, fat tissue, blood, heart, nerve tissue, muscle, somatic cells through genetic reprogramming
1  Current Developments and Future Perspectives of Tissue Engineering and Regenerative Medicine 5

[33]. iPSC were first discovered by Takahashi for cell attachment, growth, and differentiation.
and Yamanaka in 2006 through the reprogram- Therefore, an ideal biomaterial should be bio-
ming of adult mouse fibroblasts by ­overexpressing compatible and support tissue developments by
four transcription factors, Oct-4, Sox-2, c-Myc, appropriate control of cell attachment, prolifera-
and Klf4 [34]. Pluripotent stem cells can be tion, migration, and differentiation [44]. When
obtained without the use of embryos. Recently, a biomaterials are implanted in the body, they
few studies have shown the possibility of repro- should readily integrate with host tissues without
gramming human cells, as they show similar inducing unfavorable inflammation. Biomaterial
morphology, gene expression, and surface mark- scaffolds possess porous structure to facilitate
ers as human embryonic stem cells [35–37]. cell migration and induce vascularization, as well
These results have opened a new era in TERM, as as provide structural support. Biodegradable bio-
they can be utilized as personalized cell sources, materials specifically should provide mechanical
not only for regenerating damaged tissues or support in early tissue development and should
organs of the patients but also for establishing degrade at a controlled rate over time, thereby
patient-specific platforms in drug discovery. In promoting new tissue formation.
addition, a recent study reported that human Numerous biomaterials with specific proper-
iPSC-derived smooth muscle precursor cells ties have been developed and applied in TERM
improve urethral sphincter function in a rat research. Selection of biomaterials is extremely
model. As such these cell sources have received important for the successful outcome of research
much attention in urology [38]. Issues related to studies. Biomaterials are traditionally classified
incomplete reprograming and teratoma forma- according to their origin: synthetic and natural
tion still remain to be solved, but research of polymer [44, 45]. Biodegradable synthetic poly-
iPSC has seen a rapid and exponential growth mers such as poly-lactic acid (PLA), poly-­
[39–42]. glycolic acid (PGA), poly-lactic-co-glycolic acid
Stem cells have been also found in urine [28]. (PLGA), and poly-caprolactone (PCL) have been
Urine-derived stem cells have high self-renewal widely used in TERM studies due to several
and expansion capacity and low telomerase activ- advantages. These polymers are easy to obtain
ity and secrete paracrine factors. These cells have and handle as compared to natural polymers,
multipotent differentiation potential, including although synthetic polymers are generally not as
osteogenic, chondrogenic, myogenic, adipo- biocompatible as natural polymers. Biodegradable
genic, neurogenic, and endothelial differentia- synthetic polymers can be manufactured in a
tion. Urine-derived stem cells have become a large scale with reproducible physical and chem-
promising cell source in urology, because the ori- ical properties, and their degradation rates are
gin of these cells is from the urinary tract system, controllable. Natural polymers that are frequently
and they can be differentiated into bladder cells used in TERM include collagen, gelatin, alginate,
[23, 28, 43]. fibrin, silk, chitosan, and hyaluronic acids.
Although naturally derived materials are difficult
to control their degradation rates, they have
1.2.2 Biomaterials advantages of possessing biologic recognition
and biocompatibility.
Biomaterials are an essential element in TERM Recently, injectable biodegradable hydrogels,
research. Biomaterials can be used alone as scaf- such as collagen, gelatin, alginate, fibrin, hyal-
folds or used with cells as tissue constructs. uronic acid, and polyethylene glycol (PEG), have
Biomaterials serve as a bridge to fill defect sites been extensively used in TERM research.
and promote new tissue formations by inducing Hydrogels can cross-link, absorb water up to
the body’s ability to regenerate. Biomaterials can 1000 times their dry weight, and provide micro-
also deliver cells into the body to facilitate rapid environments similar to that of native tissues
regeneration. Biomaterials provide environment [46]. They can be injected in combination with
6 J.H. Kim and J.J. Yoo

cells and bioactive molecules, such as growth and heart scaffolds has been tried preclinically to
factors or genes. Despite these advantages in achieve functional tissues for whole-organ engi-
TERM research, the use of hydrogels is limited neering [58–60].
due to the weak mechanical properties and rapid
degradation [47]. As such, various types of syn-
thetic hydrogels and hybrid/composite biomate- 1.2.3 Vascularization
rials have been investigated in order to improve
these properties [46, 48]. Vascularization is a key factor in the successful
Current approaches in biomaterials research outcomes of tissue regeneration. Tissue survival
have focused on developing smart biomaterials primarily depends on oxygen and nutrient supply
that provide bioactive functions. Smart biomate- from host blood vessels in vivo. Diffusion dis-
rials have been investigated with the purpose of tance of oxygen and nutrients is generally
accelerating or enhancing tissue regeneration by 200 μm, so it is difficult for implanted cells that
controlling biological activities of cells and are more than 200 μm away from host vessels to
releasing regulatory factors [49, 50]. survive [61–63]. Therefore, vascularization of
Consequently, a strategy of in situ tissue regen- volumetric, three-dimensional (3D) tissue con-
eration has gained an increasing interest in the structs is necessary to the cells for a long-term
field. This strategy uses the body’s own regenera- survival. As such, several vascularization strate-
tive capacity by activating and recruiting host gies have been investigated in engineered tissues
stem/progenitor cells into the scaffold implant. such as the urethra [64], ureter [9], bone [65],
For facilitating migration, recruitment, homing, cardiac tissue [66], skeletal muscle [67], skin
growth, and differentiation of host cells, func- [68], and lung [69], to overcome the current chal-
tional scaffolding systems containing bioactive lenges associated with diffusion limitation.
molecules, including growth factors, peptides, A common approach for vascularization in
and drugs, have been used. By controlling spatio- TERM is to stimulate vascular ingrowth into the
temporal release of the bioactive molecules con- tissue constructs from adjacent host tissues.
tained in the scaffolding system, tissues can be Angiogenic/vasculogenic growth factors, includ-
regenerated by using only the host cells [51–53]. ing vascular endothelial growth factor (VEGF),
This strategy eliminates ex vivo cell manipula- basic fibroblast growth factor (bFGF), and
tions, which are labor intensive and time-­ platelet-­
derived growth factor (PDGF), have
consuming and require enormous resources. been used to promote angiogenesis [63, 70].
Decellularized tissues and organs have been VEGF has been widely used in tissue engineer-
used as scaffold biomaterials. Decellularization ing applications in urology; for instance, VEGF-­
is the process that removes cellular components binding biomaterials were used to promote
from donor tissues and organs, while retaining angiogenesis that resulted in tissue regeneration
extracellular matrices. Decellularized scaffolds such as urethral and bladder tissues [71, 72].
preserve the architectural structures and shape of Stem/progenitor cells have been implanted for
organs and tissues. Due to lack of cellular com- inducing vascularization in the target tissues to
ponents, immunorejection can be minimized. be regenerated [73, 74]. These cells can release
Therefore, use of decellularized scaffolds has angiogenic factors or partly differentiate into vas-
become an alternative strategy to allogeneic cular endothelial cells following implantation.
organ transplantation [54]. In urology, decellular- Stem cells, vascular cells, and growth factors can
ized urinary bladder matrix and small intestinal be combined in various ways to achieve synergis-
submucosa have been investigated extensively tic effects [70, 75, 76].
for tissue reconstruction. These approaches have These strategies, however, would not be suf-
been successfully translated into the clinic for ficient to form volumetric vascularized tissues of
bladder augmentations [8, 12, 55–57]. Moreover, clinically relevant size. 3D scaffolds implanted
recellularization of decellularized lung, kidney, in vivo take time to be fully vascularized, despite
1  Current Developments and Future Perspectives of Tissue Engineering and Regenerative Medicine 7

the use of angiogenic factors. This is due to the Bioreactors have been applied in a variety of tis-
transient activity of factors that induce angio- sues, including cardiovascular, musculoskeletal,
genesis, and the rate of neovascularization is skin, and bladder tissues [85–89]. In urology,
only ~5 μm/h [77]; thus, the angiogenic activ- bioreactors have been used to culture engineered
ity cannot be maintained long-term [63]. tissues under the condition mimicking the filling
Consequently, the cells residing in the core pressures of bladder. For instance, human urothe-
region of the implanted scaffolds are unable to lial cells seeded on extracellular matrix scaffolds
receive necessary oxygen and nutrients in a were exposed to cyclical filling pressures in the
timely manner. To overcome the limitations urinary bladder bioreactor system. This resulted
associated with delayed vascularization within in a significant increase of urothelial cell growth
the 3D scaffolds, a strategy of pre-vasculariza- as compared with the static culture conditions
tion has been extensively investigated. This [90]. In another example, 3D engineered muscle
strategy involves in vitro as well as in situ forma- constructs were subjected to uniaxial cyclic bio-
tion of vascular structures within the scaffolds reactors, which resulted in improved organiza-
prior to implantation [78]. The rationale for this tion of unidirectional myotubes [91]. Bioreactors
strategy is that preformed vascular structure also have been used in cardiovascular tissue engi-
would be readily integrated with the host vascu- neering to perfuse sufficient oxygen and nutrients
lar system following implantation, resulting in a to engineered tissue constructs [92]. In addition,
rapid perfusion of blood into the implanted scaf- preconditioning of cellular vascular grafts by a
folds. Recent advances in scaffold fabrication pulsatile bioreactor could maintain cellular via-
techniques, such as microfabrication and 3D bility and improve mechanical properties [93].
printing, have enabled the development of 3D Recent advances in computer engineering,
scaffolds that possess complex and perfusable mechanical engineering, and materials science
vascular structures [79–81]. facilitated in designing of more sophisticated
Another vascularization strategy is to facili- bioreactors that simulate physiological environ-
tate vascularization by controlling the properties ments of tissues or organs [94]. With the advances
of microstructure of biomaterials. Highly con- in bioreactor technology, it is expected that its
trolled porous scaffolds facilitate the formation utility will be expanded to large, complex engi-
of vascular networks as well as enhancing supply neered tissue applications such as the kidney,
of oxygen and nutrients [82]. The use of decel- liver, and heart.
lularized tissue scaffolds presents with an advan-
tage due to the preservation of inherent vascular
structures [69, 83]. While establishing adequate 1.2.5 N
 ew and Innovative
vascularization of 3D volumetric scaffolds still Technologies for TERM
remains a challenge, continued development of Applications
innovative strategies using advanced techniques
may lead to a solution that could accelerate the Recent research works in the field of TERM have
translation of various TERM therapies to patients. shown rapid technological evolutions that target
clinical applications via multidisciplinary
research. These technological evolutions reboost
1.2.4 Bioreactors research areas that had previously been stagnated
by innovative solutions and convergence tech-
Bioreactors have been used in TERM to improve nologies. Some of these include scaffold fabrica-
structural and functional maturation of engi- tion methods, monitoring and evaluating tools,
neered tissues in vitro. Bioreactors provide bio- modeling, and preservation. Utilization of these
mechanical forces such as compression, shear technologies will further enable the field to
stress, and pulsatile flow under real-time moni- develop TERM therapies for complex tissue and
tored, highly controlled conditions [84, 85]. organ systems.
8 J.H. Kim and J.J. Yoo

3D printing, an additive manufacturing tech- image acquisition techniques have been devel-
nology, is one of the most explored scaffold fab- oped at the cell, molecular or tissue level [102].
rication technologies currently in TERM [95]. It These techniques allow researchers to collect
has become a powerful tool to fabricate clinically accurate morphological and functional informa-
relevant 3D scaffolds and tissue constructs in a tion without compromising an ongoing study. To
precise manner with high reproducibility. With this end, various cell tacking technologies and
this technology, precise fabrication is possible for related tissue monitoring methods have been
complex tissue and organs of the human body as introduced to track implanted or injected cells
well as scaffolding tailored for the geometry of [102]. Noninvasive magnetic resonance imaging
patients’ lesions. This technology also has been (MRI) has been used to assess tissue functions
used to fabricate surgical models such as kidney, associated with the movement of the body, hemo-
liver, bone, and cardiovascular tissues [96, 97]. In dynamics, and metabolism [102, 103]. For
urology, 3D printing has been used to fabricate instance, to monitor and evaluate stem cell hom-
bladder scaffolds that are later covered with the ing and tissue regeneration effects in vivo, mag-
patient’s own cells [98, 99]. Recently, 3D print- netic nanoparticle-absorbed mesenchymal stem
ing technology has rapidly improved so that vari- cells were injected, and the cells were tracked
ous types of cells as well as biomaterials can be with MRI [104]. Optical imaging techniques are
printed. 3D bioprinting technology has allowed also used in noninvasive real-time measurements
for the fabrication of living tissues and organs of the tissue. With those tools, a series of tissue
having complex structures [99, 100]. Various 3D formation processes such as gene expression, dif-
bioprinting methods, such as inkjet printing, ferentiation, apoptosis, and vascularization from
microextrusion printing, and laser printing, have implantation to remodeling can be investigated
been explored [99]. These bioprinting modalities [102, 103]. These monitoring methods progress
have been modified and combined in various in conjunction with the advancement of relevant
ways to produce tissue constructs effectively. nanoengineering, molecular biology, and com-
With the remarkable improvement of 3D bio- puter science technologies. In the future, nonin-
printing technology, 3D human-scale living tis- vasive image acquisition methods are expected to
sues have been fabricated [101]. While 3D contribute to the analysis of tissue function in
bioprinting technology holds promise as a tool to smaller scale, making it more reproducible, less
generate complex tissue constructs, several issues invasive, faster, more precise, and economical.
remain to be solved, including the development Modeling technology is becoming increas-
of clinically usable software and development of ingly important in TERM. 3D computer simula-
diverse bioinks as well as improvement of bio- tions of cell behaviors predict not only physical
printers that could be reliably used in the clinic. structures but also dynamic interactions between
In the future, one can envision treating a patient cells and environments. “BioSPICE” is open-­
with a tissue deformation. Medical data such as source software to predict spatiotemporal
CT or MRI can be converted into a 3D CAD behaviors of cells, and “cellular automata” sim-
model to generate a patient-specific tissue con- ulate cellular dynamics, including migration
struct using a bioprinter for immediate surgical and differentiation in two-dimensional (2D)
reconstruction in the operating room. space [105, 106]. These technologies might be
Another research area that is gaining increas- essential in the design of improved tissue engi-
ing attention is developing tools for monitoring neering products, as tissue development depends
and evaluation of TERM products. Histological on cellular phenotypes and behavior patterns. In
analysis using sample tissues is the most com- the near future, multi-scale modeling from the
mon method to assess tissue implants. Animals submolecular to the tissue level could be applied
are often sacrificed for tissue collections, and no to provide a framework for bioreactor design
further follow-up will be possible. To overcome and bioinspired tissue engineering constructs
this limitation, noninvasive high-resolution for TERM.
1  Current Developments and Future Perspectives of Tissue Engineering and Regenerative Medicine 9

Cells and tissues’ preservation technology is to patients. However, several challenges still
essential in clinical applications and commercial- remain, and these must be addressed in order to
ization of TERM products. The most common bring more complex tissue therapies to the clinic
technology used in TERM is cryopreservation. [3, 4, 115].
Successful cryopreservation protocols have been One practical challenge that hampers clinical
developed for different cell types with parame- translation is building a clinically relevant-sized
ters that control cryopreservation speed and the composite tissue constructs [98, 116]. In the clin-
type of cryoprotectant chemicals [107, 108]. ical situation, composite tissue damage is more
While the goal of cryopreservation is to preserve common rather than single tissue damage. For
cells for tissue fabrication of engineered tissue instance, a patient with severe facial trauma fre-
products, it is becoming increasingly popular for quently has composite tissue injuries involving
individuals to preserve reproductive tissues for the skin, muscle, nerve, and bone, which should
potential infertility or chemotherapy. To this end, all be reconstructed for functional restoration.
appropriate preservation facilities are required to Considering the clinical relevance, current
preserve cells and tissues without loss of their research should focus on developing implantable
fertility over a long period of time [109]. More composite tissue constructs for patients.
recently, a testicular tissue preservation system Accordingly, the composite tissue constructs
has been established for utilizing spermatogonial should be able to combine at least two different
stem cells, which follows good tissue practice tissue types in a single contiguous construct and
(GTP) protocol of US Food Drug Administration mimic the structural and mechanical characteris-
[110]. Establishing preservation protocols for tics of each tissue type. However, fabricating
various cell types is crucial for the fabrication composite tissue constructs remains a technolog-
and commercialization of cellular composite tis- ical challenge. As previously noted, 3D bioprint-
sue constructs. ing and decellularization/recellularization
technologies have shown the potential of fabri-
cating complex, composite, human-scale tissues
1.3 Challenges and Future [60, 101, 117]. In addition to these technologies,
Perspectives other fabrication methods that can produce com-
posite tissue constructs need to be developed.
Over the past decades, a considerable research The issue associated with identifying ideal
progress has been made to bring TERM technol- cell sources should also be considered. Tissues
ogies toward clinical translation. Consequently, and organs in the body are composed of multiple
several TERM products have been successfully cell types. As such, several types of cells have
translated to human clinical trials, some of which been used to engineer composite tissue constructs
achieved favorable outcomes involving func- to mimic the cellular composition, tissue anat-
tional improvement. For instance, tissue-­ omy, and the function of native tissues [98, 116].
engineered tubularized urethras using autologous However, placing all cell types into a space-­
smooth muscle and epithelial cells were confined construct may not be as feasible. In
implanted into urethral defect sites, and that led addition, ex vivo cell preparation requires enor-
to functional tissue restoration for up to 6 years in mous labor force, time, facility, and resources. To
5 patients [111]. In another example, various facilitate accelerated translation of engineered
types of cells have been implanted in patients for tissues into the clinic, ex vivo cell manipulation
corneal regeneration [112] and for treating myo- should be simplified and improved. Thus, the
cardial infarction [59, 113, 114]. With the suc- selection and placement of cells for target-tissue
cessful clinical translation of initial TERM regeneration should be carefully addressed.
therapies, it is expected that TERM research will Furthermore, in situ tissue regeneration through
continue to focus on developing engineered tis- host cell recruitment may gain increased atten-
sue constructs and therapies that can be translated tion in the future, as this strategy would not need
10 J.H. Kim and J.J. Yoo

ex vivo cell manipulations [51–53]. Further p­ rocesses need to be performed in a GMP (good
development of this strategy will accelerate clini- manufacturing practice) facility. Monitoring and
cal translation of many cell-based therapies. evaluating cells and scaffolds should also be
Integration of implanted engineered con- developed for each processing step. Additionally,
structs with host tissue is another critical consid- optimized and standardized protocols compatible
eration for successful translational outcomes. with ethical and safety regulations should be
Specifically, integration of engineered tissues established and adhered to. Regular and thorough
with host nervous system is critically important discussions between researchers, physicians,
in achieving target tissue function. In engineered engineers, manufacturers, enterprisers, and legis-
bladder tissue, for instance, appropriate innerva- lators will contribute to the successful develop-
tion is directly related to voiding function [10]. In ment of TERM product road map, definition of
engineered muscle tissues, failure of innervation required infrastructure/protocols for each step,
results in abnormal contraction and skeletal mus- and a system of regulation [121].
cle atrophy following implantation in vivo [118].
With the significance of innervation on the func- Conclusion
tional restoration, biological factors, including The field of TERM has achieved a remarkable
growth factors, neurotransmitters, and neuronal progress over the years. TERM is a transla-
cells, have been applied to the engineered tissues tional academic field for developing clinical
[10, 119, 120]. However, the use of those factors applications into the clinic, which may pro-
should be further investigated, in terms of clinical vide a valuable opportunity to overcome the
relevance and effectiveness. Meanwhile, enor- limitations of conventional therapies. Multiple
mous efforts have been made to create vascular- translational products have been applied in
ized engineered tissue constructs for implant patients, and these technologies are expected
survival. However, vascularization of clinically to be available to a wider population in the
relevant-sized constructs and functional integra- coming years. While recent technological
tion with host vasculature still remain a chal- developments have addressed some of the
lenge. Therefore, developing methods to achieve limitations that hamper the translational prog-
rapid vascular and neuronal integration of the ress, a majority of engineering challenges still
engineered tissues is critical. remain a problem. Continued development of
To date, the available commercial products in innovative and technological advances must
TERM are mostly for skin and cartilage regener- be made in order to translate complex and
ation. Additional efforts are needed to diversify composite tissue and organ therapies to the
and commercialize TERM products. However, a clinic.
solid understanding on the manufacturing of
TERM products has been limited, and a road map Acknowledgments The authors thank Margaret van-
has yet to be developed. Therefore, developing an Schaayk for editorial assistance with this manuscript.
infrastructure for TERM manufacturing system
that target clinical applications and commercial-
ization is necessary. In addition, systems for References
quality control should also be established through
1. Kemp P. History of regenerative medicine: look-
standardization of manufacturing processes. ing backwards to move forwards. Regen Med.
Manufactured products should be economically 2006;1(5):653–69.
competitive. For instance, manufacturing of 2. Vacanti CA. The history of tissue engineering. J Cell
TERM products using cells requires multiple Mol Med. 2006;10(3):569–76.
3. Mao AS, Mooney DJ. Regenerative medicine: cur-
processing steps, including tissue biopsy, cell rent therapies and future directions. Proc Natl Acad
isolation and expansion, scaffold design and fab- Sci U S A. 2015;112(47):14452–9.
rication, cell seeding, construct preconditioning, 4. Fisher MB, Mauck RL. Tissue engineering and
and preservation and storage. This series of regenerative medicine: recent innovations and the
1  Current Developments and Future Perspectives of Tissue Engineering and Regenerative Medicine 11

transition to translation. Tissue Eng Part B Rev. 22. Tachibana M, Amato P, Sparman M, Gutierrez NM,
2013;19(1):1–13. Tippner-Hedges R, Ma H, et al. Human embryonic
5. Gemmiti C. Tissue engineering/regenerative medi- stem cells derived by somatic cell nuclear transfer.
cine ventures should invest early in market research Cell. 2013;153(6):1228–38.
to understand the future market's needs. Tissue Eng 23. Liu G, Pareta RA, Wu R, Shi Y, Zhou X, Liu H, et al.
Part B-Re. 2013;19(2):97–8. Skeletal myogenic differentiation of urine-­derived stem
6. Vacanti JP. Tissue engineering: from bench to bedside cells and angiogenesis using microbeads loaded with
via commercialization. Surgery. 2008;143(2):181–3. growth factors. Biomaterials. 2013;34(4):1311–26.
7. Martin I, Smith T, Wendt D. Bioreactor-based road- 24. Kobolak J, Dinnyes A, Memic A, Khademhosseini
map for the translation of tissue engineering strat- A, Mobasheri A. Mesenchymal stem cells: iden-
egies into clinical products. Trends Biotechnol. tification, phenotypic characterization, biological
2009;27(9):495–502. properties and potential for regenerative medicine
8. Caione P, Boldrini R, Salerno A, Nappo SG. Bladder through biomaterial micro-engineering of their
augmentation using acellular collagen biomatrix: a niche. Methods. 2016;99:62–8.
pilot experience in exstrophic patients. Pediatr Surg 25. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW,
Int. 2012;28(4):421–8. Katz AJ, et al. Multilineage cells from human adi-
9. de Jonge PK, Simaioforidis V, Geutjes PJ, Oosterwijk pose tissue: implications for cell-based therapies.
E, Feitz WF. Recent advances in ureteral tissue engi- Tissue Eng. 2001;7(2):211–28.
neering. Curr Urol Rep. 2015;16(1):465. 26. Aust L, Devlin B, Foster SJ, Halvorsen YD, Hicok
10. Horst M, Madduri S, Gobet R, Sulser T, Milleret V, K, du Laney T, et al. Yield of human adipose-­
Hall H, et al. Engineering functional bladder tissues. derived adult stem cells from liposuction aspirates.
J Tissue Eng Regen Med. 2013;7(7):515–22. Cytotherapy. 2004;6(1):7–14.
11. de Kemp V, de Graaf P, Fledderus JO, Ruud Bosch 27. Rhie JW. Adipose-derived stem cells: characteriza-
JL, de Kort LM. Tissue engineering for human ure- tion and clinical application. J Korean Med Assoc.
thral reconstruction: systematic review of recent lit- 2012;55(8):757–69.
erature. PLoS One. 2015;10(2):e0118653. 28. Qin D, Long T, Deng J, Zhang Y. Urine-derived stem
12. Atala A, Bauer SB, Soker S, Yoo JJ, Retik AB. Tissue-­ cells for potential use in bladder repair. Stem Cell
engineered autologous bladders for patients needing Res Ther. 2014;5(3):69.
cystoplasty. Lancet. 2006;367(9518):1241–6. 29. Dziadosz M, Basch RS, Young BK. Human amniotic
13. Carr LK, Robert M, Kultgen PL, Herschorn S, fluid: a source of stem cells for possible therapeutic
Birch C, Murphy M, et al. Autologous muscle use. Am J Obstet Gynecol. 2016;214(3):321–7.
derived cell therapy for stress urinary inconti- 30. De Coppi P, Bartsch G Jr, Siddiqui MM, Xu T,
nence: a prospective, dose ranging study. J Urol. Santos CC, Perin L, et al. Isolation of amniotic stem
2013;189(2):595–601. cell lines with potential for therapy. Nat Biotechnol.
14. Orabi H, AbouShwareb T, Zhang Y, Yoo JJ, Atala 2007;25(1):100–6.
A. Cell-seeded tubularized scaffolds for reconstruc- 31. Murphy SV, Atala A. Amniotic fluid and placental
tion of long urethral defects: a preclinical study. Eur membranes: unexpected sources of highly multipo-
Urol. 2013;63(3):531–8. tent cells. Semin Reprod Med. 2013;31(1):62–8.
15. Schnabel M, Marlovits S, Eckhoff G, Fichtel 32. Chun SY, Kwon JB, Chae SY, Lee JK, Bae JS, Kim
I, Gotzen L, Vecsei V, et al. Dedifferentiation-­ BS, et al. Combined injection of three different lin-
associated changes in morphology and gene expres- eages of early-differentiating human amniotic fluid-­
sion in primary human articular chondrocytes in cell derived cells restores urethral sphincter function in
culture. Osteoarthr Cartil. 2002;10(1):62–70. urinary incontinence. BJU Int. 2014;114(5):770–83.
16. Wagers AJ. The stem cell niche in regenerative med- 33. Wilson KD, Wu JC. Induced pluripotent stem cells.
icine. Cell Stem Cell. 2012;10(4):362–9. JAMA. 2015;313(16):1613–4.
17. Bianco P, Cao X, Frenette PS, Mao JJ, Robey PG, 34. Takahashi K, Yamanaka S. Induction of pluripotent
Simmons PJ, et al. The meaning, the sense and stem cells from mouse embryonic and adult fibroblast
the significance: translating the science of mes- cultures by defined factors. Cell. 2006;126(4):663–76.
enchymal stem cells into medicine. Nat Med. 35. Shi Y. Induced pluripotent stem cells, new tools for
2013;19(1):35–42. drug discovery and new hope for stem cell therapies.
18. Mahla RS. Stem cells applications in regenerative Curr Mol Pharmacol. 2009;2(1):15–8.
medicine and disease therapeutics. Int J Cell Biol. 36. Burridge PW, Li YF, Matsa E, Wu H, Ong SG, Sharma
2016;2016:6940283. A, et al. Human induced pluripotent stem cell-­
19. Kim JH, Song YS. Current status of stem cell ther- derived cardiomyocytes recapitulate the predilection
apy in urology. Korean J Urol. 2015;56(6):409–11. of breast cancer patients to doxorubicin-induced car-
20. Fu Q, Cao YL. Tissue engineering and stem cell diotoxicity. Nat Med. 2016;22(5):547–56.
application of urethroplasty: from bench to bedside. 37. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-­
Urology. 2012;79(2):246–53. Bourget J, Frane JL, Tian S, et al. Induced pluripo-
21. King NM, Perrin J. Ethical issues in stem cell research tent stem cell lines derived from human somatic
and therapy. Stem Cell Res Ther. 2014;5(4):85. cells. Science. 2007;318(5858):1917–20.
12 J.H. Kim and J.J. Yoo

38. Wang Z, Wen Y, Li YH, Wei Y, Green M, Wani P, et al. of three-dimensional matrix scaffolds. Annu Rev
Smooth muscle precursor cells derived from human Biomed Eng. 2011;13:27–53.
pluripotent stem cells for treatment of stress urinary 55. Schaefer M, Kaiser A, Stehr M, Beyer HJ. Bladder
incontinence. Stem Cells Dev. 2016;25(6):453–61. augmentation with small intestinal submucosa leads
39. Lister R, Pelizzola M, Kida YS, Hawkins RD, Nery to unsatisfactory long-term results. J Pediatr Urol.
JR, Hon G, et al. Hotspots of aberrant epigenomic 2013;9(6 Pt A):878–83.
reprogramming in human induced pluripotent stem 56. Zhang F, Liao L. Tissue engineered cystoplasty aug-
cells. Nature. 2011;471(7336):68–73. mentation for treatment of neurogenic bladder using
40. Das AK, Pal R. Induced pluripotent stem cells small intestinal submucosa: an exploratory study.
(iPSCs): the emergence of a new champion in stem J Urol. 2014;192(2):544–50.
cell technology-driven biomedical applications. 57. Joseph DB, Borer JG, De Filippo RE, Hodges SJ,
J Tissue Eng Regen Med. 2010;4:413–21. McLorie GA. Autologous cell seeded biodegradable
41. O'Malley J, Woltjen K, Kaji K. New strategies to scaffold for augmentation cystoplasty: phase II study
generate induced pluripotent stem cells. Curr Opin in children and adolescents with spina bifida. J Urol.
Biotechnol. 2009;20(5):516–21. 2014;191(5):1389–95.
42. Vierbuchen T, Ostermeier A, Pang ZP, Kokubu Y, 58. Uygun BE, Soto-Gutierrez A, Yagi H, Izamis ML,
Sudhof TC, Wernig M. Direct conversion of fibro- Guzzardi MA, Shulman C, et al. Organ reengineer-
blasts to functional neurons by defined factors. ing through development of a transplantable recel-
Nature. 2010;463:1035–41. lularized liver graft using decellularized liver matrix.
43. Zhang Y, McNeill E, Tian H, Soker S, Andersson Nat Med. 2010;16(7):814–20.
KE, Yoo JJ, et al. Urine derived cells are a potential 59. Guyette JP, Gilpin SE, Charest JM, Tapias LF, Ren
source for urological tissue reconstruction. J Urol. X, Ott HC. Perfusion decellularization of whole
2008;180(5):2226–33. organs. Nat Protoc. 2014;9(6):1451–68.
44. Keane TJ, Badylak SF. Biomaterials for tissue 60. Kitahara H, Yagi H, Tajima K, Okamoto K,
engineering applications. Semin Pediatr Surg. Yoshitake A, Aeba R, et al. Heterotopic transplan-
2014;23(3):112–8. tation of a decellularized and recellularized whole
45. Dawson E, Mapili G, Erickson K, Taqvi S, Roy porcine heart. Interact Cardiovasc Thorac Surg.
K. Biomaterials for stem cell differentiation. Adv 2016;22(5):571–9.
Drug Deliv Rev. 2008;60(2):215–28. 61. Folkman J, Hochberg M. Self-regulation of growth in
46. Hunt JA, Chen R, van Veen T, Bryan N. Hydrogels three dimensions. J Exp Med. 1973;138(4):745–53.
for tissue engineering and regenerative medicine. 62. Helmlinger G, Yuan F, Dellian M, Jain RK. Interstitial
J Mater Chem B. 2014;2(33):5319–38. pH and pO2 gradients in solid tumors in vivo: high-­
47. El-Sherbiny IM, Yacoub MH. Hydrogel scaf- resolution measurements reveal a lack of correlation.
folds for tissue engineering: Progress and chal- Nat Med. 1997;3(2):177–82.
lenges. Global cardiology science & practice. 63. Novosel EC, Kleinhans C, Kluger PJ. Vascularization
2013;2013(3):316–42. is the key challenge in tissue engineering. Adv Drug
48. Koutsopoulos S. Self-assembling peptide nanofiber Deliv Rev. 2011;63(4–5):300–11.
hydrogels in tissue engineering and regenerative 64. Imbeault A, Bernard G, Rousseau A, Morissette A,
medicine: progress, design guidelines, and applica- Chabaud S, Bouhout S, et al. An endothelialized uro-
tions. J Biomed Mater Res A. 2016;104(4):1002–16. thelial cell-seeded tubular graft for urethral replace-
49. Mieszawska AJ, Kaplan DL. Smart biomaterials – ment. Can Urol Assoc J. 2013;7(1–2):E4–9.
regulating cell behavior through signaling mol- 65. Mercado-Pagan AE, Stahl AM, Shanjani Y, Yang
ecules. BMC Biol. 2010;8:59. Y. Vascularization in bone tissue engineering con-
50. Zambuzzi WF, Coelho PG, Alves GG, Granjeiro structs. Ann Biomed Eng. 2015;43(3):718–29.
JM. Intracellular signal transduction as a factor in the 66. Sun X, Altalhi W, Nunes SS. Vascularization
development of "smart" biomaterials for bone tissue strategies of engineered tissues and their applica-
engineering. Biotechnol Bioeng. 2011;108(6):1246–50. tion in cardiac regeneration. Adv Drug Deliv Rev.
51. Lee SJ, Van Dyke M, Atala A, Yoo JJ. Host cell mobi- 2016;96:183–94.
lization for in situ tissue regeneration. Rejuvenation 67. Qazi TH, Mooney DJ, Pumberger M, Geissler S,
Res. 2008;11(4):747–56. Duda GN. Biomaterials based strategies for skeletal
52. Kim JH, Jung Y, Kim BS, Kim SH. Stem cell recruit- muscle tissue engineering: existing technologies and
ment and angiogenesis of neuropeptide substance P future trends. Biomaterials. 2015;53:502–21.
coupled with self-assembling peptide nanofiber in 68. Frueh FS, Menger MD, Lindenblatt N, Giovanoli
a mouse hind limb ischemia model. Biomaterials. P, Laschke MW. Current and emerging vasculariza-
2013;34(6):1657–68. tion strategies in skin tissue engineering. Crit Rev
53. Ko IK, Lee SJ, Atala A, Yoo JJ. In situ tissue regen- Biotechnol. 2017;37:613–25.
eration through host stem cell recruitment. Exp Mol 69. Stabler CT, Caires LC Jr, Mondrinos MJ,
Med. 2013;45:e57. Marcinkiewicz C, Lazarovici P, Wolfson MR, et al.
54. Badylak SF, Taylor D, Uygun K. Whole-organ tissue Enhanced re-endothelialization of d­ ecellularized rat
engineering: decellularization and recellularization lungs. Tissue Eng Part C Methods. 2016;22(5):439–50.
1  Current Developments and Future Perspectives of Tissue Engineering and Regenerative Medicine 13

70. Rouwkema J, Khademhosseini A. Vascularization and p­ rinciples, applications and commercial constraints.
angiogenesis in tissue engineering: beyond creating Biotechnol J. 2013;8(3):298–307.
static networks. Trends Biotechnol. 2016;34(9):733–45. 86. Gardel LS, Serra LA, Reis RL, Gomes ME. Use of
71. Jia W, Tang H, Wu J, Hou X, Chen B, Chen W, et al. perfusion bioreactors and large animal models for
Urethral tissue regeneration using collagen scaffold long bone tissue engineering. Tissue Eng Part B-Re.
modified with collagen binding VEGF in a beagle 2014;20(2):126–46.
model. Biomaterials. 2015;69:45–55. 87. Tocchio A, Tamplenizza M, Martello F, Gerges I,
72. Jiang X, Xiong Q, Xu G, Lin H, Fang X, Cui D, Rossi E, Argentiere S, et al. Versatile fabrication of
et al. VEGF-loaded nanoparticle-modified BAMAs vascularizable scaffolds for large tissue engineering
enhance angiogenesis and inhibit graft shrinkage in bioreactor. Biomaterials. 2015;45:124–31.
in tissue-engineered bladder. Ann Biomed Eng. 88. Amrollahi P, Tayebi L. Bioreactors for heart valve
2015;43(10):2577–86. tissue engineering: a review. J Chem Technol Biot.
73. Shepherd BR, Enis DR, Wang F, Suarez Y, Pober 2016;91(4):847–56.
JS, Schechner JS. Vascularization and engraft- 89. Murphy SV, Atala A. Organ engineering–combining
ment of a human skin substitute using circulating stem cells, biomaterials, and bioreactors to produce
progenitor cell-derived endothelial cells. FASEB bioengineered organs for transplantation. BioEssays.
J. 2006;20(10):1739–41. 2013;35(3):163–72.
74. Levenberg S, Rouwkema J, Macdonald M, Garfein 90. Davis NF, Mooney R, Piterina AV, Callanan A,
ES, Kohane DS, Darland DC, et al. Engineering McGuire BB, Flood HD, et al. Construction and
vascularized skeletal muscle tissue. Nat Biotechnol. evaluation of urinary bladder bioreactor for uro-
2005;23(7):879–84. logic tissue-engineering purposes. Urology.
75. Kaully T, Kaufman-Francis K, Lesman A, Levenberg 2011;78(4):954–60.
S. Vascularization-the conduit to viable engineered 91. Heher P, Maleiner B, Pruller J, Teuschl AH,
tissues. Tissue Eng Part B Rev. 2009;15(2):159–69. Kollmitzer J, Monforte X, et al. A novel bioreac-
76. Kim JH, Jung Y, Kim SH, Sun K, Choi J, Kim HC, tor for the generation of highly aligned 3D skel-
et al. The enhancement of mature vessel formation etal muscle-like constructs through orientation of
and cardiac function in infarcted hearts using dual fibrin via application of static strain. Acta Biomater.
growth factor delivery with self-assembling pep- 2015;24:251–65.
tides. Biomaterials. 2011;32(26):6080–8. 92. Massai D, Cerino G, Gallo D, Pennella F, Deriu MA,
77. Utzinger U, Baggett B, Weiss JA, Hoying JB, Edgar Rodriguez A, et al. Bioreactors as engineering sup-
LT. Large-scale time series microscopy of neoves- port to treat cardiac muscle and vascular disease.
sel growth during angiogenesis. Angiogenesis. J Healthcare Eng. 2013;4(3):329–70.
2015;18(3):219–32. 93. Ahn H, Ju YM, Takahashi H, Williams DF, Yoo
78. Laschke MW, Menger MD. Prevascularization in tis- JJ, Lee SJ, et al. Engineered small diameter vas-
sue engineering: current concepts and future direc- cular grafts by combining cell sheet engineering
tions. Biotechnol Adv. 2016;34(2):112–21. and electrospinning technology. Acta Biomater.
79. Baranski JD, Chaturvedi RR, Stevens KR, Eyckmans 2015;16:14–22.
J, Carvalho B, Solorzano RD, et al. Geometric con- 94. Depprich R, Handschel J, Wiesmann HP, Jasche-­
trol of vascular networks to enhance engineered tis- Meyer J, Meyer U. Use of bioreactors in maxillofa-
sue integration and function. Proc Natl Acad Sci U S cial tissue engineering. Br J Oral Maxillofac Surg.
A. 2013;110(19):7586–91. 2008;46(5):349–54.
80. Kolesky DB, Homan KA, Skylar-Scott MA, Lewis 95. Billiet T, Vandenhaute M, Schelfhout J, Van
JA. Three-dimensional bioprinting of thick vas- Vlierberghe S, Dubruel P. A review of trends and
cularized tissues. Proc Natl Acad Sci U S A. limitations in hydrogel-rapid prototyping for tissue
2016;113(12):3179–84. engineering. Biomaterials. 2012;33(26):6020–41.
81. Lee VK, Kim DY, Ngo H, Lee Y, Seo L, Yoo SS, 96. Shafiee A, Atala A. Printing Technologies for Medical
et al. Creating perfused functional vascular chan- Applications. Trends Mol Med. 2016;22(3):254–65.
nels using 3D bio-printing technology. Biomaterials. 97. Soliman Y, Feibus AH, Baum N. 3D printing and its
2014;35(28):8092–102. urologic applications. Rev Urol. 2015;17(1):20–4.
82. Tuzlakoglu K, Reis RL. Biodegradable polymeric 98. Atala A, Kasper FK, Mikos AG. Engineering com-
fiber structures in tissue engineering. Tissue Eng plex tissues. Sci Transl Med. 2012;4(160):160rv12.
Part B Rev. 2009;15(1):17–27. 99. Murphy SV, Atala A. 3D bioprinting of tissues and
83. Quint C, Kondo Y, Manson RJ, Lawson JH, Dardik organs. Nat Biotechnol. 2014;32(8):773–85.
A, Niklason LE. Decellularized tissue-engineered 100. Ozbolat IT, Peng W, Ozbolat V. Application
blood vessel as an arterial conduit. Proc Natl Acad areas of 3D bioprinting. Drug Discov Today.
Sci U S A. 2011;108(22):9214–9. 2016;21(8):1257–71.
84. Plunkett N, O’Brien FJ. Bioreactors in tissue engi- 101. Kang HW, Lee SJ, Ko IK, Kengla C, Yoo JJ, Atala
neering. Technol Health Care. 2011;19(1):55–69. A. A 3D bioprinting system to produce human-­
85. Hansmann J, Groeber F, Kahlig A, Kleinhans scale tissue constructs with structural integrity. Nat
C, Walles H. Bioreactors in tissue engineering – Biotechnol. 2016;34(3):312–9.
14 J.H. Kim and J.J. Yoo

102. Nam SY, Ricles LM, Suggs LJ, Emelianov and long-term corneal regeneration. N Engl J Med.
SY. Imaging strategies for tissue engineering appli- 2010;363(2):147–55.
cations. Tissue Eng Part B Rev. 2015;21(1):88–102. 113. Makkar RR, Smith RR, Cheng K, Malliaras K, Thomson
103. Appel AA, Anastasio MA, Larson JC, Brey LE, Berman D, et al. Intracoronary cardiosphere-
EM. Imaging challenges in biomaterials and tissue derived cells for heart regeneration after myocardial
engineering. Biomaterials. 2013;34(28):6615–30. infarction (CADUCEUS): a prospective, randomised
104. Huang X, Zhang F, Wang Y, Sun X, Choi KY, phase 1 trial. Lancet. 2012;379(9819):895–904.
Liu D, et al. Design considerations of iron- 114. Bolli R, Chugh AR, D'Amario D, Loughran JH,
based nanoclusters for noninvasive tracking of Stoddard MF, Ikram S, et al. Cardiac stem cells in
mesenchymal stem cell homing. ACS Nano. patients with ischaemic cardiomyopathy (SCIPIO):
2014;8(5):4403–14. initial results of a randomised phase 1 trial. Lancet.
105. Hatzikirou H, Deutsch A. Cellular automata as micro- 2011;378(9806):1847–57.
scopic models of cell migration in heterogeneous 115. Dimmeler S, Ding S, Rando TA, Trounson
environments. Curr Top Dev Biol. 2008;81:401–34. A. Translational strategies and challenges in regen-
106. Kumar SP, Feidler JC. BioSPICE: a computational erative medicine. Nat Med. 2014;20(8):814–21.
infrastructure for integrative biology. OMICS. 116. Badylak SF, Weiss DJ, Caplan A, Macchiarini
2003;7(3):225. P. Engineered whole organs and complex tissues.
107. Hunt CJ. Cryopreservation of human stem cells Lancet. 2012;379(9819):943–52.
for clinical application: a review. Transfusion Med 117. Sicari BM, Rubin JP, Dearth CL, Wolf MT,
Hemother. 2011;38(2):107–23. Ambrosio F, Boninger M, et al. An acellular biologic
108. Wang Z, Qin TW. Review: vitreous cryopreservation scaffold promotes skeletal muscle formation in mice
of tissue-engineered compositions for tissue repair. and humans with volumetric muscle loss. Sci Transl
J Med Biol Eng. 2013;33(2):125–31. Med. 2014;6(234):234ra58.
109. Donnez J, Dolmans MM, Pellicer A, Diaz-Garcia C, 118. Kang SB, Olson JL, Atala A, Yoo JJ. Functional
Sanchez Serrano M, Schmidt KT, et al. Restoration of recovery of completely denervated muscle: implica-
ovarian activity and pregnancy after transplantation tions for innervation of tissue-engineered muscle.
of cryopreserved ovarian tissue: a review of 60 cases Tissue Eng Part A. 2012;18(17–18):1912–20.
of reimplantation. Fertil Steril. 2013;99(6):1503–13. 119. Ko IK, Lee BK, Lee SJ, Andersson KE, Atala A, Yoo
110. Sadri-Ardekani H, McLean TW, Kogan S, Sirintrapun JJ. The effect of in vitro formation of acetylcholine
J, Crowell K, Yousif MQ, et al. Experimental testicu- receptor (AChR) clusters in engineered muscle fibers
lar tissue banking to generate spermatogenesis in the on subsequent innervation of constructs in vivo.
future: a multidisciplinary team approach. Methods. Biomaterials. 2013;34(13):3246–55.
2016;99:120–7. 120. Morimoto Y, Kato-Negishi M, Onoe H, Takeuchi
111. Raya-Rivera A, Esquiliano DR, Yoo JJ, Lopez-­ S. Three-dimensional neuron-muscle constructs
Bayghen E, Soker S, Atala A. Tissue-engineered with neuromuscular junctions. Biomaterials.
autologous urethras for patients who need 2013;34(37):9413–9.
reconstruction: an observational study. Lancet. 121. Hunsberger J, Harrysson O, Shirwaiker R, Starly B,
2011;377(9772):1175–82. Wysk R, Cohen P, et al. Manufacturing road map for
112. Rama P, Matuska S, Paganoni G, Spinelli A, De tissue engineering and regenerative medicine tech-
Luca M, Pellegrini G. Limbal stem-cell therapy nologies. Stem Cells Transl Med. 2015;4(2):130–5.
Part II
Fundamentals of Regenerative Medicine
Biomaterials and Tissue
Engineering 2
Sang Jin Lee, James J. Yoo, and Anthony Atala

2.1 Introduction matrix (ECM) proteins secreted by the in-grow-


ing cells. Additionally, cells can be used for ther-
Tissue engineering, a major technique in regen- apy via injection either with carriers, such as
erative medicine, follows the principles of cell hydrogels, or alone [10].
biology, materials science, and engineering to A small piece of donor tissue is dissociated
develop biological substitutes that mimic ana- into individual cells when cells are used for tissue
tomical and functional features of native tissues engineering. These cells may be implanted
in order to restore and maintain normal function directly into the host, or they may be expanded in
of injured or diseased tissues [1–4]. Currently, culture and attached to a scaffold for reimplanta-
tissue engineering strategies can be divided into tion into the host. The source of donor tissue can
two major categories: cell-based and scaffold-­ be heterologous (such as bovine), allogeneic
based approaches. The classic tissue engineering (same species, different individual), or autolo-
approach attempts to create an engineered tissue gous (from the host). Ideally, both structural and
construct by combining cells with a natural and/ functional tissue replacement will occur with
or synthetic scaffold material under suitable cul- minimal complications. Autologous cells are
ture conditions. The scaffold-based tissue engi- preferential to heterologous or allogeneic cell
neering approach is more dependent on the sources because the biopsy tissue is obtained
body’s natural ability to regenerate for proper from the host, the cells are dissociated and
orientation and direction of new tissue in growth. expanded in culture, and the expanded cells are
These scaffolds can be prepared by manufactur- implanted into the original host. While an inflam-
ing artificial microenvironment derived from matory response is possible, the use of autolo-
natural or synthetic materials or by removing cel- gous cells avoids rejection and eliminates the
lular components from tissues using mechanical need for immunosuppressive medications, which
and chemical manipulation to produce collagen- have deleterious side effects.
based tissue matrices [5–9]. These scaffold mate- Currently, most tissue engineering strategies
rials slowly degrade following implantation and are dependent upon obtaining autologous cells
are subsequently replaced by the extracellular from the diseased organ of the host. Patients with
extensive end-stage organ failure, however, are
poor candidates for obtaining autologous cells
S.J. Lee (*) • J.J. Yoo • A. Atala since a tissue biopsy may not yield enough
Wake Forest Institute for Regenerative Medicine, healthy cells for expansion and transplantation.
Wake Forest School of Medicine, Medical Center
Boulevard, Winston-Salem, NC 27157, USA Moreover, primary autologous human cells can-
e-mail: sjlee@wakehealth.edu not be expanded from certain organs, such as the

© Springer Nature Singapore Pte Ltd. 2018 17


B.W. Kim (ed.), Clinical Regenerative Medicine in Urology, DOI 10.1007/978-981-10-2723-9_2
18 S.J. Lee et al.

pancreas. Stem cells are envisioned as being an tissues or organs), and synthetic biodegradable
alternative source of cells from which the desired polymers [e.g., poly(glycolic acid) (PGA),
tissue can be derived. Recovery of stem cells is poly(lactic acid) (PLA), and their copolymers].
possible from discarded human embryos, from These biomaterials have been tested with respect
fetal-related tissue (amniotic fluid or placenta), or to their biological, biochemical, and biomechani-
from adult sources (bone marrow, fat, or skin) cal properties [12]. Natural polymers and acellular
[11]. Therapeutic cloning has also played signifi- tissue matrices have the potential advantage of
cant a role in the development of the field of biological recognition; however, synthetic poly-
regenerative medicine. mers can be produced reproducibly on a large
This chapter describes biomaterials used in scale with controlled properties of their mechani-
tissue engineering, including regeneration of cal strength, degradation rate, and microstructure
specific structures (mostly soft tissues and (Table 2.1).
organs) in the body. Therapies at the cellular,
tissue, and organ levels are described, as well
as the specific challenges and applications 2.2.1 Natural Polymers
associated with each.
Natural polymers are defined as those that are
produced by the cells of a living organism.
2.2 Types of Biomaterials Particularly, structural proteins such as collagen,
laminin, elastin, and fibronectin have been used
Basically, three major classes of biomaterials have as scaffold materials for tissue engineering appli-
been utilized for tissue engineering applications: cations and as vehicles for cell delivery. Among
natural polymers (e.g., collagen and alginate), these, collagen has been found a common use as
acellular tissue matrices (e.g., decellularized a scaffold and carrier for cells in various tissue

Table 2.1  Advantages and disadvantages of biomaterials in tissue engineering


Biomaterials Advantages Disadvantages
Biodegradable synthetic polymers Easy to process Less biological properties than natural
polymers
Controlled biodegradation rate Degrade to form by-products
Controlled surface characteristics Hydrophobic nature
Tunable mechanical properties
Pathogen-free
Natural polymers Biological properties Difficult to control biodegradation rate
Mimic natural ECM structure and Poor mechanical stability
composition
Hydrophilic nature Temperature sensitive
Transfer of pathogens possible
Acellular tissue matrices Appropriate biomechanical Immune responses
properties to apply
Similar structure and composition Difficult to re-cellularize
in native tissues
Transfer of pathogens possible
Composite materials Mostly natural polymers and Limited fabrication processing
synthetic polymers
Synergic effects can be expected In some cases, poor interaction of
interface
Composition can be controlled
2  Biomaterials and Tissue Engineering 19

engineering applications [13–15], because it is acid and N-acetyl-β-D-glucosamine, is broadly


the most abundant protein in mammalian tissues distributed in the ECM and shows a critical role
as the major component of the ECM materials. in vertebrate tissue morphogenesis [26]. Due to
Collagen, which has a triple helix structure, its excellent biocompatibility and hydrodynamic
mainly provides the structural integrity of verte- features, HA has been approved for use in human
brates and organisms. Collagen can be extracted patients as viscous fluid and sheet formulations
and purified from these animal tissues by enzy- for knee pain control and surgical adhesives,
matic digestion and salt/acid extraction methods. respectively. To date, many clinical trials have
The sources of collagens are mostly animal tis- confirmed the safety and effectiveness of HA for
sues such as bovine skin and tendon, porcine skin human clinical trials [27–31]. The bioactivity of
and tendon, and rat tail. Since the use of animal- HA, like that of other carbohydrates, can be also
derived collagens has been concerned due to the improved by physical and/or chemical modifica-
possible transmission of infectious agents, the tion to modulate the cell migration, spreading,
recombinant human collagens have been pro- and multiplication.
duced [16]. Although collagen has excellent bio- Other carbohydrate polymers such as alginate,
logical functions [17, 18], its low biomechanical dextran, and chitosan have been used in various
properties and fast degradation by enzymes as biomedical applications. Alginate (alginic acid),
well as antigenicity potential have been limited composed of linear block copolymers of
for clinical applications. [1–4]-linked β-d-mannuronic acid (M) and α-l-­
Silk fibroin is an insoluble fibrous protein guluronic acid (G) covalently linked together in
obtained by silk worms (Bombyx mori), which different blocks, has been used extensively in a
contains approximately 90% amino acids, hydrogel form for cell encapsulation and drug
including glycine, alanine, and serine [19–21]. delivery [32] as well as in tissue engineering
Purified silk fibroin can be extracted from applications [33]. Alginate is ionically cross-­
degummed silk (boiling off), which refers to the linked by the addition of divalent cations (e.g.,
removal of serine. During the past decades, silk Ca2+) in aqueous solution. The production of
fibroin has been commonly used as a suture purified alginates with consistently high medical-­
material (Surusil®, Suru; Sofsilk™, Covidien). grade quality has been considered. The purity of
To date, various studies reported the use of silk the input algal source is critical, along with the
materials in nanofibers, films, scaffolds, hydro- development of validation strategies for alginate
gels, and microspheres for tissue engineering extraction and purification processes. Alginate
applications with various cell types and drug hydrogel undergoes a slow and unpredictable dis-
delivery applications [21–23]. The unique phys- solution process in vivo, mainly due to the sensi-
icochemical properties of silk include a hydro- tivity of the hydrogels toward calcium chelating
phobic structure, strong intramolecular and compounds [34]. Moreover, a key engineering
intermolecular interactions, and crystal poly- challenge in designing immunoisolation of the
morphism. Therefore, silk fibroin enables tun- alginate-based microcapsules for cell encapsula-
able and slow biodegradation kinetics without tion is that of maintaining unhampered exchange
toxic chemical cross-linkers. The hydrolysis of of nutrients, oxygen, and therapeutic molecules
silk fibroin is mainly attributed to the proteo- released by the encapsulated cells while avoiding
lytic enzymes, including part of the foreign- swelling and subsequent rupture of the alginate
body response cascade [24]. microcapsules [35]. Although many advances in
Natural carbohydrates have been utilized as alginate-based biomaterials have been informed,
hydrogels for tissue engineering applications and numerous progresses will be required for clinical
drug carrier materials for drug delivery system applications.
[25]. For instance, the hydrophilic polysaccha- Chitosan is also a linear polysaccharide of
ride hyaluronic acid (HA), which is composed of [1–4]-linked D-glucosamine and N-acetyl-D-­
repeating disaccharide units of β-D-glucuronic glucosamine residues, which is a polycationic
20 S.J. Lee et al.

material produced by the deacetylation of chitin. sion, proliferation, and differentiation [40],
Chitosan is insoluble in aqueous solution (above numerous studies have encouraged the tissue
pH 7) because of its crystalline structure, while it formation by combining cells with tissue-spe-
is fully soluble in dilute acids (below pH 5). It has cific ECM scaffolds from decellularized heart
been used in a number of gene and drug delivery [45], urinary bladder [46], liver [47], lung [48],
applications due to the abundant positive charged kidney [49], and small intestinal submucosa
functional groups. Also, its applications in tissue (SIS) [50]. Each of these decellularized tissue
engineering and regenerative medicine have been matrices maintained the biomechanical proper-
broadly investigated [36–38]. ties and structural integrity of the original tissue
or organ with minimal disruption to the ECMs.
More importantly, these ECM proteins play a
2.2.2 Acellular Tissue Matrices vital role in tissue maintenance and regeneration
of a specific tissue type, and they can modulate
ECM contributes to biomechanical/structural cell adhesion and migration, growth factor stor-
integrity and signaling and regulatory functions age and release, and progenitor cell activation
in the development, maintenance, and regenera- and differentiation [51, 52].
tion of tissues. ECM components participate in One of the most common acellular tissue
the tissue-specific control of gene expression matrices is small intestinal submucosa (SIS),
through a variety of transduction mechanisms in which is prepared by mechanical removal of
synergic effects with soluble biochemical signals tunica mucosa and muscularis, and serosal layer
provided by growth factors, cytokines, and hor- [53]. Acellular SIS contains various bioactive
mones [39–41]. Moreover, ECM is itself a molecules such as fibronectin, glycosaminogly-
dynamic structure that is actively remodeled by cans (GAGs), and growth factors [54]. Recently,
direct interactions with the cells [42]. commercially available SIS-based matrices have
Development of novel scaffolding systems that been successfully implanted in several recon-
more closely recapitulate the biological and bio- structive surgical procedures, which include her-
mechanical functions of native ECM remains an nia repair [55, 56], abdominal wall closure [57],
important area of tissue engineering approaches urinary incontinence [58, 59], bladder augmenta-
[43]. Understanding ECM’s complex functions tion [60], pelvic organ prolapse repair [61], and
in mature or regenerating tissues continues to be so on. As SIS provides high biological variability,
an important task to design a synthetic standardized protocol for preparation, and manu-
microenvironment. facturing procedure with quality control, SIS-
In the current limitations for de novo con- based tissue products can be a reliable therapeutic
struction of a true ECM mimic from purified option for clinical applications.
ECM components, decellularized tissue matrices Skeletal muscle tissue matrices have been
have been considered as an ideal scaffolding decellularized, characterized in vitro, and vali-
material due to their biological, structural, and dated in animal models [62–64]. We believe that
biomechanical similarity to native tissues and the intact ECM proteins play an important role in
their possession of tissue-specific ECM proteins tissue maintenance and reconstruction of skeletal
which remain after the decellularization process muscle tissue; furthermore, they modulate cell
[44]. As a result, these matrices have been used adhesion and migration, growth factor storage
for a number of tissue engineering applications and release, and satellite cell activation and
and have yielded an understanding of the physi- ­differentiation [51, 52]. Moreover, immune
cochemical properties of ECM as well as a responses to allogeneic or xenogeneic cell
tissue-­specific scaffold for engineering func- sources and tissues involved the recognition of
tional tissues or organs. For example, by har- major histocompatibility complex (MHC) mole-
nessing the cell-matrix interactions that are cules expressed on the surface of the cells by the
crucial for cellular behaviors, including adhe- immune cells of the host. The decellularization
2  Biomaterials and Tissue Engineering 21

process removes all cellular components, includ- 2.2.3 Synthetic Biodegradable


ing DNA, and leaves a structure comprised of Polymers
ECM materials. Thus, minimal immune rejection
response may occur with the transplantation Synthetic polymers are high molecular weight
of these allogeneic or xenogeneic nature scaf- macromolecules consisted of covalently bound
folds [64]. repeating units (monomers). These repeating
The use of native heart ECM is of special units can react with each other to form high
interest for cardiac-related cells due to the pres- molecular weight homopolymers by mainly addi-
ence of intrinsic regulatory factors for cardiac tion polymerization, step-growth polymerization,
function [65–67]. To engineer a tissue construct, and ring-opening polymerization. In tissue engi-
cells are dependent on the ECM providing tissue-­ neering, biodegradable synthetic polymers offer
specific molecular, structural, and mechanical a number of benefits as scaffolds or drug delivery
signals that regulate cell behaviors. Thus, scaf- vehicles [71, 72]. These polymers can be synthe-
fold materials for engineered cardiac tissue sized with reproducible quality controls and high
should mimic the native heart ECM in order to purity and fabricated into various shapes with
provide a natural microenvironment for the car- anticipated bulk and surface features. Specific
diac cells. Recently, incorporation of heart ECM advantages include the ability to tailor the
into a bioink formulation for bioprinting cardiac mechanical properties and degradation kinetics
tissue has been suggested [68]. In an attempt to of these polymeric materials to suit various tissue
produce such a unique ECM component, decel- engineering applications. Linear aliphatic poly(α-­
lularization technique of native tissues has been a hydroxy acids), such as PGA, PLA, and their
very attractive method, while natural polymers copolymer [poly(lactide-co-glycolide) (PLGA)],
and synthetic polymers are unable to replicate the are the most commonly used biodegradable poly-
precise spatial organization of complex struc- mers in tissue engineering (Fig. 2.1) [72]. These
tures like cardiac tissue [69]. Generally, decellu- biodegradable polymers have gained popularity
larization strategy is to remove cellular due to their processing reliability, tunable
components from tissues via mechanical or mechanical properties, and controlled biodegrad-
chemical manipulation to produce ECM-rich ability, and they are Food and Drug Administration
matrices. (FDA)-approved for human use in a variety of
The high degree of evolutionary preserva- medical applications, including suture material,
tion of ECM components allows the use of implant, and drug delivery systems.
acellular matrices derived from xenogeneic The ester bonds in these polymers degrade by
sources. Various decellularized tissue matrices nonenzymatic hydrolysis, and their nontoxic
have been utilized successfully for tissue engi- degradation products are eliminated from the
neering in animal models, and several xenoge- body in the form of carbon dioxide and water.
neic products have received regulatory approval The degradation rate of these polymers can be
for clinical use [70]. These decellularized xeno- controlled by alteration of their crystallinity,
geneic medical products are being introduced molecular weight, and the copolymer ratio of
into the market. Despite many advantages, monomers. Furthermore, the degradation rates
there are concerns about the use of decellular- that can be achieved range from several weeks to
ized xenogeneic tissue matrices. These include several months. Because these polymers are
the potential for immunogenicity, the possible thermoplastics, they can be configured into a
­
presence of infectious agents, variability among three-­dimensional (3D) structure with a desired
preparations, and the inability to completely microarchitecture, shape, and dimension.
specify and characterize the bioactive mole- However, synthetic polymers generally lack
cules of the material. Many of the soluble bio- intrinsic biological properties, and their degrada-
active factors in the tissue matrices can be lost tion products may cause adverse effects or alter
during the decellularization process. local microenvironment in vivo. In addition, the
22 S.J. Lee et al.

CH3 O O O CH3 O
H2 H2
O C C O C C O C C O C C
H H
n n n m

PLA PGA PLGA

O O O O
H2 H2 H H2 H
O (CH2)2 O C C C (CH2)5 O C C C O C C C O
n n n m
CH3 CH2
Polydioxanone PCL PHBV
CH3

Fig. 2.1  Chemical structure of the commonly used biodegradable synthetic polymers in tissue engineering

surface hydrophobicity of synthetic polymers functionality have been used. The main strategy
may mediate protein denaturation in the vicinity of this hybrid biomaterial system is to achieve
of the implant and induce fibrous encapsulation synergy between the desirable biomechanical
[73, 74]. properties of one component with the biological
Several research teams have explored the syn- compatibility and physiological relevance of the
thesis of novel polymeric biomaterials that unite other component [77]. For example, natural poly-
the advantages of “smart” polymeric systems mers possess microstructures that contain chemi-
with the biological activities of proteins at the cally and structurally defined components.
chemical level. The concept of these smart poly- Combination of these materials with synthetic
meric systems was initially derived from the polymers would offer a convenient bridge
development of polymeric materials that show between chemical and biosynthetic approaches,
large conformational changes in response to often within the context of thermodynamically
microenvironmental stimuli, such as tempera- driven assembly processes [78].
ture, ionic strength, pH, or light [75]. The
responses of the polymers may include precipita-
tion or gelation, reversible adsorption on a sur- 2.3 Basic Requirements
face, collapse of a hydrogel or surface grafting, of Biomaterials in Tissue
and alternation between hydrophilic and hydro- Engineering
phobic states [76]. In most of cases, the change in
the state of the polymer is reversible. Beyond the Biomaterials replicate the biologic and biome-
physical properties of these polymers, imparting chanical features of the native ECMs found in tis-
smart biomaterials with the specific properties of sues in the body by serving as a synthetic
signaling molecules, such as ECM components microenvironment. Biomaterials provide a 3D
and growth factors, can be achieved. Hence, space for the cells to form new tissues with
smart polymeric biomaterials aim to actively par- tissue-­specific structure and function; moreover,
ticipate in the biological system in the body for they could allow for the delivery of cells and
accelerating tissue maturation and, eventually, ­bioactive molecules (such as cell adhesive pro-
regeneration. teins and growth factors), to the targeted sites in
To maximize the role of biomaterials in tissue the body [74]. As the majority of mammalian cell
regeneration, a combination of two or more mate- types are anchorage-dependent, biomaterials pro-
rials with unique properties is often selected for vide a cell adhesion substrate that can deliver
use as a scaffold. These hybrid biomaterial sys- cells to specific sites with high loading efficiency
tems that possess biological and physiochemical and homogeneity. Biomaterials can also provide
2  Biomaterials and Tissue Engineering 23

biomechanical support against in vivo forces to ture that permits the formation of neo-tissues,
maintain the predefined 3D tissue structure dur- and [3] guide the development of neo-tissues
ing the neo-tissue formation. Also, bioactive with tissue-specific functions [7]. In details, bio-
molecules, including adhesive proteins and materials can provide an adhesion substrate for
growth factors, can be loaded along with cells to transplanted cells and serve as a delivery vehicle
regulate the cellular behaviors. into the sites of interest. A large surface area-to-­
In order to minimize the host responses, volume ratio is desirable in order to allow for
including inflammation, biomaterials should be accommodation of a high number of cells, which
biodegradable and bioresorbable. Incompatible dictates the use of porous scaffolds. It is often
materials produce an inflammatory or foreign-­ desirable for neo-tissues to have a predefined
body reaction that eventually leads to rejection structure, which can be achieved by an appropri-
and/or tissue necrosis. Degradation products, if ate selection of biomaterials with controlled
produced, should be removed from the body degradable rates. In addition, biomaterials can
through the metabolic pathway at a designed rate provide a temporary mechanical support that is
that keeps the concentration of these degradation sufficient to withstand in vivo forces and main-
products in the body at a tolerable level [79]. tain a spatial structure until neo-tissue is able to
Biomaterials should also provide a tissue-specific support itself. This is essential for hard, weight-­
microenvironment in which appropriate regula- bearing tissue regeneration like the bone and car-
tion of cell behaviors (adhesion, proliferation, tilage. The stability of biomaterials depends
migration, and differentiation) can occur so that primarily on physical properties and chemical
functional neo-tissue can be formed. Cell behav- degradation [8]. The mechanical properties of
iors in the newly formed tissue have been shown biomaterials can be adjusted to suit the needs of
to be regulated by multiple interactions of the the target tissue or organ along with the fabrica-
cells with their microenvironment, including tion technologies.
interactions with cell adhesion ligands [80] and A porous architecture with interconnectivity
with soluble growth factors [81]. Since biomate- that allows the cellular ingrowth and maximizes
rials provide temporary mechanical support the oxygen and nutrient diffusion is considered as
while the cells undergo spatial tissue reorganiza- an essential scaffold fabrication requirement.
tion, the properly chosen biomaterials should Configuration of such a structure may require tis-
allow the engineered tissue to maintain sufficient sue-specific considerations, which include varia-
biomechanical integrity to support itself in early tions in pore morphology and surface
developmental stage. In late development, bio- characteristics. In addition, biomaterial should be
materials begin degradation; hence tissue growth biodegradable or amenable to long-term integra-
is unhindered [74]. tion with the host tissue. Biodegradability allows
for the gradual and orderly replacement of the
scaffold with functional tissue and prevents the
2.3.1 Scaffolding System: Template development of adverse chronic responses to the
artificial tissue structure [9]. Therefore, biomateri-
Biomaterials for cell-based tissue engineering als should be selected based on an understanding
approaches should be biodegradable and absorb- of target tissues or organs biologically, anatomi-
able without eliciting inflammatory responses cally, pathologically, and biomechanically.
that interfere with cellular function and neo-­
tissue formation; thus it has been considered to
their biological properties, biodegradability, and 2.3.2 Biodegradability/
structural and mechanical properties. In tissue Bioabsorbability
engineering, biomaterials should [1] facilitate the
localization and delivery of somatic cells to spe- The mechanism of biodegradation of synthetic
cific sites in the body, [2] maintain a 3D architec- polymeric biomaterials is mainly processed
24 S.J. Lee et al.

through passive hydrolysis, with the mono- Unlike the synthetic polymers, the biodegra-
meric acids as the final metabolized product dation of the natural polymers is mainly depen-
[73]. The degradation rates are closely related dent on the enzymatic reaction. This is the
to molecular weight, degree of crystallization, microenvironmental responsiveness of the natu-
hydrophilicity/hydrophobicity, and surface-to- ral polymers through degradation and remodel-
volume ratio. For instance, PGA is generally ing by cell-secreted enzymes [83]. The
available in a highly crystalline form, and it biodegradation rate of these natural polymers can
possesses a relatively high melting point be controlled by various cross-linking processes.
(~230 °C) and low solubility in organic sol- For instance, a collagen scaffold that is designed
vents. However, its hydrophilic nature allows for a long-term in vivo use would require a cross-­
for rapid absorption of water leading to a fast linking process to improve its resistance to the
degradation within a few weeks. In contrast, proteolytic degradation and hydrolysis and to
PLA is available in semicrystalline or amor- enhance its mechanical strength [12].
phous forms, depending on the types of the d Glutaraldehyde is a commonly used cross-linker
and l optical isomers of lactic acid present in for tissue matrices because it has amine-reactive
the molecule. D-PLA and L-PLA are both homobifunctional groups [84]. Carbodiimide
highly crystalline, whereas the racemic copoly- chemistry has been used extensively to cross-link
mer D,L-PLA is completely amorphous. The collagen scaffolds and to immobilize other pro-
presence of a methyl group makes PLA more teins to the scaffolds. Particularly, 1-ethyl-3-(3-­
hydrophobic than PGA, resulting in lower dimethylaminopropyl) carbodiimide
water absorption rates and correspondingly hydrochloride (EDC) has been used as a zero-­
slower degradation, in the order of months [10]. length cross-linker that directly binds between
Moreover, the factors affecting biodegradation collagen molecules [85].
include chemical and structural configuration, Biomaterials used for cell-based tissue engi-
fabrication parameters, sterilization methods, neering serve as an artificial ECM and bring forth
and implantation sites. For example, steriliza- biological, biochemical, and biomechanical
tion using γ-irradiation can decrease the poly- functionalities of native ECM found in tissues
mer’s Mw by 30–40%. The irradiated polymers and organs. Therefore, cell-biomaterial interac-
continue to decrease in Mw during storage at tions, such as cellular adhesion on the matrix sur-
room temperature. The decline in Mw affects face, cell maturation, ECM production, and
the mechanical properties and the biodegrada- proliferation, are critical for the success of this
tion rate of the polymers [8, 11]. strategy. ECM is known to affect cellular differ-
The degradation of polymeric biomaterials entiation and function; thus, biomaterials can be
results in erosion (loss of mass), when the deg- designed to deliver cells and bioactive molecules
radation products diffuse or dissolve into the (e.g., cytokines and growth factors) to a desig-
body fluid. Polymer erosion is generally classi- nated region for neo-tissue formation with
fied by bulk erosion and surface erosion [82]. enhanced functionality [4–6].
The polymer chain scission by the bulk erosion
is wholly occurred, while the surface erosion is
only occurred in the surface of the polymeric 2.4 Advanced Biomaterial
scaffolds. In the bulk erosion, the molecular Systems in Tissue
weight and mechanical properties of the poly- Engineering
meric scaffolds decrease in time; however, the
external dimension of the scaffolds remains Future advances in biomaterials for tissue engi-
essentially unchanged until the scaffold is fully neering rely on the development of the biomi-
degraded. In the surface erosion, the external metic materials that can utilize in vivo
dimension and mass of the scaffolds decrease in microenvironment for efficient tissue regenera-
time. tion and better integration with host tissue
2  Biomaterials and Tissue Engineering 25

(Fig. 2.2). Desirable biomimetic materials could onto biomaterial surfaces to achieve added func-
be designed as a smart scaffolding system that tionality [88]. These approaches provide an oppor-
mimics the natural biological system and inte- tunity to tailor a biomaterial surface so that it has
grates the necessary structural and biological specific affinities, site recognition properties, and
properties. Thorough understanding of biomate- controlled mobility, while the bulk properties of
rials science combined with extensive knowledge biomaterials are still maintained.
of the clinical challenges and cell biology is vital
for development of novel advanced biomaterials
to be used for tissue engineering applications. 2.4.2 Drug Delivery System

Biomaterial functionalization has also allowed


2.4.1 Surface Modification enhanced cellular interactions via delivery of bio-
active molecules from an implanted scaffold
One commonly used technique is the surface mod- [89]. Bioactive molecules, such as cytokines and
ification of polymeric biomaterials. Chemical and growth factors, are powerful regulators of bio-
physical modification of the biomaterials surface logical functions, which include the cell migra-
can increase their biological properties (Fig. 2.3). tion, proliferation, and differentiation.
Chemical procedures, such as oxidation, Incorporation of bioactive molecules into bioma-
­hydrolysis, and quaternization, change the surface terial scaffolds is a technique to improve the out-
chemistry and functionality of biomaterials.
­ come of cell-based tissue engineering. To employ
Polymerization or grafting of water-soluble poly- this technique, an understanding of the biological
mers to a biomaterial surface is usually performed activities of molecules is necessary. For example,
to improve the protein adsorption and further cell the biological activity of growth factors is
adhesion [86, 87]. In addition, bioactive mole- ­dependent not only on their presence in solution
cules, such as enzymes, drugs, proteins, peptide but also on their interactions with the surround-
sequences, and antibodies, have been immobilized ing ECM. Some growth factors are most effective

Endogenous Migration  Replication  Differentiation


stem cells

Biochemical signals
Growth factors
Cytokines
Chemokines
Ch Small molecules
em
ota
xis Genes

Physical signals
ECM proteins
Topography
Elasticity

Topography Immobilization Elasticity

Biomimetic materials to utilize microenvironment

Fig. 2.2  Schematic illustration of interactions between endogenous stem cells and biomimetic materials. Stem cell fate
in a particular microenvironment is regulated by intricate reciprocal molecular interactions with its surroundings
26 S.J. Lee et al.

Fig. 2.3  Schematic diagram of surface modification methods of Polymeric biomaterials

when released over a prolonged period of time, influenced the attachment, growth, and differen-
whereas others are more effective when delivered tiation of human embryonic stem cells (ESCs) in
in a bolus. Some factors are active while tethered unexpected ways [91]. More revolutionary devel-
to a material, whereas others are only active when opments in biomaterials science will continue to
they have been released from the biomaterial and arise as tissue engineering is combined with
are internalized into a cell. These considerations technology-­based approaches. Basic understand-
must be taken into account when designing a ing of the 3D structure of existing biological
delivery system [90]. molecules is being applied to a “bottom-up”
­
approach to generate new, self-assembling supra-
molecular architectures [92].
2.4.3 ECM-Mimetic Biomaterials Self-assembling peptides offer promise
because of the large variety of sequences that can
Development of advanced biomaterials has been be made easily by automated chemical synthesis.
extended to the design, discovery, and evaluation The potential for bioactivity, the ability to form
of biologically active materials. This required the nanofibers, and responsiveness to microenviron-
relatively straightforward chemical synthesis of mental cues are inherent in some of these bioma-
new polymeric materials, coupled with a search terials [93]. Recent advances include the design
for novel activities and examination of their per- of short peptides (e.g., heptamers) based on
formance in biological systems. By adapting the coiled-coil motifs that reversibly assemble into
combinatorial library approach that has been well nanofilaments and nanoropes, without excessive
established for synthetic peptides and small mol- aggregation [94]. These novel peptide amphiphi-
ecule drugs with novel high-throughput assays, les can be induced to self-assemble by changes in
thousands of candidate chemicals can be synthe- concentration, pH, or the level of divalent cations
sized and tested. As one example, screening pro- [95]. Branched structures can be designed to
cess of a combinatorial library derived from present bioactive sequences such as arginine-­
commercially available monomers in the acrylate glycine-­aspartic acid (RGD) to cells by nanofi-
family revealed novel synthetic polymers that brous hydrogels or as coatings on typical tissue
2  Biomaterials and Tissue Engineering 27

engineering scaffolds [96]. In addition, assembly [100–103]. This may allow selective capture and
can occur under conditions that permit the recovery of specific cell types, delivery of cells to
­entrapment of cells in the resulting nanofibrous a desired location, and modulation of enzymes
matrix [97]. The entrapped cells retain motility that influence the tissue remodeling process. The
and the ability to proliferate. Peptide-based nano- discovery of new classes of biomaterials may
fibers can be designed to present bioactive provide yet another opportunity to overcome the
sequences to cells at very high density, substan- current limitations in tissue engineering.
tially exceeding that of corresponding peptide
epitopes in biological ECM.
A pentapeptide epitope of laminin, isoleucine-­ 2.4.4 Biophysical Signals
lysine-­valine-alanine-valine (IKVAV), known to
promote neurite extension, was incorporated into Cells are exposed to a tissue-specific microenvi-
peptide amphiphiles capable of self-assembly ronment in vivo, and they respond to numerous
into nanofibers that form a hydrogel [98]. When chemical and physical stimuli. As a result, cellu-
neural progenitor cells (NPCs) capable of differ- lar functions like cell adhesion and proliferation,
entiating into neurons or glial cells were encap- protein synthesis, and cytoskeletal architecture
sulated during assembly of the nanofibers, they are critically influenced by the microenvironment
survived over a few weeks in culture. Moreover, of the cells. To maintain the phenotype expres-
even without the addition of neurotrophic factors, sion and differentiation of tissue-specific cells,
they displayed neuronal differentiation as exem- numerous cues have been applied to biomaterial
plified by the extension of large neurites that scaffolds to mimic natural ECM microenviron-
were obvious after 1 day in culture and by their ments. Cells can be directly influenced by topo-
expression of β-tubulin III. The production of graphical cues of their surrounding
neuron-like cells from neural progenitors, microenvironment. The topography of the bio-
whether dissociated or grown as clustered neuro- material surface can influence the cell adhesion
spheres, was more rapid and robust in the IKVAV-­ and proliferation, which further affects cellular
containing peptide amphiphile (PA) gels than on functions. It has been determined that natural
laminin-coated substrates or with soluble ECM has a nanoscaled fibrous structure that sup-
IKVAV. By contrast, the production of cells ports the cellular interactions and stimulates the
expressing glial fibrillary acidic protein (GFAP), cellular functions. This substrate nanoscale
a marker of astrocytic differentiation, was sup- topography provides a tool for investigating com-
pressed significantly in the IKVAV-PA hydrogels plex cell functions, such as adhesion, migration,
even when compared to growth on laminin, cytoskeleton reorganization, and cell polarization
which favors neuronal differentiation. The ability [104, 105].
to direct stem or progenitor cell differentiation Physical contact of cells on the biomaterial
via a chemically synthesized biomaterial, with- surface is essential to anchorage dependence and
out the need to incorporate growth factors, offers function. Current understanding reveals that stem
many potential advantages in tissue engineering cell fates can be significantly affected by the
and regenerative medicine. ­substrate elasticity from the surrounding micro-
Proteins have been conjugated either ran- environment, while stem cell differentiation
domly or in a site-specific manner, through engi- induced by soluble biological signals has been
neering of protein to introduce a reactive amino widely developed. For example, neural cell dif-
acid at a particular site [99]. If a conjugation site ferentiation can be induced on soft matrices (high
is introduced near the ligand-binding domain of elasticity), which mimic the mechanical proper-
protein, it has been shown that induction of a ties of the brain tissue, while myogenic and
change in conformational state of the novel poly- osteogenic differentiation require stiffer matrices
mers can serve to regulate the protein’s activity (low elasticity) that mimic muscle and
28 S.J. Lee et al.

c­ollagenous bone tissues. Soft matrices that 2.5.1 Hydrogels


mimic brain are neurogenic, stiffer matrices that
mimic muscle are myogenic, and comparatively Hydrogels have been used in injectable
rigid matrices that mimic collagenous bone prove approaches for cell therapy and tissue engineer-
osteogenic [106]. These studies have demon- ing, which offer many advantages. These
strated that specific stem cell differentiation include the repair of tissue defects or injection
can be achieved using substrates or matrices of cells for achieving cellular function [33].
with particular surface elasticity/stiffness. However, while injection therapy is simple and
Modification of the cross-­linking degree of poly- does not require invasive surgical procedures,
meric hydrogels is the basis of substrate-con- its application is limited to areas of the body
trolled elasticity. where injection is feasible. Injectable biomate-
rials usually take the form of liquids or gels,
which can be configured from synthetic and
2.5 Scaffold Fabrication natural polymers. A “hydrogel” is defined as a
Technologies 3D network swollen by water which is usually
the major component of the hydrogel system.
Biomaterials used for tissue engineering applica- Hydrogels are formed by cross-­linking water-
tions require different spatial configurations, soluble polymers to form an insoluble, hydro-
depending on the desired outcome. In tissue engi- philic polymer network. These polymer
neering, biomaterials need to be fabricated to networks are capable of absorbing water from a
facilitate regeneration of the target tissues or small fraction of their dry weight up to thou-
organs. Thus, starting with simple foams and sands of times their dry weight [107].
fibers, the porosity and pore interconnectivity of Representative hydrogels include poly(ethylene
biomaterial scaffolds has quickly become a cen- glycol) (PEG), poly(ethylene oxide) (PEO),
tral theme, with pore dimensions and physical poly(vinyl alcohol) (PVA), poly(acrylic acid)
properties being vital in promoting cell seeding, (PAA), Pluronics (PEO-PPO-PEO copolymers),
migration, proliferation, and de novo production agarose, alginate, chitosan, collagen, fibrin, gel-
of ECM molecules. Many fabrication technolo- atin, and hyaluronic acid (HA).
gies have been employed to configure biomateri- In the past decade, progress in creating 3D
als in this way for tissue-engineered scaffolds. In cellular microenvironments using hydrogels has
such instances, biomaterials provide the struc- appeared to have similar elasticity to that of
tural support for cells to proliferate and maintain native tissues. The structure and composition of
their function. To permit cell accommodation, hydrogels can be tailored to bear the suitable
biomaterials must possess adequate surface area chemical, biological, and physical cues that
for cell attachment and distribution. This is usu- encourage the development of tissue constructs
ally achieved by creating interconnecting pores [108]. For example, matrix metalloproteinase
that allow for maximum diffusion of oxygen (MMP)-sensitive PEG-based hydrogels have
and nutrients. Thus, numerous fabrication been developed to direct differentiation of plu-
technologies have been developed to create
­ ripotent cardioprogenitor cells using P19 embry-
porous scaffolds, which include solvent casting/ onal carcinoma cells [109]. In order to
particulate leaching, freeze-drying, fiber bond- systematically probe 3D microenvironmental
ing, electrospinning, phase separation, gas foam- effects on P19 embryonal carcinoma cell differ-
ing, high pressure processing, melt molding, entiation, matrix elasticity, MMP-sensitivity, and
membrane lamination, hydrocarbon templating, the concentration of a matrix-bound RGDSP
cell printing, and solid free-form fabrication peptide were modulated. Results indicated that
(SFF) methods. soft PEG-based hydrogels mimicking the
2  Biomaterials and Tissue Engineering 29

e­ lasticity of embryonic cardiac tissue could direct and particulate leaching, provide scaffolds with
cardiac commitment of pluripotent P19 cells. very high porosity but potentially low intercon-
The most common cell encapsulation system nectivity and may not allow for easy shaping
involves alginate microcapsules, which can [113]. Electrospinning has become a popular
enclose living cells or other molecules. Cell alternative fabrication method, as it can be
encapsulation techniques such as this allow allo- applied to many disciplines and is a relatively
geneic and xenogeneic cell sources to be consid- simple and inexpensive scaffold fabrication pro-
ered for clinical translation, and this is important cess. It relies on a high-voltage direct or alternat-
as autologous cells can often be limited in supply. ing current source to charge a polymer solution
In order to use allogeneic or xenogeneic cell or melt contained in a syringe [114]. Production
sources, the immune responses of the host need of nanofibers by electrospinning provides excel-
to be addressed. An immunoisolation technique lent pore interconnectivity and precise control of
using cell encapsulation permits the use of these porosity through adjustment of fiber size [115].
cell sources without the use of immunosuppres- This interconnectivity can allow for integration
sants. Cell microencapsulation can be used for of cells into the scaffold if the pore size is large
cell therapy as well as for delivering biological enough [116] and also allows for dissolution of
products. This technique consists of enclosing soluble factors and nutrients throughout the scaf-
biologically active substances within a polymeric fold. These properties closely mimic the natural
hydrogel surrounded by a semipermeable mem- properties of ECM, and the parameters of the
brane that is designed to prevent immune recog- electrospinning process can be controlled so as to
nition and subsequent rejection. Ideally, the adjust to features of targeted tissue types. For
encapsulating membrane allows the bidirectional instance, arteries are composed of a layer called
diffusion of nutrients, oxygen, and waste as well the intima, which consists of an endothelial cell
as the secretion of therapeutic products while (EC) layer lining the lumen of the artery, which
preventing immune cells and antibodies, which sits on a basal lamina. Surrounding this layer is
would normally destroy the enclosed cells, from the tunica media, which is a layer of connective
entering the capsule [110]. Pancreatic islets, tissue and smooth muscle cells (SMCs). To
hepatocytes, renal cells, parathyroid cells, and mimic this vessel structure, our lab showed that it
adrenal chromaffin cells have been immunoiso- is possible to create a bilayered vascular scaffold
lated for cell-based therapeutic delivery systems which supports EC growth on an electrospun inti-
in this manner. Several polymeric encapsulation mal layer and SMC growth on an electrospun
systems have been developed and are currently media layer (Fig. 2.4) [116].
being tested in clinical trials [111, 112]. Electrospun fibrous scaffolds can also be
functionalized by adding biochemical and bio-
mechanical cues to enhance cellular interactions;
2.5.2 Electrospinning however, specific interactions between cells and
electrospun scaffolds functionalized through sur-
While many other scaffold fabrication methods face modifications and bioactive factor incorpo-
are used in tissue engineering applications, few ration are still poorly understood. A better
can provide scaffolds with critical similarities to understanding of the specific cues that enhance
natural ECM that electrospinning of nanofibers cell adhesion, proliferation, and guidance of cells
can provide. Other fabrication processes allow seeded on a scaffold, as well as the cues that
for the inclusion of natural polymers such as col- could affect host cell infiltration, differentiation,
lagen into the scaffold, but some, such as melt and vascularization both in vitro and in vivo, is
molding, require high temperatures which may crucial for the advancement of tissue engineering
denature proteins. Others, such as solvent casting applications of electrospinning.
30 S.J. Lee et al.

a b

TRITC FITC Merge

c d e

Fig. 2.4  Bilayered electrospun PCL/collagen vascular (e) interface between outer and inner layers of bilayered
scaffolds; (a) gross appearance, (b) fluorescent images of scaffolds. Reproduced with permission from Ju et al.
bilayered scaffolds incorporated fluorescent dyes. SEM [116]
images of cross-sectional (c) outer layer, (d) entire, and

2.5.3 Computer-Aided Scaffold ing system has been utilized to build various 3D
Fabrication architectures by depositing a continuous stream
of cell-laden hydrogel [121]. These approaches
For clinical translation, a fabrication method take advantage of rapid prototyping assisted by
should provide a scaffold with appropriate shape computer-assisted design (CAD) procedures to
and size to reconstruct damaged tissues in build a tissue construct with geometric control of
patients. Computer-aided fabrication or additive its internal structure and external shape. We have
manufacturing of tissue-engineered constructs is recently showed that viable 3D heterogeneous
a rapidly emerging technology that promises to construct with multiple cell types could be cre-
accelerate the clinical translation of many tissue ated by printing the cells with hydrogel layer-by-­
engineering applications. This technology is layer [120]. This cell-printed constructs were
based on the ability to build tissue-engineered able to survive and mature into tissues with ade-
constructs with highly reproducible architecture quate vascularization when implanted in vivo
and to provide compositional variation across the (Fig. 2.5) [122].
entire scaffold through tightly controlled, In 2016, our laboratory has reported a novel
computer-­driven fabrication. To accomplish this, integrated tissue and organ printing (ITOP) sys-
a conventional inkjet printer can be used [117– tem that makes it possible to fabricate a high
120]. This thermal inkjet printer produces small strength tissue construct and precisely place vari-
air bubbles consisting of cells and a hydrogel by ous cell types within a single architecture [122,
heating, and then bubbles are collapsed to pro- 123]. This system consists of multiple nozzles
vide the pressure pulse to eject a very tiny drop of that allow delivery of multiple types of biomate-
“bioink” out of the nozzle. This printing tech- rials and cells. The goal of ITOP is to concur-
nique uses aqueous processes that have been rently deliver a supportive 3D template (mostly
shown to induce little damage to cells. In the biodegradable thermoplastic polymers) and 3D–
extrusion printing method, a pneumatic dispens- patterned deposition of cell-laden hydrogel
2  Biomaterials and Tissue Engineering 31

a b c g

d e f h

Fig. 2.5  3D multiple cell positioning: (a–c) type I pattern 6 (n = 3). (h) Cell proliferation results showed that the
and (d–f) type II pattern. (a, d) Two types of 3D pattern- number of cells continuously increased over a 15-day
ing, including cell-A (red), cell-B (blue), and PCL (green), period, and no significant differences between the control
were fabricated. (b, e) Photographs and (c, f) fluorescent and the printed constructs were observed (n = 5).
image of the 3D printed patterns. (g) Cell viability (%) Reproduced with permission from Kang et al. [122]
was over 95% on day 0 and then maintained on days 3 and

b­ iomaterials in a precise manner. This approach cedure and the low morbidity associated with it.
is borne out of the challenges of tissue engineer- Investigators are seeking alternative biomaterials
ing 3D tissue constructs. that would be safe for human use. The ideal sub-
stance for the endoscopic treatment of reflux and
incontinence should be injectable, nonantigenic,
2.6 Tissue Engineering nonmigratory, volume stable, and safe for human
Applications use. Toward this goal, long-term studies were
conducted to determine the effect of injectable
Several therapeutic approaches on the cellular chondrocytes in vivo. It was initially determined
level have been investigated for direct treatment that alginate, a liquid solution suspended with
of medical conditions or for use of implanted chondrocytes, could serve as a synthetic substrate
engineered tissue constructs. A variety of tissue for the injectable delivery and maintenance of
engineering products have been tested by injec- cartilage architecture in vivo. This system was
tion, gene delivery, implantation, encapsulation, adapted for the treatment of vesicoureteral reflux
or combined systems. Applications in tissue in a mouse model [124] and a porcine model
engineering of the several therapies are described. [125]. The use of autologous cartilage for the
treatment of vesicoureteral reflux in humans
would satisfy all the requirements for an inject-
2.6.1 B
 ulking Agents for Urinary able substance.
Incontinence Two multicenter clinical trials were conducted
and Vesicoureteral Reflux using this technology. Patients with vesicoure-
teral reflux were treated at ten centers throughout
Injectable bulking agents have been used endo- the USA. The patients had a similar success rate
scopically in the treatment of both urinary incon- as with other injectable substances, in terms of
tinence and vesicoureteral reflux. The advantages cure. Chondrocyte formation was not noted in
in treating urinary incontinence and vesicoure- patients who had treatment failure. It is supposed
teral reflux with this minimally invasive approach that the patients who were cured have a biocom-
include the simplicity of a quick outpatient pro- patible region of engineered autologous tissue
32 S.J. Lee et al.

present, rather than a foreign material [126]. model [130] and a canine model [10, 131] of
Patients with urinary incontinence were also urethral sphincter injury, and both replacement
treated endoscopically with injected chondro- of mature myotubes, as well as restoration of
cytes at three different medical centers. Phase 1 functional motor units, were noted in the regen-
trials showed an approximate success rate of 80% erating sphincteric muscle tissue. This was the
at follow-up 3 and 12 months postoperatively first demonstration of the replacement of both
[127]. Therefore, human application of cell-­ sphincter muscle tissue and its innervation by
based tissue engineering for urologic applica- the injection of MPCs. As a result, MPCs may
tions occurred successfully with the injection of be a minimally invasive solution for urinary
chondrocytes for the correction of vesicoureteral incontinence in patients with irreversible uri-
reflux in children and for urinary incontinence in nary sphincter muscle insufficiency.
adults [126, 127].

2.6.3 Endocrine Replacement


2.6.2 I njection Therapy Using
Muscle Progenitor Cells Patients, who have a testicular dysfunction and
hypogonadal disorders, are dependent on andro-
The potential use of injectable cultured primary gen replacement therapy to restore and maintain
muscle progenitor cells (MPCs) for the treatment physiological levels of serum testosterone and its
of stress urinary incontinence has been investi- metabolites, dihydrotestosterone, and estradiol
gated [128]. Fluorescence-labeled MPCs were [132]. Currently available androgen replacement
injected directly into the proximal urethra and modalities, such as testosterone tablets and cap-
lateral bladder walls of nude mice with a micro- sules, depot injections, and skin patches, may be
syringe in an open surgical procedure. Tissue associated with fluctuating serum levels and
harvested up to 35 days after injection contained complications such as fluid and nitrogen reten-
the labeled MPCs, as well as evidence of differ- tion, erythropoiesis, hypertension, and bone den-
entiation of the labeled MPCs into newly formed sity changes [133]. Since Leydig cells of the
myofibers. It was reported that a significant por- testes are the major source of testosterone in
tion of the injected MPC population persisted men, implantation of heterologous Leydig cells
in vivo. Similar techniques of sphincteric-derived or gonadal tissue fragments has previously been
MPCs have been used for the treatment of uri- proposed as a method for chronic testosterone
nary incontinence in a clinical trial [129]. The replacement [134]. However, these approaches
fact that MPCs can be labeled, survive after injec- were limited by the failure of the tissues and cells
tion, and begin the process of myogenic differen- to produce testosterone.
tiation further supports the feasibility of using Encapsulation of cells in biocompatible and
cultured cells of muscular origin as an injectable semipermeable polymeric membranes has been
bioimplant. an effective method to protect against host
The use of injectable MPCs has also been immune responses while maintaining viability
investigated for use in the treatment of urinary of the cells and allowing the secretion of
incontinence due to irreversible urethral sphinc- desired therapeutic agents [135, 136].
ter injury or maldevelopment. MPCs are the Encapsulation of Leydig cells in alginate-
quiescent satellite cells found in each myofiber poly(l-lysine) was used as a novel method for
that proliferate to form myoblasts and, eventu- testosterone delivery in vivo [132]. Elevated
ally, myotubes and new muscle tissue. stable serum testosterone levels were noted in
Previously, intrinsic MPCs have been shown to castrated adult rats over the course of the study,
play an active role in the regeneration of injured which suggests microencapsulated Leydig
striated urethral sphincter. In a subsequent cells may be a potential therapeutic modality
study, autologous MPCs were injected into a rat for testosterone supplementation.
2  Biomaterials and Tissue Engineering 33

2.6.4 Urethral Tissue Further study, however, indicated that when tubu-
larized urethral repairs with unseeded scaffolds
Various approaches have been proposed for the were attempted experimentally, adequate urethral
regeneration of urethral tissue. PGA nonwoven tissue regeneration was not achieved, and com-
meshes without cells [137, 138] or with cells plications, such as graft contracture and stricture
[139] were used to regenerate urethra tissues in formation, occurred [144]. To determine if seed-
various animal models. Acellular tissue matrices ing the scaffold with cells from the urinary tract
such as decellularized bladder submucosa [5] and could improve the results of tubularized urethral
urethral submucosa [140] have also been tested repairs, autologous rabbit bladder epithelial cells
in various animal models for urethral reconstruc- and SMCs were grown and seeded onto tubular
tion. Bladder submucosa matrix (BSM) proved to scaffolds. Entire urethra segments were then
be a suitable graft for repair of urethral defects in resected, and urethroplasties were performed
rabbits [5]. The grafts demonstrated a normal with tubularized scaffolds either seeded with
urothelial luminal lining and organized muscle cells or without cells. The tubularized scaffolds
bundles on the outer portion of the graft. These seeded with autologous cells formed neo-tissue
results were confirmed clinically in a series of which was histologically similar to native urethra
patients with a history of failed hypospadias tissue. The tubularized scaffolds without cells
reconstruction. The urethral defects in these lead to poor tissue development, fibrosis, and
patients were repaired with human BSMs [141]. stricture formation. These findings were con-
An engineered urethral tissue was created by firmed clinically when a trial using tubularized
anastomosing the matrix to the urethral plate in non-seeded scaffolds for urethral stricture repair
an onlay fashion. The size of the engineered ure- was performed in eight evaluable patients. Two
thra tissue ranged from 5 to 15 cm. At the 3 years patients with short inflammatory strictures main-
follow-up, three of the four patients had a suc- tained urethral patency. Stricture recurrence
cessful outcome in regard to cosmetic appear- developed in the other six patients within
ance and function. One patient, who had a 15 cm 3 months of surgery [145].
neo-urethra tissue formed, developed a subglanu- In 2011, Raya-Rivera et al. were able to show
lar fistula. Similar results were obtained in pedi- that synthetic polymers can also be used in ure-
atric and adult patients with primary urethral thral reconstruction when they are tubularized
stricture disease using the same BSMs [142]. and seeded with autologous cells [146]. This
Another study in 30 patients with recurrent group used PGA-PLGA scaffolds seeded with
stricture disease showed that a healthy urethral bed autologous cells derived from bladder biopsies
(two or fewer prior urethral surgeries) was needed taken from each patient. These cell-seeded scaf-
for successful urethral reconstruction using the folds were then used to repair urethral defects in
BSM grafts [143]. More than 200 pediatric and five boys. Upon follow-up evaluation, it was
adult patients with urethral disease have been suc- found that most of the boys had excellent urinary
cessfully treated in an onlay manner with BSMs. flow rates postoperatively, and voiding cystoure-
One advantage of this acellular scaffold over the thrograms indicated that these patients main-
traditional nongenital tissue grafts that have been tained wide urethral calibers. Urethral biopsies
used for urethroplasty is that the acellular scaffold revealed that the grafts had developed a normal
is “off the shelf.” Eliminating the necessity of appearing architecture consisting of urothelial
additional surgical procedures for graft harvesting and muscular tissues (Fig. 2.6).
may decrease operative time as well as the poten-
tial morbidity due to the harvest procedure.
These techniques, which employed acellular 2.6.5 Bladder
tissue scaffolds that had not been reseeded with
cells, were applied experimentally and clinically The challenges in the bladder augmentation are
in a successful manner for onlay urethral repairs. often associated with renal function and quality of
34 S.J. Lee et al.

a b c d

e f g

h i j k l

Fig. 2.6 Neo-urethra implantation and clinical out- (arrows show the abnormal margins), (f) 12 months after
comes; (a) a cell-seeded graft sutured to the normal ure- surgery (arrows show margins of tissue-engineered ure-
thral margins. Scanning electron microscopy of seeded thras), and (g) at last follow-up. Histological and immu-
scaffolds 6 days after seeding; (b) urothelial cells, (c) nohistochemical analyses; (h) H&E, (i) α-actin, (j)
smooth muscle cells, and (d) scaffold without cells. desmin, (k) mvosln, and (l) AE1/AE3. Reproduced with
Voiding cystourethrograms of a patient (e) before surgery permission from Raya-Rivera et al. [146]

life [147]. Gastrointestinal segments are most com- Even though the clinical translation of tissue
monly used for this purpose in the clinic, but they engineering and regenerative medicine is slowly
are designed to absorb specific solutes, whereas progressing, the success of tissue engineering for
bladder tissue is designed for the excretion of sol- some applications was already demonstrated
utes. Numerous studies have attempted alternative clinically. A clinical experience involving engi-
biomaterials and tissue-engineered constructs for neered bladder tissue for cystoplasty reconstruc-
bladder replacement or repair due to the problems tion was conducted starting in 1999. A small
encountered with the use of gastrointestinal seg- pilot study of seven patients was reported, using
ments. The ability to use donor tissue efficiently a collagen scaffold seeded with cells either with
and to provide the right conditions for long-term or without omentum coverage, or a combined
survival, differentiation, and growth determines the PGA/collagen scaffold seeded with cells and
success of cell transplantation strategies for bladder omental coverage. Patient’s own bladder cells
reconstruction. These principles were applied in were isolated and expanded in vitro prior to seed-
the creation of tissue-­engineered bladders in an ing on a scaffolding system of collagen and
animal model that required a subtotal cystectomy PGA. The scaffold was designed to replace the
with subsequent replacement with a tissue-engi- bladder size to improve its compliancy and patient
neered organ in beagle dogs [148]. Bladder urothe- continence (Fig. 2.7a–c). The tissue-­engineered
lial cells and SMCs were separately expanded from constructs were implanted for a period up to
an autologous bladder biopsy and seeded onto a 61 months. Our outcomes show the definitive
bladder-­shaped biodegradable polymeric scaffold. improvement of bladder compliance and capacity
The results from this study revealed the possibility while restoring physiological function (Fig. 2.7d,
of engineering bladder constructs that are anatomi- e). In addition, the engineered bladder biopsies
cally and functionally normal. showed an adequate structural architecture and
2  Biomaterials and Tissue Engineering 35

a b c

d
Pves

30 cmH20/Div

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

e
Pves

30 cmH20/Div

5 min/Div
t1 t2 t4 t5
t3 t5

f g h

i j k

Fig. 2.7 (a) Scaffold seeded with cells, (b) engineered ture, consisting of lumen lined with urothelial cells, U,
bladder anastomosed to native bladder with running 4–0 surrounded by submucosa, S, and muscle, M. (f) H&E,
PGA sutures, and (c) implant covered with fibrin glue and immunocytochemical analysis with (g) anti-­
omentum. (d) Preoperative and (e) 10 months postopera- pancytokeratin AE1/AE3 antibodies, and (h) anti-α
tive cystograms and urodynamic findings in patient with a smooth muscle actin antibodies showed presence of phe-
collagen-PGA scaffold engineered bladder. (f–h) cysto- notypically normal urothelium and smooth muscle. (i–k)
scopic biopsies of implanted engineered bladders native bladder tissue. Original magnification: ×100.
31 months after augmentation shows extent of regenera- Reproduced with permission from Atala et al. [147]
tion. Engineered bladder tissue showed tri-layered struc-
36 S.J. Lee et al.

phenotype (Fig. 2.7f–k). Although the experi- chemistry, and Western blotting. Electrical field
ence is promising in terms of showing that engi- stimulation studies in the tissue-engineered con-
neered tissues can be implanted safely, it is just a structs showed similar functional properties to
start in terms of accomplishing the goal of engi- those of normal vaginal tissue. When these con-
neering fully functional bladders. Further experi- structs were used for autologous total vaginal
mental and clinical work is being conducted. replacement, patent vaginal structures were noted
in the tissue-engineered specimens, while the
non-cell-seeded structures were noted to be ste-
2.6.6 M
 ale and Female notic. In 2014, an engineered vaginal tissue
Reproductive Organs derived from the patient’s own cells and implanted
showed normal structural and functional vari-
Reconstructive surgery is required for a wide ables with a follow-up of up to 8 years in the
variety of pathologic penile conditions such as clinic [152].
penile carcinoma, trauma, severe erectile dys- Congenital malformations of the uterus may
function, and congenital conditions such as have profound clinical implications. Patients
ambiguous genitalia, hypospadias, and epispa- with cloacal exstrophy and intersex disorders
dias. One of the major limitations of phallic may not have sufficient uterine tissue present for
reconstructive surgery is the scarcity of sufficient future reproduction. Campbell et al. performed
autologous tissue. The major components of the another type of study in which the peritoneal cav-
phallus are corporal SMCs and ECs. The creation ity of rats and rabbits were used as in vivo biore-
of autologous functional and structural corporal actors to produce uterine tissue grafts [153]. In
tissue de novo would be beneficial. Autologous this study, biomaterial templates of the appropri-
cavernosal SMCs and ECs were harvested, ate shape were implanted in the peritoneal cavi-
expanded, and seeded on decellularized tissue ties of rats or rabbits. After 2–3 weeks, the
matrices and implanted in a rabbit model [149, templates were removed, and the encapsulating
150]. The engineered corpora demonstrated myofibroblast-rich tissue that resulted from the
structural and functional parameters similar to foreign-body response to the biomaterial was
native tissue and male rabbits receiving the bilat- harvested. This tissue was then used to replace
eral implants were able to successfully impreg- resected segments of the uterus in the same ani-
nate females. This study demonstrated that mals in which the tissue was grown. At 12 weeks
neo-corpora could be engineered for total pendu- after grafting, it was reported that this novel uter-
lar penile corporal body replacement [150]. Such ine graft tissue thickened and developed the mor-
technology has considerable potential for patients phology of normal uterus. This structure included
requiring penile reconstruction. a lumen lined with endometrium, which was sur-
Several pathologic conditions, including con- rounded by several layers of SMCs (myometrium-­
genital malformations and malignancy of the like) interspersed with collagen. Importantly,
vagina, can adversely affect normal development these grafted uterine horns supported embryos to
or anatomy. Vaginal reconstruction has tradition- the late stages of gestation. The dysfunctional
ally been challenging due to the paucity of avail- uterine cervix may also benefit from tissue engi-
able native tissue. The feasibility of engineering neering strategies. Spontaneous preterm birth is a
vaginal tissue in vivo was investigated [151]. frequent complication of pregnancy, and in some
Vaginal epithelial cells and SMCs of female rab- cases, abnormalities of the cervix have been
bits were harvested, grown, and expanded in cul- implicated in this issue.
ture. These cells were seeded onto biodegradable House et al. have shown that tissue engineer-
polymer scaffolds, and the cell-seeded constructs ing techniques can be used to create 3D cervical-­
were then implanted into nude mice for up to like tissue constructs [154]. Cervical cells were
6 weeks. The presence of vaginal tissue pheno- isolated from two premenopausal women and
types was confirmed by histology, immunohisto- seeded on porous silk scaffolds. These constructs
2  Biomaterials and Tissue Engineering 37

were cultured under dynamic or static culture analysis of the collected urine-like fluid, including
conditions. After an 8 weeks culture interval, cer- urea nitrogen and creatinine levels, electrolyte lev-
vical cells had proliferated in the 3D construct els, specific gravity, and glucose concentration,
and had synthesized an ECM with biochemical revealed that the implanted renal cells possessed
constituents and morphology similar to native tis- filtration, reabsorption, and secretory capabilities.
sue, but this matrix was better formed under Histological examination of the retrieved implants
dynamic culture conditions. This study suggests revealed extensive vascularization and self-organi-
that it may be possible to engineer cervical tissue zation of the cells into glomeruli and tubule-like
for a variety of conditions. structures. A clear continuity between the glomer-
uli, the tubules, and the polycarbonate membrane
was noted that allowed the passage of urine into the
2.6.7 Kidney collecting reservoir. Immunohistochemical analysis
with renal-­specific antibodies revealed the presence
The kidney is a complex organ with multiple cell of renal proteins. RT-PCR analysis confirmed the
types and a complex functional anatomy that ren- transcription of renal-specific RNA in the cloned
ders it one of the most difficult to reconstruct specimens, and Western blot analysis confirmed the
[155]. Previous efforts in tissue engineering of presence of elevated renal-specific protein levels.
renal tissues have been directed toward the devel- Studies have shown that bovine clones harbor
opment of extracorporeal renal support systems the oocyte mitochondrial DNA [162]; therefore,
made of biological and synthetic components the mitochondrial DNA (mtDNA) of the donor egg
[156–160], and ex vivo renal replacement devices was thought to be a potential source of immuno-
are known to be life-sustaining. Obviously, logic incompatibility. Differences in mtDNA-­
patients with end-stage kidney disease (ESRD) encoded proteins expressed by cloned cells could
would benefit if these devices could be implanted stimulate a T-cell response specific for mtDNA-­
long term without the need for an extracorporeal encoded minor histocompatibility antigens when
perfusion circuit or immunosuppressive drugs. the cloned cells are implanted back into the original
We have previously applied the principles of nuclear donor [163]. We used nucleotide sequenc-
both tissue engineering and therapeutic cloning in ing of the mtDNA genomes of the clone and fibro-
an effort to produce genetically identical renal tis- blast nuclear donor to identify potential antigens in
sue in a large animal model, the cow (Bos taurus) the muscle constructs. Only two amino acid substi-
[161]. Bovine skin fibroblasts from adult Holstein tutions were noted to distinguish the clone and the
steers were obtained by ear notch, and single donor nuclear donor, and, as a result, a maximum of two
cells were isolated and microinjected into the peri- minor histocompatibility antigens could be defined.
vitelline space of donor-­enucleated oocytes (nuclear Given the lack of knowledge regarding peptide-
transfer). The resulting blastocysts were implanted binding motifs for bovine MHC class I molecules,
into progestin-­synchronized recipients to allow for reliable methods to predict the impact of these
further in vivo growth. After 12 weeks, cloned renal amino acid substitutions on bovine histocompati-
cells were harvested, expanded in vitro, and then bility are unavailable. Oocyte-derived mtDNA was
seeded on scaffolds consisting of three collagen-­ also thought to be a potential source of immuno-
coated cylindrical polycarbonate membranes. The logic incompatibility in the cloned renal cells.
ends of the three membranes of each scaffold were Maternally transmitted minor histocompatibility
connected to catheters that terminated into a collect- antigens in mice have been shown to stimulate both
ing reservoir. This created a renal neo-organ with a skin allograft rejection in vivo and cytotoxic T lym-
mechanism for collecting the excreted urinary fluid. phocyte expansion in vitro [163]. These antigens
The scaffolds with the collecting devices were could prevent the use of the cloned constructs in
transplanted subcutaneously into the same steer patients with chronic rejection of major
from which the genetic material originated and then histocompatibility-­ matched human renal trans-
retrieved at 12 weeks after implantation. Chemical plants [164]. We tested for a possible T-cell
38 S.J. Lee et al.

response to the cloned renal devices using delayed- a decellularized porcine kidney scaffold [49, 69,
type hypersensitivity testing in vivo and Elispot 165, 166]. Given the complexity of the kidney
analysis of interferon-gamma secreting T cells structure, which includes over 30 different cell
in vitro. Both analyses revealed that the cloned types, vascular network as well as a large array of
renal cells lacked evidence of a T-cell response and functional structures, the use of the underlying
suggested that rejection will not necessarily occur ECM composing the unique microenvironment is
in the presence of oocyte-­derived mtDNA. This a logical starting point for the bioengineering of a
finding may represent a step forward in overcom- transplantable whole kidney. The efficacy of cel-
ing the histocompatibility problem of stem cell lular removal and biocompatibility of the pre-
therapy [155]. These studies demonstrated that served porcine matrices, coupled with scaffold
cells derived from nuclear transfer can be success- reproducibility, are critical to the success of this
fully harvested, expanded in culture, and trans- approach. To perform the decellularization, we
planted in vivo with the use of biodegradable have designed and constructed high-throughput
scaffolds on which the single suspended cells can system for decellularization process. This system
organize into tissue structures that are genetically showed significant cellular removal (<50 ng
identical to that of the host. This was the first dem- DNA/mg dry tissue). And, decellularized organs
onstration of the use of therapeutic cloning for retained intact microarchitecture including the
regeneration of tissues in vivo. renal vasculature and essential ECM components
One such approach for whole organ bioengi- (Fig. 2.8). Our method ensures clearance of por-
neering is to combine functional renal cells with cine cellular material, which directly impacts

a 0 hours 6 hours 12 hours 24 hours 36 hours

b c

RV RA
RV

RA

Fig. 2.8 Decellularization of porcine kidney. (a) renal system by continuation of the renal artery (RA) to
Representative time lapse of decellularization process of the renal vein (RV). Histological analyses of (d–f) native
porcine kidney. Comparison of whole organ vasculature and (g–i) decellularized porcine kidney; (d, g) H&E, (e,
system between (b) native and (c) decellularized porcine h) Alcian Blue/Sirius Red, and (f, i) Masson’s trichrome.
kidney illustrating the preservation of the arterial–venous Reproduced with permission from Sullivan et al. [49]
2  Biomaterials and Tissue Engineering 39

d e f

g h i

Fig. 2.8 (continued)

immunoreactivity during transplantation, and frequency of thrombosis, stenosis, occlusion, and


preserves the ECM and cellular compatibility of infection [171, 172].
these renal scaffolds. Thus, we have developed a Tissue engineering offers an attractive
rapid decellularization method that can be scaled approach to vascular grafting, particularly for
up for use in other large organs. Our results rep- small diameter (<5 mm) vessels. The basic
resent a step toward development of a transplant- approach is to create vessels by combining autol-
able organ using tissue engineering techniques. A ogous cells with a natural and/or synthetic scaf-
preclinical large animal study for the application fold under suitable culture conditions, resulting
of this technology is currently being conducted. in a tubular construct [116] that can be implanted
in vivo. Coating the vascular lumen of synthetic
grafts with ECs has been shown to overcome
2.6.8 Blood Vessels acute thrombosis [169, 170]. The presence of a
permanent synthetic graft remains an issue, as it
Cardiovascular disease, including coronary artery may lead to chronic inflammatory responses and
and peripheral vascular diseases, is the leading other problems. A preferred solution would be
cause of mortality in the USA [167]. There the use of biodegradable scaffolds that, over time,
remains a major clinical demand to improve tech- can be replaced by cells to generate an essentially
nology for vascular grafts. Segments of autolo- normal bioengineered blood vessel. Indeed, vas-
gous vessels continue to be the standard for many cular scaffolds combined with viable cells have
revascularization procedures. Unfortunately, been shown to allow grafts to remodel when
autografts are limited in supply and dimensions, introduced in vivo.
while allografts and xenografts are limited by Previous work by our group demonstrated that
strong immunogenic response [168–170]. a decellularized porcine vessel scaffold coated
Synthetic vascular grafts have been explored for with autologous ECs could replace a small diam-
over half a century. Substances such as expanded eter blood vessel in a sheep carotid arterial inter-
polytetrafluoroethylene (ePTFE) or Dacron position model [173, 174]. The implanted
(polyethylene terephthalate fiber) work well as decellularized vascular grafts could remain pat-
large diameter (>5 mm) bypass conduits, but ent for at least 130 days. While the use of cells
such synthetics have not proven generally satis- bound to a naturally derived vascular tissue
factory for small diameter grafts, due to the high matrix demonstrates the principle of vascular
40 S.J. Lee et al.

t­issue engineering, this approach has several collagen scaffolds showed tensile properties sim-
drawbacks that would be expected to hinder clin- ilar to those of native blood vessels due to the
ical translation. These include the limited supply stiffness of collagen. Most recently, we have
of vessels of the required dimensions, inconsis- developed a bilayered scaffolding system that
tency in donor tissue composition, and potential provides different pore sizes to facilitate adequate
contamination by pathogens. Therefore, it is cellular interactions. Thus, our system allows for
essential to develop a synthetic vascular scaffold- EC adhesion onto the luminal surface and homog-
ing system that would allow for the consistent enous infiltration of SMCs into the outer layer
production of small diameter vessel substitutes of (Fig. 2.4). Moreover, this scaffold provides suffi-
any desired dimensions. cient mechanical properties that withstand physi-
A number of approaches have been attempted ologically relevant vascular conditions. Our
with the goal of producing an ideal vascular scaf- findings suggest that bilayered scaffolds may
fold. Among these, electrospinning technology, facilitate endothelialization and smooth muscle
which uses high-voltage electrostatic fields to maturation for improved vessel tissue function. A
generate nanofibers, appears particularly attrac- preclinical large animal study for the application
tive because it provides a biomimetic environ- of this fully cellularized vascular scaffold is cur-
ment that may be designed to resemble that of rently being conducted. These studies will evalu-
ECM architecture of native vasculature. ate the utility of vascular tissue engineering to
Electrospinning permits fabrication of fibrous provide platform technologies for rehabilitation
matrices of nano- to microscale. In addition, it is of patients recovering from severe and devastat-
possible to control the composition, structure, ing cardiovascular diseases. The long-term goals
dimension, and biomechanical properties of fab- are to provide alternatives to vascular grafting
ricated scaffolds. Numerous reports have docu- using tissue-engineered blood vessels derived
mented that synthetic biodegradable from the patient’s own cells. We believe that
polymer-based materials can be electrospun to completion of this study has the potential to lead
generate candidate vascular scaffolds with directly to clinical translation and address an
in vitro characteristics of strength and biocom- important unmet medical need.
patibility that appear consistent with clinical
requirements [77, 116, 175–177]. Many efforts
attempted to develop a durable biomaterial capa- 2.6.9 Skeletal Muscle
ble of resisting physiological forces and main- and Innervation
taining structural integrity until mature vascular
tissue forms in vivo. It is also important that engi- Skeletal muscle defects due to trauma or tumor
neered vessels should be compliant, resistant to ablation usually require reconstructive proce-
kinking and compression, and possess sufficient dures in order to restore normal tissue function.
tensile and shear strength to resist fraying at cut Currently, muscle pedicle flap from adjacent
edges and tearing out of sutures [178]. regions is the primary method practiced. This
With these considerations in mind, we recently option is challenged by host muscle tissue avail-
developed a vascular scaffold that is a composite ability and donor site morbidity such as func-
of poly(ε-caprolactone) (PCL) and type I colla- tional loss and volume deficiency [180, 181].
gen [77, 116]. We observed that this scaffold can Recent advances in cell therapy using myoblasts
withstand physiologically relevant vascular con- have provided an alternate therapeutic opportu-
ditions over a period of 1 month in vitro and after nity to regenerate muscle tissue for functional
implantation in vivo in a rabbit aortoiliac bypass augmentation [182, 183]. Injection of cultured
model [179]. The introduction of collagen to myoblasts has shown some efficacy [184–186];
PCL appeared to increase the structural stability however, this approach may not be suitable for
under high physiological pressure as well as cell treating large volumetric muscle loss injury
compatibility. Interestingly, the hydrated PCL/ [185]. Hence, creation of an implantable func-
2  Biomaterials and Tissue Engineering 41

tional muscle tissue that could restore muscle tis- tural characteristics and induce vascularization
sue defects may be a possible solution [187, 188]. and nerve integration in vivo [122].
An essential step in bioengineering functional Another critical component that is required to
skeletal muscle tissue construct is to mimic the successfully engineer a functional muscle tissue
structure of native tissue which is comprised of in vivo is effective innervation of the skeletal mus-
highly oriented myofibers formed from numer- cle [195]. The established contacts of tissue-­
ous fused mononucleated muscle cells [189]. It is engineered muscle constructs with host nervous
well known that structure and organization of tissue is vitally important following implantation,
myofibers dictate tissue function. Thus, muscle as improper or incomplete innervation leads to
cell alignment that permits organized myotube atrophy of muscle tissue and loss of contractile
formation is a crucial step in the musculoskeletal function [196]. When muscle cells/fibers are
myogenesis [190]. The ability to efficiently orga- implanted in the body, host nerves contact with the
nize muscle cells to form aligned myotubes muscle fibers to form neuromuscular junctions
in vitro would greatly benefit efforts in skeletal (NMJs). Denervated muscle, which is analogous
muscle tissue engineering. To date, significant to in vitro bioengineered muscle constructs, has
deficits in reconstructing complex structure with been shown to be reinnervated upon direct trans-
spatial organization have been problematic and plantation of host nerve [197]. The process of
have limited construction to primarily hollow and innervations into denervated muscle is slow, how-
simple tissue and organ structures. 3D printing ever, and substantial time is required before mus-
technology has been shown to be a viable option cle tissue is functional. Therefore, methods to
for creating a tissue-engineered construct accelerate innervations are needed. Previous
designed to regenerate or replace a damaged tis- reports indicate that the expression of acetylcho-
sue/organ [191, 192]. This 3D printing method line receptors (AChRs) and their clustering on
can precisely place cells with hydrogel-based muscle fibers are critical factors required to induce
materials in a layer-by-layer fashion, allowing it neural contacts on newly formed muscle fibers in a
to replicate the 3D complex structure of tissues or natural biological system [198–200]. For instance,
organs. Our group has used this technology to in NMJ development in the mouse, AChRs are
fabricate various tissue-like structures such as pre-patterned at sites that are used by the outgrow-
cartilage [193], bone [194], and vascularized tis- ing motor neurons to form synaptic contacts [201].
sue [120]. We have previously demonstrated our On each muscle fiber, one synaptic site (often con-
ability to print a skeletal muscle bundle using the tacted by several different nerves) becomes stabi-
3D bioprinting technology [122]. A 3D muscle lized, and the mature endplate pattern is established
construct was fabricated using muscle cells with at a later embryonic stage. During postnatal devel-
hydrogel material. After 3D bioprinting, the opment, further structural and functional changes
printed muscle structures contained muscle fiber-­ are observed. Multiple innervations of muscle
like bundles. Notably, the printed cells started fibers are reduced until motor neurons form single
stretching the shape along the longitudinal axis synaptic contacts, and embryonic AChRs are
of the printed fiber structure. Our result demon- replaced by aggregates of adult AChRs that
strates that the 3D bioprinting can mimic the tis- acquire a characteristically “pretzel-like” pattern.
sue organization in the human body for During the process of maturation of NMJs between
reconstruction; moreover, this may result in muscle fibers and motor neurons, the formation of
improved function and maturation of engineered embryonic AChR clusters on muscle fibers clearly
skeletal muscle tissue constructs. Findings from appears to be a prerequisite step that controls the
our study show that these tissue constructs were overall process leading to mature innervations of
able to maintain structural and functional charac- muscle fibers [199]. Although this well-organized
teristics in vitro. In a pilot animal study, we have process of pre-forming AChR clusters on embry-
showed that the implanted 3D bioprinted muscle onic muscle fibers is present in normal vertebrate
construct was robust enough to maintain its struc- systems, tissue-engineered muscle constructs are
42 S.J. Lee et al.

unable to provide such a sophisticated process and muscle in vivo [208]. Thus, we tested whether
remain a major concern for the use of bioengi- agrin molecules would facilitate the formation
neered muscle tissue in vivo. of AChR clusters on the bioengineered muscle
To address this issue, we investigated fibers in fibrin hydrogel constructs [209]. Our
whether prefabrication of AChR clusters on results indicate that agrin treatment increased
bioengineered, bulk muscle fibers using agrin, AChR cluster formation on the differentiated
a neural-­released trophic factor, would acceler- muscle cells, and these prefabricated AChR
ate innervation when implanted in vivo. Agrin clusters facilitated accelerated contacts with
has a critical role in inducing AChR expression dorsal root ganglion (DRG) nerve in vitro or
and their clustering on muscle cells [202–205]. with the host nerve in bioengineered muscle
Neural agrin stimulates the rapid phosphoryla- fibers treated with agrin in vivo (Fig. 2.9)
tion of a muscle-specific kinase (MuSK), [209]. Moreover, the implanted agrin-treated
which has been shown to be necessary for the muscle constructs displayed a mature myofiber
formation of the NMJ and leads to the redistri- structure with myosin heavy chain (MHC)
bution of previously unlocalized AChR pro- expression in the vicinity of the host nerve,
teins, to synaptic sites [206]. The function of indicating enhanced interaction with the host
agrin was demonstrated in an agrin-deficient nerve. It is our belief that the effective use of a
mutant mouse model, which failed to form NMJ-­ inducing factor, agrin, could induce
functional endplates [207]. This deficit was proper maturation of bioengineered muscle
overcome by gene transfer experiments and/or constructs for accelerating further host nerve
injection of recombinant agrin into rat soleus integration (innervation).

1200

a c e g 1100

1000
Count ratio (340 nm / 380 nm)

Agrin (+)
900
Agrin (-)

800

700
ACh addition
600
CPN Agrin (-)
500

100 µm 1mm 50 µm
Host tissue 400
0 10000 20000 30000

Time (ms)

b d f h
7

6
* Agrin (-)

5 Agrin (+)
Agrin (+)

# of NMJ (NF+ BTX+)


/field of View (X400)

*
3
*
2

CPN 1

100 µm 1mm 50 µm
0
Host tissue 2 wks 4 wks 8 wks
Implantation time

Fig. 2.9  Prefabrication of AChR by agrin treatment implant without agrin treatment (c). (e, f)
accelerates innervations in vitro and in vivo. (a, b) In vitro Immunofluorescent images of the harvested C2C12/fibrin
myotube formation (a) without and (b) with agrin treat- gels; higher numbers of innervated structures [neurofila-
ment; agrin treatment induced enhanced AChR expres- ment (NF)+/α-BTX+ double staining (arrowheads)] were
sion on myotubes [α-BTX+ indicated by arrows on the observed in agrin-treated group (f) than in no treated
surface of myotubes (box in b)] while there are no charac- group (e) and quantification of their number (h). Student
teristic evidence in untreated myotubes (a). (c, d) t-test, *P < 0.05, n = 3, from three individual animals. (g)
Prefabricated C2C12 myotubes in fibrin gel were In vitro functional properties of AChR expression on
implanted subcutaneously embedded with common pero- myotubes by calcium imaging; agrin-treated myotubes
neal nerve (CPN); gross appearance of the harvested displayed depolarization-induced increases in steady-­
C2C12/fibrin gel with CPN at 2 weeks after implantation; state intracellular calcium levels in the kinetic curve with
numerous large blood vessels were observed in the the sharp increase (arrow head) in agrin-treated myo-
implant treated with agrin (d), whereas there was no tubes. Reproduced with permission from Ko et al. [209]
remarkable blood vessels recruitment surrounding the
2  Biomaterials and Tissue Engineering 43

2.7 Vascularization enhanced local angiogenesis in vivo for up to


of Engineered Tissues 21 days. These studies demonstrated the delivery
of VEGF in a controlled and localized fashion
A major challenge in tissue engineering is the in vivo. This angiogenic factor delivery system
lack of proper vascularization in the engineered was applied to bone regeneration, and its potent
tissue constructs when implanted. To develop ability to enhance angiogenesis within implanted
complex tissues or organs, fully vascularized tis- scaffolds was followed by enhanced bone regen-
sue constructs should be provided because it is eration and outlines a novel approach for engineer-
critical to long-term cell survival and function of ing tissues in hypovascular environments [215].
cell constructs not only at the margin but also at As another example, a dual delivery system
the center of the tissue grafts [210]. In fact, the combining VEGF and insulin-like growth factor
growth of a new microvascular system persists as 1 (IGF-1) was developed to enhance transplanta-
one of the major limitations to the successful tion and dispersion of cultured myogenic cells
introduction of tissue engineering products to [216]. Localized delivery of VEGF and IGF-1
clinical practice [211]. Accordingly, the focus of from alginate scaffolds into injured muscle
much research in tissue engineering has evolved enhanced local angiogenesis, altered muscle fiber
to include a better understanding of angiogenesis. type, and enhanced muscle regeneration in a
Numerous efforts have been made to overcome model of severe muscle damage. Transplanting
this limitation, and attempts to enhance angio- cultured myoblasts on scaffolds that delivered
genesis within the host tissue have been pursued VEGF/IGF-1 dramatically enhanced their direct
using several approaches. participation in muscle regeneration and further
enhanced angiogenesis and the return to normal
tissue perfusion levels, as compared to growth
2.7.1 Angiogenic Factor Delivery factor delivery alone. This approach holds prom-
ise in the transplantation of many cell types used
It has been demonstrated that the delivery of to promote the regenerative response of multiple
growth factors and cytokines that play central tissues.
regulatory roles in the process of angiogenesis,
which is thought to induce ingrowth of capillaries
and blood vessels into an engineered implant, 2.7.2 Incorporation
yields diminishing hypoxia-related cell damage. of Endothelial Cells
The delivery of such angiogenic factors has been
achieved either by incorporating the desired fac- Human vascular ECs and skeletal myoblasts
tors into the scaffold biomaterials to be used or transfected with adenovirus encoding the gene
by genetic modification of the cells to be used in for VEGF were tested for regenerating a vascu-
the engineering process, which forces the cells to larized engineering muscle construct [212]. The
express factors such as vascular endothelial transfected cells were injected subcutaneously in
growth factor (VEGF), one of the most potent athymic mice and noted to form a muscle tissue
angiogenic factors [212, 213]. with neovascularization by histological and
A study evaluated controlled release of VEGF immunohistochemical analyses with mainte-
by incorporating VEGF directly into PLGA scaf- nance of their muscle volume, while engineered
folds or by incorporating VEGF encapsulated in muscle of non-transfected cells had a signifi-
PLGA microspheres into scaffolds [214]. VEGF cantly smaller mass of cells with loss of muscle
incorporated into scaffolds resulted in rapid release volume over time, less neovascularization, and
of the cytokine, whereas the ­ pre-­
encapsulated no surviving ECs. Results indicate that a combi-
group showed a delayed release. In addition, both nation of VEGF and ECs may be useful for
systems showed negligible release of VEGF into inducing neovascularization and volume preser-
the systemic circulation, yet their use led to vation in engineered tissue.
44 S.J. Lee et al.

The development of a method of formation and many of the same tools developed for evaluating
stabilization of endothelialized vessel networks the biocompatibility of traditional biodegradable
in vitro in engineered skeletal muscle tissue [217] polymers are still used to investigate the funda-
has shown promising results. A 3D multicultural mental interactions between new classes of bioma-
system consisting of myoblasts, embryonic fibro- terials and their host [221, 222].
blasts, and ECs co-seeded on highly porous, bio- It is evident from the previous examples that
degradable polymeric scaffolds. These results the diversity in biomaterials for tissue engineer-
showed that pre-­vascularization of the implants ing applications is immense. Numerous
improved angiogenesis and cell survival within the approaches to mimicking the structure and
scaffolds. Moreover, this research group empha- mechanical properties, and more importantly, the
sized that cocultures with ECs and SMCs may also function of the ECM have been devised. It is
be important for inducing differentiation of engi- imperative that these important technologies con-
neered tissues. A synthetic, biomimetic hydrogel tinue to be investigated for their ability to interact
was developed to allow the rapid formation of a in biological systems. An essential tool set has
stable and mature vascular network [218]. previously been described that will enable com-
Hydrogels were fabricated with integrin-binding prehensive evaluation of novel biomaterials in
sites and protease-sensitive substrates to mimic the their host environment. Successful regulatory
natural provisional ECM, and ECs cultured in approval of new categories of regenerative tech-
these hydrogels were organized into stable, intri- nologies entering human clinical trials will con-
cate networks of capillary-like structures. The tinue to be based on these fundamental principles
resulting structures were further stabilized by of biocompatibility and biological interaction.
recruitment of mesenchymal progenitor cells that Recent progress suggests that tissue-engineered
differentiated into a SMC lineage and deposited constructs may have an expanded clinical appli-
collagen IV and laminin. cability in the future and may represent a viable
therapeutic option for those who require tissue
replacement or repair.
2.8  onclusions and Future
C
Directions
References
Various tissue-engineered constructs are at differ-
ent stages of development, with some in current 1. Atala A. Engineering organs. Curr Opin Biotechnol.
clinical use. The tissue-engineered scaffold pro- 2009;20(5):575–92.
vides mechanical support, shape, and cell-­scale 2. Atala A. Regenerative medicine and tissue engineer-
ing in urology. Urol Clin North Am. 2009;36(2):
architecture for neo-tissue construction in vitro or 199–209. viii-ix
in vivo as seeded cells expand and organize. Most 3. Atala A. Tissue engineering of reproductive tissues
degradable biomaterials used to date comprise a and organs. Fertil Steril. 2012;98(1):21–9.
class of synthetic polyesters and/or natural poly- 4. Atala A, Kasper FK, Mikos AG. Engineering complex
tissues. Sci Transl Med. 2012;4(160):160rv12.
mers. A multitude of fabrication techniques have 5. Chen F, Yoo JJ, Atala A. Acellular collagen matrix
been devised and afford an abundance of potential as a possible “off the shelf” biomaterial for urethral
shapes, sizes, porosities, and architectures [219, repair. Urology. 1999;54(3):407–10.
220]. In addition, scaffolds should provide more 6. Chun SY, Lim GJ, Kwon TG, Kwak EK, Kim BW,
Atala A, et al. Identification and characterization
than temporary architectural structure to a devel- of bioactive factors in bladder submucosa matrix.
oping tissue construct. As cells and molecular Biomaterials. 2007;28(29):4251–6.
biology meet with materials science and biomedi- 7. Yoo JJ, Meng J, Oberpenning F, Atala A. Bladder aug-
cal engineering, new applications in regenerative mentation using allogenic bladder submucosa seeded
with cells. Urology. 1998;51(2):221–5.
medicine will benefit from interactive biomaterials 8. Eberli D, Susaeta R, Yoo JJ, Atala A. Tunica repair
that serve to orchestrate cell attachment and with acellular bladder matrix maintains corporal tissue
growth, as well as tissue morphogenesis. However, function. Int J Impot Res. 2007;19(6):602–9.
2  Biomaterials and Tissue Engineering 45

9. Kim BS, Atala A, Yoo JJ. A collagen matrix derived 26. Spicer AP, Tien JY. Hyaluronan and morpho-
from bladder can be used to engineer smooth muscle genesis. Birth Defects Res C Embryo Today.
tissue. World J Urol. 2008;26(4):307–14. 2004;72(1):89–108.
10. Eberli D, Aboushwareb T, Soker S, Yoo JJ, Atala 27. Beck DE, Cohen Z, Fleshman JW, Kaufman HS, van
A. Muscle precursor cells for the restoration of Goor H, Wolff BG. A prospective, randomized, mul-
irreversibly damaged sphincter function. Cell ticenter, controlled study of the safety of Seprafilm
Transplant. 2012;21(9):2089–98. adhesion barrier in abdominopelvic surgery of the
11. Joo S, Ko IK, Atala A, Yoo JJ, Lee SJ. Amniotic intestine. Dis Colon Rectum. 2003;46(10):1310–9.
fluid-derived stem cells in regenerative medicine 28. Diamond MP. Reduction of adhesions after uterine
research. Arch Pharm Res. 2012;35(2):271–80. myomectomy by Seprafilm membrane (HAL-F): a
12. Pariente JL, Kim BS, Atala A. In vitro biocompat- blinded, prospective, randomized, multicenter clini-
ibility evaluation of naturally derived and synthetic cal study. Seprafilm adhesion study group. Fertil
biomaterials using normal human bladder smooth Steril. 1996;66(6):904–10.
muscle cells. J Urol. 2002;167(4):1867–71. 29. Kemper F, Gebhardt U, Meng T, Murray
13. Meaney Murray M, Rice K, Wright RJ, Spector C. Tolerability and short-term effectiveness of
M. The effect of selected growth factors on human hylan G-F 20 in 4253 patients with osteoarthritis of
anterior cruciate ligament cell interactions with a the knee in clinical practice. Curr Med Res Opin.
three-dimensional collagen-GAG scaffold. J Orthop 2005;21(8):1261–9.
Res. 2003;21(2):238–44. 30. Waddell DD, Bricker DC. Hylan G-F 20 toler-
14. Patino MG, Neiders ME, Andreana S, Noble B, ability with repeat treatment in a large orthopedic
Cohen RE. Collagen as an implantable mate- practice: a retrospective review. J Surg Orthop Adv.
rial in medicine and dentistry. J Oral Implantol. 2006;15(1):53–9.
2002;28(5):220–5. 31. Waddell DD, Bricker DC. Clinical experience with
15. van Susante JLC, Pieper J, Buma P, van Kuppevelt the effectiveness and tolerability of hylan G-F 20 in
TH, van Beuningen H, van Der Kraan PM, et al. 1047 patients with osteoarthritis of the knee. J Knee
Linkage of chondroitin-sulfate to type I collagen scaf- Surg. 2006;19(1):19–27.
folds stimulates the bioactivity of seeded chondro- 32. Tonnesen HH, Karlsen J. Alginate in drug delivery
cytes in vitro. Biomaterials. 2001;22(17):2359–69. systems. Drug Dev Ind Pharm. 2002;28(6):621–30.
16. Yang C, Hillas PJ, Baez JA, Nokelainen M, Balan 33. Gutowska A, Jeong B, Jasionowski M. Injectable gels
J, Tang J, et al. The application of recombinant for tissue engineering. Anat Rec. 2001;263(4):342–9.
human collagen in tissue engineering. BioDrugs. 34. Boontheekul T, Kong HJ, Mooney DJ. Controlling
2004;18(2):103–19. alginate gel degradation utilizing partial oxida-
17. Friess W. Collagen--biomaterial for drug delivery. tion and bimodal molecular weight distribution.
Eur J Pharm Biopharm. 1998;45(2):113–36. Biomaterials. 2005;26(15):2455–65.
18. Lee CH, Singla A, Lee Y. Biomedical applications of 35. Zimmermann H, Zimmermann D, Reuss R, Feilen
collagen. Int J Pharm. 2001;221(1–2):1–22. PJ, Manz B, Katsen A, et al. Towards a medically
19. Kasoju N, Bora U. Silk fibroin in tissue engineering. approved technology for alginate-based microcap-
Adv Healthc Mater. 2012;1(4):393–412. sules allowing long-term immunoisolated transplan-
20. Talukdar S, Nguyen QT, Chen AC, Sah RL, tation. J Mater Sci Mater Med. 2005;16(6):491–501.
Kundu SC. Effect of initial cell seeding density 36. Khor E, Lim LY. Implantable applications of chitin
on 3D-engineered silk fibroin scaffolds for articu- and chitosan. Biomaterials. 2003;24(13):2339–49.
lar cartilage tissue engineering. Biomaterials. 37. Shi C, Zhu Y, Ran X, Wang M, Su Y, Cheng
2011;32(34):8927–37. T. Therapeutic potential of chitosan and its
21. Altman GH, Diaz F, Jakuba C, Calabro T, Horan RL, derivatives in regenerative medicine. J Surg Res.
Chen J, et al. Silk-based biomaterials. Biomaterials. 2006;133(2):185–92.
2003;24(3):401–16. 38. Miranda SC, Silva GA, Hell RC, Martins MD,
22. Li G, Li Y, Chen G, He J, Han Y, Wang X, et al. Alves JB, Goes AM. Three-dimensional culture
Silk-based biomaterials in biomedical textiles of rat BMMSCs in a porous chitosan-gelatin scaf-
and fiber-based implants. Adv Healthc Mater. fold: a promising association for bone tissue engi-
2015;4(8):1134–51. neering in oral reconstruction. Arch Oral Biol.
23. Yucel T, Lovett ML, Kaplan DL. Silk-based bioma- 2011;56(1):1–15.
terials for sustained drug delivery. J Control Release. 39. Jones PL, Schmidhauser C, Bissell MJ. Regulation
2014;190:381–97. of gene expression and cell function by extra-
24. Wang Y, Rudym DD, Walsh A, Abrahamsen L, Kim cellular matrix. Crit Rev Eukaryot Gene Expr.
HJ, Kim HS, et al. In vivo degradation of three-­ 1993;3(2):137–54.
dimensional silk fibroin scaffolds. Biomaterials. 40. Juliano RL, Haskill S. Signal transduction from the
2008;29(24–25):3415–28. extracellular matrix. J Cell Biol. 1993;120(3):577–85.
25. Coviello T, Matricardi P, Marianecci C, Alhaique 41. Reid L, Morrow B, Jubinsky P, Schwartz E,
F. Polysaccharide hydrogels for modified release Gatmaitan Z. Regulation of growth and differentia-
formulations. J Control Release. 2007;119(1):5–24. tion of epithelial cells by hormones, growth factors,
46 S.J. Lee et al.

and substrates of extracellular matrix. Ann N Y 56. Ansaloni L, Catena F, Coccolini F, Gazzotti F,
Acad Sci. 1981;372:354–70. D'Alessandro L, Pinna AD. Inguinal hernia repair
42. Birkedal-Hansen H. Proteolytic remodeling with porcine small intestine submucosa: 3-year
of extracellular matrix. Curr Opin Cell Biol. follow-up results of a randomized controlled trial
1995;7(5):728–35. of Lichtenstein's repair with polypropylene mesh
43. Lutolf MP, Hubbell JA. Synthetic biomaterials as versus Surgisis inguinal hernia matrix. Am J Surg.
instructive extracellular microenvironments for mor- 2009;198(3):303–12.
phogenesis in tissue engineering. Nat Biotechnol. 57. Karpelowsky JS, Thomas G, Shun A. Definitive
2005;23(1):47–55. abdominal wall closure using a porcine intestinal sub-
44. Hodde J. Naturally occurring scaffolds for soft mucosa biodegradable membrane in pediatric trans-
tissue repair and regeneration. Tissue Eng. plantation. Pediatr Transplant. 2009;13(3):285–9.
2002;8(2):295–308. 58. Wiedemann A, Otto M. Small intestinal submucosa
45. Ott HC, Matthiesen TS, Goh SK, Black LD, Kren for pubourethral sling suspension for the treatment
SM, Netoff TI, et al. Perfusion-decellularized of stress incontinence: first histopathological results
matrix: using nature's platform to engineer a bioarti- in humans. J Urol. 2004;172(1):215–8.
ficial heart. Nat Med. 2008;14(2):213–21. 59. Trost L, Elliott D. Small intestinal submucosa ure-
46. Eberli D, Atala A, Yoo JJ. One and four layer acel- thral wrap at the time of artificial urinary sphincter
lular bladder matrix for fascial tissue reconstruction. placement as a salvage treatment option for patients
J Mater Sci Mater Med. 2010;22(3):741–51. with persistent/recurrent incontinence follow-
47. Baptista PM, Siddiqui MM, Lozier G, Rodriguez ing multiple prior sphincter failures and erosions.
SR, Atala A, Soker S. The use of whole organ decel- Urology. 2012;79(4):933–8.
lularization for the generation of a vascularized liver 60. Caione P, Boldrini R, Salerno A, Nappo SG. Bladder
organoid. Hepatology. 2010;53(2):604–17. augmentation using acellular collagen biomatrix: a
48. Ott HC, Clippinger B, Conrad C, Schuetz C, pilot experience in exstrophic patients. Pediatr Surg
Pomerantseva I, Ikonomou L, et al. Regeneration Int. 2012;28(4):421–8.
and orthotopic transplantation of a bioartificial lung. 61. Chaliha C, Khalid U, Campagna L, Digesu GA, Ajay
Nat Med. 2010;16(8):927–33. B, Khullar V. SIS graft for anterior vaginal wall pro-
49. Sullivan DC, Mirmalek-Sani SH, Deegan DB, lapse repair--a case-controlled study. Int Urogynecol
Baptista PM, Aboushwareb T, Atala A, et al. J Pelvic Floor Dysfunct. 2006;17(5):492–7.
Decellularization methods of porcine kidneys for 62. Zhang J, Hu ZQ, Turner NJ, Teng SF, Cheng WY,
whole organ engineering using a high-throughput Zhou HY, et al. Perfusion-decellularized skeletal
system. Biomaterials. 2012;33(31):7756–64. muscle as a three-dimensional scaffold with a vascu-
50. Sandusky GE, Lantz GC, Badylak SF. Healing com- lar network template. Biomaterials. 2016;89:114–26.
parison of small intestine submucosa and ePTFE 63. Wassenaar JW, Boss GR, Christman
grafts in the canine carotid artery. J Surg Res. KL. Decellularized skeletal muscle as an in vitro
1995;58(4):415–20. model for studying drug-extracellular matrix inter-
51. DeQuach JA, Mezzano V, Miglani A, Lange S, actions. Biomaterials. 2015;64:108–14.
Keller GM, Sheikh F, et al. Simple and high yield- 64. Fishman JM, Lowdell MW, Urbani L, Ansari T,
ing method for preparing tissue specific extracel- Burns AJ, Turmaine M, et al. Immunomodulatory
lular matrix coatings for cell culture. PLoS One. effect of a decellularized skeletal muscle scaffold in
2010;5(9):e13039. a discordant xenotransplantation model. Proc Natl
52. Stern MM, Myers RL, Hammam N, Stern KA, Acad Sci U S A. 2013;110(35):14360–5.
Eberli D, Kritchevsky SB, et al. The influence of 65. Duan Y, Liu Z, O'Neill J, Wan LQ, Freytes DO,
extracellular matrix derived from skeletal muscle Vunjak-Novakovic G. Hybrid gel composed of
tissue on the proliferation and differentiation of native heart matrix and collagen induces cardiac dif-
myogenic progenitor cells ex vivo. Biomaterials. ferentiation of human embryonic stem cells without
2009;30(12):2393–9. supplemental growth factors. J Cardiovasc Transl
53. Badylak S, Liang A, Record R, Tullius R, Hodde Res. 2011;4(5):605–15.
J. Endothelial cell adherence to small intestinal 66. Wassenaar JW, Gaetani R, Garcia JJ, Braden RL,
submucosa: an acellular bioscaffold. Biomaterials. Luo CG, Huang D, et al. Evidence for mechanisms
1999;20(23–24):2257–63. underlying the functional benefits of a myocardial
54. Kim MS, Ahn HH, Shin YN, Cho MH, Khang G, matrix hydrogel for post-MI treatment. J Am Coll
Lee HB. An in vivo study of the host tissue response Cardiol. 2016;67(9):1074–86.
to subcutaneous implantation of PLGA- and/or por- 67. Singelyn JM, Sundaramurthy P, Johnson TD, Schup-­
cine small intestinal submucosa-based scaffolds. Magoffin PJ, Hu DP, Faulk DM, et al. Catheter-­
Biomaterials. 2007;28(34):5137–43. deliverable hydrogel derived from decellularized
55. Helton WS, Fisichella PM, Berger R, Horgan S, Espat ventricular extracellular matrix increases endog-
NJ, Abcarian H. Short-term outcomes with small enous cardiomyocytes and preserves cardiac func-
intestinal submucosa for ventral abdominal hernia. tion post-myocardial infarction. J Am Coll Cardiol.
Arch Surg. 2005;140(6):549–60. discussion 60-2 2012;59(8):751–63.
2  Biomaterials and Tissue Engineering 47

68. Pati F, Jang J, Ha DH, Won Kim S, Rhie JW, Shim 85. Everaerts F, Torrianni M, Hendriks M, Feijen
JH, et al. Printing three-dimensional tissue ana- J. Biomechanical properties of carbodiimide
logues with decellularized extracellular matrix bio- crosslinked collagen: influence of the forma-
ink. Nat Commun. 2014;5:3935. tion of ester crosslinks. J Biomed Mater Res A.
69. Arenas-Herrera JE, Ko IK, Atala A, Yoo 2008;85(2):547–55.
JJ. Decellularization for whole organ bioengineer- 86. Lee SJ, Khang G, Lee YM, Lee HB. The effect of
ing. Biomed Mater. 2013;8(1):014106. surface wettability on induction and growth of neu-
70. Gilbert TW, Sellaro TL, Badylak SF. Decellularization rites from the PC-12 cell on a polymer surface. J
of tissues and organs. Biomaterials. 2006;27(19): Colloid Interface Sci. 2003;259(2):228–35.
3675–83. 87. Lee JH, Khang G, Lee JW, Lee HB. Interaction
71. Uhrich KE, Cannizzaro SM, Langer RS, Shakesheff of different types of cells on polymer surfaces
KM. Polymeric systems for controlled drug release. with wettability gradient. J Colloid Interface Sci.
Chem Rev. 1999;99(11):3181–98. 1998;205(2):323–30.
72. Vert M. Aliphatic polyesters: great degradable poly- 88. Mansur HS, Orefice RL, Vasconcelos WL, Lobato
mers that cannot do everything. Biomacromolecules. ZP, Machado LJ. Biomaterial with chemically engi-
2005;6(2):538–46. neered surface for protein immobilization. J Mater
73. Gunatillake PA, Adhikari R. Biodegradable syn- Sci Mater Med. 2005;16(4):333–40.
thetic polymers for tissue engineering. Eur Cell 89. Chai C, Leong KW. Biomaterials approach to
Mater. 2003;5:1–16. expand and direct differentiation of stem cells. Mol
74. Kim BS, Mooney DJ. Development of biocompat- Ther. 2007;15(3):467–80.
ible synthetic extracellular matrices for tissue engi- 90. Sakiyama-Elbert SE, Panitch A, Hubbell
neering. Trends Biotechnol. 1998;16(5):224–30. JA. Development of growth factor fusion pro-
75. Galaev IY, Mattiasson B. 'Smart' polymers and teins for cell-triggered drug delivery. FASEB J.
what they could do in biotechnology and medicine. 2001;15(7):1300–2.
Trends Biotechnol. 1999;17(8):335–40. 91. Anderson DG, Levenberg S, Langer R. Nanoliter-­
76. Hoffman AS, Stayton PS, Bulmus V, Chen G, scale synthesis of arrayed biomaterials and applica-
Chen J, Cheung C, Founder’s Award, Society for tion to human embryonic stem cells. Nat Biotechnol.
Biomaterials, et al. Sixth world biomaterials con- 2004;22(7):863–6.
gress 2000, Kamuela, HI, may 15–20, 2000. Really 92. Zhao X, Zhang S. Fabrication of molecular mate-
smart bioconjugates of smart polymers and receptor rials using peptide construction motifs. Trends
proteins. J Biomed Mater Res. 2000;52(4):577–86. Biotechnol. 2004;22(9):470–6.
77. Lee SJ, Lim GJ, Lee JW, Atala A, Yoo JJ. In vitro 93. Fairman R, Akerfeldt KS. Peptides as novel smart
evaluation of a poly(lactide-co-glycolide)-col- materials. Curr Opin Struct Biol. 2005;15(4):453–63.
lagen composite scaffold for bone regeneration. 94. Wagner DE, Phillips CL, Ali WM, Nybakken GE,
Biomaterials. 2006;27(18):3466–72. Crawford ED, Schwab AD, et al. Toward the devel-
78. Chaikof EL, Matthew H, Kohn J, Mikos AG, opment of peptide nanofilaments and nanoropes
Prestwich GD, Yip CM. Biomaterials and scaf- as smart materials. Proc Natl Acad Sci U S A.
folds in reparative medicine. Ann N Y Acad Sci. 2005;102(36):12656–61.
2002;961:96–105. 95. Hartgerink JD, Beniash E, Stupp SI. Peptide-­
79. Bergsma JE, Rozema FR, Bos RR, Boering G, de amphiphile nanofibers: a versatile scaffold for the
Bruijn WC, Pennings AJ. In vivo degradation and preparation of self-assembling materials. Proc Natl
biocompatibility study of in vitro pre-degraded as-­ Acad Sci U S A. 2002;99(8):5133–8.
polymerized polyactide particles. Biomaterials. 96. Guler MO, Hsu L, Soukasene S, Harrington DA,
1995;16(4):267–74. Hulvat JF, Stupp SI. Presentation of RGDS epi-
80. Hynes RO. Integrins: versatility, modulation, and topes on self-assembled nanofibers of branched
signaling in cell adhesion. Cell. 1992;69(1):11–25. peptide amphiphiles. Biomacromolecules.
81. Deuel TF. Polypeptide growth factors: roles in nor- 2006;7(6):1855–63.
mal and abnormal cell growth. Annu Rev Cell Biol. 97. Beniash E, Hartgerink JD, Storrie H, Stendahl JC,
1987;3:443–92. Stupp SI. Self-assembling peptide amphiphile nano-
82. Gopferich A, Tessmar J. Polyanhydride deg- fiber matrices for cell entrapment. Acta Biomater.
radation and erosion. Adv Drug Deliv Rev. 2005;1(4):387–97.
2002;54(7):911–31. 98. Silva GA, Czeisler C, Niece KL, Beniash E,
83. Dang JM, Leong KW. Natural polymers for gene Harrington DA, Kessler JA, et al. Selective differ-
delivery and tissue engineering. Adv Drug Deliv entiation of neural progenitor cells by high-epit-
Rev. 2006;58(4):487–99. ope density nanofibers. Science. 2004;303(5662):
84. Bedran-Russo AK, Pereira PN, Duarte WR, 1352–5.
Drummond JL, Yamauchi M. Application of cross- 99. Hoffman AS. Bioconjugates of intelligent poly-
linkers to dentin collagen enhances the ultimate ten- mers and recognition proteins for use in diag-
sile strength. J Biomed Mater Res B Appl Biomater. nostics and affinity separations. Clin Chem.
2007;80(1):268–72. 2000;46(9):1478–86.
48 S.J. Lee et al.

100. Stayton PS, Shimoboji T, Long C, Chilkoti A, Chen 116. Ju YM, Choi JS, Atala A, Yoo JJ, Lee SJ. Bilayered
G, Harris JM, et al. Control of protein-ligand rec- scaffold for engineering cellularized blood vessels.
ognition using a stimuli-responsive polymer. Nature. Biomaterials. 2010;31(15):4313–21.
1995;378(6556):472–4. 117. Cui X, Boland T. Human microvasculature fabri-
101. El-Sayed ME, Hoffman AS, Stayton PS. Smart poly- cation using thermal inkjet printing technology.
meric carriers for enhanced intracellular delivery of Biomaterials. 2009;30(31):6221–7.
therapeutic macromolecules. Expert Opin Biol Ther. 118. Boland T, Xu T, Damon B, Cui X. Application of
2005;5(1):23–32. inkjet printing to tissue engineering. Biotechnol J.
102. Shimoboji T, Larenas E, Fowler T, Hoffman AS, 2006;1(9):910–7.
Stayton PS. Temperature-induced switching of 119. Xu T, Jin J, Gregory C, Hickman JJ, Boland T. Inkjet
enzyme activity with smart polymer-enzyme conju- printing of viable mammalian cells. Biomaterials.
gates. Bioconjug Chem. 2003;14(3):517–25. 2005;26(1):93–9.
103. Stayton PS, Ding Z, Hoffman AS. Smart polymer-­ 120. Xu T, Zhao W, Zhu JM, Albanna MZ, Yoo JJ, Atala
streptavidin conjugates. Methods Mol Biol. A. Complex heterogeneous tissue constructs con-
2004;283:37–43. taining multiple cell types prepared by inkjet print-
104. Bettinger CJ, Langer R, Borenstein JT. Engineering ing technology. Biomaterials. 2012;34(1):130–9.
substrate topography at the micro- and nanoscale 121. Fedorovich NE, Swennen I, Girones J, Moroni L,
to control cell function. Angew Chem Int Ed Eng. van Blitterswijk CA, Schacht E, et al. Evaluation
2009;48(30):5406–15. of photocrosslinked Lutrol hydrogel for tis-
105.
Ross AM, Jiang Z, Bastmeyer M, Lahann sue printing applications. Biomacromolecules.
J. Physical aspects of cell culture substrates: 2009;10(7):1689–96.
topography, roughness, and elasticity. Small. 122. Kang HW, Lee SJ, Ko IK, Kengla C, Yoo JJ, Atala
2012;8(3):336–55. A. A 3D bioprinting system to produce human-­
106. Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix scale tissue constructs with structural integrity. Nat
elasticity directs stem cell lineage specification. Biotechnol. 2016;34(3):312–9.
Cell. 2006;126(4):677–89. 123. Merceron TK, Burt M, Seol YJ, Kang HW, Lee SJ,
107. Hoffman AS. Hydrogels for biomedical applica- Yoo JJ, et al. A 3D bioprinted complex structure for
tions. Adv Drug Deliv Rev. 2002;54(1):3–12. engineering the muscle-tendon unit. Biofabrication.
108. Cushing MC, Anseth KS. Materials science. Hydrogel 2015;7(3):035003.
cell cultures. Science. 2007;316(5828):1133–4. 124. Atala A, Cima LG, Kim W, Paige KT, Vacanti JP,
109. Kraehenbuehl TP, Zammaretti P, Van der Vlies AJ, Retik AB, et al. Injectable alginate seeded with
Schoenmakers RG, Lutolf MP, Jaconi ME, et al. chondrocytes as a potential treatment for vesicoure-
Three-dimensional extracellular matrix-directed teral reflux. J Urol. 1993;150(2 Pt 2):745–7.
cardioprogenitor differentiation: systematic modu- 125. Atala A, Kim W, Paige KT, Vacanti CA, Retik
lation of a synthetic cell-responsive PEG-hydrogel. AB. Endoscopic treatment of vesicoureteral reflux
Biomaterials. 2008;29(18):2757–66. with a chondrocyte-alginate suspension. J Urol.
110. Orive G, Gascon AR, Hernandez RM, Igartua M, 1994;152(2 Pt 2):641–3. discussion 4
Luis PJ. Cell microencapsulation technology for 126. Diamond DA, Caldamone AA. Endoscopic correc-
biomedical purposes: novel insights and challenges. tion of vesicoureteral reflux in children using autol-
Trends Pharmacol Sci. 2003;24(5):207–10. ogous chondrocytes: preliminary results. J Urol.
111. King A, Strand B, Rokstad AM, Kulseng B, 1999;162(3 Pt 2):1185–8.
Andersson A, Skjak-Braek G, et al. Improvement 127. Bent AE, Tutrone RT, McLennan MT, Lloyd LK,
of the biocompatibility of a­lginate/poly-l-lysine/ Kennelly MJ, Badlani G. Treatment of intrinsic
alginate microcapsules by the use of epimer- sphincter deficiency using autologous ear chon-
ized alginate as a coating. J Biomed Mater Res A. drocytes as a bulking agent. Neurourol Urodyn.
2003;64(3):533–9. 2001;20(2):157–65.
112. Klock G, Pfeffermann A, Ryser C, Grohn P, Kuttler B, 128. Chancellor MB, Yokoyama T, Tirney S, Mattes
Hahn HJ, et al. Biocompatibility of mannuronic acid- CE, Ozawa H, Yoshimura N, et al. Preliminary
rich alginates. Biomaterials. 1997;18(10):707–13. results of myoblast injection into the urethra and
113. Yang S, Leong KF, Du Z, Chua CK. The design bladder wall: a possible method for the treat-
of scaffolds for use in tissue engineering. Part ment of stress urinary incontinence and impaired
I. Traditional factors. Tissue Eng. 2001;7(6):679–89. detrusor contractility. Neurourol Urodyn.
114. Greiner A, Wendorff JH. Electrospinning: a fas- 2000;19(3):279–87.
cinating method for the preparation of ultrathin
129. Strasser H, Marksteiner R, Margreiter E,
fibers. Angew Chem Int Ed Eng. 2007;46(30): Pinggera GM, Mitterberger M, Frauscher F, et al.
5670–703. Autologous myoblasts and fibroblasts versus col-
115. Pham QP, Sharma U, Mikos AG. Electrospinning of lagen for treatment of stress urinary incontinence
polymeric nanofibers for tissue engineering applica- in women: a randomised controlled trial. Lancet.
tions: a review. Tissue Eng. 2006;12(5):1197–211. 2007;369(9580):2179–86.
2  Biomaterials and Tissue Engineering 49

130. Yiou R, Yoo JJ, Atala A. Restoration of functional ogous urethras for patients who need reconstruction: an
motor units in a rat model of sphincter injury by observational study. Lancet. 2011;377(9772):1175–82.
muscle precursor cell autografts. Transplantation. 147. Atala A, Bauer SB, Soker S, Yoo JJ, Retik AB. Tissue-­
2003;76(7):1053–60. engineered autologous bladders for patients needing
131. Eberli D, Andersson KE, Yoo JJ, Atala A. A canine cystoplasty. Lancet. 2006;367(9518):1241–6.
model of irreversible urethral sphincter insuffi- 148. Oberpenning F, Meng J, Yoo JJ, Atala A. De novo
ciency. BJU Int. 2009;103(2):248–53. reconstitution of a functional mammalian urinary
132. Machluf M, Orsola A, Boorjian S, Kershen R, Atala bladder by tissue engineering. Nat Biotechnol.
A. Microencapsulation of Leydig cells: a system 1999;17(2):149–55.
for testosterone supplementation. Endocrinology. 149. Kwon TG, Yoo JJ, Atala A. Autologous penile cor-
2003;144(11):4975–9. pora cavernosa replacement using tissue engineering
133. Bardin CW, Swerdloff RS, Santen RJ. Androgens: techniques. J Urol. 2002;168(4 Pt 2):1754–8.
risks and benefits. J Clin Endocrinol Metab. 150. Chen KL, Eberli D, Yoo JJ, Atala A. Bioengineered
1991;73(1):4–7. corporal tissue for structural and functional res-
134. Tai J, Johnson HW, Tze WJ. Successful trans- toration of the penis. Proc Natl Acad Sci U S A.
plantation of Leydig cells in castrated inbred rats. 2010;107(8):3346–50.
Transplantation. 1989;47(6):1087–9. 151. De Filippo RE, Yoo JJ, Atala A. Engineering of vag-
135. De Vos P, De Haan B, Van Schilfgaarde R. Effect inal tissue in vivo. Tissue Eng. 2003;9(2):301–6.
of the alginate composition on the biocompatibility 152. Raya-Rivera AM, Esquiliano D, Fierro-Pastrana
of alginate-polylysine microcapsules. Biomaterials. R, Lopez-Bayghen E, Valencia P, Ordorica-Flores
1997;18(3):273–8. R, et al. Tissue-engineered autologous vaginal
136. Tai IT, Sun AM. Microencapsulation of recombi- organs in patients: a pilot cohort study. Lancet.
nant cells: a new delivery system for gene therapy. 2014;384(9940):329–36.
FASEB J. 1993;7(11):1061–9. 153. Campbell GR, Turnbull G, Xiang L, Haines M,
137. Bazeed MA, Thuroff JW, Schmidt RA, Tanagho Armstrong S, Rolfe BE, et al. The peritoneal cav-
EA. New treatment for urethral strictures. Urology. ity as a bioreactor for tissue engineering visceral
1983;21(1):53–7. organs: bladder, uterus and vas deferens. J Tissue
138. Olsen L, Bowald S, Busch C, Carlsten J, Eriksson Eng Regen Med. 2008;2(1):50–60.
I. Urethral reconstruction with a new synthetic 154. House M, Sanchez CC, Rice WL, Socrate S,
absorbable device. An experimental study. Scand J Kaplan DL. Cervical tissue engineering using silk
Urol Nephrol. 1992;26(4):323–6. scaffolds and human cervical cells. Tissue Eng A.
139. Atala A, Vacanti JP, Peters CA, Mandell J, Retik 2010;16(6):2101–12.
AB, Freeman MR. Formation of urothelial structures 155. Auchincloss H, Bonventre JV. Transplanting cloned
in vivo from dissociated cells attached to biodegrad- cells into therapeutic promise. Nat Biotechnol.
able polymer scaffolds in vitro. J Urol. 1992;148(2 2002;20(7):665–6.
Pt 2):658–62. 156. Amiel GE, Yoo JJ, Atala A. Renal therapy using
140. Sievert KD, Bakircioglu ME, Nunes L, Tu R, tissue-­engineered constructs and gene delivery.
Dahiya R, Tanagho EA. Homologous acellular World J Urol. 2000;18(1):71–9.
matrix graft for urethral reconstruction in the rab- 157. Aebischer P, Ip TK, Panol G, Galletti PM. The bio-
bit: histological and functional evaluation. J Urol. artificial kidney: progress towards an ultrafiltration
2000;163(6):1958–65. device with renal epithelial cells processing. Life
141. Atala A, Guzman L, Retik AB. A novel inert collagen Support Syst. 1987;5(2):159–68.
matrix for hypospadias repair. J Urol. 1999;162(3 Pt 158. Humes HD, Buffington DA, MacKay SM, Funke
2):1148–51. AJ, Weitzel WF. Replacement of renal function in
142. El-Kassaby AW, Retik AB, Yoo JJ, Atala A. Urethral uremic animals with a tissue-engineered kidney. Nat
stricture repair with an off-the-shelf collagen matrix. Biotechnol. 1999;17(5):451–5.
J Urol. 2003;169(1):170–3. discussion 3 159. Joki T, Machluf M, Atala A, Zhu J, Seyfried NT,
143. El-Kassaby A, AbouShwareb T, Atala A. Randomized Dunn IF, et al. Continuous release of endostatin
comparative study between buccal mucosal and acel- from microencapsulated engineered cells for tumor
lular bladder matrix grafts in complex anterior ure- therapy. Nat Biotechnol. 2001;19(1):35–9.
thral strictures. J Urol. 2008;179(4):1432–6. 160. Lanza RP, Hayes JL, Chick WL. Encapsulated cell
144. De Filippo RE, Yoo JJ, Atala A. Urethral replacement technology. Nat Biotechnol. 1996;14(9):1107–11.
using cell seeded tubularized collagen matrices. J 161. Lanza RP, Chung HY, Yoo JJ, Wettstein PJ,
Urol. 2002;168(4 Pt 2):1789–92. discussion 92-3 Blackwell C, Borson N, et al. Generation of histo-
145. le Roux PJ. Endoscopic urethroplasty with unseeded compatible tissues using nuclear transplantation. Nat
small intestinal submucosa collagen matrix grafts: a Biotechnol. 2002;20(7):689–96.
pilot study. J Urol. 2005;173(1):140–3. 162.
Steinborn R, Schinogl P, Zakhartchenko V,
146. Raya-Rivera A, Esquiliano DR, Yoo JJ, Lopez-­ Achmann R, Schernthaner W, Stojkovic M, et al.
Bayghen E, Soker S, Atala A. Tissue-engineered autol- Mitochondrial DNA heteroplasmy in cloned ­cattle
50 S.J. Lee et al.

produced by fetal and adult cell cloning. Nat pun polycaprolactone-­collagen scaffolds in vascular
Genet. 2000;25(3):255–7. reconstruction. Biomaterials. 2009;30(4):583–8.
163. Fischer Lindahl K, Hermel E, Loveland BE, Wang 180. Bach AD, Beier JP, Stern-Staeter J, Horch
CR. Maternally transmitted antigen of mice: a RE. Skeletal muscle tissue engineering. J Cell Mol
model transplantation antigen. Annu Rev Immunol. Med. 2004;8(4):413–22.
1991;9:351–72. 181. DiEdwardo CA, Petrosko P, Acarturk TO, DiMilla
164. Yard BA, Kooymans-Couthino M, Reterink T, van PA, LaFramboise WA, Johnson PC. Muscle tissue
den Elsen P, Paape ME, Bruyn JA, et al. Analysis of engineering. Clin Plast Surg. 1999;26(4):647–56.
T cell lines from rejecting renal allografts. Kidney ix-x
Int Suppl. 1993;39:S133–8. 182. Cossu G, Biressi S. Satellite cells, myoblasts and
165. Abolbashari M, Agcaoili SM, Lee MK, Ko IK, other occasional myogenic progenitors: possible ori-
Aboushwareb T, Jackson JD, et al. Repopulation of gin, phenotypic features and role in muscle regen-
porcine kidney scaffold using porcine primary renal eration. Semin Cell Dev Biol. 2005;16(4–5):623–31.
cells. Acta Biomater. 2016;29:52–61. 183. Cao B, Deasy BM, Pollett J, Huard J. Cell ther-
166. Kim IH, Ko IK, Atala A, Yoo JJ. Whole kidney apy for muscle regeneration and repair. Phys Med
engineering for clinical translation. Curr Opin Organ Rehabil Clin N Am. 2005;16(4):889–907.
Transplant. 2015;20(2):165–70. 184. Wernig A, Zweyer M, Irintchev A. Function of
167. Ross R. The pathogenesis of atherosclerosis: a per- skeletal muscle tissue formed after myoblast trans-
spective for the 1990s. Nature. 1993;362(6423):801–9. plantation into irradiated mouse muscles. J Physiol.
168. Bujan J, Garcia-Honduvilla N, Bellon JM. Engineering 2000;522(Pt 2):333–45.
conduits to resemble natural vascular tissue. Biotechnol 185. Cesar M, Roussanne-Domergue S, Coulet B, Gay
Appl Biochem. 2004;39(Pt 1):17–27. S, Micallef JP, Chammas M, et al. Transplantation
169. Edelman ER. Vascular tissue engineering: designer of adult myoblasts or adipose tissue precursor
arteries. Circ Res. 1999;85(12):1115–7. cells by high-density injection failed to improve
170. Gong Z, Niklason LE. Blood vessels engineered reinnervated skeletal muscles. Muscle Nerve.
from human cells. Trends Cardiovasc Med. 2008;37(2):219–30.
2006;16(5):153–6. 186. DeRosimo JF, Washabaugh CH, Ontell MP, Daood
171. Greenwald SE, Berry CL. Improving vascular grafts: MJ, Watchko JF, Watkins SC, et al. Enhancement
the importance of mechanical and haemodynamic of adult muscle regeneration by primary myoblast
properties. J Pathol. 2000;190(3):292–9. transplantation. Cell Transplant. 2000;9(3):369–77.
172. Swain TW 3rd, Calligaro KD, Dougherty 187. Payumo FC, Kim HD, Sherling MA, Smith LP,
MD. Management of infected aortic prosthetic Powell C, Wang X, et al. Tissue engineering skeletal
grafts. Vasc Endovasc Surg. 2004;38(1):75–82. muscle for orthopaedic applications. Clin Orthop
173. Amiel GE, Komura M, Shapira O, Yoo JJ, Yazdani Relat Res. 2002;(403 Suppl):S228–42.
S, Berry J, et al. Engineering of blood vessels from 188. Powell CA, Smiley BL, Mills J, Vandenburgh
acellular collagen matrices coated with human endo- HH. Mechanical stimulation improves tissue-­
thelial cells. Tissue Eng. 2006;12(8):2355–65. engineered human skeletal muscle. Am J Phys Cell
174. Kaushal S, Amiel GE, Guleserian KJ, Shapira Physiol. 2002;283(5):C1557–65.
OM, Perry T, Sutherland FW, et al. Functional 189. Choi JS, Lee SJ, Christ GJ, Atala A, Yoo JJ. The
small-diameter neovessels created using endothe- influence of electrospun aligned poly(epsilon-­
lial progenitor cells expanded ex vivo. Nat Med. caprolactone)/collagen nanofiber meshes on the
2001;7(9):1035–40. formation of self-aligned skeletal muscle myotubes.
175. Drilling S, Gaumer J, Lannutti J. Fabrication of burst Biomaterials. 2008;29(19):2899–906.
pressure competent vascular grafts via electrospin- 190. Wakelam MJ. The fusion of myoblasts. Biochem J.
ning: effects of microstructure. J Biomed Mater Res 1985;228(1):1–12.
A. 2009;88(4):923–34. 191. Derby B. Printing and prototyping of tissues and
176. Lee SJ, Yoo JJ, Lim GJ, Atala A, Stitzel J. In vitro scaffolds. Science. 2012;338(6109):921–6.
evaluation of electrospun nanofiber scaffolds for 192. Jones N. Science in three dimensions: the print revo-
vascular graft application. J Biomed Mater Res A. lution. Nature. 2012;487(7405):22–3.
2007;83(4):999–1008. 193. Xu T, Binder KW, Albanna MZ, Dice D, Zhao W,
177. Stitzel J, Liu J, Lee SJ, Komura M, Berry J, Soker S, Yoo JJ, et al. Hybrid printing of mechanically and
et al. Controlled fabrication of a biological vascular biologically improved constructs for cartilage tis-
substitute. Biomaterials. 2006;27(7):1088–94. sue engineering applications. Biofabrication.
178. Baguneid MS, Seifalian AM, Salacinski HJ, Murray 2012;5(1):015001.
D, Hamilton G, Walker MG. Tissue engineering of 194. De Coppi P, Bartsch G Jr, Siddiqui MM, Xu T,
blood vessels. Br J Surg. 2006;93(3):282–90. Santos CC, Perin L, et al. Isolation of amniotic stem
179. Tillman BW, Yazdani SK, Lee SJ, Geary RL, cell lines with potential for therapy. Nat Biotechnol.
Atala A, Yoo JJ. The in vivo stability of electros- 2007;25(1):100–6.
2  Biomaterials and Tissue Engineering 51

195. Koning M, Harmsen MC, van Luyn MJ, Werker agrin on skeletal muscle fibers in vivo. J Cell Biol.
PM. Current opportunities and challenges in skeletal 2001;153(7):1441–52.
muscle tissue engineering. J Tissue Eng Regen Med. 209. Ko IK, Lee BK, Lee SJ, Andersson K-E, Atala A,
2009;3(6):407–15. Yoo JJ. The effect of in vitro formation of acetyl-
196. Kingham PJ, Terenghi G. Bioengineered nerve choline receptor (AChR) clusters in engineered
regeneration and muscle reinnervation. J Anat. muscle fibers on subsequent innervation of con-
2006;209(4):511–26. structs in vivo. Biomaterials. 2013;34(13):3246–55.
197. Kang SB, Olson JL, Atala A, Yoo JJ. Functional doi:10.1016/j.biomaterials.2013.01.029.
recovery of completely denervated muscle: implica- 210. Folkman J, Hochberg M. Self-regulation of growth in
tions for innervation of tissue-engineered muscle. three dimensions. J Exp Med. 1973;138(4):745–53.
Tissue Eng A. 2012;18(17–18):1912–20. 211. Nomi M, Atala A, Coppi PD, Soker S. Principals of
198. Bian W, Bursac N. Soluble miniagrin enhances neovascularization for tissue engineering. Mol Asp
contractile function of engineered skeletal muscle. Med. 2002;23(6):463–83.
FASEB J. 2012;26(2):955–65. 212. De Coppi P, Delo D, Farrugia L, Udompanyanan K,
199. Kummer TT, Misgeld T, Sanes JR. Assembly of Yoo JJ, Nomi M, et al. Angiogenic gene-modified
the postsynaptic membrane at the neuromuscu- muscle cells for enhancement of tissue formation.
lar junction: paradigm lost. Curr Opin Neurobiol. Tissue Eng. 2005;11(7–8):1034–44.
2006;16(1):74–82. 213. Schuch G, Machluf M, Bartsch G Jr, Nomi M,
200. Wu H, Xiong WC, Mei L. To build a synapse: signal- Richard H, Atala A, et al. In vivo administration of
ing pathways in neuromuscular junction assembly. vascular endothelial growth factor (VEGF) and its
Development. 2010;137(7):1017–33. antagonist, soluble neuropilin-1, predicts a role of
201. Witzemann V. Development of the neuromuscular VEGF in the progression of acute myeloid leukemia
junction. Cell Tissue Res. 2006;326(2):263–71. in vivo. Blood. 2002;100(13):4622–8.
202. Bach AD, Beier JP, Stark GB. Expression of Trisk 214. Ennett AB, Kaigler D, Mooney DJ. Temporally
51, agrin and nicotinic-acetycholine receptor regulated delivery of VEGF in vitro and in vivo. J
epsilon-­
subunit during muscle development in a Biomed Mater Res A. 2006;79(1):176–84.
novel three-dimensional muscle-neuronal co-culture 215. Kaigler D, Wang Z, Horger K, Mooney DJ,
system. Cell Tissue Res. 2003;314(2):263–74. Krebsbach PH. VEGF scaffolds enhance angio-
203. Bentzinger CF, Barzaghi P, Lin S, Ruegg genesis and bone regeneration in irradiated osseous
MA. Overexpression of mini-agrin in skeletal defects. J Bone Miner Res. 2006;21(5):735–44.
muscle increases muscle integrity and regenerative 216. Borselli C, Cezar CA, Shvartsman D, Vandenburgh HH,
capacity in laminin-alpha2-deficient mice. FASEB J. Mooney DJ. The role of multifunctional delivery scaf-
2005;19(8):934–42. fold in the ability of cultured myoblasts to promote mus-
204. Glass DJ, Bowen DC, Stitt TN, Radziejewski C, cle regeneration. Biomaterials. 2011;32(34):8905–14.
Bruno J, Ryan TE, et al. Agrin acts via a MuSK 217. Levenberg S, Rouwkema J, Macdonald M, Garfein
receptor complex. Cell. 1996;85(4):513–23. ES, Kohane DS, Darland DC, et al. Engineering
205. Lin S, Landmann L, Ruegg MA, Brenner HR. The vascularized skeletal muscle tissue. Nat Biotechnol.
role of nerve- versus muscle-derived factors in 2005;23(7):879–84.
mammalian neuromuscular junction formation. J 218. Moon JJ, Saik JE, Poche RA, Leslie-Barbick JE, Lee SH,
Neurosci. 2008;28(13):3333–40. Smith AA, et al. Biomimetic hydrogels with pro-angio-
206. DeChiara TM, Bowen DC, Valenzuela DM, genic properties. Biomaterials. 2010;31(14):3840–7.
Simmons MV, Poueymirou WT, Thomas S, et al. 219. Tsang VL, Bhatia SN. Fabrication of three-­
The receptor tyrosine kinase MuSK is required for dimensional tissues. Adv Biochem Eng Biotechnol.
neuromuscular junction formation in vivo. Cell. 2007;103:189–205.
1996;85(4):501–12. 220. Weigel T, Schinkel G, Lendlein A. Design and prep-
207. Gautam M, Noakes PG, Moscoso L, Rupp F, aration of polymeric scaffolds for tissue engineering.
Scheller RH, Merlie JP, et al. Defective neuromus- Expert Rev Med Devices. 2006;3(6):835–51.
cular synaptogenesis in agrin-deficient mutant mice. 221. Williams DF. A model for biocompatibility and its
Cell. 1996;85(4):525–35. evaluation. J Biomed Eng. 1989;11(3):185–91.
208. Bezakova G, Helm JP, Francolini M, Lomo 222. Remes A, Williams DF. Immune response in bio-
T. Effects of purified recombinant neural and muscle compatibility. Biomaterials. 1992;13(11):731–43.
Cell Technologies
3
So Young Chun

3.1  vailable Stem Cell Types


A then donated for research purposes. Embryonic
for Urology Field stem cells are described as “pluripotent,” mean-
ing that they can generate all the different types
3.1.1 Overview of cells. Embryonic stem cells can be obtained
from the blastocyst, a very early stage of devel-
Stem cells are undifferentiated biological cells opment that consists of a mostly hollow ball of
that can differentiate into specialized cells and approximately 150–200 cells. At this stage,
can divide to produce more stem cells. When a there are no organs, just an “inner cell mass”
stem cell divides, each new cell has the potential from which embryonic stem cells can be
either to remain a stem cell or become another obtained. Human embryonic stem cells are
type of cell with a more specialized function. derived primarily from blastocysts that were
They are found in multicellular organisms. In created by in vitro fertilization (IVF) for assisted
mammals, there are two broad types of stem reproduction. The fertilized cells that immedi-
cells: embryonic stem cells, which are isolated ately arise in the first few divisions are “totipo-
from the inner cell mass of blastocysts, and adult tent.” This means that they can generate a viable
(somatic) stem cells, which are found in various embryo. Then these cells transition to become
tissues. In adult organisms, stem cells and pro- pluripotent.
genitor cells act as a repair system for the body, Adult (somatic) stem cells are found in most
replenishing adult tissues. In a developing major organs and tissues and are currently being
embryo, stem cells can differentiate into all the isolated from many tissues in the body. The
specialized cells—ectoderm, endoderm, and methods of isolation and culture are dependent
mesoderm—but also maintain the normal turn- on the cell source and lineage. Many isolation
over of regenerative organs, such as blood, skin, and purification protocols involve flow cytometry
or intestinal tissues. and cell sorting. Positive and negative sorting for
Embryonic stem cells are derived from cell surface markers can quickly generate
embryos. Most embryonic stem cells are derived enriched populations.
from eggs that have been fertilized in vitro and Tissue-specific stem cells, referred to as
“adult” or “somatic” stem cells, are already
somewhat specialized and can produce some or
S.Y. Chun all of the mature cell types found within the par-
BioMedical Research Institute, ticular tissue or organ. Because of their ability to
Kyungpook National Univ. Hospital,
Deagu, 41940, USA generate multiple, organ-specific cell types, they
e-mail: soyachun@hotmail.com are described as “multipotent.” Tissue-specific

© Springer Nature Singapore Pte Ltd. 2018 53


B.W. Kim (ed.), Clinical Regenerative Medicine in Urology, DOI 10.1007/978-981-10-2723-9_3
54 S.Y. Chun

stem cells have been found in several organs that and local factors, the cell acquires a more special-
need to continuously replenish themselves, such ized function after cell division and becomes a
as the blood, skin, and gut. These types of stem less potent stem cell or becomes a fully differen-
cells represent a very small population. tiated cell.
There are three known accessible sources of Two mechanisms exist to ensure that a stem
autologous adult stem cells in humans: bone mar- cell population is maintained: asymmetric repli-
row, which requires extraction by harvesting, that cation, a stem cell divides into one mother cell
is, drilling into bone; adipose tissue, which that is identical to the original stem cell and
requires extraction by liposuction; and blood, another daughter cell that is differentiated; and
which requires extraction through apheresis, stochastic differentiation, when one stem cell
wherein blood is drawn from the donor and develops into two differentiated daughter cells,
passed through a machine that extracts the stem another stem cell undergoes mitosis and produces
cells and returns other portions of the blood to the two stem cells identical to the original.
donor. Stem cells can also be taken from amni- Stem cells can differentiate into embryonic
otic fluid through amniocentesis, umbilical cord and extraembryonic cell types. Such cells can
blood just after birth, and voided urine. construct a complete, viable organism. These
Adult stem cells are frequently used in various cells are produced from the fusion of an egg and
medical therapies. Stem cells can now be artifi- sperm cell. Cells produced by the first few divi-
cially grown and differentiated into specialized sions of the fertilized egg are also totipotent.
cell types with characteristics consistent with Pluripotent stem cells are the descendants of
cells of various tissues. Research on adult stem totipotent cells and can differentiate into nearly
cells has generated a great deal of excitement. all cells. Multipotent stem cells can differentiate
Scientists have found adult stem cells in many into a number of cell types, but only those of a
more tissues. This finding has led researchers and closely related family of cells. Oligopotent stem
clinicians to ask whether adult stem cells could cells can differentiate into only a few cell types.
be used for transplants. In fact, stem cells from Unipotent cells can produce only one cell type,
bone marrow have been used in transplants for their own [1].
more than 40 years. Scientists now have evidence
that stem cells exist in the brain and the heart, two
locations where adult stem cells were not at first 3.1.3 S
 imilarities and Differences
expected to reside. If the differentiation of adult Between Embryonic and Adult
stem cells can be controlled in the laboratory, Stem Cells
these cells may become the basis of
transplantation-­based therapies. Human embryonic and adult stem cells each have
advantages and disadvantages regarding potential
use for cell-based regenerative therapies. One
3.1.2 Stem Cell Properties major difference between adult and embryonic
stem cells is their different abilities in the number
Stem cells are distinguished from other cell types and type of differentiated cell types they can
by two important characteristics: First, they are become. Embryonic stem cells can become all
unspecialized cells capable of renewing them- cell types of the body because they are pluripo-
selves through cell division. Second, under cer- tent. Adult stem cells are thought to be limited to
tain physiologic or experimental conditions, they differentiating into different cell types of their
can be induced to become tissue-specific cells tissue of origin.
with special functions. In vivo, stem cell division Embryonic stem cells can be grown relatively
can either result in two identical daughter cells, easily in culture. Adult stem cells are rare in
or one of the cells can acquire more specialized mature tissues, so isolating these cells from an
functions. Depending on stem cell type, location, adult tissue is challenging, and methods to
3  Cell Technologies 55

expand their numbers in cell culture have not yet where age, disease, or trauma has led to tissue
been worked out. Tissues derived from embry- damage or dysfunction.
onic and adult stem cells may differ in the likeli-
hood of being rejected after transplantation. It is ES Cell Markers
not yet known for certain whether tissues derived A human embryonic stem cell is also defined by
from embryonic stem cells would cause trans- the expression of several transcription factors and
plant rejection, since relatively few clinical trials cell surface proteins. The transcription factors
have tested the safety of transplanted cells Oct4, Nanog, and Sox2 form the core regulatory
derived. network that ensures the suppression of genes
Adult stem cells are currently believed less that lead to differentiation and the maintenance
likely to initiate rejection after transplantation. of pluripotency [4]. The cell surface antigens
This is because a patient’s own cells, it could be most commonly used to identify hES cells are the
expanded in culture, differentiated, and then rein- glycolipids stage-specific embryonic antigens 3
troduced into the patient. The use of adult stem and 4 and the keratan sulfate antigens Tra-1-60
cells derived from the patient’s own adult stem and Tra-1-81. By using human embryonic stem
cells would mean that the cells are less likely to cells to produce specialized cells like nerve cells
be rejected by the immune system. This repre- or heart cells in the lab, scientists can gain access
sents a significant advantage, as immune rejec- to adult human cells without taking tissue from
tion can be circumvented only by continuous patients. They can then study these specialized
administration of immunosuppressive drugs. adult cells in detail to try and catch complications
of diseases or to study cell reactions to poten-
3.1.3.1 Embryonic Stem Cells, ES Cells tially new drugs. The molecular definition of a
stem cell includes many more proteins and con-
Pluripotency tinues to be a topic of research [5].
ES cells are pluripotent stem cells derived from
the inner cell mass of a blastocyst, an early-stage Proliferation and Differentiation
preimplantation embryo. Human embryos reach Under defined conditions, embryonic stem cells
the blastocyst stage 4–5 days post fertilization, at are capable of propagating themselves indefi-
which time they consist of 50–150 cells. ES cells nitely in an undifferentiated state and have the
are pluripotent and give rise during development capacity when provided with the appropriate sig-
to all derivatives of the three primary germ lay- nals to differentiate, presumably via the forma-
ers: ectoderm, endoderm, and mesoderm. In tion of precursor cells, to almost all mature cell
other words, they can develop into each of the phenotypes [6]. This allows embryonic stem cells
more than 200 cell types of the adult body when to be employed as useful tools for both research
given sufficient and necessary stimulation for a and regenerative medicine, because they can pro-
specific cell type. Human ES cells measure duce limitless numbers of themselves for contin-
approximately 14 μm, and mouse ES cells are ued research or clinical use.
closer to 8 μm [2, 3].
Pluripotency distinguishes embryonic stem Stemness Maintenances
cells from adult stem cells; while embryonic Nearly all research to date has made use of mouse
stem cells can generate all cell types in the body, embryonic stem cells or human embryonic stem
adult stem cells are multipotent and can produce cells derived from the early inner cell mass. Both
only a limited number of cell types. If the plu- have the essential stem cell characteristics, yet
ripotent differentiation potential of embryonic they require very different environments in order
stem cells could be harnessed in vitro, it might be to maintain an undifferentiated state. Mouse ES
a means of deriving cell or tissue types virtually cells are grown on a layer of gelatin as an extra-
to order. This would provide a radical new treat- cellular matrix and require the presence of leuke-
ment approach to a wide variety of conditions mia inhibitory factor (LIF). Human ES cells are
56 S.Y. Chun

grown on a feeder layer of mouse embryonic have also been differentiated to natural killer
fibroblasts (MEFs) and require the presence of cells and bone tissue. Studies involving ES are
basic fibroblast growth factor (bFGF or FGF-2) also underway to provide an alternative treatment
[7]. Without optimal culture conditions or genetic for diabetes.
manipulation, embryonic stem cells will rapidly
differentiate. Application to Genetic Disorders
This has been done either by genetically manipu-
Potential Clinical use lating the cells or more recently by deriving dis-
Current research focuses on differentiating ES eased cell lines identified by prenatal genetic
into a variety of cell types for eventual use as cell diagnosis. This approach may very well prove
replacement therapies. Some of the cell types that invaluable at studying disorders such as fragile X
have or are currently being developed include syndrome, cystic fibrosis, and other genetic mal-
cardiomyocytes, neurons, hepatocytes, bone adies that have no reliable model system.
marrow cells, islet cells, and endothelial cells [8]. Yury Verlinsky developed prenatal diagnosis
However, the derivation of such cell types from testing methods to determine genetic and chro-
ESs is not without obstacles, and hence current mosomal disorders a month and a half earlier
research is focused on overcoming these than standard amniocentesis. The techniques
barriers. are now used by many pregnant women and
Besides in the future becoming an important prospective parents, especially those couples
alternative to organ transplants, ES are also being with a history of genetic abnormalities, when
used in field of toxicology and as cellular screens the risk of genetically related disorders is
to uncover new chemical entities that can be higher. In addition, by allowing parents to
developed as small molecule drugs. select an embryo without genetic disorders,
Studies have shown that cardiomyocytes they have the potential of saving the lives of
derived from ES are validated in vitro models to siblings that already had similar disorders and
test drug responses and predict toxicity profiles diseases using cells from the disease-free off-
[8]. ES-derived cardiomyocytes have been shown spring [9].
to respond to pharmacological stimuli and hence
can be used to assess cardiotoxicity. Repair of DNA Damage
ES-derived hepatocytes are also useful models ES cells use a different strategy to deal with
that could be used in the preclinical stages of double-­strand breaks (DSBs). Because ES cells
drug discovery. However, the development of give rise to all of the cell types of an organism
hepatocytes from ESC has proven to be challeng- including the cells of the germ line, mutations
ing, and this hinders the ability to test drug arising in ES cells due to faulty DNA repair are
metabolism. Therefore, current research is focus- a more serious problem than in differentiated
ing on establishing fully functional ES-derived somatic cells. Consequently, robust mechanisms
hepatocytes with stable phase I and II enzyme are needed in ES cells to repair DNA damages
activity [7]. accurately and, if repair fails, to remove those
Researchers have also differentiated ESC into cells with unrepaired DNA damages. Thus,
dopamine-producing cells with the hope that mouse ES cells predominantly use high fidelity
these neurons could be used in the treatment of homologous recombinational repair (HRR) to
Parkinson’s disease [4]. Recently, the develop- repair DSBs. This type of repair depends on the
ment of ESC after somatic cell nuclear transfer of interaction of the two sister chromosomes
olfactory ensheathing cells to a healthy oocyte formed during S phase and present together dur-
has been recommended for neuro-degenerative ing the G2 phase of the cell cycle. HRR can
diseases [5]. The same group also has advocated accurately repair DSBs in one sister chromo-
the use of olfactory ensheathing cells for demye- some by using intact information from the other
linating diseases like multiple sclerosis. ESCs sister chromosome. Cells in the G1 phase of the
3  Cell Technologies 57

cell cycle have only one copy of each chromo- must be carefully monitored for changes in
some. Mouse ES cells lack a G1 checkpoint and phenotype, gene expression, and karyotype.
­
do not undergo cell cycle arrest upon acquiring Following differentiation, the cells are subjected
DNA damage. Rather they undergo programmed to sorting by flow cytometry for further purifica-
cell death (apoptosis) in response to DNA dam- tion. ESCs are predicted to be inherently safer
age. Apoptosis can be used as a fail-safe strategy than iPS cells because they are not genetically
to remove cells with unrepaired DNA damages modified with genes such as c-Myc that are
in order to avoid mutation and progression to linked to cancer. Nonetheless, ESC expresses
cancer. Consistent with this strategy, mouse ES very high levels of the iPS-inducing genes, and
stem cells have a mutation frequency about 100- these genes including Myc are essential for ESC
fold lower than that of isogenic mouse somatic self-renewal and pluripotency.
cells [10].
3.1.3.2 Reprogrammed Stem Cells
Preparation Method for hES Cells One of the hottest topics in stem cell research
Human embryonic stem cells can be derived today is the study of induced pluripotent stem
from donated embryos, or additionally they can cells (iPSCs). These are adult cells (e.g., skin
also be extracted from cloned embryos using a cells) that are “reprogrammed” to become plu-
cell from a patient and a donated egg. The inner ripotent, i.e., behave like an embryonic stem cell.
cell mass, from the blastocyst stage of the While these iPSCs share many of the same char-
embryo, is separated from the trophectoderm, acteristics of embryonic stem cells, including the
the cells that would differentiate into extraem- ability to give rise to all the cell types, the origi-
bryonic tissue. Immunosurgery, the process in nal iPSCs were produced by using viruses to
which antibodies are bound to the trophecto- insert extra copies of three to four genes known
derm and removed by another solution, and to be important in embryonic stem cells. It is not
mechanical dissection are performed to achieve yet completely understood how these three to
separation. The resulting inner cell mass cells four “reprogramming” genes are able to induce
are plated onto cells that will supply support. pluripotency. In addition, recent studies have
The inner cell mass cells attach and expand fur- focused on alternative ways of reprogramming
ther to form a human embryonic cell line, which cells using methods that are safer for use in clini-
are undifferentiated. These cells are fed daily cal settings.
and are enzymatically or mechanically sepa-
rated every 4–7 days. For differentiation to Cell Sources for Reprogramming
occur, the human embryonic stem cell line is The most frequently used sources for repro-
removed from the supporting cells to form gramming are blood cells and fibroblasts, and
embryoid bodies, co-cultured with a serum con- taking cells from urine is less invasive. The lat-
taining necessary signals, or grafted in a three- ter method does not require a biopsy or blood
dimensional scaffold [2]. sampling. Urine-derived stem cells had been
differentiated into endothelial, osteogenic,
Difficulties chondrogenic, adipogenic, skeletal myogenic,
One potential disadvantage of ESCs is that they and neurogenic lineages, without forming tera-
can form teratomas, tumors containing cell types tomas [12]. Their epigenetic memory is suited
from all three germ layers [11]. Studies suggest to reprogramming into iPSCs. However, few
that teratoma formation may be a result of chro- cells appear in urine, only low conversion effi-
mosomal changes that occur in some ES cells ciencies had been achieved, and the risk of bac-
during culture. As length of time in culture and terial contamination is relatively high. Another
the number of passages increase, cells are more promising source of cells for reprogramming is
likely to display chromosomal instability and mesenchymal stem cells derived from human
mosaicism within the population. ES cell lines hair follicles.
58 S.Y. Chun

Classification by Potency iPSC properties are very similar to ESCs. iPSCs


Induced stem cells (iSC) are stem cells derived have been shown to support the development of
from somatic, reproductive, pluripotent, or other all-iPSC mice using a tetraploid (4n) embryo, the
cell types by deliberate epigenetic reprogram- most stringent assay for developmental
ming. They are classified as totipotent (iTC), plu- potential.
ripotent (iPSC), or progenitor (multipotent,
iMSC, also called an induced multipotent pro- Rejuvenation Mechanism
genitor cell, iMPC, or unipotent, iUSC) accord- An important advantage of iPSC over ESC is that
ing to their developmental potential and degree they can be derived from adult cells, rather than
of dedifferentiation. Progenitors are obtained from embryos. Therefore, it became possible to
by so-called direct reprogramming or direct obtain iPSC from adult and even elderly patients.
differentiation. Reprogramming somatic cells to iPSC leads to
rejuvenation. It was found that reprogramming
iPSC Strategy leads to telomere lengthening and subsequent
Although additional research is needed, iPSCs shortening after their differentiation back into
are already useful tools for drug development and fibroblast-like derivatives. Thus, reprogramming
modeling of diseases, and scientists hope to use leads to the restoration of embryonic telomere
them in transplantation medicine. Viruses are length and hence increases the potential number
currently used to introduce the reprogramming of cell divisions [13].
factors into adult cells, and this process must be
carefully controlled and tested before the tech- Immune Response
nique can lead to useful treatment for humans. In However, because of the dissonance between
animal studies, the virus used to introduce the rejuvenated cells and the surrounding niche of
stem cell factors sometimes causes cancers. the recipient’s older cells, the injection of his own
Researchers are currently investigating non-viral iPSC usually leads to an immune response, which
delivery strategies. In any case, this breakthrough can be used for medical purposes or the forma-
discovery has created a powerful new way to tion of tumors such as teratoma. The reason has
“dedifferentiate” cells whose developmental been hypothesized to be that some cells differen-
fates had been previously assumed to be deter- tiated from ESC and iPSC in vivo continue to
mined. In addition, tissues derived from iPSCs synthesize embryonic protein isoforms. So, the
will be a nearly identical match to the cell donor immune system might detect and attack cells that
and thus probably avoid rejection by the immune are not cooperating properly [14].
system. The iPSC strategy creates pluripotent
stem cells that, together with studies of other Teratoma Formation Mechanism
types of pluripotent stem cells, will help research- Teratoma formation by pluripotent stem cells
ers learn how to reprogram cells to repair dam- may be caused by low activity of PTEN enzyme,
aged tissues in the human body. reported to promote the survival of a small popu-
lation (0, 1–5% of total population) of highly
Reprogramming Factors tumorigenic, aggressive, teratoma-initiating
Human somatic cells are made pluripotent by embryonic-like carcinoma cells during differen-
transducing them with factors that induce pluri- tiation. The survival of these teratoma-initiating
potency (OCT 3/4, SOX2, Klf4, c-Myc, NANOG, cells is associated with failed repression of Nanog
and LIN28). This results in the production of as well as a propensity for increased glucose and
iPSCs, which can differentiate into any cells of cholesterol metabolism [15]. These teratoma-­
the three embryonic germ layers (mesoderm, initiating cells also expressed a lower ratio of
endoderm, ectoderm). Reprogramming mecha- p53/p21 when compared to nontumorigenic cells.
nisms are thus linked, rather than independent In connection with the above safety problems,
and are centered on a small number of genes. the use iPSC for cell therapy is still limited.
3  Cell Technologies 59

However, they can be used for a variety of other (donor of nuclei), because of mitochondrial DNA
purposes—including the modeling of disease, in cytoplasm, as a donor of oocytes, instead of the
screening of drugs, and toxicity testing of various patient’s mitochondrial DNA. This reduces their
drugs. value as a source for autologous stem cell trans-
It is interesting to note that the tissues grown plantation therapy, until it is not clear how it can
from iPSCs, placed in the “chimeric” embryos in induce an immune response of the patient upon
the early stages of mouse development, practi- treatment.
cally do not cause an immune response (after the This technologic may have far-reaching clini-
embryos have grown into adult mice) and are cal applications for overcoming cytoplasmic
suitable for autologous transplantation. At the defects in human oocytes. For example, the tech-
same time, full reprogramming of adult cells nology could prevent inherited mitochondrial
in vivo within tissues by transitory induction of disease from passing to future generations.
the four factors Oct4, Sox2, Klf4, and c-Myc in Mitochondrial genetic material is passed from
mice results in teratomas emerging from multiple mother to child. The nucleus from one human
organs. Furthermore, partial reprogramming of egg has been transferred to another, including its
cells toward pluripotency in vivo in mice demon- mitochondria, creating a cell that could be
strates that incomplete reprogramming entails regarded as having two mothers. The eggs were
epigenetic changes (failed repression of then fertilized and the resulting embryonic stem
Polycomb targets and altered DNA methylation) cells carried the swapped mitochondrial DNA. As
in cells that drive cancer development [16]. evidence that the technique is safe, the author of
this method points to the existence of the healthy
3.1.3.3 Reprogramming Methods monkeys that are now more than 4 years old and
Several techniques are widely recognized: trans- are the product of mitochondrial transplants
plantation of nuclei taken from somatic cells into across different genetic backgrounds [19].
an oocyte lacking its own nucleus, fusion of
somatic cells with pluripotent stem cells, and Induced Totipotent Cells without SCNT
transformation of somatic cells into stem cells, Recently some researchers succeeded to get the
using the genetic material encoding reprogram- totipotent cells without the aid of SCNT. Totipotent
ming protein factors, recombinant proteins; cells were obtained using the epigenetic factors
microRNA, a synthetic, self-replicating polycis- such as oocyte germinal isoform of histone.
tronic RNA and low-molecular weight biologi- Reprogramming in vivo, by transitory induction of
cally active substances [17]. the four factors Oct4, Sox2, Klf4, and c-Myc in
mice, confers totipotency features. Intraperitoneal
Somatic Cell Nuclear Transfer injection of such in vivo iPS cells generates
Induced totipotent cells can be obtained by repro- embryo-like structures that express embryonic and
gramming somatic cells with somatic cell nuclear extraembryonic markers [20]. Transplantation of
transfer (SCNT). The process involves sucking pluripotent/embryonic stem cells into the body of
out the nucleus of a somatic cell and injecting it adult mammals usually leads to the formation of
into an oocyte that has had its nucleus removed teratomas, which can then turn into a malignant
[18]. Totipotent cells can be generated by SCNT tumor teratocarcinoma. However, putting terato-
using dermal fibroblasts nuclei from both a carcinoma cells into the embryo at the blastocyst
middle-­aged 35 years-old male and an elderly, stage caused them to become incorporated in the
75 years-old male, suggesting that age-associated cell mass and often produced a normal healthy chi-
changes are not necessarily an impediment to meric animal.
SCNT-based nuclear reprogramming of human
cells. Unfortunately, the cells generated by this Reprogramming with Reagents
technology potentially are not completely pro- The risk of cancer creates the need to develop
tected from the immune system of the patient methods for safer cell lines suitable for clinical
60 S.Y. Chun

use. An alternative approach is so-called “direct n­euronal cells. Thus, transcriptional activation
reprogramming”—transdifferentiation of cells and epigenetic remodeling of endogenous master
without passing through the pluripotent state transcription factors are sufficient for conversion
[21]. The basis for this approach was that between cell types. The rapid and sustained acti-
5-­azacytidine—a DNA demethylation reagent— vation of endogenous genes in their native chro-
can cause the formation of myogenic, chondro- matin context by this approach may facilitate
genic, and adipogenic clones in the immortal cell reprogramming with transient methods that avoid
line of mouse embryonic fibroblasts and that the genomic integration and provides a new strategy
activation of a single gene, later named MyoD1, for overcoming epigenetic barriers to cell fate
is sufficient for such reprogramming. Compared specification.
with iPSC whose reprogramming requires at
least 2 weeks, the formation of induced progeni- Antibody-Based Reprogramming
tor cells sometimes occurs within a few days, and The researchers discovered that GCSF-­
the efficiency of reprogramming is usually many mimicking antibody can activate a growth-­
times higher. This reprogramming does not stimulating receptor on marrow cells in a way
always require cell division. The cells resulting that induces marrow stem cells that normally
from such reprogramming are more suitable for develop into white blood cells to become neural
cell therapy because they do not form teratomas. progenitor cells [23]. The technique enables
researchers to search large libraries of antibodies
Single Transcription Factor and quickly select the ones with a desired bio-
Originally only early embryonic cells could be logical effect.
coaxed into changing their identity. Mature cells
are resistant to changing their identity once Conditionally Reprogrammed Cells
they’ve committed to a specific kind. However, a Schlegel and Liu [24] demonstrated that the com-
brief expression of a single transcription factor, bination of feeder cells and a Rho kinase inhibi-
ELT-7 GATA, can convert the identity of fully tor induces normal and tumor epithelial cells
differentiated, specialized non-endodermal cells from many tissues to proliferate indefinitely
of the pharynx into fully differentiated intestinal in vitro. This process occurs without the need for
cells in intact larvae and adult roundworm transduction of exogenous viral or cellular genes.
Caenorhabditis elegans with no requirement for These cells have been termed “conditionally
a dedifferentiated intermediate. reprogrammed cells (CRC).” The induction of
CRCs is rapid and results from reprogramming
CRISPR-Mediated Activator of the entire cell population. CRCs do not express
The cell fate can be effectively manipulated by high levels of protein characteristic of iPSCs or
directly activating specific endogenous gene embryonic stem cells. This induction of CRCs is
expression with CRISPR-mediated activator. reversible and feeders allow the cells to differen-
When dCas9 is combined with transcription acti- tiate normally. CRC technology can generate
vators, it can precisely manipulate endogenous 2 × 106 cells in 5–6 days from needle biopsies
gene expression. Using this method, Wei et al. and can generate cultures from cryopreserved tis-
enhanced the expression of endogenous Cdx2 sue and from fewer than four viable cells. CRCs
and Gata6 genes by CRISPR-mediated activa- retain a normal karyotype and remain
tors, and thus directly converted mouse embry- nontumorigenic.
onic stem cells into two extraembryonic lineages,
i.e., typical trophoblast stem cells and extraem- Lineage-Specific Enhancers
bryonic endoderm cells [22]. An analogous Differentiated macrophages can self-renew in tis-
approach was used to induce activation of the sues and expand long term in culture. Under
endogenous Brn2, Ascl1, and Myt1 genes to con- ­certain conditions macrophages can divide with-
vert mouse embryonic fibroblasts to induced out losing features they have acquired while
3  Cell Technologies 61

s­ pecializing into immune cells. The macrophages genesis and plays a key role in stem cell
achieve this by activating a gene network similar proliferation.
to the one found in embryonic stem cells. Single-­ Changes in outer membrane protein glyco-
cell analysis revealed that, in vivo, proliferating sylation are markers of cell states connected in
macrophages can derepress a macrophage-­ some way with pluripotency and differentia-
specific enhancer repertoire associated with a tion. The glycosylation change is apparently
gene network controlling self-renewal. This hap- not just the result of the initialization of gene
pened when concentrations of two transcription expression, but performs as an important gene
factors named MafB and c-Maf were naturally regulator involved in the acquisition and main-
low or were inhibited for a short time. Genetic tenance of the undifferentiated state. For
manipulations that turned off MafB and c-Maf in example, activation of glycoprotein ACA,
the macrophages caused the cells to start a self-­ linking glycosylphosphatidylinositol on the
renewal program. The similar network also con- surface of the progenitor cells in human
trols embryonic stem cell self-renewal but is peripheral blood, induces increased expres-
associated with distinct embryonic stem cell-­ sion of genes Wnt, Notch-1, BMI1, and
specific enhancers. HOXB4 through a signaling cascade PI3K/
Akt/mTor/PTEN and promotes the formation
Indirect Lineage Conversion of a self-renewing population of hematopoi-
Indirect lineage conversion is a reprogramming etic stem cells. Furthermore, dedifferentiation
methodology in which somatic cells transition of progenitor cells induced by ACA-dependent
through a plastic intermediate state of partially signaling pathway leads to ACA-induced plu-
reprogrammed cells (pre-iPSC), induced by brief ripotent stem cells, capable of differentiating
exposure to reprogramming factors, followed by in vitro into cells of all three germ layers.
differentiation in a specially developed chemical
environment (artificial niche) [25]. This method Physical Approach
could be both more efficient and safer, since it Cell adhesion protein E-cadherin is indispensable
does not seem to produce tumors or other unde- for a robust pluripotent phenotype. During repro-
sirable genetic changes and results in much gramming for iPS cell generation, N-cadherin
greater yield than other methods. can replace function of E-cadherin. These func-
tions of cadherins are not directly related to adhe-
Outer Membrane Glycoprotein sion because sphere morphology helps
A common feature of pluripotent stem cells is the maintaining the “stemness” of stem cells.
specific nature of protein glycosylation of their Moreover, sphere formation, due to forced
outer membrane. That distinguishes them from growth of cells on a low attachment surface,
the most nonpluripotent cells. The glycans on the sometimes induces reprogramming.
stem cell surface respond rapidly to alterations in Physical cues, in the form of parallel micro-
cellular state and signaling and are therefore ideal grooves on the surface of cell-adhesive sub-
for identifying even minor changes in cell popu- strates, can replace the effects of small molecule
lations. Many stem cell markers are based on cell epigenetic modifiers and significantly improve
surface glycan epitopes including the widely reprogramming efficiency. The mechanism relies
used markers SSEA-3, SSEA-4, Tra 1–60, and on the mechanomodulation of the cells’ epigene-
Tra 1–81 [2]. Suila et al. [26] speculate that tic state. Specifically, “decreased histone deacet-
human stem cells, extracellular O-GlcNAc and ylase activity and upregulation of the expression
extracellular O-LacNAc, play a crucial role in the of WD repeat domain 5 (WDR5)—a subunit of
fine-tuning of Notch signaling pathway, a highly H3 methyltranferase—by microgrooved surfaces
conserved cell signaling system, that regulates lead to increased histone H3 acetylation and
cell fate specification, differentiation, left–right methylation.” Nanofibrous scaffolds with aligned
asymmetry, apoptosis, somitogenesis, and angio- fiber orientation produce effects similar to those
62 S.Y. Chun

produced by microgrooves, suggesting that Adenoviruses [31] and Sendai virus, an RNA
changes in cell morphology may be responsible virus [32], have been used as alternative vectors
for modulation of the epigenetic state [27]. to transduce transcription factors because they do
Substrate rigidity is also one of the important not integrate into the genomic DNA.
biophysical cue influencing neural induction and Sendai virus (SeV) vectors do not integrate
subtype specification. For example, soft sub- into the host genome or alter the genetic informa-
strates promote neuroepithelial conversion while tion of the host cell [33]. The host cell can be
inhibiting neural crest differentiation in a BMP4-­ cleared of the vectors and reprogramming factor
dependent manner. Mechanistic studies revealed genes (i.e., Oct4, Sox2, Klf4, and c-Myc) by
a multi-targeted mechanotransductive process exploiting the cytoplasmic nature of SeV and the
involving mechanosensitive Smad phosphoryla- functional temperature sensitivity mutations
tion and nucleocytoplasmic shuttling, regulated introduced into the key viral proteins. The repro-
by rigidity-dependent Hippo/YAP activities and gramming efficiency may vary among different
actomyosin cytoskeleton integrity and contractil- cell types (~0.01–1%).
ity [28]. Viral-mediated introduction of transcription
A new method has been developed that turns factors is very inefficient. A more efficient
cells into stem cells faster and more efficiently by method was found to be the combination of lenti-
“squeezing” them using 3D microenvironment viruses and microRNAs (miRNAs) to reprogram
stiffness and density of the surrounding gel. The cells [34]. These small RNAs bind to mRNA and
technique can be applied to a large number of either inhibit translation or cause degradation of
cells to produce stem cells for medical purposes transcripts. miRNA clusters, including miR-290-
on an industrial scale [29]. 295 and miR-302-­ 367, have been shown to
enhance reprogramming of somatic cells into
3.1.3.4 iPSCs Generate Systems iPSCs.
iPSCs were first generated by the introduction of
the transcription factors Oct3/4, Sox2, Klf4, and miRNA System
c-Myc in cells maintained in culture conditions Another approach was the use of miRNA mimics
used for ESC [30]. Various introduction systems to enhance viral-mediated transduction of tran-
of these transcription factors have since been scription factors. miRNA mimics are double-­
used by other investigators. stranded modified RNAs that mimic mature
miRNAs. miRNA mimics do not require a vector;
Viral System they can be transfected directly into cells. The
Most iPSCs have been generated using retroviral combined use of transcription factors and miRNA
and lentiviral vectors to introduce transcription mimics produces more homogeneous iPSC clones
factors into cells. However, there are concerns [35]. An advantage of using miRNA mimics with
with using these viral vectors. Previous studies of transcription factors is that the transcription factor
retroviral infection of cells had suggested that c-Myc, an oncogene, is not required.
retroviruses are silenced in these cells. Silencing
is an epigenetic process that suppresses transcrip- Chemical Compound System
tion. However, it was demonstrated that silencing Several chemical compounds that modulate
was often incomplete and viral genes could still enzymes controlling epigenetic modifications
be expressed. A major consideration for using have been evaluated for increasing the efficiency
retroviruses to generate iPSCs is that the viruses of transduction by transcription factors. DNA
integrate into the host DNA. Depending on the methyltransferase and histone deacetylase
integration site, integration can have deleterious (HDAC) inhibitors were shown to potentiate the
effects on the cells, altering gene expression efficacy of transduction. The HDAC inhibitor
and increasing the risk of tumor formation. valproic acid was the most effective, increasing
3  Cell Technologies 63

reprogramming efficiency by 100-fold [36]. As 4. High transfection efficiency due to oriP/



with the miRNA mimics, the use of valproic acid EBNA-1-mediated nuclear import and reten-
eliminates the need to transduce with the onco- tion of vector DNA allows iPSC derivation in
gene c-Myc. a single transfection.
5. The episomal vectors are removed from the
Kinase Inhibitor System iPSCs without any further manipulation due
A number of other inhibitors of kinases, such as to the silencing of the viral promoter driving
the glycogen synthase kinase-3 (GSK3) inhibitor EBNA-1 expression and the loss of the epi-
CHIR99021 and the MEK inhibitor PD0325901, somes due to defects in vector synthesis and
and other enzymes that are in pathways involved partitioning.
in pluripotency have also been shown to enhance 6. Do not require the use of small molecules for
the efficiency of reprogramming [37]. reprogramming.

Plasmid System Lipofectamine System


Plasmid expression vectors have been used to Lipofectamine transfection was developed to
introduce transcription factors [38], but the effi- unleash the power of stem cells by providing a
ciency is low, and occasionally expression plas- highly efficient (40–70%), cost-effective, deliver
mids can integrate into genomic DNA. DNA into difficult-to-transfect stem cells, alter-
native method for electroporation. This advanced
Episome System lipid nanoparticle technology minimizes the
This system contains an optimized mixture of stress on cells caused by electroporation, simpli-
three episomal vectors with an oriP/EBNA-1 fies the reprogramming workflow, and enables
(Epstein-Barr nuclear antigen-1) backbone for advanced gene editing technologies.
delivering the reprogramming genes, Oct4, Sox2,
Lin28, L-Myc, and Klf4. High transfection effi- 3.1.3.5 Mesenchymal Stem Cells, MSCs
ciency due to oriP/EBNA-1-mediated nuclear MSCs are non-hematopoietic stem cells found
import and retention of vector DNA allows iPSC in most tissues of the body. The largest popula-
derivation in a single transfection. In addition, tions are in bone marrow and cord blood, with
silencing of the viral promoter driving EBNA-1 Wharton’s jelly, found in the umbilical cord,
expression and the loss of the episomes at a rate being a rich source. MSCs are heterogeneous,
of ~5% per cell cycle due to defects in vector have multilineage potential, and are capable of
synthesis and partitioning allow the removal of differentiating into multiple cell types includ-
episomal vectors from the iPSCs without any ing adipocytes, chondrocytes, osteoblasts, and
additional manipulation. cardiomyocytes.
Advantages of reprogramming with episome MSCs are the most commonly used cell type
are the following: in clinical trials for stem cell therapy.
Advantages of MSCs include: 1) they can be
1. As a transgene-free and viral-free reprogram- harvested from several tissues; 2) they have
ming system, it is a safe alternative to other multilineage potential; 3) subpopulations easily
reprogramming methods such as lentiviral isolated by cell sorting can be given; 4) they
delivery for all stages of iPSC research. can attenuate inflammatory responses and
2. It allows the reprogramming of a variety of secrete many bioactive molecules such as
somatic cell types and provides flexibility in growth factors and cytokines (including
somatic cell selection. ­angiogenic cytokines); 5) allogeneic (nonself)
3. It is optimized for feeder-free reprogram-
transplantation usually evokes a minimal
ming, enabling defined and feeder-free immune response; and 6) they may activate
reprogramming. local stem cells and repair mechanisms.
64 S.Y. Chun

Morphology Adipose tissue is becoming an important


MSC is derived from human bone marrow show- source of MSCs because adipocytes are easily
ing fibroblast-like morphology. MSCs contain a accessible and present in relatively high numbers
large, round nucleus with a prominent nucleolus, in the body. It has been estimated that 1 g of adi-
which is surrounded by finely dispersed chroma- pose tissue yields 5000 MSCs, whereas bone
tin particles, giving the nucleus a clear appear- marrow aspirate contains 100–1000 MSCs per
ance. The remainder of the cell body contains a ml [40].
small amount of Golgi apparatus, rough endo-
plasmic reticulum, mitochondria, and polyribo- Identification Markers
somes. The cells, which are long and thin, are A cell can be classified as an MSC if it shows
widely dispersed, and the adjacent extracellular plastic adherent properties under normal cul-
matrix is populated by a few reticular fibrils [39]. ture conditions and has a fibroblast-like mor-
phology. In fact, some argue that MSCs and
Cell Sources fibroblasts are functionally identical [40, 41].
MSCs were originally isolated from bone mar- Furthermore, MSCs can undergo osteogenic,
row stroma but were subsequently found to be adipogenic, and chondrogenic differentiation
present in most tissues of the body including cord ex vivo. The cultured MSCs also express on
blood, adipose tissue, skin, and periodontal liga- their surface CD44, CD73, CD90, and CD105
ments, which attach teeth to the jaw. while lacking the expression of CD11b, CD14,
The youngest, most primitive MSCs can be CD19, CD34, CD45, CD79a, and HLA-DR sur-
obtained from umbilical cord tissue, namely, face markers [42].
Wharton’s jelly and the umbilical cord blood.
However, MSCs are found in much higher con- Differentiation Capacity
centration in the Wharton’s jelly compared to The standard test to confirm multipotency is dif-
cord blood. The umbilical cord is easily obtained ferentiation of the cells into osteoblasts, adipo-
after a birth and is normally thrown away and cytes, and chondrocytes as well as myocytes and
poses no risk for collection. The cord MSCs neurons. MSCs have been seen to even differenti-
have more primitive properties than other adult ate into neuron-like cells [43], but there is linger-
MSCs obtained later in life, which might make ing doubt whether the MSC-derived neurons are
them a useful source of MSCs for clinical functional. The degree to which the culture will
applications. differentiate varies among individuals and how
Another rich source for mesenchymal stem differentiation is induced, e.g., chemical vs.
cells is the developing tooth bud of the mandibu- mechanical; and it is not clear whether this varia-
lar third molar. While considered multipotent, tion is due to a different amount of “true” pro-
they may prove to be pluripotent. They eventu- genitor cells in the culture or variable
ally form enamel, dentin, blood vessels, dental differentiation capacities of individuals’ progeni-
pulp, and nervous tissues, a minimum of 24 other tors. The capacity of cells to proliferate and dif-
different unique end organs. Because of ease in ferentiate is known to decrease with the age of
collection at 8–10 years of age before calcifica- the donor, as well as the time in culture. Likewise,
tion and minimal to no morbidity, they probably whether this is due to a decrease in the number of
constitute a major source for research and multi- MSCs or a change to the existing MSCs is not
ple therapies. known.
Amniotic fluid also has been shown to be a
rich source of stem cells. As many as 1 in 100 Immunomodulatory Effects
cells collected during amniocentesis have been Numerous studies have demonstrated that human
shown to be a pluripotent mesenchymal stem cell MSCs avoid allorecognition, interfere with den-
(see detailed information at Sect. 3.1.3.6). dritic cell and T-cell function, and generate a local
3  Cell Technologies 65

immunosuppressive microenvironment by secret- have few of the problems of allogeneic stem


ing cytokines [44]. It has also been shown that the cells, such as immune incompatibility or the risk
immunomodulatory function of human MSC is of communicable disease transmission. There
enhanced when the cells are exposed to an inflam- have been several clinical trials approved for the
matory environment characterized by the presence therapeutic use of cord blood, including the treat-
of elevated local interferon-gamma levels [45]. ment of pediatric stroke and cerebral palsy.
Cord blood contains a large number of stem
Homogenous Isolation cells, therefore, substantial numbers of cells
The majority of modern culture techniques still can be easily obtained without many passages.
take a colony-forming unit-fibroblast (CFU-F) This has three advantages: 1) the decreased
approach, where raw unpurified bone marrow or time in culture and number of cell divisions
Ficoll-purified bone marrow mononuclear cell is reduces the potential for chromosomal changes;
plated directly into cell culture plates or flasks. 2) cord blood stem cells, although they can give
Mesenchymal stem cells, but not red blood cells rise to all three germ layers, do not seem to be
or hematopoietic progenitors, are adherent to tis- tumorigenic; 3) the ability to rapidly generate
sue culture plastic within 24–48 h. large numbers of cells affords the use of high
Other flow cytometry-based methods allow the numbers of cells in therapies, which may
sorting of bone marrow cells for specific surface increase efficacy and reduce the waiting time
markers, such as STRO-1 [46]. STRO-1+ cells are for treatment.
generally more homogenous and have higher rates
of adherence and higher rates of proliferation, but Amniotic Stem Cells [48]
the exact differences between STRO-1+ cells and Amniotic stem cells are the mixture of stem cells
MSCs are not clear. Methods of immunodepletion that can be obtained from the amniotic fluid as
using such techniques as MACS have also been well as the amniotic membrane. They can develop
used in the negative selection of MSCs. into various tissue types including skin, cartilage,
The supplementation of basal media with fetal cardiac tissue, nerves, muscle, and bone. The cells
bovine serum or human platelet lysate is common also have potential medical applications, espe-
in MSC culture. Prior to the use of platelet lysates cially in organ regeneration (Figs. 3.1 and 3.2).
for MSC culture, the pathogen inactivation pro- The stem cells are usually extracted from the
cess is recommended to prevent pathogen trans- amniotic sac by amniocentesis which occurs
mission [47]. without harming the embryos. The use of amni-

3.1.3.6 Cord Blood and Amniotic


Fluid-Derived Stem Cells
Cord blood and amniotic fluid contain several
different types of stem cells, with the predomi-
nant cell type being HSC. The various cell types
have different potencies, ranging from ESC-like
cells to multipotent stem cells.

Cord Blood Stem Cells


Cord blood (and placental blood) has several
benefits for clinical therapy. The tissue source is
freely and widely available, with no extra risk to
the donor and no ethical issues. Banking of cord
blood can provide future stem cells for treatment
of the donor later in life. These stem cells would Fig. 3.1  Amniotic stem cells
66 S.Y. Chun

Fig. 3.2  Differentiation potency of amniotic fluid stem cells into muscle, neuron, endothelium, bone, fat, and
chondrocyte

otic fluid stem cells is therefore generally consid- because cells lacking c-Kit also display differen-
ered to lack the ethical problems associated with tiation potential. Culture conditions may also be
the use of cells from embryos. The majority of adjusted to promote the growth of a particular
stem cells present in the amniotic fluid share cell type.
many characteristics, which suggests they may Amniotic fluid highly contained MSCs and
have a common origin. several techniques have been described for
This cell was confirmed that the amniotic fluid MSCs isolation. They usually involve the
contains a heterogeneous mixture of multipotent removal of amniotic fluid by amniocentesis,
cells after it was demonstrated that they were and their distinction from other cells may be
able to differentiate into cells from all three germ based on their morphology or other character-
layers but they could not form teratomas follow- istics. Human leukocyte antigen testing has
ing implantation into immunodeficient mice. been utilized to confirm that the MSCs are
This characteristic differentiates them from from the fetus and not from the mother.
embryonic stem cells but indicates similarities Comparison of amniotic fluid-­derived MSCs to
with adult stem cells. However, fetal stem cells bone marrow-derived ones showed that the for-
attained from the amniotic fluid are more stable mer has a higher expansion potential in cul-
and more plastic than their adult counterparts ture. However, the cultured amniotic
making it easier for them to be reprogrammed to fluid-derived MSCs have a similar phenotype
a pluripotent state. to both adult bone marrow-derived MSCs and
A variety of techniques has been developed MSCs originating from second-trimester fetal
for the isolation and culturing of amniotic stem tissue(Fig. 3.3).
cells. One of the more common isolation meth- As like mesenchymal stem cells, embryonic-­
ods involves the removal of amniotic fluid by like stem cells also are abundant in the amniotic
amniocentesis. The cells are then extracted from fluid. Embryonic-like stem cells were originally
the fluid based on the presence of c-Kit. Several identified using markers common to embryonic
variations of this method exist. There is some stem cells such as nuclear Oct4, CD34, vimentin,
debate whether c-Kit is a suitable marker to dis- alkaline phosphatase, stem cell factor, and c-Kit.
tinguish amniotic stem cells from other cell types However, these markers were not n­ecessarily
3  Cell Technologies 67

concomitantly expressed. In addition, all of these into muscle, adipogenic, osteogenic, nephro-
markers can occur on their own or in some com- genic, neural, and endothelial cells, this did not
bination in other types of cells. The pluripotency necessarily occur from a homogenous population
of these embryonic-like stem cells remains to be of undifferentiated cells. Evidence in favor of
fully established. Although those cells which their embryonic stem cell nature is the cells’ abil-
expressed the markers were able to differentiate ity to produce clones (Figs. 3.4 and 3.5).

control 2.05% CD44 97.89% CD45 4.34%


140

140
140
Counts

Counts
Counts
MI MI MI
0

0
100 101 102 103 104 100 101 102 103 104 100 101 102 103 104
FL2-H FL2-H FL2-H

CD73 99.78% CD90 99.71% CD105 99.52% C-kit 68.15%


140

140

140
140
Counts

Counts

Counts

Counts
MI MI MI MI
0

0
100 101 102 103 104 100 101 102 103 104 100 101 102 103 104 100 101 102 103 104
FL2-H FL2-H FL2-H FL2-H
SSEA4 26.59% HLA ABC 99.31% HLA DR 3.58%
140

140

140
Counts

Counts

Counts

MI MI
0

100 101 102 103 104 100 101 102 103 104
FL2-H 100 101 102 103 104
FL2-H FL2-H

Fig. 3.3  Mesenchymal cell markers expression analysis of amniotic stem cells by FACS

p1 p2 p3 p4 p5 p6 p7 p8 p9 p10

GAPDH

Oct-4

FGF-5

SCF

Vimentin

Fig. 3.4 Pluripotency CK-18


marker expression of
amniotic fluid stem cell GATA-4
to the passages
68 S.Y. Chun

AFSC BMSC CBSC

Oct-4

SCF

vimentin

FGF-5

CK18

GAPDH

Passage 6 7 8 9 10 6 7 8 9 10 6 7 8 9 10

Fig. 3.5  Comparison of pluripotency marker expression among amniotic fluid, bone marrow, and umbilical cord-­
derived stem cell

3.2 Applications 9. Plate to T175 flask, and add complete MEF


medium to make up a final volume of 30 mL.
3.2.1 Stem Cell Culture 10. The dissociated MEFs will attach to the flask
and begin to divide overnight.
3.2.1.1 Human Embryonic Stem Cell 11. Replace the medium the next day with an
Culture Protocol equal volume of fresh complete MEF
medium.
Isolation of Primary Mouse Embryo 12. 3–4 days after, rinse the cells with Ca2+/

Fibroblasts Mg2+ free PBS.
1. Dissect out the uterus of mouse (pregnant 13. Add 3 mL of 0.05% trypsin/EDTA solution
day 13.5, ICR) with sterile forceps and scis- per flask. Inactivate the trypsin solution by
sors, and place it into a Petri dish with PBS. adding 5 mL complete MEF medium.
2. Isolate the embryos and transfer into a sec- 14. Count the number of viable cells using try-
ond Petri dish with PBS. pan blue exclusion and a hemocytometer.
3. Remove the heads, livers, intestines, heart, Viability should be between 90 and 95%.
and all viscera. 15. Centrifuge the cell suspension at room tem-
4. Transfer the carcasses to a fresh Petri dish, perature for 4 min at 200 g.
and mince with a blade. 16. Aspirate medium and resuspend the cells in
5. Add 5.0 mL of 0.25% trypsin/EDTA per ten complete MEF medium (freezing concentration,
fetuses, and triturate through a 10 mL pipette. 2.4 × 107/mL). Add an equal volume of fetal
6. Transfer the solution to a 10 mL syringe with bovine serum with 20% DMSO to get 1x final
18 G needle, slowly push the solution stocks. Dispense 1 mL per cryovial and freeze.
through the needle (two times), and collect in
a 50 mL conical tube. Preparation of MEF-Conditioned Medium,
7. Incubate for 15 min at 37°C, triturating three MEF-CM
or four times using a 10 mL pipette. 1. Plate 4 × 106 mitomycin C-treated MEFs in a
8. Add an equal volume of complete MEF T75 flask coated with 0.5% gelatin, in com-
medium. plete MEF medium.
3  Cell Technologies 69

2. The next day, replace the MEF medium with medium without antibiotics. Seed cells at
37.5 ml 20% KSR hESC medium (containing 4 × 106 cells (10 ml) per 100 mm dish, and
4 ng/ml bFGF), and incubate for 24 h at 37°C, incubate overnight at 37°C, 5% CO2.
5% CO2. 2. Transfection to 293FT cells: Dilute 9 μg of
3. Collect MEF-CM from the flasks after 24 h Virapower packaging mix (pLP1, pLP2, and
and 0.22 μM filter sterilize. MEF-CM can be pLP/VSVG mixture) and 3 μg of pLenti6/
used fresh or can be frozen. Collect MEF-CM UbC encoding the mouse Slc7a1 gene in
for up to 7 days using this procedure. 1.5 ml of OPTI-MEM I, and mix gently. In a
separate tube, dilute 36 μl of Lipofectamine
2000 in 1.5 ml of OPTI-MEM I. Mix gently.
Microdissection Passaging of hESCs After 5 min incubation, combine the diluted
1. Warm MEF-CM to 37°C in water bath, and DNA with the diluted Lipofectamine 2000.
add freshly thawed 1 M ß-mercaptoethanol to During 20 min incubation, remove the
a final concentration of 0.1 mM. medium from 293FT dishes, and add 9 ml of
2. Assess the undifferentiated colonies from dif- fresh medium to each dish. Then add 3 ml of
ferentiated colonies. Use a fine pointed tool the DNA-Lipofectamine 2000 complexes in
(fire drawn Pasteur pipette needles), and score each dish. Twenty-four hours after transfec-
four to five times across a colony and perpen- tion, aspirate the medium containing the
dicular to that another four to five times. This transfection cocktail, and add 10 ml of fresh
creates a grid that cuts the colony into pieces. FP medium.
3. Use a P1000 pipette tip to lift the colony
3. Collection of virus-containing supernatant:

pieces, and put them into MEF-CM. 48 h after transfection, collect the supernatant
of the 293FT culture with a 10 ml disposable
3.2.1.2 Human-Induced Pluripotent syringe, and then filtrate it with a 0.45 μm
Stem Cell Generation Protocol pore size cellulose acetate filter.

Preparation of Fibroblasts Lentiviral Infection


1. Disinfect skin surface three times with povi- 1. Seeding fibroblasts: Adjust the concentration
done iodine, and administer lidocaine. to 8 × 104 cells/ml by adding appropriate vol-
2. Push a disposable biopsy punch against the ume of FP medium. Seed the cells at 8 × 105
skin, deep enough to reach the fatty layer of cells (10 ml of cell suspension) per 100 mm
subcutaneous tissue. dish. Incubate overnight at 37°C, 5% CO2.
3. Put the tissue into a 100 mm dish containing 2. Transduction of fibroblasts: Replace medium
FP medium (DMEM containing 10% FBS with 10 ml/dish of the virus-containing super-
and 50 units and 50 mg/ml penicillin and natant, supplemented with 4 μg/ml Polybrene.
streptomycin), and cut it into 1 mm pieces. 3. Twenty-four hours after transduction, aspirate
4. Transfer them to a 35 mm dish, put the cover- off the virus-containing medium, wash cells,
slip onto the tissues, and then add 2 ml of FP and add 10 ml of fresh FP medium.
medium.
5. Incubate at 37°C, 5% CO2. Leave the dish Preparation of SNL Feeder Cells
untouched for a week, and later change the 1. Thawing SNL cells: Suspend the cells with
medium twice a week. 10 ml of SNL medium, and transfer to a
6. When cells cover 30–50% of the dish, it’s gelatin-­
coated 100 mm dish. Incubate the
time to transfer the cells to a larger dish. cells in a 37°C, 5% CO2 incubator, until the
cells become 80–90% confluent.
Lentivirus Production 2. Mitomycin C-inactivation of SNL cells: Add
1. Passaging 293FT cells: Adjust the concentra- 0.3 ml of 0.4 mg/ml mitomycin C solution
tion to 4 × 105 cells per milliliter with 293FT directly to the culture medium of SNL dish,
70 S.Y. Chun

and incubate 2.25 h at 37°C, 5% CO2. Then a 15 ml conical tube. Add 5 μl of 8 mg/ml
wash the cells twice with 10 ml of PBS. Add Polybrene solution into the filtrated virus-­
0.5 ml of 0.25% trypsin/1 mM EDTA, and let containing medium. Make a mixture of equal
sit for 1 min at room temperature. Neutralize parts of the medium containing OCT3/4,
the trypsin by adding 5 ml of SNL medium, SOX2, KLF4, and c-MYC retroviruses.
and seed the cells at 1.5 × 106 cells per 100 mm Aspirate the medium from fibroblast dishes,
dish. and add 10 ml per dish of the Polybrene/
virus-containing medium. Incubate the cells
Preparation of Plat-E Cells for 4 h.
1. Thawing Plat-E cells: Suspend the cells with 6. Day 5–10: After 4 h–overnight culture, aspi-
10 ml of FP medium, and transfer to a 100 mm rate the medium from the transduced fibro-
dish. Incubate the cells in a 37°C, 5% CO2 blasts, and add 10 ml per dish of fresh FP
incubator. The next day, replace the medium medium.
with 1 μg/ml of puromycin and 10 μg/ml of 7. Day 11 Replating transduced fibroblasts
blasticidin S and incubate until 80–90% onto mitomycin C-treated SNL feeder:
confluent. Aspirate the culture medium and wash with
2. Passage of Plat-E cells: Seed cells on new 10 ml per dish of PBS. Add 1 ml per dish of
100 mm dishes at 1:4–1:6 dilution. Cells 0.05% trypsin/0.53 mM EDTA, add 9 ml per
should become confluent within 2–3 days. dish of FP medium, and suspend the cells.
Count cell numbers, and adjust the concen-
Generation of iPS Cells tration to 5 × 103 or 5 × 104 cells/ml.
1. Day 1 Plat-E preparation: Count cell number 8. Day 12~: Colonies should become visible
and adjust the concentration to 3.6 × 105 2–3 weeks after the retroviral infection. They
cells/ml with FP medium. can be picked up at around day 30.
2. Day 2 Retrovirus production (transfection 9. Picking up the iPS colonies: Pick colonies
into Plat-E cells): Transfer 0.3 ml of OPTI-­ using a P2 or P10 Pipetman L, transfer it into
MEM I into a 1.5 ml tube. Deliver 27 μl of the 96-well plate with 20 μl ES medium, and
transfection reagent into the prepared tube, add 180 μl. Pipet up and down to break up
and incubate for 5 min. Add 9 μg of pMXs the colony to small clumps, transfer cell sus-
plasmid DNA (respectively encoding Oct3/4, pension into 24-well plates with mitomycin
Sox2, Klf4, and c-Myc) drop by drop, and C-treated SNL feeder cells, add 300 μl per
incubate for 15 min. Add the DNA and incu- well of human ES medium, and incubate
bate overnight. until the cells reach 80–90% confluence.
3. Day 3 Retrovirus production: Aspirate the 10. Passaging of iPS cells: After washing with
transfection reagent-containing medium and PBS, add 0.1 ml per well of CTK solution
add 10 ml of fresh FP medium. and incubate at 37°C for 2–5 min. Washing
4. Preparation of fibroblasts: When human with PBS, add 0.5 ml of human ES medium
fibroblasts expressing mouse Slc7a1 gene and suspend the cells to small clumps of
reach to 80–90% confluence, aspirate 20–30 cells by pipetting. Transfer to a 6-well
medium and wash once with 10 ml of plates with mitomycin C-treated SNL feeder
PBS. Add 1 ml per dish of 0.05% tryp- cells, add 1.5 ml of human ES medium, and
sin/0.53 mM EDTA, add 9 ml of the FP incubate until cells become 80–90% conflu-
medium per plate, and suspend the cells to a ent in 6-well plates.
single cell. Count cell numbers to 8 × 104 11. Expansion of iPS cells: After washing with
cells/ml with FP medium. PBS, add 0.5 ml per well of CTK solution
5. Day 4 Retroviral infection: 48 h post-­ and incubate at 37°C for 2–5 min. Washing
transfection, collect the medium from each with PBS, add 2 ml of human ES medium.
Plat-E dish, filter it through a 0.45 μm pore Detach iPS cells by using cell scraper and
size cellulose acetate filter, and transfer into break up the colonies to small clumps by
3  Cell Technologies 71

pipetting. Transfer the cell suspension to a 7. Recover the cells and incubate them for 5 min
60 mm dish with SNL feeder. Add 2 ml at 37°C in a 5% CO2 with 0.125% trypsin/
human ES medium, and incubate until cells EDTA solution. Then, collect cells and trans-
reach 80–90% confluence. fer to a 15 ml conical tube and centrifuge at
250 g for 5 min. Discard the supernatant and
3.2.1.3 Mesenchymal Stem Cell Culture resuspend the cellular pellet in 10 ml of HG-­
Protocol DMEM. Culture cell suspension in two 75 cm2
culture flasks (5 ml in each flask), and main-
Mesenchymal Stem Cell Isolation tain the culture until 90% confluence. Repeat
from Human Amniotic Membrane steps until the 9th passage or subculture.
1. Prepare phosphate-buffered saline solution
(PBS), Dulbecco’s Modified Eagle’s Mesenchymal Stem Cell Isolation
Medium, and high glucose (HG-DMEM), from Adipose Tissue
and add 10% fetal bovine serum (FBS), 1% 1. Wash ~250 mL of fat 3–5 times with PBS for
antibiotic-­antimycotic solution, and a colla- 5 min in each wash, add collagenase, and
genase II solution at 100 U/ml in HG-DMEM incubate for 1–4 h at 37°C on a shaker.
without FBS or antibiotic-antimycotic 2. Add 10% FBS to neutralize collagenase, and
solution. centrifuge at 800 g for 10 min.
2. Sterilize the surgical instruments by autoclav- 3. Aspirate floating adipocytes, lipids, and liquid,
ing (standard scissors, simple dissecting for- leaving stromal vascular fraction (SVF) pellet,
ceps, fine point and toothed forceps, and a resuspend in 160 mM NH4Cl, and incubate for
scalpel handle). 10 min at room temperature. Centrifuge at
3. Hold the umbilical cord, detach the amniotic 400 g for 10 min at room temperatures.
membrane (AM) to obtain a fragment of 4. Layer cells on Percoll gradient, and centrifuge
approximately 5 cm2, and remove residual at 1000 g for 30 min at room temperature.
blood with three washes of 5 ml of PBS. Make Wash cells twice with PBS and centrifuge at
a small cut in the AM with the scalpel to gen- 400 g for 10 min each wash. Resuspend cell
erate fragments of approximately 0.5 cm2. pellet in PBS and filter cells through 100 μM
4. Incubate the AM with 5 ml of 0.125% tryp- nylon mesh. Pass cells through 400 μM mesh.
sin/0.5 mM EDTA solution at 37°C for 30 min Centrifuge at 400 g for 10 min.
in a 50 ml conical tube at 37°C in a 5% CO2. 5. Resuspend cell pellet in 40% FBS/DMEM

Centrifuge at 250 g and 25°C for 5 min and and plate, and incubate at 37°C, 5% CO2 incu-
then remove the trypsin solution. bator overnight.
5. Add 15 ml of collagenase type II solution and
incubate for 2 h at 37°C. Immediately after Mesenchymal Stem Cell Isolation
digestion, add 15 ml of PBS and centrifuge at from Cord Blood
250 g and 25°C for 10 min. Discard the super- 1. Dilute cord blood with RPMI Medium 1640
natant and wash the cellular pellet with 15 ml with a 3:1 ratio (three-part cord blood to one
of PBS solution and centrifuge at 250 g and part RPMI).
25°C for 15 min. Discard the supernatant. 2. Using Ficoll-Paque, isolate mononuclear cells
Resuspend the cellular pellet in 10 ml of HG-­ (MNCs) by density gradient centrifugation at
DMEM by shaking gently. 400 g for 30 min at room temperature.
6. Seed 1 × 104 cells/cm2 in culture flask, add 3. Transfer MNCs to new centrifuge tube and
HG-DMEM, and incubate the cells at 37°C in add PBS with a 1:3 ratio (1 part MNCs to 3
a 5% CO2. Eliminate the nonadherent cell by parts PBS). Centrifuge at 400 g for 10 min at
changing the culture medium after 5–7 days. room temperature.
Change the culture medium every 3 days for 4. Remove supernatant and resuspend cells by
an additional 7–10 days to obtain a cellular adding culture medium and plate. Incubate at
confluence of 90%. 37°C, 5% CO2 incubator overnight.
72 S.Y. Chun

Mesenchymal Stem Cell Isolation 2. The medium was replaced twice a week and
from Amniotic Fluid nonadherent cells were discarded.
1. Collect amniotic fluid from 8 s-trimester
3. After 2 weeks one colony of adherent

pregnancies. fibroblast-­like cells was noticed in one of the
2. Four the AF samples were centrifuge 4 mL of seeded specimens. When the colony reached
amniotic fluid for 10 min at 300 g. the approximate size of 5 cm2, cells were
3. Pellets were resuspended and cultured in
detached and seeded in a new flask in GM.
DMEM with low glucose supplemented with 4. Cells reached confluence after 10 days.

15% fetal bovine serum and antibiotics. Following first confluence, cells were pas-
4. Culture flasks were incubated at 37°C, 5% saged regularly.
CO2.
5. Medium was changed every 3–4 days until
day 14. 3.2.2 S
 tem Cell Characteristic
6. Treatment with trypsin/EDTA was applied to Analysis Methods
detach the cells from the bottom of the flasks
(passage 0). Cell number and vitality were The characterization of stem cell has two pur-
determined and analyzed by flow cytometry poses: monitoring the genomic integrity of the
using specific CD73+/CD44+/CD45 cell sur- cells and tracking the expression of proteins asso-
face markers. ciated with pluripotency. Genomic analysis is
necessary to ensure stem cells maintained in cul-
Mesenchymal Stem Cell Isolation ture have not undergone chromosomal changes
from Mouse Bone Marrow through chromosomal loss or duplication or
1. Prepare six 2 weeks-old BALB/c mice; MSCs changes in their epigenetic profiles. Proteomic
were collected from femoral and tibial bone analysis ensures that the cells are expressing the
marrow of three mice by inserting a 26-gauge factors necessary to maintain pluripotency.
syringe at bone cavity, washing it with 10 ml Confirmation of the differentiated state by
of Knockout DMEM. analyzing key genetic and protein markers
2. After centrifuging at 1200 rpm for 10 min, ensures identification and propagation of the
bone marrow cells were resuspended in 1 ml correct cell type. The general analysis methods
at the corresponding culture media for cell for stem cells identification are karyotyping,
counting and cell feasibility verification single-­nucleotide polymorphism (SNP) analy-
3. Cultures were maintained at 37°C with 5% sis, ­epigenetic profiling, flow cytometry, immu-
CO2. nocytochemistry, RT-qPCR, western blotting,
4. After 72 h of culture, the medium was
biomarker analysis, and teratoma formation
refreshed in an interval of 3–4 days. in vivo.

Mesenchymal Stem Cells Isolated 3.2.2.1 Culture-Related Factors


from Peripheral Blood Culture-Related Factors are media composition,
1. The peripheral blood-derived MSCs (PB-­
cell density, feeder cell type/density, growth fac-
MSCs) were obtained from mononuclear cells tors/additives, feeder-free culture, passage
by density gradient centrifugation, plated at a method, number of passages, freezing and thaw-
density of 4 × 105/cm2 in 25 cm2 flasks in ing protocols, and microbial contamination
growth medium (GM) containing 10% fetal
bovine serum (FBS) and 100 units/ml penicil- Possible Changes
lin/streptomycin (pen/strep) in high-glucose The types of possible changes are chromosomal,
Dulbecco’s Modified Eagle’s Medium and phenotype/morphology, differentiation, pluripo-
cultured in a humidified atmosphere at 37°C tency loss, epigenetic changes, tumorigenesis,
with 5% CO2. and loss of self-renewal ability.
3  Cell Technologies 73

A number of techniques are used for genetic Giemsa staining, known as G-band karyotyping
characterization of stem cells: karyotyping, fluo- or G-banding; other methods include R-banding
rescence in situ hybridization (FISH), compara- (reverse Giemsa staining), C-banding (constitu-
tive genomic hybridization, single-nucleotide tive heterochromatin staining), Q-banding (quin-
polymorphism (SNP) analysis, and epigenetic acrine staining), and T-banding (telomeric
profiling. Some types of stem cells, particularly staining). Changes in banding patterns are used
ESCs and iPSCs, are more prone to being geneti- to identify abnormalities.
cally unstable and should be observed frequently
for chromosomal changes. Fluorescence In Situ Hybridization (FISH)
Stem cells express both unique and specific In FISH, fluorescently labeled overlapping DNA
combinations of transcription factors, cell surface fragments are hybridized to interphase or meta-
proteins, and cytoplasmic proteins. Techniques phase chromosomes. The chromosomes are for-
used for stem cell analysis and characterization malin fixed onto a surface and chemically
include flow cytometry, array-based analysis of denatured. Fluorescence microscopy is used to
the transcriptome, immunocytochemistry, west- detect and analyze the chromosomes. In fast
ern blots, and biomarker analysis. FISH, chromosomes are denatured with high
Different classes and types of stem cells are temperatures instead of chemically.
characterized by different combinations of markers. Spectral karyotyping (SKY) and multiplex
In addition to confirming that stem cell lines are fluorescence in situ hybridization (M-FISH) are
stable, markers can be used for screening during the commonly used forms of FISH for detection of
reprogramming of somatic cells into iPSCs or to changes. Both are hybridization-based combina-
follow the progression of stem cell differentiation. torial techniques using fluorescently labeled DNA
probes. Each chromosome is labeled with a differ-
Colony-Forming Unit-Fibroblastic (CFU-F) ent combination of fluorophores specific for that
CFU-F assays were performed by plating MSCs chromosome, giving a unique spectral signature
(P3 to P6) in 6-well plates at 10, 50, and 100 for each chromosome. Chromosomes are identi-
cells/well in GM, in two replicas. After 14 days fied by their unique signatures, and then the out-
under standard culture conditions, the cells were put is pseudo-colored to aid in visualization. The
fixed with ice-cold methanol and stained with difference between SKY and M-FISH is how the
0.3% crystal violet. The number of visible colo- fluorescent signals are collected and processed.
nies (more than 50 cells) was counted. These methods can both detect translocations
within and between chromosomes more accu-
Karyotyping rately than traditional karyotyping; [50] however,
Karyotyping is the examination of cellular chro- they cannot detect inversions or duplications and
mosome number and morphology. Differences in deletions of less than approximately 5 Mb.
appearance include size, position of centromeres, The main disadvantage of FISH for general
and changes in banding patterns. Stem cells in screening of stem cells is that probes are required
culture can accumulate changes including chro- for the length of all the chromosomes. FISH is
mosomal abnormalities. Alterations in the most commonly used for confirmation of the find-
genome can lead to changes in gene expression ings of other techniques using probes for a specific
and cellular functions of stem cells [49] and region of a chromosome. Deletions and insertions
increase the risk of the stem cells being tumori- greater than ~20 kb are detected. Fast Fish is usu-
genic. Thus, it is crucial to monitor a culture for ally used for detecting specific aneuploidies that
the any chromosomal abnormalities, particularly are commonly found in iPSCs and ESCs.
in stem cells intended for therapeutic use.
Traditional karyotyping uses dye to stain the Comparative Genomic Hybridization
chromosomes of a metaphase cell in distinct Comparative genomic hybridization can both
banding patterns. The most common method is determine chromosomal ploidy and detect copy
74 S.Y. Chun

number variations (CNVs), which are variable somes is lost, this results in LOH. Cultured hESC
numbers of copies of segments of DNA. In this have been demonstrated to lose heterozygosity
technique, DNA samples from the test stem cell with passage. LOH can be associated with an
population and from a reference population are increase in tumorigenic potential.
each labeled with different probes. When this Aneuploidy, unbalanced translocations, dele-
technique was first developed, reference and test tions, and duplications can be detected with SNP
DNA were denatured then hybridized to meta- arrays. Inversions are not detected and low levels
phase chromosomes. Using microscopy and fluo- of mosaicism may confound interpretation. As
rescence measurements, the chromosomes were with comparative genomic hybridization, due to
scanned for any differences in fluorescent signals variability between individuals, choice of refer-
between the test and reference samples [51]. This ence material is important.
technique detects and locates CNVs without
requiring any knowledge of the particular Epigenetic Profiling
sequence. A stem cell’s pluripotency is dictated to a large
The disadvantage of using whole metaphase extent by its epigenetic profile. DNA methylation
chromosomes is that the technique is not much and histone modification regulate the accessibil-
more sensitive than G-banding. The adoption of ity of DNA to the transcriptional machinery and
techniques using microarrays with genomic thus regulate gene expression. Analysis of DNA
clones has increased detection sensitivity and methylation patterns is performed by treating
efficiency. Comparative genomic hybridization DNA with bisulfite under controlled conditions
does not detect balanced translocations, inver- such that all cytosines are converted to uracils
sions, or small gains or losses. Both mosaicism in while leaving methylated cytosines unchanged.
a stem cell population and the presence of repeti- After conversion, the DNA can be analyzed for
tive regions can complicate interpretation of global methylation patterns using a chip array,
results when using arrays. Due to the variability PCR, or DNA sequencing. Chromatin immuno-
in some chromosomal regions among individu- precipitation (ChIP) analyzes patterns of histone
als, the preferred reference is either an early pas- modifications by cross-linking the DNA to his-
sage of the stem cells or another DNA sample tones. Cross-linked chromatin is then sheared
extracted from the source of the stem cells. and purified using antibodies against a specific
protein or histone modification. Analysis of the
SNP Analysis cross-linked regions can be performed using real-­
Single-nucleotide polymorphisms (SNPs), single time qPCR, microarrays, or DNA sequencing.
base-pair mutations within a region of DNA, are Quantitative assessment of chromatin struc-
the most common type of genetic variation. SNPs ture in cultured cells can also be performed using
may accumulate in a stem cell population over specific epigenetics tools. Chromatin is digested
time and can lead to phenotypic changes that in the presence or absence of nuclease, and then
influence survival or growth. These genetic the genomic DNA is purified and quantified.
changes may result in unwanted outcomes such Chromatin structure is assessed via real-time
as loss of pluripotency or gain of tumorigenicity. PCR, by comparing results against an epigeneti-
SNP genotyping with high-density oligonucle- cally silenced gene to discriminate open, actively
otide arrays can specifically identify a stem cell transcribed chromatin regions from closed, tran-
line’s origin and monitor its genomic integrity. scriptionally silent regions. This quantifies the
SNP arrays are used for detecting loss of het- impact of epigenetic events such as DNA meth-
erozygosity (LOH). Normally for a chromosome ylation and histone modification on gene expres-
pair, since the chromosomes come from two par- sion regulation through chromatin state changes.
ents, there are differences between the sequences Epigenetic profiling has shown that iPSCs can
of the two chromosomes including many SNPs. retain some epigenetic memory. As cells differ-
When one copy of a region in a pair of chromo- entiate during development, there are epigenetic
3  Cell Technologies 75

changes accompanying the changes in gene particular proteins, information can be obtained
expression patterns. When a somatic cell is repro- about relative expression levels and the subcel-
grammed to become an iPSC, it has been lular localization of the target proteins.
observed in some iPSC cell lines that not all the Immunohistochemistry is also used to visualize
epigenetic changes that occurred during differen- stem cells in tissues, including the engraftment of
tiation are reversed during dedifferentiation. stem cells after transplantation.
In animal models, stem cells can be trans-
Flow Cytometry duced to express a protein that can be easily
Flow cytometry is a commonly used technique tracked. The visualization of a protein that is not
used for the analysis of stem cells. Fluorescently expressed in a given tissue or organism permits
tagged monoclonal antibodies can be used to dis- discrimination between transplanted stem cells
tinguish specific cell populations, and cell sorting and any resident stem cells. Green fluorescent
can be used to physically separate different stem protein (GFP) can be tracked either directly by
cell populations. For example, when a population green fluorescence.
of somatic cells is being reprogrammed, iPSCs
can be separated from cells either not repro- RT-PCR, RT-qPCR, and Digital PCR
grammed or not fully reprogrammed. Another Reverse transcription quantitative PCR
common use of cell sorting is during mainte- (RT-qPCR) and RT-digital PCR provide rapid,
nance and expansion of stem cell culture. The sensitive, and quantitative methods for monitor-
development of mosaic populations is common ing the gene expression profile of any cell popu-
in stem cells stocks. Sorting can be used to lation. The levels of transcription factors such as
remove unwanted cell types and retain homoge- Oct4, NANOG, SOX2, and other synergistic fac-
neity of the population. tors determine whether pluripotency is main-
There are a large number of well-­characterized tained. Multiplexed RT-qPCR assays and panels
cell surface markers that can be used both to provide an efficient way to screen and quantify
determine cellular phenotypes and to separate transcription factors, kinases, and other mole-
mixed populations. Additionally, stem cells cules involved in both maintaining pluripotency
express a number of cell surface proteins that are and in differentiation. The ability to do multiplex
used as markers of pluripotency and lineage. For transcriptome analysis increases the sensitivity of
example, the glycolipid antigens SSEA-3 and determining the status of cells.
SSEA-4 and the keratin sulfate antigens TRA- Digital PCR can increase the accuracy of mea-
1-­60 and TRA-1-81 are commonly used antigens surements with lower amounts of input nucleic
for identifying and sorting ESC populations [52]. acid. Droplet digital PCR allows for absolute
Fixed stem cells can also be sorted based on the quantification of transcripts and detection of rare
expression of the transcription factors such as sequences. In addition to being useful for tran-
Oct3/4 and NANOG. As stem cells differentiate, scriptome analysis, digital PCR can accurately
the loss of these markers and expression of new detect CNVs and SNPs.
markers can be used to track the lineages and
level of differentiation of the population. Western Blotting
Western blotting is useful for determining the
Immunocytochemistry success of transfection experiments. When a
and Immunohistochemistry gene is either introduced into the cell or knocked
Immunocytochemistry is predominantly used to down using antisense RNA or RNAi molecules,
confirm the presence or absence of protein detection and quantitation of the presence and
expression in stem cells. Cells are incubated with level of protein expression can be analyzed by
antibodies targeting specific proteins prior to flu- western blotting. Importantly, western blots can
orescent imaging. In addition to providing infor- be used to study the effects of transfection on
mation about the numbers of cells expressing downstream protein expression.
76 S.Y. Chun

Biomarker Analysis s­ ystems biology approach to developmental biol-


Biological processes can be controlled by protein ogy emphasizes the importance of investigating
interactions, relative protein levels, and post- how developmental mechanisms interact to pro-
translational protein modification. Not only can duce predictable patterns.
the presence or absence of proteins or signaling A few conserved types of molecular processes
peptides be assessed, but relative levels can be are often involved in the cellular mechanisms that
determined. This technique can be used to evalu- control these switches. The major types of molec-
ate pluripotency, lineages, and differentiation. ular processes that control cellular differentiation
The ability to multiplex allows for fine analysis involve cell signaling. The signal molecules that
of stem cells and differentiation pathways. convey information from cell to cell during the
Biological reactions and signaling networks can control of cellular differentiation are called
be monitored in real time. The use of bead-based growth factors. The specific signal transduction
multiplex immunoassays allows for simultaneous pathways share the following general steps. A
monitoring of protein, cytokines, and chemo- ligand produced by one cell binds to a receptor in
kines and can also assess the phosphorylation the extracellular region of another cell. The shape
states of many proteins involved in signal of the cytoplasmic domain of the receptor
transduction. changes, and the receptor acquires enzymatic
activity. The receptor then catalyzes reactions
Embryoid Body and Teratoma Formation that phosphorylate other proteins, activating
Embryoid body (EB) and teratoma formation are them. A cascade of phosphorylation reactions
used as indicators of stemness. Both will differ- eventually activates a dormant transcription fac-
entiate and contain cells from all three germ lay- tor or cytoskeletal protein, thus contributing to
ers, demonstrating pluripotency. EBs will develop the differentiation process in the target cell [54].
spontaneously in ESC or iPSC cultures upon Signal induction refers to cascades of signaling
withdrawal of factors that maintain pluripotency, events.
when cultures become very dense or when sus- Other important mechanisms fall under the
pended in media that promotes formation of EBs. category of asymmetric cell divisions.
Generation of teratomas is a method for func- Asymmetric cell divisions can occur because of
tional analysis of pluripotent stem cells in vivo. asymmetrically expressed maternal cytoplasmic
Teratomas are tumors containing differentiated determinants [55]. The distinct daughter cells are
and undifferentiated cells from all three germ created during cytokinesis because of an uneven
layers. distribution of regulatory molecules in the parent
cell. A well-studied example of pattern formation
by asymmetric divisions is body axis patterning
3.2.3 Cellular Differentiation in Drosophila. RNA molecules are an important
Mechanisms type of intracellular differentiation control
signal.
Specialized cell in an organism expresses a sub-
set of all the genes that constitute the genome. 3.2.3.1 Epigenetics in Stem Cell
Each cell type is defined by its particular pattern Differentiation
of regulated gene expression. Cell differentiation Since each cell possesses the same genome,
is thus a transition of a cell from one cell type to determination of cell type must occur at the level
another, and it involves a switch from one pattern of gene expression. While the regulation of gene
of gene expression to another. Cellular differen- expression can occur through cis- and trans-­
tiation during development can be understood as regulatory elements including a gene’s promoter
the result of a gene regulatory network. A regula- and enhancers, the problem arises as to how this
tory gene and its cis-regulatory modules are expression pattern is maintained over numerous
nodes in a gene regulatory network [53]. The generations of cell division. As it turns out,
3  Cell Technologies 77

e­ pigenetic processes play a crucial role in regu- thought that these factors’ alterations in chroma-
lating the decision to adopt a stem, progenitor, or tin structure, such as histone modification and
mature cell fate. DNA methylation, can restrict or permit the tran-
scription of target genes.
Epigenetic Control In the process of gene silencing, Polycomb
One of the important gene regulatory networks repressive complex 2 catalyzes the di- and tri-
for determination of cell fate is of epigenetic pro- methylation of histone H3 lysine 27 (H3K27me2/
cesses. A paper by Lister et al. [56] is reported on me3) [57]. By binding to the H3K27me2/3-­
aberrant epigenomic programming in human-­ tagged nucleosome, PRC1 catalyzes the mono-­
induced pluripotent stem cells. As induced plu- ubiquitinylation of histone H2A at lysine 119
ripotent stem cells (iPSCs) are thought to mimic (H2AK119Ub1), blocking RNA polymerase II
embryonic stem cells in their pluripotent proper- activity and resulting in transcriptional suppres-
ties, few epigenetic differences should exist sion. PcG knockout ES cells do not differentiate
between them. To test this prediction, the authors efficiently into the three germ layers, and dele-
conducted whole-genome profiling of DNA tion of the PRC1 and PRC2 genes leads to
methylation patterns in several human embryonic increased expression of lineage-affiliated genes
stem cells (ESCs), iPSC, and progenitor cell and unscheduled differentiation. Presumably,
lines. PcG complexes are responsible for transcription-
Adipose cells, lung fibroblasts, and foreskin ally repressing differentiation and development-­
fibroblasts were reprogrammed into induced plu- promoting genes.
ripotent state with the OCT4, SOX2, KLF4, and PcG proteins are recruited to promoters of
MYC genes. Patterns of DNA methylation in pluripotency transcription factors. PcG-deficient
ESCs, iPSCs, somatic cells were compared. ES cells can begin differentiation but cannot
Results observed significant resemblance in maintain the differentiated phenotype.
methylation levels between embryonic and Simultaneously, differentiation and development-­
induced pluripotent cells. Around 80% of CG promoting genes are activated by trithorax group
dinucleotides in ESCs and iPSCs were methyl- (TrxG) chromatin regulators and lose their
ated; the same was true of only 60% of CG dinu- repression. TrxG proteins are recruited at regions
cleotides in somatic cells. In addition, somatic of high transcriptional activity, where they
cells possessed minimal levels of cytosine meth- ­catalyze the trimethylation of histone H3 lysine 4
ylation in non-CG dinucleotides, while induced and promote gene activation through histone
pluripotent cells possessed similar levels of acetylation. PcG and TrxG complexes engage in
methylation as embryonic stem cells. Thus, con- direct competition and are thought to be func-
sistent with their respective transcriptional activi- tionally antagonistic, creating at differentiation
ties, DNA methylation patterns, at least on the and development-promoting loci what is termed
genomic level, are similar between ESCs and a “bivalent domain” and rendering these genes
iPSCs. Thus, epigenetic processes are heavily sensitive to rapid induction or repression.
involved in cell fate determination, as seen from Gene expression regulation is more progressed
the similar levels of cytosine methylation between through DNA methylation, in which the DNA
induced pluripotent and embryonic stem cells, methyltransferase-mediated methylation of cyto-
consistent with their respective patterns of sine residues in CpG dinucleotides maintains
transcription. heritable repression by controlling DNA accessi-
bility [58]. The majorities of CpG sites in embry-
Mechanisms of Epigenetic Regulation onic stem cells are unmethylated and appear to be
Three transcription factors, OCT4, SOX2, and associated with H3K4me3-carrying nucleo-
NANOG, are highly expressed in undifferenti- somes. Upon differentiation, a small number of
ated embryonic stem cells and are necessary for genes, including OCT4 and NANOG, are meth-
the maintenance of their pluripotency [57]. It is ylated and their promoters repressed to prevent
78 S.Y. Chun

their further expression. Consistently, DNA and Notch signaling is involved in the prolifera-
methylation-deficient embryonic stem cells rap- tion and self-renewal of stem cells. Sonic hedge-
idly enter apoptosis upon in vitro differentiation. hog, in addition to its role as a morphogen, also
The DNA sequence of most cells of an organ- promotes embryonic stem cell differentiation and
ism is the same, but the binding patterns of tran- the self-renewal of somatic stem cells [60].
scription factors and the corresponding gene
expression patterns are different. To a large Matrix Elasticity Effect on Differentiation
extent, differences in transcription factor binding Stem cells are known to migrate from their niches
are determined by the chromatin accessibility of to regenerate variety of tissues by adhering to
their binding sites through histone modification new extracellular matrices (ECM) and differenti-
and/or pioneer factors. In particular, it is impor- ate. The ductility of these microenvironments is
tant to know whether a nucleosome is covering a unique to different tissue types. The ECM sur-
given genomic binding site or not. Recent studies rounding brain, muscle, and bone tissues range
have elucidated the role of nucleosome position- from soft to stiff. The transduction of the stem
ing during stem cell development [59]. cells into these cells types is not directed solely
by chemokine cues and cell to cell signaling. The
Cell Signaling in Epigenetic Control elasticity of the microenvironment can also affect
The cell signaling can give influence to the epi- the differentiation of mesenchymal stem cells.
genetic processes governing differentiation. The When MSCs are placed on substrates of the same
first major candidate is Wnt signaling pathway. stiffness as brain, muscle, and bone ECM, the
The Wnt pathway is involved in all stages of dif- MSCs take on properties of those respective cell
ferentiation, and the ligand Wnt3a can substitute types [62].
for the overexpression of c-Myc in the generation Matrix sensing requires the cell to pull against
of induced pluripotent stem cells [60]. On the the matrix at focal adhesions, which triggers a
other hand, disruption of ß-catenin, a component cellular mechano-transducer to generate a signal
of the Wnt signaling pathway, leads to decreased to be informed what force is needed to deform
proliferation of neural progenitors. the matrix. To determine the key players in
The second candidate of epigenetic regulators matrix-elasticity-driven lineage specification in
of cellular differentiation is growth factors. These MSCs, different matrix microenvironments were
morphogens are crucial for development and mimicked. Researchers have obtained some
include bone morphogenetic proteins, transform- ­success in inducing stem cell-like properties in
ing growth factors (TGFs) and fibroblast growth HEK 239 cells by providing a soft matrix without
factors (FGFs). TGFs and FGFs have been shown the use of diffusing factors [63]. The stem cell
to sustain expression of OCT4, SOX2, and properties appear to be linked to tension in the
NANOG by downstream signaling to Smad pro- cells’ actin network. One identified mechanism
teins. Depletion of growth factors promotes the for matrix-induced differentiation is tension-
differentiation of ESCs, while genes with biva- induced proteins, which remodel chromatin in
lent chromatin can become either more restrictive response to mechanical stretch [64].
or permissive in their transcription.
As other pathways, cytokine leukemia inhibi-
tory factors are associated with the maintenance 3.2.4 S
 tem Cell Therapy
of mouse ESCs in an undifferentiated state. This and Research
is achieved through its activation of the Jak-­
STAT3 pathway, which has been shown to be Stem cells become a major part of regenerative
necessary and sufficient toward maintaining medicine. There are an increasing number of
mouse ESC pluripotency [61]. Retinoic acid can stem cell therapies being researched and thou-
induce differentiation of human and mouse ESCs, sands of clinical trials.
3  Cell Technologies 79

3.2.4.1 Transplantation of Stem Cells tractable models for the study of all aspects of
Transplantation of stem cells into the body entails cell biology. Stem cell models are being used to
assessment of either morphologic repair or resto- study many cell functions including: mechanisms
ration of function. There are a number of require- and signals for stem cell proliferation (symmetric
ments for a successful stem cell therapy: 1) easy and asymmetric), signals that trigger migration,
to obtain and can readily be sorted and character- signals and pathways involved in differentiation,
ized; 2) will survive in the body, remaining via- pathways for differentiation to different pheno-
ble, and not trigger a severe immune response; 3) types, sequences of epigenetic changes during
able to proliferate, preferably in the target tissue; differentiation into different lineages and cell
4) can differentiate into the target tissue; 5) can types, cell functions of stem and somatic cells
integrate into the targeted tissue; 6) engrafted under normal nonstressed conditions, changes in
cells function appropriately; and 7) will at least cell functions under stress, changes in cell func-
partially restore function and repair damaged tions upon stimulation, apoptosis, cell death,
area. functions and behaviors of stem cells, roles of
Blood stem cells are currently the most fre- stem cells in normal development, growth, and
quently used stem cells for therapy. For more repair.
than 50 years, doctors have been using bone mar-
row transplants to transfer blood stem cells to 3.2.4.3 For Disease
patients, and more advanced techniques for col- The generation of stem cell models from somatic
lecting blood stem cells are now being used to cells provides the ability to develop stem cell
treat leukemia, lymphoma, and several inherited lines for many diseases. Comparison of normal
blood disorders. Umbilical cord blood, like bone and disease stem cells will identify disease
marrow, is often collected as a source of blood phenotype(s), investigate the development of the
stem cells and in certain cases is being used as an disease phenotype, profile control and disease
alternative to bone marrow transplantation. cells at transcriptome and protein levels to reveal
Additionally, some bone, skin, and corneal dis- differences, measure functional changes, deter-
eases or injuries can be treated by grafting tissues mine which cell types are affected by the disease,
that are derived from or maintained by stem cells. and identify disease biomarkers and potential
These therapies have also been shown to be safe new diagnostics.
and effective. The major advantage of iPS cells is that they
Other stem cell treatments are still at very early are a very good way to make pluripotent stem cell
experimental stages. For example, the mesenchy- lines that are specific to a disease or even to an
mal stem cell, found throughout the body includ- individual patient. Disease-specific stem cells are
ing in the bone marrow, can be directed to become powerful tools for studying the cause of a partic-
bone, cartilage, fat, and possibly even muscle. In ular disease and then for testing drugs or discov-
certain experimental models, these cells also have ering other approaches to treat or cure that
some ability to modify immune functions. These disease. The development of patient-specific
abilities have created considerable interest in stem cells is also very attractive for cell therapy,
developing ways of using mesenchymal stem cells as these cell lines are from the patient themselves
to treat a range of musculoskeletal abnormalities, and may minimize some of the serious complica-
cardiac disease, and some immune abnormalities tions of rejection and immunosuppression that
such as graft-­versus-­host disease following bone can occur.
marrow transplant.
3.2.4.4 For Drug Discovery
3.2.4.2 For Research The ability to generate large populations of cells
Although much of the initial focus has been on with defined phenotypes is having an impact on
stem cell models for disease, stem cells provide drug discovery and testing. Stem cell-derived
80 S.Y. Chun

model systems can be used for all aspects of stem cells, are painstaking and difficult processes.
drug discovery, including: screening compounds Pluripotent stem cells, such as embryonic stem
for changes in known biomarkers, screening cells, can be grown indefinitely in the lab and have
compounds or pools of compounds for changes the advantage of having the potential to become
in disease phenotype, evaluating the any cell in the body, but these processes are again
mechanism(s) of alteration of disease by lead very complex and must be tightly controlled. iPS
compounds, toxicity screening, screening dif- cells, while promising, are also limited by these
ferent cell lines to ensure applicability to a concerns. In both cases, considerable work
whole population rather than small subpopula- remains to be done to ensure that these cells can be
tions, safety and effects in nontarget cell types, isolated and used safely and routinely. In addition,
in vitro clinical trials, and reduced animal test- it is very important to have a close match between
ing (and associated costs). the donor tissue and the recipient; the more closely
Researchers are able to grow up differentiated the tissue matches the recipient, the lower the risk
cell lines and then test new drugs on each cell of rejection. Being able to avoid the life-long use
type to examine possible interactions in vitro of immunosuppressants would also be preferable.
before performing in vivo studies. This is critical The discovery of iPS cells has opened the door to
in the development of drugs for use in veterinary develop patient-specific pluripotent stem cell lines
research because of the possibilities of species-­ that can later be developed into a needed cell type
specific interactions. The hope is that having without the problems of rejection and immuno-
these cell lines available for research use will suppression that occur from transplants from unre-
reduce the need for research animals used lated donors. Also, a system for delivering the
because effects on human tissue in vitro will pro- cells to the right part of the body must be devel-
vide insight not normally known before the ani- oped. Once in the right location, the new cells
mal testing phase. must then be encouraged to integrate and function
together with the body’s other cells.
3.2.4.5 Clinical Translation
Clinical translation is the process used to turn sci- 3.2.4.7 Conservation
entific knowledge into real-world medical treat- Stem cells are being explored for use in conserva-
ments. Researchers take what they have learned tion efforts. Spermatogonial stem cells have been
about how a tissue usually works and what goes harvested from a rat and placed into a mouse
wrong in a particular disease or injury and use this host, and fully mature sperm were produced with
information to develop new ways to diagnose, the ability to produce viable offspring. Currently
stop, or fix what goes wrong. Before being mar- research is underway to find suitable hosts for the
keted or adopted as standard of care, most treat- introduction of donor spermatogonial stem cells.
ments are tested through clinical trials. Sometimes, If this becomes a viable option for conservation-
in attempting new surgical techniques or where the ists, sperm can be produced from high genetic
disease or condition is rare and does not have a quality individuals who die before reaching sex-
large enough group of people to form a clinical ual maturity, preserving a line that would other-
trial, certain treatments might be tried on one or wise be lost [65].
two people, a form of testing sometimes referred
to as innovative medicine. In recent years, stem Glossary
cell clinics have emerged that treat patients with Adult stem cell See somatic stem cell
their own bone marrow or adipose-­derived adult Astrocyte: A type of supporting (glial) cell
stem cells as part of clinical trials or FDA- found in the nervous system
authorized same-day outpatient IRB programs. Blastocoel: The fluid-filled cavity inside the
blastocyst, an early, preimplantation stage of the
3.2.4.6 Challenges developing embryo
Identifying, isolating, and growing the right kind Blastocyst: A very early embryo that has the
of stem cell, particularly in the case of rare adult shape of a ball and consists of approximately
3  Cell Technologies 81

150–200 cells. It contains the inner cell mass, interaction of a cell’s genes with the physical and
from which embryonic stem cells are derived, chemical conditions outside the cell, usually
and an outer layer of cells called the trophoblast through signaling pathways involving proteins
that forms the placenta embedded in the cell surface
Bone marrow stromal cells: A population of Directed differentiation: The manipulation of
cells found in bone marrow that are different stem cell culture conditions to induce differentia-
from blood cells tion into a particular cell type
Bone marrow stromal stem cells (skeletal DNA: Deoxyribonucleic acid, a chemical
stem cells): A multipotent subset of bone marrow found primarily in the nucleus of cells. DNA car-
stromal cells able to form bone, cartilage, and ries the instructions or blueprint for making all
stromal cells that support blood formation, fat, the structures and materials the body needs to
and fibrous tissue function. DNA consists of both genes and non-­
Cell-based therapies: Treatment in which stem gene DNA in between the genes
cells are induced to differentiate into the specific Ectoderm: The outermost germ layer of cells
cell type required to repair damaged or destroyed derived from the inner cell mass of the blastocyst;
cells or tissues gives rise to the nervous system, sensory organs,
Cell culture: Growth of cells in vitro in an arti- skin, and related structures
ficial medium for research Embryo: The early developing organism; this
Cell division: Method by which a single cell term denotes the period of development between
divides to create two cells. There are two main the fertilized egg and the fetal stage
types of cell division depending on what happens Embryonic bodies: Rounded collections of
to the chromosomes: mitosis and meiosis cells that arise when embryonic stem cells are
Cell line: Cells that can be maintained and cultured in suspension. Embryonic bodies con-
grown in a dish outside of the body tain cell types derived from all three germ layers
Chromosome: A structure consisting of DNA Embryonic germ cells: Pluripotent stem cells
and regulatory proteins found in the nucleus of that are derived from early germ cells (those that
the cell. The DNA in the nucleus is usually would become sperm and eggs). Embryonic germ
divided up among several chromosomes. The cells are thought to have properties similar to
number of chromosomes in the nucleus varies embryonic stem cells
depending on the species of the organism. Embryonic stem cell: Cells derived from
Humans have 46 chromosomes very early in development, usually the inner
Clinical translation: The process of using sci- cell mass of a developing blastocyst. These
entific knowledge to design, develop, and apply cells are self-renewing (can replicate them-
new ways to diagnose, stop, or fix what goes selves) and pluripotent (can form all cell types
wrong in a particular disease or injury found in the body)
Clone (v): To generate identical copies of a Embryonic stem cell line: Embryonic stem
region of a DNA molecule or to generate geneti- cells, which have been cultured under in vitro
cally identical copies of a cell or organism; (n) conditions that allow proliferation without differ-
the identical molecule, cell, or organism that entiation for months to years
result from the cloning process Endoderm: The innermost layer of the cells
Culture medium: The liquid that covers cells derived from the inner cell mass of the blastocyst;
in a culture dish and contains nutrients to nourish it gives rise to lungs, other respiratory structures,
and support the cells. Culture medium may also and digestive organs or generally “the gut”
include growth factors added to produce desired Enucleated: Having had its nucleus removed
changes in the cells Epigenetic: The process by which regulatory
Differentiation: The process whereby an proteins can turn genes on or off in a way that can
unspecialized embryonic cell acquires the fea- be passed on during cell division
tures of a specialized cell such as a heart, liver, or Feeder layer: Cells used in co-culture to main-
muscle cell. Differentiation is controlled by the tain pluripotent stem cells. For human embryonic
82 S.Y. Chun

stem cell culture, typical feeder layers include disease. These are done outside of a typical clini-
mouse embryonic fibroblasts (MEFs) or human cal trial framework
embryonic fibroblasts that have been treated to In vitro: Latin for “in glass”; in a laboratory
prevent them from dividing dish or test tube; an artificial environment
Fertilization: The joining of the male gamete In vitro fertilization: A procedure in which an
(sperm) and the female gamete (egg) egg cell and sperm cells are brought together in a
Fetus: In humans, the developing human from dish to fertilize the egg. The fertilized egg will
approximately 8 weeks after conception until the start dividing and, after several divisions, forms
time of its birth the embryo that can be implanted into the womb
Gamete: An egg (in the female) or sperm (in of a woman and give rise to pregnancy
the male) cell. See also somatic cell Inner cell mass (ICM): The cluster of cells
Gastrulation: The process in which cells pro- inside the blastocyst. These cells give rise to the
liferate and migrate within the embryo to trans- embryo and ultimately the fetus. The ICM may
form the inner cell mass of the blastocyst stage be used to generate embryonic stem cells
into an embryo containing all three primary germ Long-term self-renewal: The ability of stem
layers cells to replicate themselves by dividing into the
Gene: A functional unit of heredity that is a same nonspecialized cell type over long periods
segment of DNA found on chromosomes in the (many months to years) depending on the specific
nucleus of a cell. Genes direct the formation of an type of stem cell
enzyme or other protein Meiosis: The type of cell division a diploid
Germ layers: After the blastocyst stage of germ cell undergoes to produce gametes (sperm
embryonic development, the inner cell mass of or eggs) that will carry half the normal chromo-
the blastocyst goes through gastrulation, a period some number. This is to ensure that when fertil-
when the inner cell mass becomes organized into ization occurs, the fertilized egg will carry the
three distinct cell layers, called germ layers. The normal number of chromosomes rather than
three layers are the ectoderm, the mesoderm, and causing aneuploidy (an abnormal number of
the endoderm chromosomes)
Hematopoietic stem cell: A stem cell that Mesenchymal stem cells: Mesenchymal stem
gives rise to all red and white blood cells and cells were originally discovered in the bone
platelets marrow, but have since been found throughout
Human embryonic stem cell (hESC): A type the body and can give rise to a large number of
of pluripotent stem cell derived from early-­stage connective tissue types such as bone, cartilage,
human embryos, up to and including the blasto- and fat
cyst stage. hESCs are capable of dividing without Mesoderm: Middle layer of a group of cells
differentiating for a prolonged period in culture derived from the inner cell mass of the blastocyst;
and are known to develop into cells and tissues of it gives rise to bone, muscle, connective tissue,
the three primary germ layers kidneys, and related structures
Induced pluripotent stem (iPS) cell: Induced Microenvironment: The molecules and com-
pluripotent cells (iPS cells) are stem cells that pounds such as nutrients and growth factors in
were engineered (“induced”) from nonpluripotent the fluid surrounding a cell in an organism or in
cells to become pluripotent. In other words, a cell the laboratory, which play an important role in
with a specialized function (e.g., a skin cell) that determining the characteristics of the cell
has been “reprogrammed” to an unspecialized Mitosis: The type of cell division that allows a
state similar to that of an embryonic stem cell population of cells to increase its numbers or to
Innovative medicine: Treatments that are per- maintain its numbers. The number of chromo-
formed on a small number of people and are somes in each daughter cell remains the same in
designed to test a novel technique or treat a rare this type of cell division
3  Cell Technologies 83

Multipotent stem cells: Stem cells that can This smaller cell is called the first polar body.
give rise to several different types of specialized The first polar body usually degenerates. The
cells, but in contrast to a pluripotent stem cell, are ovum, or larger cell, then divides again, produc-
restricted to a certain organ or tissue types. For ing a second polar body with half the amount of
example, blood stem cells are multipotent cells chromosomes but almost no cytoplasm. The sec-
that can produce all the different cell types that ond polar body splits off and remains adjacent to
make up the blood but not the cells of other the large cell, or oocyte, until it (the second polar
organs such as the liver or brain body) degenerates. Only one large functional
Neural stem cell: A stem cell found in adult oocyte, or egg, is produced at the end of meiosis
neural tissue that can give rise to neurons and Preimplantation: With regard to an embryo,
glial (supporting) cells. Examples of glial cells preimplantation means that the embryo has not
include astrocytes and oligodendrocytes yet implanted in the wall of the uterus. Human
Neurons: Nerve cells, the principal functional embryonic stem cells are derived from preim-
units of the nervous system. A neuron consists of plantation-stage embryos fertilized outside a
a cell body and its processes: an axon and one or woman’s body (in vitro)
more dendrites. Neurons transmit information to Proliferation: Expansion of the number of
other neurons or cells by releasing neurotrans- cells by the continuous division of single cells
mitters at synapses into two identical daughter cells
Oligodendrocyte: A supporting cell that pro- Regenerative medicine: A field of medicine
vides insulation to nerve cells by forming a devoted to treatments in which stem cells are
myelin sheath (a fatty layer) around axons induced to differentiate into the specific cell type
Parthenogenesis: The artificial activation of an required to repair damaged or destroyed cell popu-
egg in the absence of a sperm; the egg begins to lations or tissues. (See also cell-based therapies)
divide as if it has been fertilized Reproductive cloning: The process of using
Passage: In cell culture, the process in which somatic cell nuclear transfer (SCNT) to produce
cells are disassociated, washed, and seeded into a normal, full grown organism (e.g., animal)
new culture vessels after a round of cell growth genetically identical to the organism (animal)
and proliferation. The number of passages a line that donated the somatic cell nucleus. In mam-
of cultured cells has gone through is an indication mals, this would require implanting the resulting
of its age and expected stability embryo in a uterus where it would undergo nor-
Pluripotent stem cells: Stem cells that can mal development to become a live independent
become all the cell types that are found in an being. The first mammal to be created by repro-
implanted embryo, fetus, or developed organism. ductive cloning was Dolly the sheep, born at the
Embryonic stem cells are pluripotent stem cells. Roslin Institute in Scotland in 1996. See also
Scientists demonstrate pluripotency by providing somatic cell nuclear transfer (SCNT)
evidence of stable developmental potential, even Self-renewal: The process by which a cell
after prolonged culture, to form derivatives of all divides to generate another cell that has the same
three embryonic germ layers from the progeny of potential
a single cell and to generate a teratoma after Stem cells: Cells that have both the capacity to
injection into an immunosuppressed mouse self-renew (make more stem cells by cell division)
Polar body: A polar body is a structure pro- and to differentiate into mature, specialized cells
duced when an early egg cell, or oogonium, Signals: Internal and external factors that con-
undergoes meiosis. In the first meiosis, the oogo- trol changes in cell structure and function. They
nium divides its chromosomes evenly between can be chemical or physical in nature
the two cells but divides its cytoplasm unequally. Somatic cell: Any body cells other than gam-
One cell retains most of the cytoplasm, while the etes (egg or sperm); sometimes referred to as
other gets almost none, leaving it very small. “adult” cells
84 S.Y. Chun

Somatic cell nuclear transfer (SCNT): A tech- Tissue-specific stem cells (also known as adult
nique that combines an enucleated egg and the or somatic stem cells): Stem cells found in differ-
nucleus of a somatic cell to make an embryo. ent tissues of the body that can give rise to some
SCNT can be used for therapeutic or reproduc- or all of the mature cell types found within the
tive purposes, but the initial stage that combines particular tissue or organ from which they came,
an enucleated egg and a somatic cell nucleus is i.e., blood stem cells can give rise to all the cells
the same. See also therapeutic cloning and repro- that make up the blood, but not the cells of organs
ductive cloning such as the liver or brain
Somatic (adult) stem cell: A relatively rare Totipotent stem cells: Stem cells that, under
undifferentiated cell found in many organs and the right conditions, are wholly capable of gener-
differentiated tissues with a limited capacity for ating a viable embryo (including the placenta)
both self-renewal (in the laboratory) and differ- and, for humans, exist until about 4 days after
entiation. Such cells vary in their differentiation fertilization, prior to the blastocyst stage from
capacity, but it is usually limited to cell types in which embryonic stem cells are derived
the organ of origin. This is an active area of Transdifferentiation: The process by which
investigation stem cells from one tissue differentiate into cells
Stem cells: Cells with the ability to divide for of another tissue
indefinite periods in culture and to give rise to Trophoblast: The outer cell layer of the blas-
specialized cells tocyst. It is responsible for implantation and
Stromal cells: Connective tissue cells found in develops into the extraembryonic tissues,
virtually every organ. In bone marrow, stromal including the placenta, and controls the
cells support blood formation exchange of oxygen and metabolites between
Subculturing: Transferring cultured cells, with mother and embryo
or without dilution, from one culture vessel to Umbilical cord blood stem cells: Stem cells
another collected from the umbilical cord at birth that can
Surface markers: Proteins on the outside sur- produce all of the blood cells in the body. Cord
face of a cell that are unique to certain cell types blood is currently used to treat patients who have
and that can be visualized using antibodies or undergone chemotherapy to destroy their bone
other detection methods marrow due to cancer or other blood-related
Teratoma: A multi-layered benign tumor that disorders
grows from pluripotent cells injected into mice Undifferentiated: A cell that has not yet devel-
with a dysfunctional immune system. Scientists oped into a specialized cell type
test whether they have established a human
embryonic stem cell (hESC) line by injecting
putative stem cells into such mice and verifying
that the resulting teratomas contain cells derived References
from all three embryonic germ layers
1. Schöler R. The potential of stem cells, vol. 28.
Therapeutic cloning: The process of using Farnham: Ashgate Publishing; 2007.
somatic cell nuclear transfer (SCNT) to pro- 2. Thomson IE, Shapiro J, Waknitz SS, et al. Blastocysts
duce cells that exactly match a patient. By embryonic stem cell lines derived from human.
combining a patient’s somatic cell nucleus and Science. 1998;282:1145–7.
3. Thomson JA. Homologous recombination in
an enucleated egg, a scientist may harvest human embryonic stem cells. Nat Biotechnol.
embryonic stem cells from the resulting 2003;21:319–21.
embryo that can be used to generate tissues 4. Parish CL, Arenas E. Stem-cell-based strategies for
that match a patient’s body. This means the tis- the treatment of Parkinson's disease. Neurodegener
Dis. 2007;4:339–47.
sues created are unlikely to be rejected by the 5. Abdul MB. Designer’s microglia with novel deliv-
patient’s immune system. See also somatic cell ery system in neurodegenerative diseases. Med
nuclear transfer (SCNT) Hypotheses. 2014;83(4):510–2.
3  Cell Technologies 85

6. Nichols Y, Chambers J, Smith IA. BMP induction 23. Zhang H, Wilson IA, Lerner RA. Selection of

of id proteins suppresses differentiation and sustains antibodies that regulate phenotype from intracel-
embryonic stem cell self-renewal in collaboration lular combinatorial antibody libraries. PNAS.
with STAT3. Cell. 2003;115:281–92. 2012;109:15728–33.
7. Söderdahl T, Küppers-Munther B, Heins N, et al. 24. Liu X, Ory V, Chapman S, et al. ROCK inhibitor and
Glutathione transferases in hepatocyte-like cells feeder cells induce the conditional reprogramming of
derived from human embryonic stem cells. Toxicol In epithelial cells. Am J Pathol. 2012;180:599–607.
Vitro. 2007;21:929–37. 25. Kurian L, Sancho-Martinez I, Nivet E, et al.

8. Davila JC, Cezar GG, Thiede M, et al. Use and Conversion of human fibroblasts to angioblast-like
application of stem cells in toxicology. Toxicol Sci. progenitor cells. Nat Methods. 2012;10:77–83.
2004;79:214–23. 26. Suila H, Hirvonen T, Ritamo I, et al. Extracellular
9. Kuliev A, Verlinsky Y. Place of preimplantation o-linked n-acetylglucosamine is enriched in stem cells
diagnosis in genetic practice. Am J Med Genet A. derived from human umbilical cord blood. BioRes
2005:134A;105–10. Open Access. 2014;3:39–44.
10. Tichy ED, Pillai R, Deng L, et al. Mouse embryonic 27. Downing TL, Soto J, Morez C, et al. Biophysical reg-
stem cells, but not somatic cells, predominantly use ulation of epigenetic state and cell reprogramming.
homologous recombination to repair double-strand Nat Mater. 2013;12:1154–62.
DNA breaks. Stem Cells. 2010;19:1699–711. 28. Sun Y, Koh AY, Meng V, et al. Hippo/YAP-­

11. Knoepfler PS. Deconstructing stem cell tumorigenic- mediated rigidity-dependent motor neuron differen-
ity: a roadmap to safe regenerative medicine. Stem tiation of human pluripotent stem cells. Nat Mater.
Cells. 2009;27:1050–6. 2014;13:599–604.
12. Bharadwaj S, Liu G, Shi Y, et al. Multipotential differ- 29. Caiazzo M, Okawa Y, Ranga A, et al. Defined three-­
entiation of human urine-derived stem cells: potential dimensional microenvironments boost induction of
for therapeutic applications in urology. Stem Cells. pluripotency. Nat Mater. 2016;15:344–52.
2013;31:1840–56. 30. Takahashi K, Yamanaka S. Induction of pluripotent
13. Yehezkel S, Rebibo-Sabbah A, Segev Y, et al.
stem cells from mouse embryonic and adult fibroblast
Reprogramming of telomeric regions during the gen- cultures by defined factors. Cell. 2006;124:663–76.
eration of human induced pluripotent stem cells and 31. Stadtfeld M, Nagaya M, Utikal J, et al. Induced plu-
subsequent differentiation into fibroblast-like deriva- ripotent stem cells generated without viral integration.
tives. Epigenetics. 2011;6:63–75. Science. 2008;322:949–9.
14. Dabir DV, Hasson SA, Setoguchi K, et al. A small 32. Seki T, Yuasa S, Fukuda K. Generation of induced
molecule inhibitor of redox-regulated protein translo- pluripotent stem cells from a small amount of
cation into mitochondria. Dev Cell. 2013;25:81–92. human peripheral blood using combination of
15. Lindgren AG, Natsuhara K, Tian E, et al. Loss of pten activated T cells and Sendai virus. Nat Protoc.
causes tumor initiation following differentiation of 2012;7:718–28.
murine pluripotent stem cells due to failed repression 33. Fusaki N, Ban H, Nishiyama A, et al. Efficient induc-
of Nanog. PLoS One. 2011;6:e16478. tion of transgene-free human pluripotent stem cells
16. Ohnishi K, Semi K, Yamamoto T, et al. Premature using a vector based on Sendai virus, an RNA virus
termination of reprogramming in vivo leads to cancer that does not integrate into the host genome. Proc Jpn
development through altered epigenetic regulation. Acad Ser B Phys Biol Sci. 2009;85:348–62.
Cell. 2014;156:663–77. 34. Anokye-Danso F, Trivedi CM, Juhr D, et al. Highly
17. Yamanaka S, Blau H. Nuclear reprogramming to
efficient miRNA-mediated reprogramming of mouse
a pluripotent state by three approaches. Nature. and humansomatic cells to pluripotency. Sell Stem
2010;465:704–12. Cell. 2011;8:376–88.
18. Tachibana M, Amato P, Sparman M, et al. Human 35. Judson RL, Babiarz JE, Venere M, et al. Embryonic
embryonic stem cells derived by somatic cell nuclear stem cell-specific microRNAs promote induced pluri-
transfer. Cell. 2013;153:1228–38. potency. Nat Biotechnol. 2009;27:459–61.
19. Paull D, Emmanuele V, Weiss K, et al. Nuclear
36. Huangfu D, Maehr R, Guo W, et al. Induction of
genome transfer in human oocytes eliminates mito- pluripotent stem cells by defined factors is greatly
chondrial DNA variants. Nature. 2012;493:632–7. improved by small-molecule compounds. Nat
20. Shinagawa T, Takagi T, Tsukamoto D, et al. Histone Biotechnol. 2008;26:795–7.
variants enriched in oocytes enhance reprogramming 37. Zhang M, Zhang J, Chen X, et al. Glycogen synthase
to induced pluripotent stem cells. Cell Stem Cell. kinase 3 promotes p53 mRNA translation viaphos-
2014;14:217–27. phorylation of RNPC1. Genes Dev. 2012;27:2246–58.
21. Han DW, Tapia N, Hermann A, et al. Direct repro- 38. Okita K, Nakagawa M, Hyenjong H, et al. Generation
gramming of fibroblasts into neural stem cells by of mouse induced pluripotent stem cells without viral
defined factor. Cell Stem Cell. 2012;10:465–72. vectors. Science. 2008;322:949–53.
22. Wei S, Zou Q, Lai S, et al. Conversion of embryonic 39. Brighton CT, Hunt RM. Early histological and ultra-
stem cells into extraembryonic lineages by CRISPR-­ structural changes in medullary fracture callus. J
mediated activators. Sci Rep. 2016;6:19648. Bone Joint Surg. 1991;73:832–47.
86 S.Y. Chun

40. Haniffa MA, Collin MP, Buckley CD, et al.


52. Adewumi O. Characterization of human embryonic
Mesenchymal stem cells: the fibroblasts’ new clothes? stem cell lines by the international stem cell initiative.
Haematologica. 2009;94:258–63. Nat Biotechnol. 2007;25:803–16.
41. Hematti P. Mesenchymal stromal cells and fibro-
53. Ben-Tabou S, Davidson EH. Gene regulation: gene
blasts: a case of mistaken identity? Cytotherapy. control network in development. Annu Rev Biophys
2012;14:516–21. Biomol Struct. 2007;36:191–212.
42. Dominici M, Le Blanc K, Mueller I, et al. Minimal 54. Knisely K, Gilbert SF. Developmental biology. 8th ed.
criteria for defining multipotent mesenchymal Sunderland, Mass: Sinauer Associates; 2009. p. 147.
stromal cells. The international society for cel- 55. Rudel S. The evolution of developmental mecha-

lular therapy position statement. Cytotherapy. nisms. Dev Biol. 2003;264:15–37.
2006;8:315–7. 56. Lister R. Hotspots of aberrant epigenomic repro-

43. Jiang Y, Jahagirdar BN, Reinhardt RL, et al.
gramming in human induced pluripotent stem cells.
Pluripotency of mesenchymal stem cells derived from Nature. 2011;471:68–73.
adult marrow. Nature. 2002;418:41–9. 57. Christophersen NS, Helin K. Epigenetic control of
44.
Ryan JM, Barry FP, Murphy JM, Mahon embryonic stem cell fate. J Exp Med. 2010;207:2287–95.
BP. Mesenchymal stem cells avoid allogeneic rejec- 58.
Meissner A. Epigenetic modifications in plu-
tion. J Inflamm. 2005;2:8. ripotent and differentiated cells. Nat Biotechnol.
45. Ryan JM, Barry F, Murphy JM, Mahon BP. Interferon-γ 2010;28:1079–88.
does not break, but promotes the immunosuppressive 59. Teif VB, Vainshtein Y, Caudron-Herger M, et al.

capacity of adult human mesenchymal stem cells. Genome-wide nucleosome positioning during embry-
Clin Exp Immunol. 2007;149:353–63. onic stem cell development. Nat Struct Mol Biol.
46. Gronthos S, Graves SE, Ohta S, Simmons PJ. The 2012;19:1185–92.
STRO-1+ fraction of adult human bone mar- 60. Mohammad HP, Baylin SB. Linking cell signal-

row contains the osteogenic precursors. Blood. ing and the epigenetic machinery. Nat Biotechnol.
1994;84:4164–73. 2010;28:1033–8.
47. Iudicone P, Fioravanti D, Bonanno G, et al. Pathogen-­ 61. Niwa H, Burdon T, Chambers I, Smith A. Self-renewal
free, plasma-poor platelet lysate and expansion of pluripotent embryonic stem cells is mediated via
of human mesenchymal stem cells. J Transl Med. activation of STAT3. Genes Dev. 1998;12:2048–60.
2014;12:28. 62. Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix
48. Fauza D. Amniotic fluid and placental stem cells. Best elasticity directs stem cell lineage specification. Cell.
Pract Res Clin Obstet Gynaecol. 2004;18:877–91. 2006;126:677–89.
49.
Inzunza J. Comparative genomic hybridization 63. Guo J, Wang Y, Sachs F, Meng F. Actin stress

and karyotyping of human embryonic stem cells in cell reprogramming. Proc Natl Acad Sci.
reveals the occurrence of an isodicentric X chromo- 2014;111:E5252–61.
some after long-term cultivation. Mol Hum Reprod. 64. Guilak F, Cohen DM, Estes BT, et al. Control of stem
2004;10:461–6. cell fate by physical interactions with the extracellular
50. Liehr T. Multicolor fish probe sets and their applica- matrix. Cell Stem Cell. 2009;5:17–26.
tions. Histol Histopathol. 2004;19:229–37. 65. Dobrinski A, Travis J. Germ cell transplantation

51. Kallioniemi A. Comparative genomic hybridization for the propagation of companion animals, non-­
for molecular cytogenetic analysis of solid tumors. domestic and endangered species. Reprod Fertil Dev.
Science. 1992;258:818–21. 2007;19:732–9.
Bioreactors for Regenerative
Medicine in Urology 4
In Kap Ko, Anthony Atala, and James J. Yoo

4.1 Introduction in vivo, and (3) severe fibrosis resulting from for-
eign body reactions and other immune responses
The urogenital system is generally composed of that inhibit tissue function. Therefore, an alterna-
the urinary and reproductive tissues and organs. tive treatment is needed to address these
The primary function of the urinary system, limitations.
which includes the kidneys, ureters, bladder, and Recent advances in tissue engineering and
urethra, is to produce and excrete urine. The male regenerative medicine have offered promising
reproductive system consists of the penis and tes- approaches for the treatment of damaged tissues
tes, while the female reproductive organs include and organs in the urogenital system [7–9]. Several
the vagina and uterus. Congenital disorders, strategies incorporate cell-based therapies that
infections, tumors, and defects in the urogenital use either a single cell-type (cell therapy) or engi-
system may result in tissue and organ damage or neered implantable tissue constructs (bioengi-
the complete loss of function [1]. Currently, these neering of functional tissue). Recent
conditions may be treated using autografts of developments in cell biology, stem cell biology,
non-urological tissue, such as the skin and and cell manipulation technology have enabled
mucosa. However, this method is limited by the identification, characterization, and expan-
donor site morbidity and poor survival of the sion of therapeutic cells for use in treatments.
grafted tissue [2]. Complicated injuries require Potential cell sources include tissue-specific pri-
extensive reconstructive surgery, which often mary cells, adult stem cells, and pluripotent stem
incorporates biomaterial-derived products [3–6]. cells. The engineering of functional urogenital
For instance, disorders in the pelvic muscles have tissue constructs in vitro by culturing tissue-­
been clinically treated with natural and synthetic specific cells seeded on a template (e.g., scaffold)
material-based slings and meshes; however, this and then implanting the resulting construct
treatment is limited by several issues, including in vivo may provide a solution to current unmet
(1) a tendency to perforate the other urogenital medical needs [7–9]. In order to engineer func-
organs, (2) retraction of the graft due to shrinkage tional tissues in vitro, the appropriate selection of
cells, scaffold material and structure, and biolog-
ical and mechanical cues is of the utmost impor-
tance. Scaffolding systems play a significant role
I.K. Ko (*) • A. Atala • J.J. Yoo in instructing cellular behavior and function, and
Wake Forest Institute for Regenerative Medicine,
they need to be properly selected in order to pro-
Wake Forest School of Medicine, Medical Center
Boulevard, Winston-Salem, NC 27157, USA vide an appropriate environment for cells.
e-mail: iko@wakehealth.edu Naturally derived or synthetically prepared

© Springer Nature Singapore Pte Ltd. 2018 87


B.W. Kim (ed.), Clinical Regenerative Medicine in Urology, DOI 10.1007/978-981-10-2723-9_4
88 I.K. Ko et al.

­materials [10–12] have been fabricated into scaf- Such bioreactor design enables the control of
folds for three-dimensional (3D) cell culture. environmental conditions for cell culture, thus
Additionally, biological (e.g., cytokines, growth allowing cells within the scaffold to undergo
factors) and mechanical cues need to be applied proper maturation and differentiation under
to the newly formed tissues, thus promoting more physiological environments that are similar to
functional and tissue-specific characteristics. in vivo conditions [13]. In this book chapter,
While numerous studies have produced func- basic components and design of bioreactor sys-
tional engineered tissue constructs that have been tems will be discussed, and potential uses of
tested in preclinical studies, the successful appli- bioreactor systems toward specific urogenital
cation of these technologies for clinical transla- applications will be presented.
tion requires several components, including a
large-scale system for producing therapeutic
cells, a well-designed three-dimensional (3D) 4.2 Fundamental Design
culture system with sufficient oxygen and nutri- and Types of Bioreactor
ent distribution, and appropriate biological and Systems
mechanical stimulation to the tissue constructs
that have been fabricated in vitro. Automated and 4.2.1 S
 pinner Flask Bioreactor
well-established bioreactor systems may provide System
a comprehensive solution.
Bioreactor systems have been used for Spinner flask bioreactors have been used for the
industrial fermentation, food processing, and culture of single cells (e.g., suspension cells)
the production of biological and pharmaceuti- and small-sized tissue constructs (Fig. 4.1a). To
cal drugs by mass culture [13]. Recently, biore- provide efficient and homogeneous mass trans-
actor-based mass tissue culture has been fer to cells during culture, culture medium is
successfully established as a method of pro- agitated by a stir bar, and the agitation speed
ducing sufficient numbers of therapeutic cells, can be easily controlled by altering the mag-
while conventional cell culture on culture netic forces used by the device [16]. Several
dishes is still incapable of producing the types of mammalian cells have been tested in
required cell quantities [14]. Bioreactor sys- spinner flask bioreactors for efficient cell
tems have also been used to produce 3D tissue expansion, including neural stem cells [17],
constructs in vitro [15]. Several conditions of hematopoietic stem cells [18], embryonic stem
the bioreactor system, including pH, tempera- cells [19], and induced pluripotent stem cells
ture, and nutrient and oxygen distribution in (iPSCs) [20, 21] (Fig.4.1b–d). 3D engineered
the culture medium, need to be controlled in an tissue constructs, such as cartilage tissue con-
efficient manner in order to maintain cell via- structs [22], have also been successfully cre-
bility and support cellular maturation [13]. For ated using this type of bioreactor. While the
instance, efficient delivery of nutrients and spinner flask bioreactor has been successfully
oxygen to seeded cells during bioreactor cul- applied to in vitro cell culture, uncontrolled
ture is crucial for cell survival. Appropriate convection flow within the system may affect
mechanical stimulation is another essential cue cell viability, so flow parameters need to be
that allows preconditioning of the newly well adjusted and controlled within specific cell
formed tissue under physiological conditions. culture systems [16].
4  Bioreactors for Regenerative Medicine in Urology 89

a b

60 100

c d
50
80

Cell concentration
40

(x104 cells/ml)

Viability (%)
60

30
40
20

20
10

0 0
0 2 4 6
Time (day)

Fig. 4.1  Spinner-flask bioreactor. (a) Schematic design rene beads and cell aggregations (c) and cell growth and
[13]. (b) Human iPSC cultures in a spinner-flask bioreac- viability during the culture (d) [21]. Reprinting of each
tor and the cultured iPSCs on the Matrigel-coated polysty- image was permitted from publishers

4.2.2 R
 otating Wall Vessel (RWV) connected to an oxygen and gas supply. The
Bioreactor System gravitational force experienced by single cells is
minimal (approximately 10−2 g), thus reducing
Unlike the spinner flask bioreactor, the rotating damage to cells and promoting cell viability and
wall vessel (RWV) bioreactor is designed to pro- tissue maturation during in vitro culture. The effi-
vide low shear stress and a homogeneous dynamic ciency of RWV bioreactors for in vitro cell cul-
cell culture environment [22] (Fig. 4.2a). The ture has been evaluated through histological,
RWV bioreactor system was originally devel- biochemical, and biomechanical analysis of sev-
oped by scientists at the National Aeronautics eral cell types that have been cultured in RWV
and Space Agency (NASA) for cell culture under bioreactors, including tumor cells [23, 24] and
low- or no-gravity conditions for targeted use in stem cells [25, 26] (Fig. 4.2b–d), and engineered
space. Briefly, the RWV system consists of tissue constructs, such as cartilage tissue con-
­culture medium that is horizontally rotated while structs [27, 28].
90 I.K. Ko et al.

a b

Fd
Fc
D

Fg

c d SOX1 GATA6 BRACHYURY


a b

c d

Fig. 4.2  Rotating wall vessel bioreactor. (a) Schematic using several markers (d) [26]. Reprinting of each image
design [13]. (b) A rotary bioreactor used for embryoid was permitted from publishers
body (EB) formation (c) and characterization of EBs

4.2.3 Hollow Fiber System material type (i.e., cellulose or synthetic mate-
rials), have been modified to improve dialysis
One representative application of hollow fiber efficiency [31].
bioreactors is used as a hemodialysis mem-
brane (dialyzer) for treating patients with kid-
ney failure [29]. A dialyzer contains a bundle 4.2.4 Hollow Fiber-Incorporated
of hollow fibers (≈10,000 fibers), each with an Perfusion Bioreactor
inner diameter of approximately 200 μm. As a
patient’s blood is perfused through the fibers, Recent progress in cell culture techniques and
blood waste is removed through the fiber mem- stem cell research has offered great promise in
brane, and the filtered blood is returned to the the field of cell-based therapies, and large-scale
patient. Ever since the first dialysis membrane production of therapeutic cells is required to
was developed by Lipps et al. using cellulose- address massive needs for clinical translation. To
derived hollow fibers [30], various types of this end, hollow fiber bioreactors have been used
materials have been developed to improve to scale up mammalian cell culture [32]. When
blood dialysis efficiency [31]. Dialysis effi- compared with stirred flask culture, cell culture
ciency primarily depends on the biocompati- within the hollow fiber system has several advan-
bility of the hollow fibers, which is influenced tages. Hollow fibers provide a large surface area
by the membrane components and flow proper- for cell inoculation, and cells can be seeded and
ties within the dialyzer. In addition, several cultured within the hollow fibers, thus creating
fiber parameters, such as diameter, molecular cultures with high cell densities. Appropriate
weight cutoff (MWCO), wall thickness, and ­perfusion through the hollow fibers is needed to
4  Bioreactors for Regenerative Medicine in Urology 91

efficiently provide oxygen and nutrients to seeded are developing in order to incorporate viable cells
cells. During bioreactor culture, shear stress must and thereby support metabolic function. One
be maintained at an appropriate level that does early study, performed by Sussman et al., used
not harm the cells in order to preserve cellular hollow fiber systems to temporarily support
viability during long-term cell culture. patients with liver failure [41]. The bioartificial
Hybridoma cell lines have been used to test the liver developed in this study, known as the extra-
feasibility of large-scale antibody production corporeal liver-assist device (ELAD®), was
[32], and recent studies have reported in vitro designed to seed immortalized liver cells into the
mass production of therapeutic cells [33] and hollow fibers of the system, maintain cell viabil-
stem cells [34], including embryonic stem cells ity, and provide metabolic function to blood per-
(ESCs) [35] and mesenchymal stem cells (MSCs) fused throughout the system. After ELAD®
[36, 37]. The hollow fiber system, in particular, showed promises in numerous preclinical trials
has been used as an appropriate environment for [42], the safety and efficiency of ELAD® in sup-
stem cell differentiation [34]. Several types of porting liver function were confirmed in clinical
cells, including hematopoietic stem cells (HSCs) trials [41], and the device is currently available
[38], ESCs [39], and iPSCs [40], have been effi- for liver failure patients [43] (Fig. 4.3b).
ciently differentiated into blood cells [38], Likewise, kidney-assist devices using viable
dopamine-­producing neurons [39], and hepato- kidney cells have been developed to address the
cytes [40], respectively. limitations of blood dialysis in the conventional
Another application of the hollow fiber-­ hemodialysis system [44]. To support renal bio-
incorporated perfusion system includes the logical functions that cannot be replicated by
development of bioartificial organs as a promis- conventional blood dialysis, Humes et al. [45]
ing strategy for treating organ failures. One developed a bioartificial kidney device by seed-
example is the use of bioartificial livers to tempo- ing pig-derived proximal renal cells into a hollow
rarily support liver function. In end-stage liver fiber kidney-assist device and implanting the
failure, hemodialysis and liver-assist systems are device into dogs with renal failure (Fig. 4.3a).
limited in their ability to remove toxins from the The bioartificial kidney system was able to facili-
blood and cannot replicate several other func- tate improved filtration capability, plasma param-
tions of the liver. However, liver-assist systems eters, and metabolism compared to a hemodialysis

a b a HepatAssistTM b Extracorporeal Liver Assist Device (ELAD)®


ELAD
bioreactors
extracapillary space
Plasma

fiber wall
Bioreactor

proximal tubule cells


luminal space Glucose
pump
heat
Charcoal
exchanger
Whole column Whole
blood blood Heat
pump 2
5-7 ml/min exchanger
5-10
mm Hg
ultrafiltrate RAD cartridge
pressure
reservoir c Modular Extracorporeal Liver Support b AMC-Bioartificial Liver (AMC-BAL)
monitor
heat
10-25 processed Oxygenation
exchanger
mm Hg ultrafiltrate
Cell Module

(urine)
Post hemofiter blood
Plasma

Bioreactor

(into RAD ECS)


Post RAD blood
ultrafiltrate
(into RAD replacement
luminal space) fluid
hemofilter
Detox Module

pump 3
70-80 ml/min Whole Oxygenation
Whole
blood
blood

pump 1
80 ml/min

Venous blood
Reservoir Albumin dialysis solution Low-flux dialysis filter
Waste Filter Plasmapheresis device
Dialysis solution High-flux dialysis filter Oxygenator

Fig. 4.3  Hollow fiber-incorporated perfusion bioreactor. (a) An extracorporeal perfusion unit for bioartificial kidney
[45], (b) various types of bioartificial livers [43]. Reprinting of each image was permitted from publishers
92 I.K. Ko et al.

system that did not incorporate cells. These and a majority of early studies were conducted in
results demonstrate that it is possible to develop 3D culture systems without in vitro bioreactor
an artificial kidney by incorporating viable renal systems. In an early study, we developed an engi-
cells into the conventional kidney dialysis system neered bladder tissue construct using dog-derived
and that such a cell-based artificial kidney may bladder-derived cells [51]. We used canine-­
contribute to the improvement of renal function derived allogeneic submucosa as a scaffold and
in kidney failure patients. then seeded urogenital and smooth muscle cells
into the scaffold for in vitro culture. The implan-
tation of this autologous cell-seeded bladder con-
4.3  pplication of Bioreactors
A struct resulted in an approximately 99% increase
for the Engineering in bladder urodynamic capacity, while the
of Urogenital Constructs implantation of cell-free scaffolds increased
bladder size by only 30%. Functional and histo-
As described previously, bioreactor systems have logical analysis demonstrated that the implanta-
been applied to the reconstruction of 3D engi- tion of the cell-seeded construct facilitated
neered tissues, and numerous types of urogenital normal bladder compliance, and the seeded cells
constructs, including bladder, urethra, and kidney showed normal cellular organization and pheno-
constructs, have been fabricated in vitro for typic properties along the harvested host tissue.
implantation in vivo. While most early studies These results demonstrate that a small biopsy
did not use bioreactor systems for 3D culture, the (≈1 cm2) is sufficient to obtain enough cells for
need to develop functional large-scale tissue con- engineering bladder tissue and that the implanta-
structs that mimic in vivo environments has tion of cell-seeded tissue constructs produces
necessitated the creation of well-designed biore- more bladder augmentation than the implantation
actor systems. of cell-free scaffolds [51].
The most critical challenge in the implanta-
tion of cell-seeded constructs is to accelerate host
4.3.1 Bladder vascularization that is able to provide seeded
cells with sufficient nutrients and oxygen for bet-
End-stage bladder disease is frequently treated ter cell survival within the implant. Schoeller
using grafts of native gastrointestinal segments; et al. tested whether pre-vascularization of the
however, the incorporation of the graft tissue into scaffold matrix would enhance bladder recon-
the host urinary tract often causes several compli- struction [49]. To achieve pre-vascularization,
cations, including metabolic disturbances, the authors utilized native host regenerative capa-
increase of mucosa production, and malignant bilities to fabricate a capsule pouch with host
diseases. Alternatively, natural and synthetic bio- vascularization. Pre-vascularized scaffolds were
materials have been used to address these issues, created by implanting silicone blocks into the
but several disadvantages, such as foreign body groins of rats for 1 week. Autologous urothelial
reactions, perforation of adjacent urogenital tis- cells were then seeded into these vascularized
sues, severe fibrosis, and poor host tissue integra- capsules, and the cell-seeded constructs were
tion, still remain unresolved [46]. Cell-based transposed into bladder wall defect sites. The
approaches utilizing viable bladder cells have group treated with the pre-vascularized cell-­
allowed researchers to recreate bladderlike struc- seeded scaffolds experienced a higher survival
tures in vitro, thus enabling functional bladder rate than the scaffold-only (80% mortality) and
reconstruction by in vivo implantation. Numerous saline groups (100% mortality). Histological
studies have reported the creation of bioengi- analysis of the cell-seeded constructs with pre-
neered bladderlike constructs using different fabricated capsule structures showed multilay-
types of scaffolds, such as those made from bio- ered urothelial cell lining along the transplantation
logical [47–51] and synthetic materials [7, 52], site. These results suggest that it is possible to
4  Bioreactors for Regenerative Medicine in Urology 93

pre-vascularize cell-seeded constructs using the muscle cells were cultured, seeded, and matured
host’s regenerative capacities, which may also within collagen-based scaffolds for several
contribute to accelerating urogenital tissue regen- weeks. The engineered bladder constructs were
eration in vivo [49]. Another study utilizing a then implanted into myelomeningocele patients
pre-vascularization strategy for the engineering using an omental wrap. In follow-up studies, we
of bladder constructs was conducted by found that bladder functions, such as leak point
Schultheiss et al. [47], who seeded autologous pressures and compliance, were improved in
urothelial and smooth muscle cells onto scaffolds patients who were implanted with our engineered
made of decellularized bowel segments. To pro- bladder constructs. These results suggest that our
vide the constructs with pre-vascularization bioengineered bladderlike construct can be used
capabilities, endothelial progenitor cells were to treat patients who need cystoplasty.
also incorporated into the engineered bladder Since the bladder tissue repeatedly undergoes
constructs, which underwent 3D in vitro culture dynamic circumstances (such as fill-void cycles)
before implantation. The implantation study daily, reconstruction of functional bladder tissue
revealed that bladder constructs seeded with in vitro is desirable in order to retain structural
endothelial cells efficiently prevented blood stability against mechanical stimulation [53]. To
thrombosis when compared to a control without this end, mechanical preconditioning can be
pre-vascularization capabilities, indicating that applied to the bladder construct to create a func-
pre-vascularization of an engineered bladderlike tional bladder that has mechanical properties sim-
construct can be used for the reconstruction of a ilar to those of bladders in in vivo environments
defective bladder. [15, 53] (Fig. 4.4a). Several approaches have been
More recently, our group successfully devel- used in order to improve the mechanical proper-
oped an approach to create engineered bladder ties of engineered bladder constructs. One early
constructs containing autologous bladder cell study attempted to test the feasibility of simulat-
sources for treating patients with end-stage blad- ing normal bladder physiology using a bioreactor
der disease [7]. Autologous urothelial and smooth system [54]. Wallis et al. developed a bioreactor

a b Regulator with
adjusting screw

Clamps Manometer

Seal

Cell-seeded UBM

Pressure

Media

Cell-seeded
UBM scaffold

Fig. 4.4  In vitro bioreactor systems for engineering bladder tissue constructs. (a) [15] and (b) [55]. Reprinting of each
image was permitted from publishers
94 I.K. Ko et al.

system that simulated mechanical conditions the implanted bladders developed morphologi-
occurring in normal bladder tissue, thereby allow- cally normal bladder structure, suggesting that
ing cell-seeded scaffolds to undergo cyclic the in vivo environment may be used as a func-
mechanical stimulation. The scaffolds, which tioning bioreactor system to reconstruct bladder
were seeded with bladder cells, were based on tissue for treating bladder augmentation
acellular porcine matrix or commercially avail- cystoplasty.
able small intestinal submucosa (SIS). In another study, Kajbafzadeh et al. used
Histological analysis demonstrated that the native bladder tissue as an in vivo bioreactor to
seeded urothelial and smooth muscle cells were determine appropriate scaffolding systems for
evident during bioreactor culture and that cellular bladder reconstruction [58]. They implanted sev-
structure aligned along the direction of applied eral scaffolds (such as pericardium, biofilm, and
pressure. The authors suggest that the bioreactor polyglycolic acid (PGA) scaffolds or a combina-
developed in this study was successful in promot- tion of each type of scaffold) between bladder
ing the formation of mechanically sound con- mucosa layers for several weeks and examined
structs and could be used to test the role of the immunological response and vascularization
mechanical stimulation on bladderlike tissue [54]. of the implants. Histological analysis revealed
In another study, Davis et al. [55] tested the that PGA-coated pericardium was the most effec-
effects of physiological bladder dynamics on cell tive scaffold in terms of reducing inflammation
viability using a bioreactor system (Fig. 4.4b). To and promoting efficient vascularization. This
engineer a bladderlike structure, they seeded result suggests that a combination of biodegrad-
human urothelial cells into a porcine bladder able materials with acellular matrices will pro-
matrix and compared cell viability and prolifera- duce appropriate scaffolding systems that
tive capability in the resulting construct with that optimize cell attachment in vitro and bladder tis-
of constructs grown in conventional static cul- sue reconstruction in vivo [58].
ture. Cell viability analysis demonstrated that
dynamic culture in the bioreactor significantly
improved cell viability and proliferation after 4 4.3.2 Urethra and Ureter
days of culture. The authors suggest that a biore-
actor system that provides mechanical stimula- Urethral defect is a common disease that occurs
tion is beneficial in recreating functional bladder secondary to urinary injuries, and treatment of
tissue in vitro. Similar studies using different bio- these defects is limited by a shortage of implant-
reactors have consistently confirmed these results able tissue [59]. Recent advances in tissue engi-
[25, 56]. neering and regenerative medicine have offered
Several studies have also utilized the microen- alternative solutions by producing functional ure-
vironments of native tissues or organs as “in vivo thral constructs in vitro. Although the cell-free
bioreactors” that retain their original regenerative scaffolding system has potential for use in treat-
capabilities for cell proliferation. While a few ing urethral defects [60], several reports suggest
studies have already used the in vivo environment that the use of cell-based urethral constructs may
for pre-vascularization of scaffolding systems provide better results without any graft failure
[49], Campbell et al. [57] used the peritoneal cav- and stricture formation. In an early study, De
ity as an “in vivo bioreactor” to bioengineer vis- Filippo et al. developed a 3D culture method to
ceral organs, such as the bladder and uterus. To engineer urethral constructs. Autologous rabbit
produce tissue grafts, they implanted appropriate bladder cells were expanded and seeded into
templates for different organs into rats or rabbits bladder submucosa, and the engineered urethral
for 2–3 weeks. The produced tissue, which was constructs were implanted into urethral defects in
found to contain a myofibroblast-rich cell popu- rabbits. To examine the effects of the seeded cells
lation, was then harvested and reimplanted into on urethral recovery, some animals were
the bladders (of what animals?). After 14 months, implanted with the scaffold alone. Histological
4  Bioreactors for Regenerative Medicine in Urology 95

and molecular analysis demonstrated that the then placed into the bioreactor, where cyclic
seeded cells formed normal urethral structures mechanical stimulation was applied to produce
1 month after implantation, and the neoforma- extended constructs. Histological analysis
tions retained epithelial and smooth muscle cell showed higher cell densities within these
phenotypes, as determined by Western blotting. extended constructs compared to those not given
Organ bath analysis confirmed contractility of mechanical stimulation (Fig. 4.5b). An in vivo
the harvested urethral tissue, indicating efficient study using dogs also revealed that implantation
neural integration. However, implantation of the of the extended constructs resulted in improved
scaffold alone caused graft failure, characterized urethral functional outcomes and normal struc-
by strictures and poor tissue formation. These tural morphology similar to that of autologous
results suggest that a tubularized tissue construct urethral tissue (Fig. 4.5c–d). These results sug-
with bladder cells can be fabricated and that such gest that mechanical stimulation in combination
a construct may contribute to the reconstruction with the use of appropriate cell types is necessary
of defective urethral tissues. Different cell for engineering functional urethral constructs and
sources have also been used to engineer urethral that mechanically stimulated implants better
tissue constructs. Since the epidermis seems to facilitate urethral reconstruction following
play an important role in the reconstruction of implantation.
urethral tissue, Li et al. [61, 62] established a cell Various approaches for reconstructing ureteral
culture method for the expansion of oral kerati- tissues have been developed in order to treat ure-
nocytes. They isolated keratinocytes from the teral defects and lesions due to surgical dissec-
epidermis of rabbits, incorporated the keratino- tion and traumatic injury. While a few studies
cytes into constructs seeded with various other have utilized cell-free scaffolding systems, such
cell types for engineering urethral tissues, and as biologic scaffolds [63, 64], more recent trials
implanted the engineered constructs into urethral have focused on cell-based strategies to engineer
mucosa defect sites. Histological analysis showed ureteral tissue constructs for replacement of ure-
that the constructs with keratinocytes facilitated teral defects [65, 66]. In several trials, functional
better integration with host epithelium than those ureteral tissues preconditioned with mechanical
without keratinocytes. In fact, animals that were stimulation have been fabricated using bioreactor
given the implants without keratinocytes devel- systems. Vardar et al. developed a flow bioreactor
oped inflammation, resulting in graft failure. system that mimics normal flow in the human
These results suggest that the incorporation of ureter [67]. Cyclic mechanical stimuli were
epidermal layers into urethral constructs is help- applied to collagen-based scaffolds seeded with
ful for promoting efficient mucosal integration human urothelial and smooth muscle cells, and
with the host [62]. phenotypic changes were examined by histologi-
In addition to the formation of epidermal lay- cal and molecular analysis. Dynamic culture
ers within engineered urethral constructs, under mechanical stimulation was found to sig-
mechanical stimulation has been applied to cre- nificantly improve tissue formation and facilitate
ate mature, functional, urethral constructs. Fu normal muscle tissue-specific phenotypes. These
et al. developed a robust bioreactor system that results also emphasize the importance of mechan-
permits cyclic mechanical stimulation (Fig. 4.5a) ical cues for the reconstruction of functional ure-
[59]. To create their constructs, they incorporated teral tissue, where a flow bioreactor system is
several cell types, such as adipose-derived stem necessary to provide cyclic mechanical stimula-
cells (ADSC) and oral epithelial cells, for muscu- tion. In another study using a bioreactor system,
lar and mucosal layers, respectively. The epithe- an automated bioreactor system able to control
lial cells were purified to obtain cells with higher physiological conditions and cell culture param-
proliferative capacity. Both types of cells were eters, such as pH and temperature, was estab-
seeded onto fibrous PGA mesh using a layer lished and used to produce tubular 3D tissue
seeding technique. The seeded construct was constructs [68].
96 I.K. Ko et al.

Peristaltic pump
a b

HE
Reaction Fluid collection bottle
chamber Culture media
PGA scaffold

AE1/AE3
HE AE1/AE3

c d
Un-extension

Un-extension

Un-extension
Extension

Extension

Extension

Fig. 4.5 (a) An in vitro bioreactor system for engineering Urethrography (left) and the harvested tissue images
urethral constructs [59]. (b) Gross and histological images (right). (d) Histological evaluation of the harvested tissue.
of the engineered urethral tissue construct in vitro. (c) Reprinting of each image was permitted from publisher

4.3.3 Kidney focused on the engineering of kidney tissue con-


structs using renal cells and scaffolding systems
Kidney transplantation is the only definitive treat- without bioreactor systems. Collagen-­based bio-
ment for end-stage renal disease, but the lack of materials have often been used as scaffolds due to
transplantable kidneys has resulted in an increase their biocompatibility and mechanical properties
in the number of patients waiting for treatment. when seeded with renal epithelial and mesangial
Recent advances in the field of regenerative medi- cells [69] and neonatal renal cells [70]. Our group
cine and tissue engineering have enabled scien- has also developed a collagen gel-­based culture
tists to recreate kidney tissue constructs in vitro, system to create 3D renal constructs using pri-
and preclinical trials have been performed to test mary renal cells [71]. To improve the mechanical
the feasibility of restoring renal function using properties of the engineered constructs, we used
these functional constructs. Important compo- materials such as hyaluronic acid [72], synthetic
nents of this treatment strategy include (1) the polycarbonate material [73], and PGA scaffolds
development of scaffolding systems, (2) the estab- [74]. When the engineered renal constructs were
lishment of kidney cell culture systems and 3D tested in preclinical studies, the implants main-
culture conditions, and (3) the designing of tained renal-specific structures [73, 74] and, inter-
in vitro bioreactor systems. Early efforts have estingly, produced urine-like fluids [73]. More
4  Bioreactors for Regenerative Medicine in Urology 97

recently, the development of induced pluripotent produce recellularized whole-kidney constructs


stem cells (iPSCs) has enabled the recreation of [79–81, 83–85].
kidney tissues in vivo [75–78]. By combining an An early study confirmed the importance of
appropriate scaffolding system with iPSCs, bioreactor culture for repopulation of a whole-­
researchers can produce large-scale renal con- kidney construct. Ross et al. developed a bioreac-
structs in vitro that can be used to treat renal fail- tor system to recellularize acellular rat
ure in clinical trials. Although the use of whole-kidney constructs using ESCs [81]. The use
implantable segments is a promising treatment for of ESCs for this purpose is advantageous due to
kidney failure, engineered segments may not able the high proliferative capacity and multi-­
to replicate functions performed by a whole-­sized differentiative ability of ESCs; however, several
kidney. To this end, an interesting approach to ethical and safety issues need to be addressed prior
producing organ-sized kidney constructs has been to using ESCs in clinical trials [86]. To examine
developed for whole-kidney transplantation. cellular viability and tissue formation, the authors
The concept of engineering whole-kidney con- first cultured ESCs seeded within whole-kidney
structs originated from utilizing native kidney scaffolds under static conditions. However, most
architecture as a cell-seeding template [79–81]. of the cells underwent apoptosis within a few days
The engineering process is composed of two tech- of culture, so the authors then established a perfu-
niques. First, cellular components are removed sion culture system based on a bioreactor system.
from the native kidney tissue in order to eliminate The perfusion culture system was equipped with
potential immune responses when the kidney con- a peristaltic pump, a tubing, a pressure monitor-
struct is implanted into a normal, non-­ ing system, and a cyclic beating system that
immunocompromised host [82]. This process, maintained constant physiologic pressure
called “decellularization,” is usually completed by (120/80 mmHg) with periodic beating (270–300
dynamically perfusing detergents through the beats/min), constant CO2, and constant tempera-
entire kidney tissue. The optimal decellularization ture. Over 10 days of perfusion culture, the whole-
process completely removes cell components, kidney constructs, which were seeded with ESCs
maintains an intact extracellular matrix, and pre- through the renal artery or ureter, maintained via-
serves vascular integrity. Preservation of the intact ble cells and facilitated proliferation of seeded
vascular network is particularly critical because it ESCs within tubular, vascular, and glomerular
allows for efficient blood perfusion following structures. A majority of the ESCs lost their pluri-
implantation [79, 82]. The decellularized kidney potency during bioreactor culture, implying that
scaffold is then recellularized in order to produce a they may have undergone renal differentiation. In
functional renal scaffold. In the recellularization a subsequent study, the authors also demonstrated
process, cells with kidney phenotypes are repopu- that the seeded ESCs were able to differentiate
lated within the acellular collagen-based kidney into the endothelial cell lineage [83]. Interestingly,
scaffold. When undifferentiated stem cells are the attachment of mouse ESCs induced remodel-
used to recellularize the kidney scaffolds, the ing of the basement membrane in the rat-derived
seeded cells need to undergo the differentiation kidney matrix, suggesting a possible strategy for
process for renal lineages. The use of a bioreactor clinical xenotransplantation. Overall, these results
is essential in the recellularization process for the suggest that acellular whole-kidney scaffolds can
creation of functional whole-kidney constructs be repopulated with pluripotent cells using a biore-
in vitro. Unlike the previously mentioned trials actor system with simulated physiological condi-
that produced small scaffold segments with low tions in order to form renal tissue.
cell-seeding densities, whole-kidney scaffolds are Other studies focus on monitoring physical
seeded with a high number of cells and therefore and biochemical function during perfusion biore-
require efficient perfusion to maintain cellular actor culture. The examination of viable and
viability throughout the entire scaffold. Several functional kidney constructs for implantation is
types of bioreactors have thus been developed to preferably done in a noninvasive manner. Uzarski
98 I.K. Ko et al.

et al. developed a bioreactor system that allowed delivered via the renal artery and ureter, respec-
them to examine renal function in terms of cell tively. The seeded kidney constructs were then
viability and renal-specific behaviors [84] (ref). cultured in a bioreactor system, where constant
Using the bioreactor system, the authors were flow was maintained through the renal artery at
able to measure decellularization efficiency in a 1.5 ml/min and 5% CO2 was applied with passive
noninvasive manner, monitor (arterial?) pressure drainage through the renal vein and ureter.
changes due to the level of recellularization, and Histological and functional analysis demonstrated
evaluate recellularization efficiency by measur- that the seeded kidney constructs induced partial
ing cell viability (resazurin) and renal function re-endothelialization and renal tubule formation,
(alumin, kidney injury molecule-1) through sam- and tissue formation was confirmed by endothelial
pling the culture medium. The results demon- and renal functional tests. When the kidney con-
strate that this bioreactor system may be struct was orthotopically implanted in a rat, can-
successfully used to scale up and broaden the nulation between the engineered construct and the
applications of whole-kidney technology. host animal was successfully performed, blood
In addition to in vitro recreation of whole-­ perfusion was maintained for several hours, and
kidney constructs, the possibility of implanting production of urine-like fluid was confirmed.
whole-kidney constructs that were created in vitro These results suggest that the bioengineering of a
needs to be tested prior to clinical translation. whole-kidney construct is possible and that the
Song et al. fabricated a whole-kidney construct developed kidney construct may be translatable
using rat-derived kidney tissue and tested it in a for use in clinical trials. For clinical translation,
preclinical rat study [80]. They developed a biore- several issues need to be addressed, including (1)
actor system (Fig. 4.6a) to use endothelial and kid- scaling-up, (2) functional and homogeneous re-­
ney cells to recellularize kidney scaffolds made endothelialization, and (3) kidney-specific recel-
from decellularized native rat, pig, and human lularization of engineered kidney constructs using
renal tissue. The endothelial and kidney cells were clinically available cell sources [87].

a b d f

a A B b A B c b

Ra C Ra
a
K -40 mm Hg K c e

U U
g

Flow
d c
3-way
connector
Pump

Pulse dampner

Fig. 4.6  In vitro bioreactor systems for engineering kid- and (c) perfusion bioreactor system for recellularization
ney constructs. (a) In vitro recellularization of a rat kidney of a whole-pig-kidney construct [85]. Reprinting of each
scaffold with endothelial and kidney cells [80]. (b) High-­ image was permitted from publisher
throughput system for pig kidney decellularization [90]
4  Bioreactors for Regenerative Medicine in Urology 99

To address these challenges, our group first ­tubular structures within the kidney constructs
worked on the decellularization of clinical-sized [85]. Cells were seeded through an extravascular
kidney scaffolds, particularly those from pig-­ route by direct injection into the kidney paren-
derived kidney sources. Pig and human kidneys are chyma, and the seeded construct was maintained
similarly sized, and porcine-derived tissue materi- in the bioreactor system to promote the formation
als, such as the heart valve [88] and small intestinal of renal structures. The newly formed tubules
submucosa (SIS) [89], have been used in the clinic. showed functional characteristics, such as elec-
Through decellularization of pig kidneys, we suc- trolyte and protein readsorption, amino acid
cessfully developed acellular kidney scaffolds that transport capability, and erythropoietin produc-
maintained vascular architecture and an intact tion. These results suggest that our repopulation
extracellular matrix (ECM) [90] (Fig. 4.6b). One method in combination with bioreactor culture
critical challenge in developing this technology was will help promote the formation of tubular struc-
maintaining long-term blood flow so that perfused tures within the recellularized porcine kidney
blood would be able to provide nutrients and oxy- constructs. The ongoing work is investigating the
gen to the seeded kidney cells following scaffold long-term implantation of engineered whole-­
implantation. To provide antithrombogenic capac- kidney constructs that are seeded with autolo-
ity to the acellular kidney scaffolds, we tested a gous endothelial and renal cells.
method for re-­endothelialization of the porcine kid-
ney constructs using a bioreactor system [79]. Our
bioreactor system allowed static and dynamic cul- 4.4 Future Perspectives
ture for functional and efficient re-­endothelialization
of the acellular kidney constructs, as confirmed by To date, the application of bioreactors in the fields
in vitro and in vivo studies. Since we observed of regenerative medicine and tissue engineering
endothelial cell detachment, which caused further has focused on mass cell culture and the creation
blood clots on the vascular surface, we introduced of functional urological tissue constructs in vitro.
CD31 antibody conjugation technology into the Dynamic cell culture in bioreactors has enabled
vascular lumen in order to better maintain re-­ better tissue grafting and functional outcomes
endothelialization of the constructs. We hypothe- in vivo. To apply bioreactor systems for clinical
sized that the conjugated CD31 antibody would translation, several things need to be considered
firmly retain endothelial cells by interacting with (Fig. 4.7) [91]. First, bioreactor systems need to
the antigen on the endothelial cells. Our in vivo produce engineered functional tissue products in a
implantation study demonstrated that re-­consistent manner. One possible method is the
endothelialization in combination with CD31 anti- development of sensor-based bioreactors that may
body conjugation facilitated better blood perfusion minimize variations in terms of product controls.
than re-endothelialization alone, indicating the fea- Second, in terms of regulatory aspects, bioreactor
sibility of using this anticoagulation strategy when systems for clinical purposes need to satisfy safety
implanting whole-kidney constructs [79]. guidelines and offer traceability. The overall
In a parallel study, we also developed a recel- processes of bioreactor systems need to be in
lularization method for creating functional renal compliance with clear specifications. For com-
constructs [85]. To test the feasibility of expand- mercialization of engineered tissue products, an
ing cells from a clinically relevant source, we automated bioreactor system needs to be built for
established a culture method that allowed us to cost-effective, standardized production and scal-
expand primary renal cells isolated from human ing-up of engineered tissue constructs [91].
renal tissues (Fig. 4.6c) [71]. Based on histologi- Other applications of bioreactors for urologi-
cal analysis, a majority of the cell population cal regenerative medicine include (1) a microar-
demonstrated proximal tubular phenotypes. We ray bioreactor for drug screening (Fig. 4.8a) [92,
used these proximal tubular cells to develop a 93] and (2) a microfluidic bioreactor for mainte-
recellularization method to efficiently form nance of reproductive tissue (e.g., sperm)
100 I.K. Ko et al.

a b
Disposable bioreactor Graphical programming interface
GO
Verification of core biology platform 1

Identification and Validation of


Specification of Assessment of
Corrections validation of methods to
clinical target culture parameter
biological determine cell
application sensitivity
markers identity & purity
Industry
linkage

Verification of pre-clinical platform 2

Design of Reproducible Risk analysis &


Corrections Preliminary
scaleable and graft engineering mitigation for
assessment of
streamlined and QC techniques bioreactor-based
for animal studies economic model
bioprocesses graft production
Industry
linkage

Automated instrument Translation to clinical platform 3

Implementation Review of clinical Update & deploy Training of


Corrections of potency plan by clinical and regulatory and surgeons and
biomarkers for regulatory reimbursement education of
quality control advisory group strategy patients/clinics

Detachable cell output vial and Commercial bioreactor-based tissue product


integrated transport container

TRENDS in Biotechnology TRENDS in Biotechnology

Fig. 4.7  Translation of tissue engineering strategies [91]. engineered products into the clinic. Reprinting of each
(a) Potential model of a commercial bioreactor-based sys- image was permitted from publisher
tem. (b) Proposed road map for translating ­bioreactor-­based

Monolayer ECM-coated porous


microcarrier beads
a
Clinical tumor
biopsy from patient
RX
Sereening in
2
Slow-turning patient-specific Identification
lateral vessel (STLV) tumor models of optimal drug
for patient

Bead movement Genetic profiling for


in STLV precision medicine
Filling port oncology
Docking point RX RX RX RX
1 2 3 1
and air supply
Drug choices based Best ‘guess’ drug
Gas exchange Sampling ports with on genetic profile/ therapy
membrane luer lock faucets tumor mutations
(central core)
b Tissue
Skin biopsy from engineer 3D
patient target
Lung organoid
mucosal Basement system
epithelium membrane
Collagen
Basement coating
membrane Porous Generate iPSC; differentiate
bead into tissue of interest
Collagen
coating RX RX RX RX
Porous 1 2 3 3
bead Screen drugs for Identification of best
onboard system treatment for individual

Fig. 4.8  Various urological applications using bioreactor s­ ystem to use biofabricated tissues in personalized medi-
systems. (a) Rotating wall vessel bioreactor system for cine [98]. Reprinting of each image was permitted from
studying host-pathogen interactions using organotypic 3D publisher
cell culture models [93]. (b) Microfluidic bioreactor
4  Bioreactors for Regenerative Medicine in Urology 101

[94, 95]. For example, there has been particular 13. Martin I, Wendt D, Heberer M. The role of biore-
actors in tissue engineering. Trends Biotechnol.
interest in using bioreactors for drug screening
2004;22:80–6.
and developing patient-specific treatments [96– 14. Martin Y, Vermette P. Bioreactors for tissue mass cul-
98] (Fig. 4.8b). More active research thrusts ture: design, characterization, and recent advances.
focused on new applications of bioreactors for Biomaterials. 2005;26:7481–503.
15. Farhat WA, Yeger H. Does mechanical stimulation
treating urological diseases, and complications
have any role in urinary bladder tissue engineering?
will be expected in the future. World J Urol. 2008;26:301–5.
16. Ismadi MZ, Hourigan K, Fouras A. Experimental

Acknowledgment  I wish to thank Ms. Alicia Lee for edi- characterization of fluid mechanics in a spinner flask
torial assistance with this manuscript. bioreactor. PRO. 2014;2:753–72.
17. Kallos MS, Sen A, Behie LA. Large-scale expansion
of mammalian neural stem cells: a review. Med Biol
Eng Comput. 2003;41:271–82.
References 18. Cabrita GJ, Ferreira BS, da Silva CL, Goncalves

R, Almeida-Porada G, Cabral JM. Hematopoietic
1. Ko IK, Atala A, Yoo JJ. Tissue engineering of the stem cells: from the bone to the bioreactor. Trends
urogenital system. In: Tissue engineering: principles Biotechnol. 2003;21:233–40.
and practices. Taylor & Francis Group, vol. 34: CRC 19. Zandstra PW, Bauwens C, Yin T, Liu Q, Schiller
Press; 2012. p. 31–9. H, Zweigerdt R, Pasumarthi KB, Field LJ. Scalable
2. Ninkovic M, Dabernig W. Flap technology for recon- production of embryonic stem cell-derived cardio-
­
structions of urogenital organs. Curr Opin Urol. myocytes. Tissue Eng. 2003;9:767–78.
2003;13:483–8. 20. Ismadi MZ, Gupta P, Fouras A, Verma P, Jadhav S,
3. Keys T, Badlani G. The scientific rationale for using Bellare J, Hourigan K. Flow characterization of a
biomaterials in stress urinary incontinence and pelvic spinner flask for induced pluripotent stem cell culture
organ prolapse. Curr Urol Rep. 2011;12:393–5. application. PloS One. 2014;9:e106493.
4. Comiter CV. Surgery insight: management of failed 21. Ashok P, Fan Y, Rostami MR, Tzanakakis
sling surgery for female stress urinary incontinence. ES. Aggregate and microcarrier cultures of human
Nat Clin Pract Urol. 2006;3:666–74. pluripotent stem cells in stirred-suspension systems.
5. Devaseelan P, Fogarty P. The role of synthetic mesh Methods Mol Biol. 2016;1502:35–52.
in the treatment of pelvic organ prolapse. Obstet 22. Unsworth BR, Lelkes PI. Growing tissues in micro-
Gynaecol. 2009;11:1–9. gravity. Nat Med. 1998;4:901–7.
6. Baessler K, Maher CF. Mesh augmentation during 23. Rhee HW, Zhau HE, Pathak S, Multani AS, Pennanen
pelvic-floor reconstructive surgery: risks and benefits. S, Visakorpi T, Chung LW. Permanent phenotypic and
Curr Opin Obstet Gynecol. 2006;18:560–6. genotypic changes of prostate cancer cells cultured in
7. Atala A, Bauer SB, Soker S, Yoo JJ, Retik AB. Tissue-­ a three-dimensional rotating-wall vessel. In Vitro Cell
engineered autologous bladders for patients needing Dev Biol Anim. 2001;37:127–40.
cystoplasty. Lancet. 2006;367:1241–6. 24. Licato LL, Prieto VG, Grimm EA. A novel preclinical
8. Raya-Rivera A, Esquiliano DR, Yoo JJ, Lopez-­ model of human malignant melanoma utilizing bio-
Bayghen E, Soker S, Atala A. Tissue-engineered autol- reactor rotating-wall vessels. In Vitro Cell Dev Biol
ogous urethras for patients who need reconstruction: Anim. 2001;37:121–6.
an observational study. Lancet. 2011;377:1175–82. 25. Wei X, Li DB, Xu F, Wang Y, Zhu YC, Li H, Wang
9. Raya-Rivera AM, Esquiliano D, Fierro-Pastrana R, KJ. A novel bioreactor to simulate urinary blad-
Lopez-Bayghen E, Valencia P, Ordorica-Flores R, der mechanical properties and compliance for blad-
Soker S, Yoo JJ, Atala A. Tissue-engineered autolo- der functional tissue engineering. Chin Med J.
gous vaginal organs in patients: a pilot cohort study. 2011;124:568–73.
Lancet. 2014;384:329–36. 26. Lei X, Deng Z, Duan E. Uniform Embryoid body
10. Khodabukus A, Baar K. Regulating fibrinolysis to production and enhanced Mesendoderm differ-
engineer skeletal muscle from the C2C12 cell line. entiation with murine embryonic stem cells in a
Tissue engineering part C. Methods. 2009;15:501–11. rotary suspension bioreactor. Methods Mol Biol.
11. Huang YC, Dennis RG, Larkin L, Baar K. Rapid for- 2016;1502:63–75.
mation of functional muscle in vitro using fibrin gels. 27. Vunjak-Novakovic G, Martin I, Obradovic B, Treppo
J Appl Physiol. 2005;98:706–13. S, Grodzinsky AJ, Langer R, Freed LE. Bioreactor
1 2. Levenberg S, Rouwkema J, Macdonald M, Garfein cultivation conditions modulate the composition and
ES, Kohane DS, Darland DC, Marini R, van mechanical properties of tissue-engineered cartilage.
Blitterswijk CA, Mulligan RC, D'Amore PA, Langer J Orthopaedic Res. 1999;17:130–8.
R. Engineering vascularized skeletal muscle tissue. 28. Pei M, Solchaga LA, Seidel J, Zeng L, Vunjak-­
Nat Biotechnol. 2005;23:879–84. Novakovic G, Caplan AI, Freed LE. Bioreactors
102 I.K. Ko et al.

mediate the effectiveness of tissue engineering scaf- 43. Struecker B, Raschzok N, Sauer IM. Liver sup-

folds. FASEB J. 2002;16:1691–4. port strategies: cutting-edge technologies. Nat Rev
29. Sakai K. Dialysis membranes for blood purification. Gastroenterol Hepatol. 2014;11:166–76.
Front Med Biol Eng. 2000;10:117–29. 44. Jansen J, Fedecostante M, Wilmer MJ, van den Heuvel
30. Lipps B, Stewart RD, Perkins H, Holmes GW,
LP, Hoenderop JG, Masereeuw R. Biotechnological
McLain EA, Rolfs MR, Oja P. The hollow fiber challenges of bioartificial kidney engineering.
artificial kidney. Trans Am Soc Artif Intern Organs. Biotechnol Adv. 2014;32:1317–27.
1967;13:200–7. 45. Humes HD, Buffington DA, MacKay SM, Funke

31. Clark WR. Hemodialyzer membranes and con-
AJ, Weitzel WF. Replacement of renal function in
figurations: a historical perspective. Semin Dial. uremic animals with a tissue-engineered kidney. Nat
2000;13:309–11. Biotechnol. 1999;17:451–5.
32. Patankar D, Oolman T. Wall-growth hollow-fiber
46. Gleeson MJ, Griffith DP. The use of alloplas-

reactor for tissue culture: I. Preliminary experiments tic biomaterials in bladder substitution. J Urol.
Biotechnology and bioengineering. 1990;36:97–103. 1992;148:1377–82.
33. Storm MP, Sorrell I, Shipley R, Regan S, Luetchford 47. Schultheiss D, Gabouev AI, Cebotari S, Tudorache
KA, Sathish J, Webb S, Ellis MJ. Hollow fiber biore- I, Walles T, Schlote N, Wefer J, Kaufmann PM,
actors for in vivo-like mammalian tissue culture. J Vis Haverich A, Jonas U, Stief CG, Mertsching
Exp. 2016;111:53431. H. Biological vascularized matrix for bladder tissue
34. Wung N, Acott SM, Tosh D, Ellis MJ. Hollow fibre engineering: matrix preparation, reseeding technique
membrane bioreactors for tissue engineering applica- and short-term implantation in a porcine model. J
tions. Biotechnol Lett. 2014;36:2357–66. Urol. 2005;173:276–80.
35. Roberts I, Baila S, Rice RB, Janssens ME, Nguyen K, 48. Danielsson C, Ruault S, Basset-Dardare A, Frey

Moens N, Ruban L, Hernandez D, Coffey P, Mason P. Modified collagen fleece, a scaffold for trans-
C. Scale-up of human embryonic stem cell culture plantation of human bladder smooth muscle cells.
using a hollow fibre bioreactor. Biotechnol Lett. Biomaterials. 2006;27:1054–60.
2012;34:2307–15. 49. Schoeller T, Lille S, Stenzl A, Ninkovic M, Piza

36. Nold P, Brendel C, Neubauer A, Bein G, Hackstein H, Otto A, Russell RC, Wechselberger G. Bladder
H. Good manufacturing practice-compliant animal-­ reconstruction using a prevascularized capsular tissue
free expansion of human bone marrow derived seeded with urothelial cells. J Urol. 2001;165:980–5.
mesenchymal stroma cells in a closed hollow-fiber-­ 50. Kanematsu A, Yamamoto S, Noguchi T, Ozeki M,
based bioreactor. Biochem Biophys Res Commun. Tabata Y, Ogawa O. Bladder regeneration by bladder
2013;430:325–30. acellular matrix combined with sustained release of
37. Usuludin SB, Cao X, Lim M. Co-culture of stro- exogenous growth factor. J Urol. 2003;170:1633–8.
mal and erythroleukemia cells in a perfused hollow 51. Yoo JJ, Meng J, Oberpenning F, Atala A. Bladder
fiber bioreactor system as an in vitro bone marrow augmentation using allogenic bladder submucosa
model for myeloid leukemia. Biotechnol Bioeng. seeded with cells. Urology. 1998;51:221–5.
2012;109:1248–58. 52. Pattison MA, Wurster S, Webster TJ, Haberstroh

38. Housler GJ, Miki T, Schmelzer E, Pekor C, Zhang KM. Three-dimensional, nano-structured PLGA scaf-
X, Kang L, Voskinarian-Berse V, Abbot S, Zeilinger folds for bladder tissue replacement applications.
K, Gerlach JC. Compartmental hollow fiber capillary Biomaterials. 2005;26:2491–500.
membrane-based bioreactor technology for in vitro 53. Korossis S, Bolland F, Ingham E, Fisher J, Kearney J,
studies on red blood cell lineage direction of hema- Southgate J. Review: tissue engineering of the urinary
topoietic stem cells. Tissue Eng Part C Methods. bladder: considering structure-function relationships
2012;18:133–42. and the role of mechanotransduction. Tissue Eng.
39. Yamazoe H, Iwata H. Efficient generation of dopa- 2006;12:635–44.
minergic neurons from mouse embryonic stem 54. Wallis MC, Yeger H, Cartwright L, Shou Z, Radisic M,
cells enclosed in hollow fibers. Biomaterials. Haig J, Suoub M, Antoon R, Farhat WA. Feasibility
2006;27:4871–80. study of a novel urinary bladder bioreactor. Tissue
40. Amimoto N, Mizumoto H, Nakazawa K, Ijima H, Eng Part A. 2008;14:339–48.
Funatsu K, Kajiwara T. Hepatic differentiation of 55.
Davis NF, Mooney R, Piterina AV, Callanan
mouse embryonic stem cells and induced pluripotent A, McGuire BB, Flood HD, McGloughlin
stem cells during organoid formation in hollow fibers. TM. Construction and evaluation of urinary bladder
Tissue Eng Part A. 2011;17:2071–8. bioreactor for urologic tissue-engineering purposes.
41. Sussman NL, Kelly JH. Extracorporeal liver support: Urology. 2011;78:954–60.
cell-based therapy for the failing liver. Am J Kidney 56. Davis NF, Callanan A. Development of a bladder
Dis. 1997;30:S66–71. bioreactor for tissue engineering in urology. Methods
42. Kelly JH, Koussayer T, He D, Chong MG, Shang TA, Mol Biol. 2016;1502:213–21.
Whisennand HH, Sussman NL. Assessment of an 57. Campbell GR, Turnbull G, Xiang L, Haines M,

extracorporeal liver assist device in anhepatic dogs. Armstrong S, Rolfe BE, Campbell JH. The peritoneal
Artif Organs. 1992;16:418–22. cavity as a bioreactor for tissue engineering visceral
4  Bioreactors for Regenerative Medicine in Urology 103

organs: bladder, uterus and vas deferens. J Tissue Eng tion of human kidney structures for renal cell therapy.
Regen Med. 2008;2:50–60. Nephrol Dial Transplant. 2012;27:3082–90.
58. Kajbafzadeh AM, Esfahani SA, Sadeghi Z, Elmi A, 72. Rosines E, Johkura K, Zhang X, Schmidt HJ,

Monajemzadeh M. Application of different scaffolds Decambre M, Bush KT, Nigam SK. Constructing
for bladder wall regeneration: the bladder as a natural kidney-like tissues from cells based on programs
bioreactor. Tissue Eng Part A. 2012;18:882–7. for organ development: toward a method of in vitro
59. Fu Q, Deng CL, Zhao RY, Wang Y, Cao Y. The effect tissue engineering of the kidney. Tissue Eng Part A.
of mechanical extension stimulation combined with 2010;16:2441–55.
epithelial cell sorting on outcomes of implanted 73. Yoo JJ, Ashkar S, Atala A. Creation of functional
tissue-engineered muscular urethras. Biomaterials. kidney structures with excretion of kidney-like fluid
2014;35:105–12. in vivo. Pediatrics. 1996;98(suppl):605.
60. le Roux PJ. Endoscopic urethroplasty with unseeded 74. Kim SS, Park HJ, Han J, Choi CY, Kim BS. Renal
small intestinal submucosa collagen matrix grafts: a tissue reconstitution by the implantation of renal
pilot study. J Urol. 2005;173:140–3. segments on biodegradable polymer scaffolds.
61. Li C, Xu Y, Song L, Fu Q, Cui L, Yin S. Preliminary Biotechnol Lett. 2003;25:1505–8.
experimental study of tissue-engineered urethral 75. Taguchi A, Kaku Y, Ohmori T, Sharmin S, Ogawa M,
reconstruction using oral keratinocytes seeded on Sasaki H, Nishinakamura R. Redefining the in vivo
BAMG. Urol Int. 2008;81:290–5. origin of metanephric nephron progenitors enables
62. Li C, Xu YM, Song LJ, Fu Q, Cui L, Yin S. Urethral generation of complex kidney structures from plu-
reconstruction using oral keratinocyte seeded bladder ripotent stem cells. Cell Stem Cell. 2014;14:53–67.
acellular matrix grafts. J Urol. 2008;180:1538–42. 76. Sharmin S, Taguchi A, Kaku Y, Yoshimura Y, Ohmori
63. Dahms SE, Piechota HJ, Nunes L, Dahiya R, Lue TF, T, Sakuma T, Mukoyama M, Yamamoto T, Kurihara
Tanagho EA. Free ureteral replacement in rats: regen- H, Nishinakamura R. Human induced pluripotent
eration of ureteral wall components in the acellular stem cell-derived podocytes mature into vascularized
matrix graft. Urology. 1997;50:818–25. glomeruli upon experimental transplantation. J Am
64. Osman Y, Shokeir A, Gabr M, El-Tabey N, Mohsen Soc Nephrol. 2016;27:1778–91.
T, El-Baz M. Canine ureteral replacement with long 77. Lam AQ, Freedman BS, Morizane R, Lerou PH,

acellular matrix tube: is it clinically applicable? J Valerius MT, Bonventre JV. Rapid and efficient dif-
Urol. 2004;172:1151–4. ferentiation of human pluripotent stem cells into
65. Shen J, Fu X, Ou L, Zhang M, Guan Y, Wang
intermediate mesoderm that forms tubules expressing
K, Che Y, Kong D, Steinhof G, Li W, Yu Y, Ma kidney proximal tubular markers. J Am Soc Nephrol.
N. Construction of ureteral grafts by seeding urothe- 2014;25:1211–25.
lial cells and bone marrow mesenchymal stem cells 78. Morizane R, Monkawa T, Itoh H. Differentiation of
into polycaprolactone-­lecithin electrospun fibers. Int murine embryonic stem and induced pluripotent stem
J Artif Organs. 2010;33:161–70. cells to renal lineage in vitro. Biochem Biophys Res
66. Shi JG, Fu WJ, Wang XX, Xu YD, Li G, Hong BF, Commun. 2009;390:1334–9.
Wang Y, Du ZY, Zhang X. Tissue engineering of ure- 79. Ko IK, Abolbashari M, Huling J, Kim C, Mirmalek
teral grafts by seeding urothelial differentiated hAD- Sani SH, Moradi M, Orlando G, Jackson JD,
SCs onto biodegradable ureteral scaffolds. J Biomed AbouShwareb T, Soker S, Yoo JJ, Atala A. Enhanced
Mater Res A. 2012;100:2612–22. re-endothelialization of acellular kidney scaffolds for
67. Vardar E, Engelhardt EM, Larsson HM, Mouloungui whole organ engineering via antibody conjugation of
E, Pinnagoda K, Hubbell JA, Frey P. Tubular com- vasculatures. Technology. 2014;2:243–53.
pressed collagen scaffolds for ureteral tissue engi- 80. Song JJ, Guyette JP, Gilpin SE, Gonzalez G, Vacanti
neering in a flow bioreactor system. Tissue Eng Part JP, Ott HC. Regeneration and experimental orthotopic
A. 2015;21:2334–45. transplantation of a bioengineered kidney. Nat Med.
68. Seifarth V, Gossmann M, Janke HP, Grosse JO,
2013;19:646–51.
Becker C, Heschel I, Artmann GM, Temiz Artmann 81. Ross EA, Williams MJ, Hamazaki T, Terada N,

A. Development of a bioreactor to culture tissue engi- Clapp WL, Adin C, Ellison GW, Jorgensen M, Batich
neered ureters based on the application of tubular CD. Embryonic stem cells proliferate and differen-
OPTIMAIX 3D scaffolds. Urol Int. 2015;95:106–13. tiate when seeded into kidney scaffolds. J Am Soc
69. Wang PC, Takezawa T. Reconstruction of renal glo- Nephrol. 2009;20:2338–47.
merular tissue using collagen vitrigel scaffold. J 82.
Arenas-Herrera JE, Ko IK, Atala A, Yoo
Biosci Bioeng. 2005;99:529–40. JJ. Decellularization for whole organ bioengineering.
70. Lu SH, Lin Q, Liu YN, Gao Q, Hao T, Wang Y, Zhou Biomed Mater. 2013;8:014106.
J, Wang H, Du Z, Wu J, Wang CY. Self-assembly of 83. Ross EA, Abrahamson DR, John PS, Clapp WL,
renal cells into engineered renal tissues in collagen/ Williams MJ, Terada N, Hamazaki T, Ellison GW,
Matrigel scaffold in vitro. J Tissue Eng Regen Med. Batich CD. Mouse stem cells seeded into decel-
2012;6:786–92. lularized rat kidney scaffolds endothelialize and
71.
Guimaraes-Souza NK, Yamaleyeva LM, remodel basement membranes. Organogenesis.
AbouShwareb T, Atala A, Yoo JJ. In vitro reconstitu- 2012;8:49–55.
104 I.K. Ko et al.

84. Uzarski JS, Bijonowski BM, Wang B, Ward HH,


egies into clinical products. Trends Biotechnol.
Wandinger-Ness A, Miller WM, Wertheim JA. Dual-­ 2009;27:495–502.
purpose bioreactors to monitor noninvasive physi- 92. Radtke AL, Herbst-Kralovetz MM. Culturing and

cal and biochemical markers of kidney and liver applications of rotating wall vessel bioreactor derived
scaffold recellularization. Tissue Eng C Methods. 3D epithelial cell models. J Vis Exp. 2012;62:3868.
2015;21:1032–43. 93. Barrila J, Radtke AL, Crabbe A, Sarker SF, Herbst-­
85.
Abolbashari M, Agcaoili SM, Lee MK, Ko Kralovetz MM, Ott CM, Nickerson CA. Organotypic
IK, Aboushwareb T, Jackson JD, Yoo JJ, Atala 3D cell culture models: using the rotating wall ves-
A. Repopulation of porcine kidney scaffold sel to study host-pathogen interactions. Nat Rev
using porcine primary renal cells. Acta Biomater. Microbiol. 2010;8:791–801.
2016;29:52–61. 94. Komeya M, Kimura H, Nakamura H, Yokonishi
86. Martin GR. Isolation of a pluripotent cell line from T, Sato T, Kojima K, Hayashi K, Katagiri K,
early mouse embryos cultured in medium conditioned Yamanaka H, Sanjo H, Yao M, Kamimura S, Inoue
by teratocarcinoma stem cells. Proc Natl Acad Sci U K, Ogonuki N, Ogura A, Fujii T, Ogawa T. Long-
S A. 1981;78:7634–8. term ex vivo maintenance of testis tissues produc-
87. Kim IH, Ko IK, Atala A, Yoo JJ. Whole kidney engi- ing fertile sperm in a microfluidic device. Sci Rep.
neering for clinical translation. Curr Opin Organ 2016;6:21472.
Transplant. 2015;20:165–70. 95. McCormack MC, McCallum S, Behr B. A novel

88. Lawton JS, Moazami N, Pasque MK, Moon MR,
microfluidic device for male subfertility screening. J
Damiano RJ Jr. Early stenosis of Medtronic mosaic Urol. 2006;175:2223–7. discussion 2227
porcine valves in the aortic position. J Thorac 96. Schneider M, Schuler J, Hofflin R, Korzeniewski N,
Cardiovasc Surg. 2009;137:1556–7. Grullich C, Roth W, Teber D, Hadaschik B, Pahernik
89. Iannotti JP, Codsi MJ, Kwon YW, Derwin K, Ciccone S, Hohenfellner M, Duensing S. Phenotypic drug
J, Brems JJ. Porcine small intestine submucosa aug- screening and target validation for improved person-
mentation of surgical repair of chronic two-tendon alized therapy reveal the complexity of phenotype-­
rotator cuff tears. A randomized, controlled trial. J genotype correlations in clear cell renal cell
Bone Joint Surg Am. 2006;88:1238–44. carcinoma. Urol Oncol. 2014;32:877–84.
90. Sullivan DC, Mirmalek-Sani SH, Deegan DB,
97. Skardal A, Devarasetty M, Forsythe S, Atala A, Soker
Baptista PM, Aboushwareb T, Atala A, Yoo S. A reductionist metastasis-on-a-chip platform for
JJ. Decellularization methods of porcine kidneys for in vitro tumor progression modeling and drug screen-
whole organ engineering using a high-throughput sys- ing. Biotechnol Bioeng. 2016;113:2020–32.
tem. Biomaterials. 2012;33:7756–64. 98. Skardal A, Shupe T, Atala A. Organoid-on-a-chip and
91. Martin I, Smith T, Wendt D. Bioreactor-based road- body-on-a-chip systems for drug screening and disease
map for the translation of tissue engineering strat- modeling. Drug Discov Today. 2016;21:1399–411.
3D Bioprinting for Tissue
Engineering 5
Sujin Noh, Noehyun Myung, Myeongji Park,
Seulgi Kim, Sung-Uk Zhang,
and Hyun-Wook Kang

Abbreviations HUVECs Human umbilical vein endothelial


cells
BMSC Bone marrow stem cells HUVSMC Human umbilical vein smooth
CTZ Cetirizine HCl muscle cells
DAT Decellularized adipose tissue IBU Ibuprofen
DPD Diphenylhydramine HCl ihMSCs Immortalized human mesenchymal
ECM Endothelial cell medium stem cells
FM Fibroblast medium MSCs Mesenchymal stromal cells
GelMA Gelatin methacrylate MTX Methotrexate
GS Gentamicin sulfate MWNTs Multi walled carbon nanotubes
HA Hyaluronic acid n-HA nano-Hydroxyapatite
hASCs Human adipose tissue-derived NHDFs Normal human dermal fibroblasts
mesenchymal stem cells NHLFs Normal human lung fibroblasts
hCMPCs Human cardiac-derived cardio- PCL Polycaprolactone
myocyte progenitor cells PDLLA Poly(d,l-lactic acid or d,l-lactide)
hESCs Human embryonic stem cells PDMS Polydimethylsiloxane
HFF-1 Neonatal human foreskin fibroblasts PLGA Poly(lactic-co-glycolic acid)
hiPSCs Human induced pluripotent stem cells PLLA Poly-l-lactide
PU Polyurethane
PVA Polyvinyl alcohol
SCs Schwann cells
SF-PGA Silk fibroin-poly glutamic acid
SF-PLL Silk fibroin-poly-l-lysine
SMCM Smooth muscle cell medium
SMCs Smooth muscle cells
S. Noh • N. Myung • M. Park • S. Kim • H.-W. Kang (*)
School of Life Science, Ulsan National Institute of
Science and Technology, 50 UNIST-gil, Ulju-gun, 5.1 Introduction
44919 Ulsan, South Korea
e-mail: hkang@unist.ac.kr
Tissue engineering has become a promising tech-
S.-U. Zhang
Department of Mechanical Engineering, Dong-Eui
nology for the functional recovery of tissues or
University, 176 Eomgwangro, Busanjin-gu, organs damaged by accidents or diseases.
47340 Busan, South Korea According to the 2014 annual report of the

© Springer Nature Singapore Pte Ltd. 2018 105


B.W. Kim (ed.), Clinical Regenerative Medicine in Urology, DOI 10.1007/978-981-10-2723-9_5
106 S. Noh et al.

Scientific Registry of Transplant Recipients Recently, bioprinting has emerged as a tech-


(SRTR), an imbalance remains between the sup- nology within tissue engineering; it is a two-­
ply and demand for organ transplantation [1]. The dimensional (2D) or three-dimensional (3D)
shortage of donor organs has resulted in the devel- fabrication method that uses living cells, bioma-
opment of alternatives, such as artificial mechani- terials, and bioactive molecules. Using bioprint-
cal organs, xenotransplantation, and tissue ing technology, tissue- or organ-like architectures,
engineering. Using artificial mechanical organs composed of multiple cell types, can be pro-
may produce many unwanted side effects and duced. Moreover, such printing processes can be
markedly reduces patient quality of life. automated using computer-aided design tem-
Xenotransplantation uses organs from transgenic plates. Many researchers working in regenerative
animals with reduced capacity. This technology medicine have tried to develop biomimetic archi-
has the intrinsic problem of an acute immune tectures with this technology. In this chapter, cur-
response after transplantation. To address this, rent bioprinting technologies and their
immunosuppressive therapy is used with xeno- applications are introduced.
transplantation but the potential for spreading ani-
mal viruses and long-term psychological problems
are still concerns. Tissue engineering, a candidate 5.2 Bioprinting Techniques
technology to avoid these problems, provides a
solution for repairing injured organs by artificially Bioprinting techniques can be categorized into
regenerating the damaged organs (Fig. 5.1). three types: jetting-, extrusion-, and laser-based
Moreover, engineered tissues and organs can be bioprinting (LBB). In this section, the three tech-
used for drug testing and screening and as person- niques are described in terms of their working
alized medicine. However, this approach still has principles, components, advantages, disadvan-
many obstacles to overcome to achieve these tages, and applicable bio-inks.
goals, such as vascularization, scale-up, and com- Table 5.1 summarizes achievable specifica-
posite tissue regeneration [2, 3]. tions for each bioprinting technology, in terms of

Cell isolation
Cell culture

Biopsy

Mechanical Chemical
stimulus stimulus Cell proliferation

Implantation

Scaffold

Tissue growth

Fig. 5.1  The classical tissue engineering approach


Table 5.1  Summary of bioprinting techniques
Jetting-based bioprinting Extrusion-based Laser-based printing
Printing technique bioprinting Stereo-lithography Laser-assisted bioprinting References
Resolution or droplet size <85 μm 5 μm ~ <1.2 μm 8.7–140 μm [22, 30, 34, 42, 43]
Printing speed 10 μm/s–50 mm/s 2–5 mm/s 50 μm/s–5,000 mm/s 200–2,000 mm/s (laser scanning [13, 24, 35, 36, 43,
(laser scanning rates) rates) 45, 46]
Cell viability >89.8% 70.0–93.0% >85.0% 63.8–96.0% [8, 19, 30, 41, 44,
47]
Cell density 106 cells/mL <107 cells/mL 106 cells/mL 107–108 cells/mL [13, 36, 48, 49]
Material Hydrogel, media, cells, Hydrogel, cell aggregates Photocurable hydrogel, Hydrogel, cell aggregates (w or w/o [5–7, 13–15, 26,
proteins and drug (w/ or w/o media), cells, and bioceramics media), peptides, and bioceramics 27, 35, 38, 41, 50,
5  3D Bioprinting for Tissue Engineering

bioceramics, proteins and 51]


drug
Material viscosity 1–18 mPa/s 30,000–50,000 mPa/s 2.05–5,000 mPa/s 40–120 mPa/s [8, 12, 27, 36, 52,
53]
Gelation method Ionic, thermal, enzymatic, Ionic, thermal, Photo crosslinking Ionic and pH crosslinking [10, 13, 41, 42, 47,
and photo crosslinking enzymatic, photo, 54–60]
chemical and pH
crosslinking
Advantages Low printer cost, control Capacity to extrude cells Control pore Nozzle-free, no clogging and wide [4, 25]
droplet size, availability to up to high densities and distribution, pore size range of viscosity
many biomaterials and wide range of viscosity and shape of a scaffold,
control cell concentration high resolution and less
by changing drop density effect of temperature
during the printing
process
Disadvantages Frequent nozzle clogging, Extrusion pressure and Limited printing Long preparation time, poor cell [4, 61]
mechanical and thermal crosslinkers decrease cell materials positioning/targeting, contamination
stress to cells, difficulty in viability by metallic residues and high cost
cell encapsulation,
limitations of material
viscosity, requirement for
crosslinkers which are
cytotoxic and low cell
density than other
techniques
107
108 S. Noh et al.

resolution, printing speed, cell viability and the printer head. The actuator is triggered by
­density, materials, applicable viscosity, gelation applying a voltage pulse, so that the actuator
method, and advantages and disadvantages. deforms and squeezes ink out of the nozzle. The
third method is solenoid valve-based bioprinting.
The valve is controlled by a microcontroller, so
5.2.1 Jetting-Based Bioprinting that the size of droplets can be changed by the
valve-open duration.
Jetting-based bioprinting generates and delivers Jetting-based bioprinting has several advan-
small droplets to a desired site to produce cell tages. First, this method is relatively inexpensive
patterning; it is also called “drop-on-demand” in comparison with extrusion- based bioprinting
printing (Fig. 5.2). This method is easy to develop and laser-based bioprinting. Second, the printed
because it needs only minor modifications from a concentration of cells can be adjusted readily by
“traditional” inkjet printer. The first modification controlling for the density of droplets. Finally,
is to replace the ink with a bio-ink; the second is this methodology can use a variety of biomateri-
that an elevator is placed under the printing head als as bio-ink [4]. For example, Horváth et al. [5]
to achieve a layer-by-layer process for the con- developed an in vitro air–blood tissue barrier
struction of a 3D structure. Thus, a jetting-based using hy926 endothelial cells and A549 epithelial
bio-printer consists essentially of the printing cells with Matrigel. Scoutaris et al. [7] reported
head and elevator. The printing head produces printing results with cetirizine HCl (CTZ),
droplets using a thermal piezoelectric actuator, or diphenhydramine HCl (DPD), and ibuprofen
a solenoid valve. The first method involves a (IBU).
heater attached to the printer head. The heater However, jetting-based bioprinting has an
generates thermal energy using electric current intrinsic limitation in terms of the biomaterials;
pulses. The energy induces vapor bubbles within these should have a low viscosity (1–18 mPa/s)
the ink. Then, these bubbles push the ink out of because the technique requires the generation of
the nozzle, so that droplets can be generated. The small droplets [8]. The viscosity also affects the
second method uses a piezoelectric actuator on speed of the process and the cell viability. To cre-

Piezoelectric Thermal Solenoid Valve

S
Solenoid
Piezoelectric Heater Valve
actuator
Vapor
bubble

Fig. 5.2  Jetting-based bioprinting (using piezoelectric pressure (left), thermally induced air-pressure (middle), and
solenoid valve (right))
5  3D Bioprinting for Tissue Engineering 109

ate a rigid scaffold using low-viscosity materials, printer consists of a cartridge, deposition stage,
a time-consuming process for crosslinking is and printer head, composed of a syringe and noz-
typically required after printing. When droplets zle. According to the dispensing system,
are created from the printer head, cell viability extrusion-­based bioprinting is categorized into
can be influenced by thermal or mechanical two types: pneumatic and mechanical extrusion
stress. Moreover, the available cell density is lim- systems (Fig. 5.3). Pneumatic extrusion-based
ited, because nozzle clogging is an issue with this bioprinting prints bio-ink out of the nozzle by
technology [4]. applying air pressure. The amount of bio-ink is
Recently, several new methods have been intro- adjusted by controlling the air pressure. This
duced to overcome these difficulties. Kador et al. method can use relatively high-viscosity materi-
[9] suggested a design for a retinal model pro- als, in comparison with jetting-based methods.
duced by thermal inkjet printing. They overcame Mechanical extrusion-based bioprinting uses a
the limitations of the electrospinning approach, in piston or screw for printing. Piston-based bio-
terms of cell positioning, by combined use of jet- printing controls the volume of ink extruded
ting-based bioprinting. Xu et al. [10] prepared directly. Screw-based dispensing can produce
functional cartilage tissue with electrospinning. much higher forces, so the system can handle
The electrospinning technique was used to address materials of much higher viscosity.
the disadvantages of jetting-­based bioprinting in As mentioned above, extrusion-based bio-
terms of mechanical properties. Moreover, Binder printing can handle high-viscosity materials, of
[11] developed an in situ skin bioprinter for about 30,000–50,000 mPa/s [12]. Thus, high
enhancing the healing of skin wounds. They concentrations of cells can be used in such a sys-
directly printed skin cells, including fibroblasts tem. A cell concentration of ~1 × 107 cells/mL
and keratinocytes, on a skin wound. has been reported [13]. Moreover, a wide range
of biomaterials has been used in extrusion-based
systems. Luo et al. [14] created a scaffold with
5.2.2 Extrusion-Based Bioprinting alginate, Pluronic F127, and a bioceramic
(Ca7Si2P2O16) powder for bone tissue engineer-
Extrusion-based bioprinting is a technique to cre- ing. Latza et al. [15] studied a device for wear-
ate 3D cellular constructs by dispensing continu- able electronics using sucker ring teeth (SRT;
ous filaments with a cell-laden hydrogel. The from squid) protein, water, and glycerol. Yang

Pneumatic Piston Screw

inlet

Fig. 5.3 Extrusion-­
based bioprinting (using
pneumatic (left) and
mechanical (piston
(middle), and screw
(right)) pressure)
110 S. Noh et al.

et al. [13] produced osteochondral biphasic grafts 5.2.3.1 Stereolithography (SLA)


using alginate, cartilage-derived ECM, PLGA, SLA produces 3D structures with a scanning laser
hydroxyapatite (HA), and cells. beam on a photo-curable material. Thus, the sys-
Recently, various functional bio-inks have tem consists primarily of a laser and optics, a pho-
been developed for extrusion-based bioprinting. topolymer container, and an elevator. The laser
Rees et al. [17] suggested a nanocellulose (car- beam irradiates the photopolymer. Galvanometric
boxymethylation and periodate oxidation)-based mirrors and lenses are used to control the position
bio-ink that could prevent bacterial growth. The of the beam and its focusing. Transferred photon
material was used to print a wound dressing. energy initiates the photo-­curing process and a 2D
Highley et al. [16] developed a supramolecular pattern is generated by scanning the beam. Then, a
self-healing hydrogel, which was cured by apply- 3D structure can be produced, by stacking the pat-
ing a physical stimulus. The hydrogel was terns layer-by-layer.
restored when the physical stimulus was removed. SLA technology has good spatial resolution,
The material could be used to construct complex of <1.2 μm [22]. Recently, higher resolutions
structures with high resolution. Cho’s group used have been obtained using two-photon polymer-
a decellularization process to develop tissue-­ ization (TPP). TPP uses a femtosecond laser to
specific bio-inks with adipose, cartilage, and obtain a high-intensity near-infrared beam that
heart tissue. These bio-inks provided enhanced induces localized excitation by two-photon
biocompatibility for tissue growth in a 3D struc- absorption. This then induces nano-scale photo-­
ture [18]. polymerization. Haske et al. [23] achieved a reso-
Extrusion-based scaffold-free bioprinting has lution of 65 nm. Thus, SLA can control the shape
also been described, in which cell aggregates of a scaffold, its pore distribution, and overall
having spherical or rod shapes are printed directly size very precisely. Other advantages of SLA
with no additional biomaterials [20, 21]. Using technology include the speed and temperature of
the bio-printed cell aggregates, organ-specific the fabrication process. The speed is high because
biological features could be mimicked. Moreover, the laser scanning rate can be up to 5000 mm/s
tissue-like cell densities can be achieved. For [24]. The temperature during the fabrication is
example, Itoh et al. [20] introduced a tubular lower than with other techniques, so that cells are
structure with multicellular spheroids. However, less affected [25]. SLA can use several types of
the structure did not easily stand by itself withoutbiomaterials. Lee et al. [26] introduced a custom-
supporting materials, so a fine architecture could ized poly(propylene fumarate)/diethyl fumarate
not be formed. To overcome this, Kucukgul et al. photopolymer and built 3D scaffolds for bone tis-
[21] introduced a new process. They defined a sue engineering. Jansen et al. [27] created porous
designed structure and empty spaces. In the structures with a well-defined gyroid architecture
empty spaces, a supporting structure was printed for tissue engineering using pentaerythritol
with agarose gel. Then, computer-designed struc- tetra-­
­ acrylate resin, ceramic suspensions, and
tures were plotted with the rod-shaped cell PDLLA3-FAME/NVP resins. However, the num-
aggregates. ber of applicable biomaterials for SLA is still
small in comparison with the other techniques,
because SLA requires a photocurable polymer.
5.2.3 Laser-Based Bioprinting (LBB) Recently, Shanjani et al. [28] reported a hybrid
printing system combining SLA and extrusion-
The LBB technique involves constructing a struc- based bioprinting to allow use of “softer” materi-
ture by solidifying or depositing bio-ink with als as the bio-ink.
photon energy. This technique can be divided Recently, researchers have studied building 3D
into stereolithography (SLA) (Fig. 5.4) and laser-­ cellular scaffolds using SLA [29–31]. Soman et al.
assisted bioprinting (LAB) (Fig. 5.5). [29] created an in vitro model with PEG material
5  3D Bioprinting for Tissue Engineering 111

Fig. 5.4 Laser-based
bioprinting
(stereolithography Laser pulse
(SLA))

Solidified bioink

Liquid bioink

Laser pulse

Quartz support
Energy-
absorbing
layer
Built pressure

Fig. 5.5 Laser-based
bioprinting (laser-­
assisted bioprinting
(LAB))

and cancer cells to mimic a metastatic process. by culturing and imaging with Chinese hamster
SLA has also been used to construct microfluidic ovary (CHO) cell on the chip.
chips, because the method is cost-­efficient and the
process can be automated. Shallan et al. [31] 5.2.3.2 Laser-Assisted Bioprinting
developed a complex monolithic device using a (LAB)
transparent resin. Au et al. [32] evaluated a com- LAB involves primarily a laser, bio-ink-coated
mercially available SLA system to produce com- ribbon, and the collector substrate. Lasers can be
puter-designed microfluidic chips. It was validated divided into pulsed and continuous devices. The
112 S. Noh et al.

ribbon consists of a glass or quartz plate, a metal mL [36]. However, the ribbon is used over a rela-
layer, typically of gold, silver, or titanium and an tively long time and high-precision cell pattern-
energy absorbing layer (interface layer) made of ing cannot be guaranteed. LAB is expensive
a sacrificial hydrogel. The bio-ink is then coated because of the laser and it has limitations in terms
on the energy absorbing layer. The collector sub- of scaling up the structure [37].
strate is coated with biopolymer and cell culture Several bio-inks have been introduced for use
medium to enhance adhesion of the cells. with LAB technology. Pagès et al. [35] created 3D
The laser pulse is focused and irradiated on constructs for complex tissue substitutes using
the bio-ink-coated ribbon using the optics. Then, collagen and cells in medium. Dinca et al. [38]
the energy absorbing layer is expanded thermally introduced a controllable self-assembly technique
and generates vapor bubbles to form and jet drop- for peptides for use in biosensors and tissue engi-
lets of bio-ink. The droplets fall on the receiving neering. Cheng et al. [39] developed a unique pro-
substrates so that the designed 2D cell pattern is cess to coat HA powders on titanium substrates.
created. The two most common methods of LAB Recently, several advanced processes have
are absorbing film-assisted laser-induced forward been reported for LAB. Catros et al. [40] opti-
transfer (AFA-LIFT) and matrix-assisted pulsed mized experimental conditions using inorganic
laser evaporation direct writing (MAPLE-DW). nano-hydroxyapatite (n-HA) and human osteo-
AFA-LIFT is modified from LIFT for bioprint- progenitor cells. They showed that cell viability,
ing. AFA-LIFT uses a sacrificial metal, such as proliferation and phenotype were maintained
gold or titanium, for the energy absorbing layer. without changes in the physicochemical proper-
It uses a high-power laser system and the metal ties of n-HA. Xiong et al. [41] printed straight
layer can induce metallization of the ejected ink. and Y-shaped tubes using a ribbon composed of
This can affect the cytotoxicity of the process. To alginate and cells in medium. Their preliminary
overcome this, MAPLE-DW uses a lower-power results suggest the technique is valuable for 3D
laser and a sacrificial hydrogel, such as Matrigel, tubular constructs and organ printing.
for the energy absorbing layer [33].
LAB uses a laser pulse, so that relatively high
resolution can be achieved, from 8.7 μm to 5.3 Applications
140  μm [31, 34]. Laser scanning rates up to
2000 mm/s have been reported; this high printing Recently, 3D bioprinting has been used widely in
speed can be advantageous [35]. LAB uses a rib- applications such as tissue engineering, body-on-­
bon, so a nozzle is not necessary. Thus, nozzle a-chip, and drug delivery systems. Table 5.2
clogging is not a concern, so materials of various shows a summary of bioprinting applications
viscosities can be used for printing. Additionally, sorted by printing methodology, cell source/pro-
the cell density can be increased, up to 108 cells/ tein, and biomaterials.

Table 5.2  Bioprinting applications for blood vessel


Printing method Cell source/ protein Material References
Extrusion-based HUVECs, SMCs, and NHDFs ECM, SMCM, FM, and gelatin [20]
Extrusion-based HCASMCs Alginate, MWCNT [64]
Extrusion-based L929 mouse fibroblasts Alginate [65]
Extrusion-based HUVECs, NHLFs Collagen, gelatin, fibrin [66]
Laser-based Breast cancer cell (MDA-MB-231 Gelatin [59]
and MCF-7) and fibroblasts
(BJ5ta)
Laser-based HUVSMCs Alginate [41]
5  3D Bioprinting for Tissue Engineering 113

5.3.1 Tissue Engineering

As mentioned above, the “traditional” scaffold-­


based approach has difficulties in terms of vascu-
larization and developing a complex tissue or
organ. In tissue engineering, 3D bioprinting is
considered a breakthrough technology. Bioprinting
processes can use multiple cell types and precisely
control scaffold geometry in developing complex
tissues. This subsection introduces artificial ves-
sels, bones, cartilage, and organs produced with
3D bioprinting technology.

5.3.1.1 Blood Vessels


Blood vessels are the part of the cardiovascular
system that transfers blood in circulation, trans-
porting nutrients, oxygen, carbon dioxide, hor-
mones, and blood cells to and from tissues. To
prepare scalable organs, the artificial formation
of blood vessels is one of the most important
issues in tissue engineering. The vessel is tube-­
Fig. 5.6 Scaffold-free vascular graft with MSCs.
shaped, having a lumen, endothelium, and base- Adapted from Itoh et al. PLoS One 2015;10(9):1–15 [20]
ment membrane. Printing a blood vessel is a
challenge because of its complexity.
Many studies have been reported on the con- that the rat femoral artery could sustain physio-
struction of artificial vessels. Itoh et al. [20] pro- logical blood pressure, from 80 to 100 mmHg
duced vascular prostheses with human umbilical and a normal level of flow was observed [63].
vein endothelial cells (HUVECs), human aortic Dolati et al. [64] used a coaxial bioprinting
smooth muscle cells (HASMCs), and human nor- process and printed vascular conduits directly.
mal dermal fibroblasts (HNDFBs) using a They prepared alginate reinforced with carbon
scaffold-­free bioprinting technique (Fig. 5.6). nanotubes and then encapsulated human coro-
The tubular tissues were implanted in the aortas nary artery smooth muscle cells (HCASMCs) in
of male F344-rnu/rnu athymic nude rats. They the material for printing. Their method had flexi-
observed endothelial cells on the inner surface of bility in terms of scale, from micro- to sub-­
the tubular tissues and normal flow rates in the millimeter. Similarly, Gao et al. [65] made a
aortas using percutaneous ultrasonography. Also, built-in micro channel structure by using layer-­
thrombosis did not occur in any rat. by-­layer printing. They used a coaxial nozzle and
Miller et al. [62] used a printable, cyto-­ printed hollow calcium alginate filaments, encap-
compatible, and sacrificial material to produce sulating L929 mouse fibroblasts.
micro-channels incorporating a hydrogel struc- Thus, arteries have been printed successfully.
ture. After printing a 3D channel network, cell-­ However, a capillary network at the single-cell
laden hydrogel was infiltrated into the sacrificial scale is much harder to print. To address this, sev-
mold and cross-linked (Fig. 5.7). Then, the mold eral researchers have studied perfusable capillary
was dissolved to produce hollow channels. An networks to induce angiogenesis. Lee et al. [66]
artificial vascular structure was produced by used HUVECs and biological matrices and
seeding HUVECs in the channel and culturing formed larger fluidic vascular channels. The cap-
them. Then, the construct was implanted in the illary network was constructed by angiogenesis,
circulatory system of a rat. The results showed induced from the printed channels.
114 S. Noh et al.

Fig. 5.7  A schematic


overview of the casting
of perfusable vascular
architectures by using
bioprinted sugar lattice.
Reprinted from
NATURE MATERIALS,
Miller JS, Stevens KR,
Print a 3D
Yang MT, Baker BM, Encapsulate Dissolve
carbohydrate-
Nguyen D-HT and lattice and living lattice in
glass lattice
Cohen DM, Rapid cells in ECM cell media
casting of patterned
vascular networks for
perfusable engineered
three-dimensional
tissues, 2012;11(7):768– Flow
74, Copyright © 2012,
Rights Managed by Living cells in
Nature Publishing monolithic ECM
Group [62] with perfusable
vascular architecture

Table 5.3  Bioprinting applications for bone


Printing method Cell source/protein Material References
Extrusion-based Osteoblast precursor cells (MC3T3-E1) PLGA and PLGA/n-HAp/β-TCP [67]
Extrusion-based Osteoblast precursor cells (MC3T3-E1) PCL, cryogel, and gelatin [68]
Extrusion-based N/A Bioceramics, Pluronic F-127, and [14]
alginate
Extrusion-based ihMSCs PLGA-PEG-PLGA triblock [84]
copolymer hydrogel
Extrusion-based Human AFSCs Gelatin, fibrinogen, HA, glycerol, [70]
and PCL
Laser-based MG-63 cells n-HA and Matrigel [40]

5.3.1.2 Bone and Cartilage They observed colonized cells attached to the


Many studies have been reported on the regenera- porous cryogel network.
tion of bone and cartilage. Tissues can be printed Luo et al. [14] described a hollow struts-­
using patient data, acquired with imaging tech- packed (HSP) bioceramic scaffold using a coax-
niques. This has been shown to be an effective ial 3D printing strategy. The HSP bioceramic
method for reconstructing structures damaged by scaffold enhanced the proliferation and attach-
osteoporosis and bone fractures, for example. ment of cells. When this structure was implanted
Roh et al. [67] produced scaffolds using oxy- in the damaged femur of a rabbit, recovery of a
gen plasma treatment and a PLGA/n-HAp/β-TCP large bone defect was observed because of the
composite. Based on the scaffold, they enhanced implant’s osteo-conductivity (Table 5.3).
the initial adhesion, proliferation, and differentia- Izadifar et al. [69] performed a layer-by-layer
tion of preosteoblasts. Van Rie et al. [68] printed process with two bio-inks and a PCL. The first bio-
scaffolds using cryogel-PCL composite and ink was composed of alginate and primary chondro-
seeded MC3T-E1 osteoblast precursor cells. cytes extracted from embryonic chick cartilage. The
5  3D Bioprinting for Tissue Engineering 115

Table 5.4  Bioprinting applications for cartilage


Printing method Cell source/ protein Material Reference
Extrusion-based Rabbit ear chondrocytes Gelatin, fibrinogen, HA, glycerol, PCL [70]
Extrusion-based Primary chondrocytes (from Polycaprolactone (PCL) and alginate [69]
embryonic chick cartilage),
ATDC5 mouse cell lines
Extrusion-based Human nasoseptal Nanocellulose-alginate [82]
chondrocyte
Extrusion-based Equine chondrocytes and Polycaprolactone (PCL), gelatin [83]
MSCs methacrylate (GelMA), and GelMA-gellan
hydrogel

second contained ATDC5 mouse cell with alginate.


The structure showed outstanding performance in
terms of proliferation, cell viability, and secretion of
cartilage ECM. Moreover, the real thickness of
human articular cartilage could be achieved.
Kang et al. [70] developed an integrated
tissue-­organ printer (ITOP) and applied it to the
regeneration of bone, cartilage, and muscle tis-
sue. They printed a cell-laden hydrogel and a bio-
degradable polymer (PCL), anchored in sacrificial
Pluronic F127, into a single structure. Their
method increased the mechanical stability of the
scaffold and overcame limitations in terms of
structural integrity, size, and shape. To obtain a
more precise structure, tissues were constructed
using medical image data (Table 5.4). They
implanted the structure in nude rats and observed
the functionality of the structure (Fig. 5.8).

5.3.1.3 Other Organs 5 mm


Faulkner-Jones et al. [71] printed human-induced
pluripotent stem cells (hiPSCs) to create an artifi-
cial liver. Gaetani et al. [72] reported a bioprinted
cardiac construct. They used human cardiomyo- Fig. 5.8  Gross appearance of a bioprinted ear construct
cyte progenitor cells (hCMPCs) mixed with algi- at 1 month after implantation. Reproduced from Kang
nate for printing. After printing the scaffold, they et al. Nat Biotechnol. 2016;34(3):312–9 [70]
observed high cell viability, of 89%, for 7 days.
They increased the expression of cardiac tran- showed a high surface area to volume ratio so it
scription factors and a sarcomeric protein through obtained high oxygen and nutrient diffusion. They
3D culture. This was an important achievement reported that the functionality of the scaffold was
with respect to the goal of creating a fully differ- fully restored by vascularization in the pores.
entiated cardiac construct. Pati et al. [74] produced biomimetic 3D adi-
Marchioli et al. [73] made a porous construct pose tissues with a bioprinting technique to
using INS1E β-cells and human and mouse islets reconstruct soft tissues. They constructed flexible
mixed with alginate/gelatin mixture. The scaffold dome-shaped structures encapsulating human
116 S. Noh et al.

adipose tissue-derived mesenchymal stem cells lack of a definite niche. Huang et al. [75] made a
(hASCs) with a decellularized adipose tissue 3D biomimetic scaffold with gelatin and algi-
(DAT) matrix-based bio-ink (Fig. 5.9). They also nate. The 3D bioprinted scaffold had micro-­
showed a viable clinical result for soft tissue environmental characteristics, so that a restrictive
regeneration in a patient-specific manner. niche could be regenerated and epidermal pro-
Damaged sweat glands cannot be recovered genitor could be induced to regenerate sweat
because of their low regenerative potential and gland cells (Table 5.5).

Oxygen and nutrient transfer

DAT gel encapsulating Cell printed construct with


cells DAT

Fig. 5.9  Cell-printed constructs were prepared by layer-­ from Biomaterials, Pati F, Ha DH, Jang J, Han HH, Rhie
by-­layer deposition of synthetic polymer and decellular- JW and Cho DW, A 3D bioprinting system to produce
ized adipose tissue (DAT) bio-ink, maintaining a porous human-scale tissue constructs with structural integrity,
structure. An open porous construct enhances nutrient and 2015;62:164–75, Copyright © 2015 Elsevier Ltd. All
oxygen transfer to the core of the construct. Reprinted rights reserved [74]

Table 5.5  Bioprinting applications for other organs


Organs Printing method Cell source/protein Material References
Skin and sweat Extrusion-based NHFF-1 Polyelectrolyte [85]
gland gelatin-chitosan
Extrusion-based Mouse derived Gelatin and alginate [75]
epidermal progenitor
cells
Laser-based NIH3T3 fibroblasts and Collagen [37]
HaCaT keratinocytes
Muscle Extrusion-based neonHFF-1 Thermoplastic PU and [86]
PCL
Pancreas Extrusion-based INISIE β-cell line Alginate/gelatin mixture [73]
(from rat insulinoma),
human and mouse islets
Heart Extrusion-based hCMPCs Alginate and Matrigel [72]
Neural tissue Extrusion-based Murine neural stem PU, PLLA, PCL, and [51]
cells PDLLA
Extrusion-based Mouse derived BMSC Collagen and agarose [87]
and SCs
Liver Jetting-based hiPSCs and hESCs Alginate [71]
Breast Extrusion-based hASCs Decellularized adipose [74]
tissue (DAT) hydrogel
5  3D Bioprinting for Tissue Engineering 117

5.3.2 Bio-Chip introduced a micro-organ device as a drug screen-


ing model.
A bio-chip is a miniaturized system for testing Snyder et al. [78] applied a precision extrusion
biological reactions; 3D bioprinting technology deposition (PED)/replica molding process to produce
has been used in the development of bio-chip sys- a cell-laden device. They simplified the fabrication
tems. This subsection introduces 3D bioprinted process and enhanced leak-resistance (up to 2.0 mL/
bio-chip systems in terms of an organoid for a min) and saw pervasive diffusion in the device. They
body-on-a-chip and biosensors. also showed that multi-nozzle printing was adequate
for a device including multiple cell types.
5.3.2.1 Body-on-a-Chip
The so-called body-on-a-chip refers to a multi-­ 5.3.2.2 Biosensor
channel microfluidic biochip that includes human A biosensor is an analytical device that detects an
cells. The body-on-a-chip will be used to simu- analyte of interest using biological components,
late human body responses. In the near future, such as protein or bacteria. Bioprinting technol-
this may become an alternative drug screening ogy has been applied in the field of biosensors.
model, replacing some animal studies. Recently, Fuchiwaki et al. [6] developed a microchip for
bioprinting technologies have been applied to enzyme-linked immunosorbent assays (ELISAs)
produce 3D organoids for studies that will ulti- using anti-C-reactive protein antibody. They
mately lead to a body-on-a-chip. developed a quartz crystal microbalance (QCM)
Chang et al. [76] developed 3D liver micro-­ sensor system to reduce background noise in a
organ devices for an in vitro drug metabolism process with jetting-based bioprinting. Drachuk
model using HepG2 liver cells and alginate. Liver et al. [79] introduced a prototype of a cell-based
tissue could be precisely patterned using bio- thin-film biosensor that was printed using a dual
printing and was then integrated with a microflu- array of silk fibroin-poly(L-lysine) (SF-PLL) and
idic system. They showed that the approach could silk fibroin-poly(glutamic acid) (SF-PGA). Each
control cellular-level differentiation and tissue-­ array included E. coli coated with different fluo-
level function. Tourlomousis and Chang [77] also rescent markers (Fig. 5.10). They sought to detect

SF-PLL
SF-PGA
E. coli cells with
different reporters

Fluorescent Mixture of
signal analytes

Fig. 5.10  Schematics of the inkjet printing process for Kaplan DL, Printed Dual Cell Arrays for Multiplexed
dual-color cell–cell arrays. Reprinted from ACS Sensing, 2015;150,410,104,710,005, Copyright © 2015
Biomaterials Science & Engineering, Drachuk I, Suntivich American Chemical Society [79]
R, Calabrese R, Harbaugh S, Kelley-Loughnane N and
118 S. Noh et al.

Table 5.6  Bioprinting applications for bio-chips


Bio-chip Printing method Cell source/ protein Material References
Body-on-a-chip Extrusion-based HepG2 cells Alginate [77]
Extrusion-based HepG2 cells Alginate, PCL [78]
Extrusion-based HepG2 cells Alginate [76]
Jetting-based Human breast Gelatin [88]
cancer cells
(MCF-7)
Biosensor Jetting-based E. coli cells SF-PLL and SF-PGA [79]
Jetting-based Anti-C-reactive Anti-C-reactive [6]
protein antibody protein antibody
aqueous solution
Laser-based Mouse, rabbit IgG Polylysine [89]
and human cDNA

multiple target analytes using a dual-type sensor shape of the drug product could be of various
and studied chemical and pharmaceutical screen- forms, such as filaments, discs, and beads.
ing and environmental monitoring with the sys- Khaled et al. [80] printed polypills including
tem (Table 5.6). multiple active drugs (Fig. 5.11). The polypills
include multiple active ingredients for specific
diseases. For example, they used aspirin, hydro-
5.3.3 Drug Delivery System chlorothiazide, atenolol, pravastatin sodium, and
ramipril for cardiovascular disease and high
Many researchers have applied 3D printing tech- blood pressure. They used a 3D printing tech-
niques to drug delivery systems. Scoutaris et al. nique to control the amounts of ingredients, and
[7] created a mucosal thin film for taste masking their distributions and geometries, so that they
using jetting-based bioprinting. As it contained could make patient-specific polypills.
the bitter substances CTZ, DPD, and IBU, the Goyanes et al. [81] used hot-melt extrusion-­
film could be used as a test to distinguish bitter based 3D printing to produce computer-designed
tastes. Many other drug–polymer solutions could drug forms, such as cubes, pyramids, cylinders,
be used so that the film could be adapted for other spheres, and a torus. Traditional manufacturing
applications. techniques, such as powder compaction, cannot
Weisman et al. [50] produced antibiotic and be used to control the shape of the drug product
chemotherapeutic drugs with pellets containing in this way. The printed drug products were com-
gentamicin sulfate (GS) and methotrexate posed of polyvinyl alcohol (PVA) and
(MTX). They verified the functionality of drug paracetamol. They measured drug release rates
products using bacteria and osteosarcoma cells according to the shape of the drug form and con-
and demonstrated the thermal stability of the firmed that the drug release rate increased as the
drug preparation made using 3D printing. The surface-to-volume ratio increased (Table 5.7).
5  3D Bioprinting for Tissue Engineering 119

Fig. 5.11 Multi-active
drug tablets (polypills).
Reprinted from Journal
of Controlled Release,
Khaled SA, Burley JC,
Alexander MR, Yang J
and Roberts CJ, 3D
printing of five-in-one
dose combination
polypill with defined
immediate and sustained
release profiles,
2015;217:308–14,
Copyright © 2015
Elsevier B.V. All rights
reserved [80]

Table 5.7  Bioprinting applications for drug delivery system


Method Cell source/ protein Material References
Extrusion-based N/A PLA, gentamicin sulfate, and [50]
methotrexate
Extrusion-based N/A Aspirin, hydrochlorothiazide, atenolol, [80]
pravastatin, and ramipril
Extrusion-based N/A Paracetamol and PVA [81]
Jetting-based N/A Three-powder blend (cornstarch, [90]
dextran, and gelatin)
Jetting-based N/A Cetirizine HCl, diphenhydramine HCl, [7]
and ibuprofen
120 S. Noh et al.

Conclusions ing multiple cell types prepared by inkjet printing


technology. Biomaterials. 2013;34(1):130–9.
Bioprinting technology can produce computer-­ 11. Binder KW. In situ bioprinting of the skin. ProQuest
designed 3D structures not only with cell-laden Dissertations Publishing. 2011;3458142.
bio-ink, but also with various biomaterials and 12. Tan EYS, Yeong WY. Concentric bioprinting of

biomolecules. This processability confers great alginate-based tubular constructs using multi-­
nozzle extrusion-based technique. Int J Bioprinting.
benefits in regenerative medicine. In this chap- 2015;1(1):49–56.
ter, we reviewed bioprinting technology and 13. Yang SS, Choi WH, Song BR, Jin H, Lee SJ, Lee
some of its applications have been introduced. SH. Fabrication of an osteochondral graft with using
According to the printing mechanism, three a solid freeform fabrication system. Tissue Eng Regen
Med. 2015;12(4):239–48.
types of bioprinting technology—jetting-, 14. Luo Y, Zhai D, Huan Z, Zhu H, Xia L, Chang J. Three-­
extrusion-, and laser-based bioprinting—were dimensional printing of hollow-struts-packed bioc-
described in terms of their components, appli- eramic scaffolds for bone regeneration. ACS Appl
cable bio-inks, and pros and cons. Although Mater Interfaces. 2015;7(43):24377–83.
15. Latza V, Guerette PA, Ding D, Amini S, Kumar A,
these bioprinting technologies are recent, many Schmidt I. Multi-scale thermal stability of a hard
researchers have reported progress in various thermoplastic protein-based material. Nat Commun.
applications, including artificial tissue/organ 2015;6:8313.
regeneration, body-on-­a-chip, biosensors, and 16. Highley CB, Rodell CB, Burdick JA. Direct 3D print-
ing of shear-thinning hydrogels into self-healing
drug delivery systems. Continued advances in hydrogels. Adv Mater. 2015;27(34):5075–9.
bioprinting may soon move this technology 17. Rees A, Powell LC, Chinga-Carrasco G, Gethin

into clinical sites and help in advancing medi- DT, Syverud K, Hill KE. 3D bioprinting of
cal technology. carboxymethylated-­periodate oxidized nanocellulose
constructs for wound dressing applications. Biomed
Res Int. 2015;2015:1–7.
18. Pati F, Jang J, Ha D-H, Won Kim S, Rhie J-W, Shim
J-H. Printing three-dimensional tissue analogues
References with decellularized extracellular matrix bioink. Nat
Commun. 2014;5:3935.
1. Hart A, Smith JM, Skeans MA, Gustafson SK, 19. Tabriz AG, Hermida MA, Leslie NR, Shu W. Three-­
Stewart DE, Cherikh WS, Wainright JL, Boyle G, dimensional bioprinting of complex cell laden algi-
Snyder JJ, Kasiske BL, Israni AK. OPTN/SRTR nate hydrogel structures. Biofabrication. 2015;7(4):
Annual Data Report 2014: Kidney. Am J Transplant. 045012.
2016;16(Suppl 2):11–46. 20. Itoh M, Nakayama K, Noguchi R, Kamohara K,

2. Nerem RM, Seliktar D. Vascular tissue engineering. Furukawa K, Uchihashi K. Scaffold-free tubular tis-
Annu Rev Biomed Eng. 2001;3:225–43. sues created by a bio-3D printer undergo remodeling
3. Derby B. Printing and prototyping of tissues and scaf- and endothelialization when implanted in rat aortae.
folds. Science. 2012;338(6109):921–6. PLoS One. 2015;10(9):1–15.
4. Murphy SV, Atala A. 3D bioprinting of tissues and 21. Kucukgul C, Ozler SB, Inci I, Karakas E, Irmak S,
organs. Nat Biotechnol. 2014;32(8):773–85. Gozuacik D. 3D bioprinting of biomimetic aortic vas-
5. Horváth L, Umehara Y, Jud C, Blank F, Petri-Fink A, cular constructs with self-supporting cells. Biotechnol
Rothen-Rutishauser B. Engineering an in vitro air- Bioeng. 2015;112(4):811–21.
blood barrier by 3D bioprinting. Sci Rep. 2015;5:7974. 22. Zhang X, Jiang XN, Sun C. Micro-stereolithography
6. Fuchiwaki Y, Tanaka M, Ooie T. Inexpensive and of polymeric and ceramic microstructures. Sensors
reliable monitoring of the microdeposition of biomol- Actuators A Phys. 1999;77(2):149–56.
ecules. Anal Lett. 2016;2719:921–8. 23. Haske W, Chen VW, Hales JM, Dong W, Barlow S,
7. Scoutaris N, Snowden M, Douroumis D. Taste Marder SR. 65 nm feature sizes using visible wave-
masked thin films printed by jet dispensing. Int J length 3-D multiphoton lithography. Opt Express.
Pharm. 2015;494(2):619–22. 2007;15(6):3426–36.
8. Demirci U, Montesano G. Single cell epitaxy by acous- 24. Alharbi N, Osman R, Wismeijer D. Effects of build
tic picolitre droplets. Lab Chip. 2007;7(9):1139–45. direction on the mechanical properties of 3D-printed
9. Kador KE, Grogan SP, Dorthé EW, Venugopalan P, complete coverage interim dental restorations. J
Malek MF, Goldberg JL. Control of retinal ganglion cell Prosthet Dent. 2016;11:760–7.
positioning and neurite growth: combining 3D printing 25. Lan PX, Lee JW, Seol YJ, Cho DW. Development of
with radial. Tissue Eng Part A. 2016;22:286–94. 3D PPF/DEF scaffolds using micro-stereolithography
10. Xu T, Zhao W, Zhu JM, Albanna MZ, Yoo JJ, Atala and surface modification. J Mater Sci Mater Med.
A. Complex heterogeneous tissue constructs contain- 2009;20(1):271–9.
5  3D Bioprinting for Tissue Engineering 121

26. Lee JW, Kang KS, Lee SH, Kim JY, Lee BK, Cho 40. Catros S, Fricain J-C, Guillotin B, Pippenger B,

DW. Bone regeneration using a microstereolithography-­ Bareille R, Remy M. Laser-assisted bioprinting for
produced customized poly(propylene fumarate)/ creating on-demand patterns of human osteoprogeni-
diethyl fumarate photopolymer 3D scaffold incorporat- tor cells and nano-hydroxyapatite. Biofabrication.
ing BMP-2 loaded PLGA microspheres. Biomaterials. 2011;3:025001.
2011;32(3):744–52. 41. Xiong R, Zhang Z, Chai W, Huang Y, Chrisey

27. Jansen J, Melchels FPW, Grijpma DW, Feijen
DB. Freeform drop-on-demand laser printing of
J. Fumaric acid monoethyl ester-functionalized poly 3D alginate and cellular constructs. Biofabrication.
(D,L-lactide )/N-vinyl-2-pyrrolidone resins for the 2015;7(4):45011.
preparation of tissue engineering Scaffolds by stereo- 42. Cui X, Breitenkamp K, Finn MG, Lotz M,

lithography. Biomacromolecules. 2009;10(2):214–20. D’Lima D. Direct human cartilage repair
28. Shanjani Y, Pan CC, Elomaa L, Yang Y. A novel using 3D bioprinting technology. Tissue Eng.
bioprinting method and system for forming hybrid 2012;18(11–12):1304–12.
tissue engineering constructs. Biofabrication. 43. Smith CM, Stone AL, Parkhill RL, Stewart RL,

2015;7(4):045008. Simpkins MW, Kachurin AM. Three-dimensional bio-
29. Soman P, Kelber JA, Lee JW, Wright TN, Vecchio assembly tool for generating viable tissue-­engineered
KS, Klemke RL. Cancer cell migration within 3D constructs. Tissue Eng. 2004;10(9):1566–76.
layer-by-layer microfabricated photocrosslinked 44. Kolesky DB, Truby RL, Gladman AS, Busbee TA,
PEG scaffolds with tunable stiffness. Biomaterials. Homan KA, Lewis JA. 3D bioprinting of vascular-
2012;33(29):7064–70. ized, heterogeneous cell-laden tissue constructs. Adv
30. Hribar KC, Meggs K, Liu J, Zhu W, Qu X, Chen Mater. 2014;26(19):3124–30.
S. Three-dimensional direct cell patterning in colla- 45. Mapili G, Lu Y, Chen S, Roy K. Laser-layered micro-
gen hydrogels with near-infrared femtosecond laser. fabrication of spatially patterned functionalized
Sci Rep. 2015;5(October):17203. tissue-­engineering scaffolds. J Biomed Mater Res -
31. Shallan AI, Smejkal P, Corban M, Guijt RM,
Part B Appl Biomater. 2005;75(2):414–24.
Breadmore MC. Cost-effective three-dimensional 46. Duan B, Hockaday LA, Kang KH, Butcher JT. 3D
printing of visibly transparent microchips within min- Bioprinting of heterogeneous aortic valve conduits
utes. Anal Chem. 2014;86(6):3124–30. with alginate/gelatin hydrogels. J Biomed Mater Res
32. Au AK, Lee W, Folch A. Mail-order microfluidics: A. 2013;101(A(5)):1255–64.
evaluation of stereolithography for the production of 47. Wang Z, Abdulla R, Parker B, Samanipour R. A sim-
microfluidic devices. Lab Chip. 2014;14(7):1294–301. ple and high-resolution stereolithography-based 3D
33. Larson C, Shepher R. 3D bioprinting technologies bioprinting system using visible light crosslinkable
for cellular engineering. In: Singh A, Gaharwar AK, bioinks. Biofabrication. 2015;7(4):1–29.
editors. Microscale technologies for cell engineering. 48. Xu T, Jin J, Gregory C, Hickman JJ, Boland T. Inkjet
Cham: Springer; 2015. printing of viable mammalian cells. Biomaterials.
34. Koch L, Kuhn S, Sorg H, Gruene M, Schlie S, Gaebel 2005;26(1):93–9.
R. Laser printing of skin cells and human stem cells. 49. Liu Tsang V. Chen a a, Cho LM, Jadin KD, Sah RL
Tissue Eng Part C Methods. 2010;16(5):847–54. and DeLong S, fabrication of 3D hepatic tissues
35. Pagès E, Rémy M, Kériquel V, Correa MM, Guillotin by additive photopatterning of cellular hydrogels.
B, Guillemot F. Creation of highly defined mesen- FASEB J. 2007;21(3):790–801.
chymal stem cell patterns in three dimensions by 50. Weisman JA, Nicholson JC, Tappa K, Jammalamadaka
laser-assisted bioprinting. J Nanotechnol Eng Med. U, Wilson CG, Mills D. Antibiotic and chemothera-
2015;6(2):021005. peutic enhanced three-dimensional printer filaments
36. Guillotin B, Souquet A, Catros S, Duocastella M, and constructs for biomedical applications. Int J
Pippenger B, Bellance S. Laser assisted bioprinting of Nanomedicine. 2015;10:357–70.
engineered tissue with high cell density and microscale 51. Hsieh FY, Lin HH, Hsu SH. 3D bioprinting of neu-
organization. Biomaterials. 2010;31(28):7250–6. ral stem cell-laden thermoresponsive biodegradable
37. Michael S, Sorg H, Peck CT, Koch L, Deiwick A, polyurethane hydrogel and potential in central ner-
Chichkov B. Tissue engineered skin substitutes cre- vous system repair. Biomaterials. 2015;71:48–57.
ated by laser-assisted bioprinting form skin-like struc- 52. Bajaj P, Marchwiany D, Duarte C, Bashir R. Patterned
tures in the dorsal skin fold chamber in mice. PLoS three-dimensional encapsulation of embryonic stem
One. 2013;8(3):e57741. cells using Dielectrophoresis and Stereolithography.
38. Dinca V, Ranella A, Farsari M, Kafetzopoulos D, Adv Healthc Mater. 2013;2(3):450–8.
Dinescu M, Popescu A. Quantification of the activity 53. Catros S, Guillotin B, Bačáková M, Fricain JC,

of biomolecules in microarrays obtained by direct laser Guillemot F. Effect of laser energy, substrate film
transfer. Biomed Microdevices. 2008;10(5):719–25. thickness and bioink viscosity on viability of endo-
39.
Cheng GJ, Pirzada D, Cai M, Mohanty P, thelial cells printed by laser-assisted bioprinting. Appl
Bandyopadhyay A. Bioceramic coating of hydroxy- Surf Sci. 2011;257(12):5142–7.
apatite on titanium substrate with Nd-YAG laser. 54. Jakab K, Norotte C, Damon B, Marga F, Neagu A,
Mater Sci Eng C. 2005;25(4):541–7. Besch-Williford CL. Tissue engineering by self-­
122 S. Noh et al.

assembly of cells printed into topologically defined 69. Izadifar Z, Chang T, Kulyk WM, Chen D, Eames
structures. Tissue Eng. 2007;14(3):110306233438005. BF. Analyzing biological performance of 3D-printed,
55. Irvine SA, Agrawal A, Lee BH, Chua HY, Low KY, cell-impregnated hybrid constructs for cartilage
Lau BC. Printing cell-laden gelatin constructs by tissue engineering. Tissue Eng Part C Methods.
­free-­form fabrication and enzymatic protein cross- 2016;22(3):173–88.
linking. Biomed Microdevices. 2015;17(1):1–8. 70. Kang H-W, Lee SJ, Ko IK, Kengla C, Yoo JJ, Atala
56. Duarte Campos DF, Blaeser A, Weber M, Jäkel J, A. A 3D bioprinting system to produce human-­
Neuss S, Jahnen-Dechent W. Three-dimensional scale tissue constructs with structural integrity. Nat
printing of stem cell-laden hydrogels submerged in Biotechnol. 2016;34(3):312–9.
a hydrophobic high-density fluid. Biofabrication. 71. Faulkner-Jones A, Fyfe C, Cornelissen D-J, Gardner
2013;5(1):015003. J, King J, Courtney A. Bioprinting of human pluripo-
57. Xu T, Binder KW, Albanna MZ, Dice D, Zhao W, Yoo tent stem cells and their directed differentiation into
JJ. Hybrid printing of mechanically and biologically hepatocyte-like cells for the generation of mini-livers
improved constructs for cartilage tissue engineering in 3D. Biofabrication. 2015;7(4):044102.
applications. Biofabrication. 2013;5(1):015001. 72. Gaetani R, Doevendans PA, Metz CHG, Alblas J,
58. Skardal A, Zhang J, Prestwich GD. Bioprinting
Messina E, Giacomello A. Cardiac tissue engineering
vessel-like constructs using hyaluronan hydrogels using tissue printing technology and human cardiac
crosslinked with tetrahedral polyethylene glycol tet- progenitor cells. Biomaterials. 2012;33(6):1782–90.
racrylates. Biomaterials. 2010;31(24):6173–81. 73. Marchioli G, van Gurp L, van Krieken PP, Stamatialis
59. Phamduy TB, Sweat RS, Azimi MS, Burow ME,
D, Engelse M, van Blitterswijk CA. Fabrication of
Murfee WL, Chrisey DB. Printing cancer cells into three-dimensional bioplotted hydrogel scaffolds for
intact microvascular networks: a model for investigat- islets of Langerhans transplantation. Biofabrication.
ing cancer cell dynamics during angiogenesis. Integr 2015;7(2):025009.
Biol (Camb). 2015;7:1068–78. 74. Pati F, Ha DH, Jang J, Han HH, Rhie JW, Cho

60. Pescosolido L, Schuurman W, Malda J, Matricardi P, DW. Biomimetic 3D tissue printing for soft tissue
Alhaique F, Coviello T. Hyaluronic acid and dextran-­ regeneration. Biomaterials. 2015;62:164–75.
based semi-IPN hydrogels as biomaterials for bio- 75. Huang S, Yao B, Xie J, Fu X. 3D bioprinted extracel-
printing. Biomacromolecules. 2011;12(5):1831–8. lular matrix mimics facilitate directed differentiation
61. Ronca A, Ambrosio L, Grijpma DW. Preparation of of epithelial progenitors for sweat gland regeneration.
designed poly(d,l-lactide)/nanosized hydroxyapa- Acta Biomater. 2016;32:170–7.
tite composite structures by stereolithography. Acta 76. Chang R, Emami K, Wu H, Sun W. Biofabrication
Biomater. 2013;9(4):5989–96. of a three-dimensional liver micro-organ as an
62. Miller JS, Stevens KR, Yang MT, Baker BM, Nguyen in vitro drug metabolism model. Biofabrication.
D-HT, Cohen DM. Rapid casting of patterned vascular 2010;2(4):045004.
networks for perfusable engineered three-­dimensional 77. Tourlomousis F, Chang RC. Numerical investigation
tissues. Nat Mater. 2012;11(7):768–74. of dynamic microorgan devices as drug screening
63. Sooppan R, Paulsen SJ, Han J, Ta AH, Dinh P,
platforms. Part I: macroscale modeling approach &
Gaffey AC. In vivo anastomosis and perfusion of a validation. Biotechnol Bioeng. 2016;113(3):612–22.
three-dimensionally-printed construct containing 78. Snyder J, Rin Son A, Hamid Q, Sun W. Fabrication
microchannel networks. Tissue Eng Part C Methods. of microfluidic manifold by precision extrusion depo-
2016;22(1):1–7. sition and replica molding for cell-laden device. J
64. Dolati F, Yu Y, Zhang Y, De Jesus AM, Sander
Manuf Sci Eng. 2015;138(4):041007.
EA, Ozbolat IT. In vitro evaluation of carbon-­ 79. Drachuk I, Suntivich R, Calabrese R, Harbaugh S,
nanotube-­ reinforced bioprintable vascular conduits. Kelley-Loughnane N, Kaplan DL. Printed dual cell
Nanotechnology. 2014;25(14):145101. arrays for multiplexed sensing. ACS Biomater Sci
65. Gao Q, He Y, Fu J, Liu A, Ma L. Coaxial nozzle-­ Eng. 2015;1:287–94.
assisted 3D bioprinting with built-in microchannels 80. Khaled SA, Burley JC, Alexander MR, Yang J,

for nutrients delivery. Biomaterials. 2015;61:203–15. Roberts CJ. 3D printing of five-in-one dose combi-
66. Lee VK, Lanzi AM, Ngo H, Yoo SS, Vincent PA, Dai nation polypill with defined immediate and sustained
G. Generation of multi-scale vascular network system release profiles. J Control Release. 2015;217:308–14.
within 3D hydrogel using 3D bio-printing technology. 81. Goyanes A, Robles Martinez P, Buanz A, Basit AW,
Cell Mol Bioeng. 2014;7(3):460–72. Gaisford S. Effect of geometry on drug release from
67. Roh H-S, Jung S-C, Kook M-S, Kim B-H. In vitro study 3D printed tablets. Int J Pharm. 2015;494(2):657–63.
of 3D PLGA/n-HAp/β-TCP composite scaffolds with 82. Markstedt K, Mantas A, Tournier I, Martínez Ávila H,
etched oxygen plasma surface modification in bone tis- Hägg D, Gatenholm P. 3D bioprinting human chondro-
sue engineering. Appl Surf Sci. 2016;388:321–30. cytes with nanocellulose-alginate bioink for cartilage
68. Van Rie J, Declercq H, Van Hoorick J, Dierick M, Van tissue engineering applications. Biomacromolecules.
Hoorebeke L, Cornelissen R. Cryogel-PCL combina- 2015;16(5):1489–96.
tion scaffolds for bone tissue repair. J Mater Sci Mater 83. Visser J, Peters B, Burger TJ, Boomstra J, Dhert WJA,
Med. 2015;26(3):124. Melchels FPW. Biofabrication of multi-material
5  3D Bioprinting for Tissue Engineering 123

a­ natomically shaped tissue constructs. Biofabrication. 87. Owens CM, Marga F, Forgacs G, Heesch CM.

2013;5(3):035007. Biofabrication and testing of a fully cellular nerve
84. Sawkins MJ, Mistry P, Brown BN, Shakesheff KM, graft. Biofabrication. 2013;5(4):045007.
Bonassar LJ, Yang J. Cell and protein compatible 88. Ling K. Bioprinting-based high-throughput fabri-
3D bioprinting of mechanically strong constructs for cation of three-dimensional MCF-7 human breast
bone repair. Biofabrication. 2015;7(3):035004. cancer cellular spheroids. Engineering. 2015;1(2):
85. Ng WL, Yeong WY, Naing MW. Polyelectrolyte
269–74.
gelatin-­
chitosan hydrogel optimized for 3D bio- 89. Duocastella M, Fernandez-Pradas JM, Morenza

printing in skin tissue engineering. Int J Bioprinting. JL, Zafra D, Serra P. Novel laser printing technique
2016;2 doi:10.18063/IJB.2016.01.009. for miniaturized biosensors preparation. Sensors
86. Merceron TK, Burt M, Seol Y-J, Kang H-W, Lee Actuators B Chem. 2010;145(1):596–600.
SJ, Yoo JJ. A 3D bioprinted complex structure for 90. Lam CXF, Mo XM, Teoh SH, Hutmacher DW. Scaffold
engineering the muscle–tendon unit. Biofabrication. development using 3D printing with a starch-based
2015;7(3):035003. polymer. Mater Sci Eng C. 2002;20(1–2):49–56.
Decellularization
6
Taekmin Kwon and Kyung Hyun Moon

6.1 Introduction sue matrices as scaffolds is very attractive,


because naturally derived polymers and synthetic
The main components used for tissue engineer- polymers are unable to replicate the precise spa-
ing are living cells, a scaffolding system based on tial organization of complex structures. These
biomaterials, bioactive factors, and appropriate biologic scaffolds made of ECM, typical in
microenvironments that facilitate cellular behav- reconstructive surgery, are used in a variety of
ior. A well-orchestrated combination of these applications, including regenerative medicine
components is of critical significance in creating strategies for increasing tissue and organ replace-
engineered tissues or organs for the development ment [4]. In in vitro studies, relying on the biore-
of functional substitutes [1]. The principal func- actor, researchers have examined the effect of
tion of a scaffolding system for tissue engineer- these scaffolds on cell proliferation and organ
ing is to provide a “template” to direct cellular structure. In in vivo implantations of decellular-
behavior, which includes cell migration, prolif- ized scaffolds, the effect of the scaffold on pro-
eration, and differentiation, and to maintain cell moting angiogenesis and regional regeneration
type-specific phenotypes [2]. Generally, three was explored (Fig. 6.1). The clinical use of decel-
classes of biomaterials are used for tissue engi- lularized scaffold blood vessels has been docu-
neering purposes: acellular tissue matrices, natu- mented for applications such as heart valves and
ral polymers, and synthetic polymers [3]. Body renal bladder. Nevertheless, the current applica-
organs are complex structures composed of tion is limited to anatomically simple organs, but
extracellular matrix (ECM) and cell ingredients. will eventually provide the foundation for com-
In the field of regenerative medicine, organs are plex organ regeneration in the future. The use of
decellularized, i.e., cellular components are decellularized scaffold in regenerative medicine
removed to produce an acellular ECM, or decel- has recently made a breakthrough. Despite varia-
lularized scaffolds. The use of decellularized tis- tions in the organ to be fashioned and used, these
scaffolds have a proven ability to promote regen-
eration. In this review, the most commonly used
decellularization methods are discussed, and
T. Kwon • K.H. Moon (*) their effect upon the resulting ECM structure and
Department of Urology, Ulsan University Hospital,
University of Ulsan College of Medicine,
composition is presented. We hope this discus-
Ulsan, South Korea sion will provide useful information regarding
e-mail: urofirst@gmail.com the decellularization methods.

© Springer Nature Singapore Pte Ltd. 2018 125


B.W. Kim (ed.), Clinical Regenerative Medicine in Urology, DOI 10.1007/978-981-10-2723-9_6
126 T. Kwon and K.H. Moon

a b c

Fig. 6.1  Schematic diagram of the liver regeneration defective part is replaced with decellularized liver scaf-
hypothesis using decellularized scaffolds. (a) Partial fold. (c) Cells in the native liver cross the suture border
resection of one hepatic lobule is performed. (b) The and regenerate on the liver scaffold. (From Yu et al. [2])

6.2 Rationale of an organ involves a combination of physical


for Decellularization and chemical processing. Physical treatment
can include agitation or sonication, mechanical
Xenogeneic and allogeneic cellular antigens massage and pressure, or a freeze–thaw pro-
are recognized as foreign by the host and there- cess. Using these methods, the cell contents are
fore induce an inflammatory response or an removed, the cell membrane is easily destroyed
immune-­ mediated rejection of the tissue. by subsequent rinsing and removal of the cell
However, the components of ECM are gener- content from the ECM. However, although
ally conserved among species and are tolerated these physical processes achieve complete
well, even by xenogeneic recipients [5, 6]. decellularization, they are insufficient, and
ECM from a variety of tissues, including heart must be combined with chemical treatment.
valves [7, 8], blood vessels [9, 10], skin [11], Enzymatic treatments, such as trypsin, ionic
nerves [12], skeletal muscle [13], tendons [14], detergents and chemical treatments, can dis-
ligaments [15], small intestinal submucosa rupt charge coupling among the cell mem-
[16], urinary bladder [17], and liver [18], has brane, cell, and extracellular component.
been reported for use in tissue engineering and Tissue is composed of both cell materials, and
regenerative medicine applications. The pur- ECM has a variable degree of miniaturization
pose of decellularization protocols, though according to the tissue source. ECM is suffi-
minimizing adverse effects on the mechanical cient to allow proper exposure of all the cells
integrity of the remaining ECM, is to effi- to the chaotropic agents. To provide a path for
ciently remove all cells and nuclear material removal from the organ, the cellular material
(Fig. 6.2). must be destroyed during the decellularization
With the purpose of removing the cells, any process. The purpose of the main part of the
of the processing steps changes the native decellularization process is to retain the origi-
three-­dimensional structure of ECM. The most nal mechanical and biological properties and
common method used for the decellularization minimize disruption.
6 Decellularization 127

a (i) (ii)

b (i) (ii)

c (i) (ii)

d (i) (ii)

TRENDS in Molecule Medicine

Fig. 6.2  Whole-organ scaffolds decellularized using per- was performed via the pulmonary artery. (ii) Cadaveric
fusion. The native extracellular matrixes (ECMs) of sheep lung before and after decellularization. (c) (i) Rat
cadaveric organs can be isolated by perfusion of the native kidney scaffold generated from cadaveric kidney using
vascular system with detergent solutions. The resulting perfusion decellularization. The abdominal aorta was can-
scaffolds are acellular, but maintain the structure of the nulated for perfusion. (ii) Porcine kidney before and after
native organ. (a) (i) Rat heart scaffold generated from a perfusion decellularization. (d) (i) Rat pancreas scaffold
cadaveric heart using perfusion decellularization. The generated from cadaveric pancreas by perfusion decellu-
ascending aorta was cannulated for perfusion. (ii) larization. The abdominal aorta was cannulated for perfu-
Cadaveric human heart before and after perfusion decel- sion. (ii) Human pancreas before and after perfusion
lularization. (b) (i) Rat lung scaffold generated from decellularization. (From Song and Ott [19])
cadaveric lung by perfusion decellularization. Perfusion
128 T. Kwon and K.H. Moon

6.3 Decellularization Protocols saline, or chelating agents, all of which facilitate


the dissociation of cells from their subjacent
The most robust and effective physical decellu- basement membrane [23]. However, the integrity
larization protocol uses a combination of chemi- of the underlying hyperfine structure and the
cals, and includes an enzymatic approach. basement membrane can been damaged by direct
Decellularization protocols, typically enzyme applications of mechanical force. The formation
treatment, surface-active agents, cytoplasmic, of barium crystals may disrupt the ultrastructural
solubilization of cellular components of the hydrostatic pressure of the ECM and requires
nucleus, and the final removal of cellular debris relatively little time compared with enzymes
from tissue, cellular components from the ECM, used to remove cells from detergents and blood
involve physical treatment or ionic solution. vessels. It may also be effective in corneal tissue
Following separation, the dissolution of the cell [25]. The temperature rises at the time of the
membrane begins. To enhance effectiveness, pressure of decellularization, but to prevent the
these steps can be combined with mechanical formation of crystals, it is possible to destroy the
stirring. Following decellularization, all the ECM because of the associated increase in
residual chemicals must be removed to avoid the entropy, which can be mitigated by a colloid such
response of chemically adverse host tissue. The as dextran [26].
efficiency of the storage of decellularization and
ECM can be assessed using one of several
methods. 6.3.2 Chemical Methods

6.3.2.1 Acids and Bases


6.3.1 Physical Methods Acids and bases cause and catalyze the hydroly-
sis of biological molecules. Peracetic acid, a
6.3.1.1 Temperature common disinfectant that by removing residual
The freeze–thaw process lyses the cells of the tis- nucleic acids has a minimal impact on the com-
sues and organs effectively; unless removed by position and structure of ECM, also serving as a
subsequent processing, the resulting membra- decellularization agent [27, 28].
nous and intracellular content remains. By using Acetic acid damages and removes collagens,
a single freeze–thaw cycle, a harmful immune thus decreasing ECM strength, but sulfated gly-
response, such as leukocyte infiltration into the cosaminoglycans (sGAG) are not affected. Bases
ECM scaffold, can be reduced [20]. Multiple (e.g., calcium hydroxide, sodium sulfide, and
freeze–thaw cycles may be used during decellu- sodium hydroxide) are used to remove hair from
larization [21] and do not significantly increase dermis samples during the early stages of decel-
the loss of ECM proteins from the tissue [22]. lularization [29].
Freeze–thaw processing does produce minor
­disruptions of the ECM ultrastructure [23] and 6.3.2.2 Hypotonic and Hypertonic
should therefore be used only when such effects Solutions
are acceptable in the final ECM product. The Hypertonic saline dissociates DNA from proteins
effect of freeze–thaw treatment on the mechani- [30]. Hypotonic solutions can readily cause cell
cal, load-bearing properties is minimal for a lysis because of simple osmotic effects, with
mechanically robust organ [24]. minimal changes in matrix molecules and archi-
tecture [31]. For a maximum osmotic effect for
6.3.1.2 Force and Pressure dipping alternately hyper- and hypotonic solution
Cells on the surface of a tissue or organ (e.g., uri- through a few cycles in the organ is common.
nary bladder, small intestine, skin, amnion) can Hypertonic and hypotonic solution also helps to
be effectively removed using mechanical abra- rinse the cell debris from within the organ after
sion in combination with enzymes, hypertonic dissolution.
6 Decellularization 129

6.3.2.3 Detergents α-galactosidase. Enzymes provide a high speci-


Ionic, non-ionic, and zwitterionic detergents solu- ficity for the removal of cell debris or undesirable
bilize cell membranes, isolating DNA from the ECM components. However, it is difficult to
protein and removing cellular material effectively effect complete cell removal in a single enzyme
from the tissue [30]. However, these agents also treatment; enzyme residues impair recellulariza-
disrupt and dissociate proteins in the ECM, as tion, and can induce a harmful immune response.
demonstrated by their use in the protein extraction Nucleases (e.g., DNase and RNase) cleave
procedures of tissue proteomics [22]. The removal nucleic acid sequences and thus help in the
of ECM proteins and DNA by detergents increases removal of nucleotides after cell lysis [26].
with exposure time [11] and varies with organ sub- Endonucleases such as benzonase [40] may be
unit, tissue type, and donor age [22]. Combining more effective than exonucleases because they
multiple detergents increases ECM protein loss cleave nucleotides mid-sequence and thereby
[32], but also allows for more complete detergent more effectively fragment DNA to prepare for its
removal from ECM after decellularization [33]. removal. Similarly, nonrestriction endonucleases
Triton X-100 can effectively remove cell residues fragment the DNA more effectively than
from thicker tissues, such as valve conduits, where sequence-dependent counterparts. Trypsin is a
enzymatic and osmotic methods are insufficient, serine protease that is used as an enzymatic
with concomitant ECM protein loss accompanied decellularization agent. However, ECM proteins
by a decreased adverse immune response in vivo such as collagens have limited resistance to tryp-
[34]. Sodium dodecyl sulfate (SDS) appears to be sin cleavage [41], and tissue exposure to trypsin
more effective than Triton X-100 at removing should therefore be used with caution. Trypsin is
nuclei from dense tissues and organs such as the slower at cell removal than detergents. Elastin
kidney and temporomandibular joint, while pre- and collagen are removed from the cell, but
serving tissue mechanics [35]. although trypsin is more destructive, it does show
better preservation of the glycosaminoglycan
6.3.2.4 Alcohols (GAG) content [42, 43]. Collagenase decellular-
Alcohol such as glycerol acid, decellularizes by ization may be employed, but only if mainte-
dehydrating and lysing the cells [29]. Phospholipids nance of the ultrastructure and of collagen is not
in valve leaflets and conduits contribute to prosthe- critical to the intended clinical applications of the
sis calcification and failure and can be extracted resulting ECM.
using alcohols [36]. In fact, isopropanol, ethanol,
and methanol are more effective than lipase at the 6.3.3.2 Nonenzymatic Agents
removal of lipids from adipose tissue in a relatively Ethylenediaminetetraacetic acid (EDTA) and
short period of time [37]. Methanol has been used in ethylene glycol tetraacetic acid (EGTA) are che-
combination with chloroform during tissue delipi- lating agents that aid cell dissociation from
dation [29]. Caution should be used when treating ECM proteins by sequestering metal ions [44].
tissues with alcohols such as ethanol and methanol Chelating agents are likely to contribute to sub-
because of their use as tissue fixatives in histology, tle disruptions in protein–protein interactions
their ability to precipitate proteins [38], and the using the same mechanism [45]. Chelating
damage they cause to the ECM ultrastructure [39]. agents alone are not sufficient for superficial
cell removal, even if agitated [23], and are thus
usually used with enzymes such as trypsin [46]
6.3.3 Biologic Agents or with detergents [20]. The effectiveness of the
chelate plus a simple hyper- or hypotonic solu-
6.3.3.1 Enzymes tion for decellularization is unknown [20].
Enzymes that have been reported in the decellu- Toxins such as latrunculin enable the researcher
larized protocol include nuclease, trypsin, to take advantage of generating a natural cyto-
collagenase, lipase, dispase, thermolysin, and
­ toxic agent for decellularization . Gillies et al.
130 T. Kwon and K.H. Moon

[47] demonstrated removal of DNA and intra- 6.5 Sterilization


cellular proteins from dense tissue, the tibialis of Decellularized Scaffolds
anterior, using only latrunculin B, hyper- and
hypotonic solutions, and DNase treatments. Clinical application of a decellularized scaffold
This method provided superior removal of DNA requires donor scaffold sterilization. The steril-
and retention of GAG to the enzymes and deter- ization process primarily eliminates endotoxins
gents for decellularization. The properties of and intact viral and bacterial DNA that may
passive mechanical testing of the ECM scaffold induce unwanted inflammation upon scaffold
were similar. implantation [57]. Decellularized scaffolds may
be sterilized using simple treatments such as
incubation in acids [58] or solvents [59], but such
6.4  hanges in the Mechanical
C methods may not provide sufficient penetration
Properties of Decellularized or may damage key ECM constituents [60].
ECM During Constructive Sterilization methods such as ethylene oxide
Remodeling exposure, gamma irradiation, and electron beam
irradiation achieve effective sterilization, but are
Facilitating the replacement of the ECM graft known to alter ECM microstructure and mechan-
with functional host tissue is considered a con- ical properties [61, 62].
structive remodeling response. ECM scaffold
changes during such a remodeling response are
dependent on factors such as the local tissue 6.5.1 Ethylene Oxide Exposure
microenvironment, the rate of scaffold degra-
dation, forces present within the mechanical Ethylene oxide sterilization does not affect cell
environment, and the rate and extent to which attachment to ECM or the stimulation of growth
the infiltrating cells deposit new ECM [48, 49]. factor secretion by fibroblasts in vitro [28].
Decellularized scaffolds retain a variety of col- Ethylene oxide may substantially change ECM
lagens, growth factors, glycosaminoglycans, mechanical properties [63] or leave them unal-
and other matrix proteins, and the arrangement tered [24, 62], and ethylene oxide treatment can
of the proteins is minimally disturbed through cause undesirable host immune responses that
the process [50–53]. Upon implantation, impair proper function of the biologic scaffold
mononuclear cells migrate into the decellular- after implantation [64]. Therefore, the use of eth-
ized scaffold and degrade it, while concomi- ylene oxide for the sterilization of decellularized
tantly depositing new site-appropriate tissue scaffolds should be considered carefully.
[54, 55]. The structural changes that occur dur-
ing in vivo remodeling of ECM scaffolds are
associated with marked changes in scaffold 6.5.2 Gamma Irradiation Exposure
strength. In the early phase of remodeling, deg-
radation occurs quite rapidly, before the newly Degradation of ECM during gamma irradiation is
deposited ECM can become fully organized. at least partially attributed to spontaneous dena-
However, once the infiltrating cells have estab- turation of key structural proteins such as colla-
lished residence and begin producing new site- gen that occurs even at relatively low doses at
specific ECM, the remodeling process body temperature [65]. As the irradiation dose
progresses over time toward a tissue construct increases, denaturation of collagen continues to
that has similar mechanical properties very increase, resulting in dose-dependent alterations
similar to those of native tissue [54, 56]. in mechanical properties [66]. Even at a dose as
6 Decellularization 131

low as 2 kGy, gamma irradiation increases tissue in vitro cytotoxicity and adverse host responses
stiffness and tensile strength, but at doss higher in vivo upon reintroduction of cells [71–73]. There
than 15 kGy, mechanical properties decrease in a are conflicting results on the effect of decellular-
dose-dependent manner [67]. Gamma irradiation ization techniques. Grauss et al. [74] found that
causes residual lipids to become cytotoxic and chemically induced decellularization using Triton
accelerates enzymatic degradation [67, 68]. X-100 or trypsin resulted in changes in the ECM
constitution, which could lead to problems in
valve functionality and cell growth and migration,
6.5.3 Supercritical Carbon Dioxide whereas Schenke-Layland et al. [75] used a decel-
lularization method, in which pulmonary valves
Supercritical carbon dioxide has become a main- were dissected from porcine hearts, placed in a
stay of the sterilization of food and pharmaceuti- solution of trypsin-EDTA, incubated at 37°C for
cals. It has recently been investigated as an 24 h, followed by a 24-h wash in phosphate-buff-
alternative method of sterilizing decellularized ered solution (PBS) , and showed acceptable
scaffolds. Supercritical carbon dioxide is versa- removal of cell components from a porcine pulmo-
tile for sterilization owing to its nonreactive nary valve. Cartmell and Dunn [14] compared the
nature, its ability to penetrate cells and tissues, its methods of decellularization for rat tail tendon
reduced energy usage, and its improved quality using three extraction chemicals (t-octylphenoxy-
of retention of heat-sensitive substrates [69]. polyethoxyethanol [Triton X-100], tri(n-butyl)
Supercritical carbon dioxide sterilization with phosphate [TnBP], and sodium dodecyl sulfate
peracetic acid sterilant causes only minor changes [SDS]). They showed that treatment of tendons
in the susceptibility of porcine acellular matrix to with 1% Triton X-100 for 24 h disrupted the col-
collagenase digestion, in tensile or tear strength, lagen fiber structure, but did not remove cells.
indicating limited alteration of the tissue struc- Treatment with 1% SDS for 24 h or 1% TnBP for
ture following supercritical carbon dioxide steril- 48 h resulted in an acellular tendon matrix with
ization [70]. However, supercritical carbon retention of near-normal structure and mechanical
dioxide sterilization is relatively new and requires properties. Woods and Gratzer [15] also compared
further investigation. the effect of decellularization using one of the
three protocols incorporating the surfactant lauryl
sulfate (SDS), Triton X-100, and/or an organic sol-
6.6  ffects and Evaluation
E vent, tri(n-­butyl)phosphate (TnBP). They showed
of Decellularized Scaffolds that all three treatments were effective at removing
cells and preserving the mechanical properties of
The major concerns of all decellularization proto- the graft, with subtle, but notable, differences.
cols remain ECM disruption, immunogenicity, Most detergents, when used as agents for decellu-
and thrombogenicity. The effectivity of decellular- larization, cause some removal of GAG from the
ization and the alterations to the ECM vary accord- scaffold, which has varying degrees of a negative
ing to the source, composition, and density of the effect on the tensile viscoelasticity of the scaffold
tissue, and to other factors. Decellularization [76]. Another factor that can influence the mechan-
agents that are very efficient at removing cellular ical properties of the scaffold is the duration of
components can also cause damage to the collagen exposure to decellularization agents [77]. There is
structure and remove growth factors and other practically no consensus regarding the effects of
important ECM components. However, because of any of the decellularization methods. Therefore, it
insufficient decellularization, residual cellular is very important to optimize the decellularization
components within ECM may contribute to method for obtaining acceptable cell removal
132 T. Kwon and K.H. Moon

according to the source, composition and density human tendon scaffolds in vivo using biolumi-
of the tissue, and other factors. nescent imaging and immunohistochemistry.
Evaluation of the residual materials and cel- They showed that reseeded cells remained via-
lular components within the decellularized ble on the implanted constructs at up to 4 weeks
ECM is a pivotal step in translating them into a using bioluminescent imaging, and histological
clinically practicable, transplantable substance. evaluation showed host cell invasion and prolif-
There are a number of methods available to eration of the repair sites. Biomechanical test-
determine the effectivity of the elimination of ing revealed no significant difference in
cellular material from tissues. Although the ultimate load to failure and a 2-mm gap force.
decellularization process cannot remove 100% This movement of host cells and human adi-
of cell material, it is crucial in quantifying cel- pose-derived stem cells into the scaffold sug-
lular components such as double-stranded DNA gests the biointegration of the allograft. Lin
(dsDNA), mitochondria, or membrane-associ- et al. [81] evaluated the biocompatibility of the
ated molecules such as phospholipids in a decellularized scaffolds using enzyme–deter-
decellularized scaffold. Crapo et al. [57] sug- gent methods for the cell removal of mouse
gested the following minimal criteria sufficient skeletal muscle tissue by histological staining
to satisfy the intent of decellularization: (1) no (sections stained with H&E, Van Gieson’s, and
visible nuclei upon histological evaluation, (2) DAPI) and DNA quantification. They showed
the remaining dsDNA content should not that mouse skeletal muscles combined with an
exceed 200 base pairs in length, and (3) the enzyme–detergent mixture (trypsin and Triton
amount of dsDNA should not exceed 50 ng/mg X-100) can yield an intact matrix devoid of
of dry weight of the material. The first criterion cells, depleting more than 93% of the nuclear
is easily evaluated using standard histological component and exhibiting comparable biome-
staining with hematoxylin and eosin or immu- chanical properties to those of native tissue,
nofluorescent methods such as DAPI or and that infiltration of inflammatory cells
Hoechst. The second and third criteria are eas- increased into the scaffold initially and then
ily evaluated using commercially available decreased gradually until day 30. Although the
dsDNA intercalators, such as PicoGreen®, above methods provide important information
propidium iodide, or bisbenzimide, and by gel regarding the effectiveness of decellularization
electrophoresis respectively. Jackson and methods, the biologic consequences of small
Simon [78] demonstrated the fate of DNA from amounts of nuclear material or cytoplasmic
allografts after transplantation using a DNA debris within the decellularized scaffold are
probe technique that clearly distinguished unclear. Crapo et al. [57] suggested that a stan-
donor cells from host cells and could also be dard for tissue decellularization might provide
utilized to evaluate whether any DNA is present numerous benefits, including (1) allowing
in the decellularized scaffold in the Spanish investigators and ECM product manufacturers
goat model. Electron microscopic methods or to evaluate the effectiveness of a protocol when
polymerase chain reaction (PCR) are usable, reporting new decellularization techniques or
but not typically used to evaluate the presence describing products composed of ECM derived
of residual nuclear material or cytoplasmic from decellularized tissue; (2) enabling congru-
debris because of the technical complexity and ous comparison of different ECM products; (3)
expense of these procedures for routine eliminating variations in cell and host responses
work [79]. Schmitt et al. [80] studied the cell to ECM products caused by variations in resid-
viability, cell migration parameters, and ual DNA, thereby facilitating interpretation and
biomechanical properties of human adipose-
­ comparison of in vitro and in vivo results; and
derived stem cells following reseeding on (4) promoting the rapid and effective develop-
6 Decellularization 133

ment of additional clinical applications for 6.8 Eight Decellularized


ECM products within the field of regenerative Scaffolds as a Platform
medicine and tissue engineering. for Bioengineered Organs

6.8.1 Liver
6.7  emoval of Residual
R
Chemicals Liver tissue engineering provides an insight into
liver regeneration; it has seen remarkable prog-
The decellularization methods involve a wide ress in recent years [83, 84]. In 2010, liver scaf-
variety of chemicals used because of their capa- folding of transferred and intact acellular liver
bility to damage cells. The strong chemicals can has been developed by perfusion of various
thoroughly remove the cell components, but the chemical detergents into the portal vein of the rat.
internal fiber scaffold structure may also be seri- These scaffolds maintain the function of the
ously damaged, whereas mild chemicals can pre- microvasculature and the three-dimensional
serve the internal fiber scaffold structure. structure of ECM components (Fig. 6.3) [84].
However, the removal effect of cell components Scaffolding of decellularized liver has demon-
is far from satisfying, which is the messy prob- strated the ability to efficiently support in vitro
lem of chemical extraction. The presence of some decellularization with subsequent perfusion of
residual decellularization agents within ECM primary liver cells [85, 86]. In in vivo microinjec-
biomaterials is toxic to host cells and may be tion of scaffolds from decellularized liver with
responsible for discouraging cellular ingrowth microscopic vascular anastomosis, the scaffold
[43, 82]. There is a need for the development of showed seeding cells. However, thrombosis
assays to quantify the presence of residual chem- ­formation was noticed shortly post-transplanta-
icals in the decellularized scaffold material and tion [87]. To address thrombus formation, hepa-
the removal methods of residual chemicals. rin was perfused in the multilayer on the inside

a b c

d e f

Fig. 6.3  Fabrication, vascular cast, light microstructure, the hepatic artery (red), and the hepatic duct (transparent).
and implantation of the decellularized liver scaffolds. (a) Scale bar 2 mm. (d) H&E staining of liver matrix shows
Decellularization of a single lobe of rat liver progressing the existence of blue-stained nuclei in intact liver, but not
under continuous detergent perfusion. Scale bar 10 mm. in (e) decellularized liver scaffold. (f) H&E staining
(b) Decellularized whole-liver scaffold with the hepatic results show the border between the liver parenchyma and
artery intact. Scale bar 20 mm. (c) Vessel corrosion cast- the implanted decellularized scaffold. Scale bar 100 μm.
ing of the microstructure of the hepatic portal vein (blue), (From Yu et al. [2])
134 T. Kwon and K.H. Moon

surface of the scaffold [88, 89]. Despite the short-­ 6.8.3 Lung
term effectiveness of this intervention, long-term
effectiveness requires further experiments. The study of lung tissue regeneration has passed
through two stages. The basic concept of the regen-
eration of the lung stem cell is possible to play the
6.8.2 Heart lung tissue, to join the synthetic material and lung
stem cells to build functional pulmonary units
The first decellularized cardiac scaffolds were (alveoli). Based on such a proposal, lung stem cells
produced from rats in 2008 [14]. These scaffolds were seeded in synthetic material in vivo and
were perfused with heart muscle cells and vascu- in vitro. The construct could not be formed, owing
lar endothelial cells in vitro to mimic the cell to the poor integration and tissue compatibility and
composition of the heart. This construct success- the possibility of complete failure of the respiratory
fully ran the pump function after transplantation function caused by surgical infection [94, 95].
[90]. Induced pluripotent stem cells (iPSCs) of Recently, lung tissue engineering has been facili-
human origin were seeded onto decellularized tated by decellularized scaffold in vivo and in vitro.
mouse heart in vitro. The seeded iPSCs migrated, Although the cellular components are removed
proliferated, and differentiated into functional during decellularization, the relevant cytokines and
cardiomyocytes after implanting, enabling the ECM are retained [96].
constructed cardiac tissues to demonstrate con-
tractility [91].
Recently, these studies focused on the repair of 6.8.4 Kidney
myocardial infarction after myocardial ischemia.
Bone marrow mesenchymal stem cells (MSCs) Porcine kidneys have been successfully decel-
were promoted to cardiac repair after myocardial lularized, proposing the possibility of using
ischemia or infarction [92]. Transplantation of these transplantable scaffolds to construct
stem cells improves the tissue condition and over- tissue-­ engineered kidney that is clinically
all cardiac function of infarcted tissue [93]. applicable (Fig. 6.4) [97]. Whole porcine kid-

a b c

Fig. 6.4  Gross appearance of a porcine kidney during the translucent. (c) After 32 h, the kidney is fully decellular-
decellularization process (a) Beginning of decellulariza- ized and is white. (From Guan et al. [97])
tion. (b) After 16 h of perfusion, the kidney became more
6 Decellularization 135

a b e f

c
g

Fig. 6.5  Fabrication and implantation of decellularized kidneys observed under a stereoscopic microscope. Scale
kidney scaffolds. (a) With continuous detergent perfusion, bar 20 mm. (e) Electron microscopy observation shows
the rat decellularized kidney scaffold shows different intact ECM in decellularized kidney scaffold. Scale bar
gross appearances. Scale bar 10 mm. (b) Casting model of 2 μm. (f) Radionuclide scanning analysis of experimental
decellularized kidney scaffolds shows intact microvessels. kidneys. (g) H&E staining shows the border between the
(c) Decellularized scaffolds were sutured to a rat that renal parenchyma and implanted decellularized scaffold.
underwent partial nephrectomy. (d) Macroscopic images Scale bar 100 μm. (From Yu et al. [2])
show longitudinal cross-sections of whole experimental

neys were decellularized and then orthotopi- 6.8.5 Pancreas


cally transplanted in vivo, and then
anticoagulant was administered as prophylaxis. Early studies on pancreatic regeneration focused
Inflammatory cells in the pericapsular region on synthetic materials such as cross-linked col-
and thrombosis occurred owing to the lack of lagen matrix liquid scaffold [100, 101]. First,
endothelial cells [98]. In recent findings, suc- decellularized pancreatic scaffolds were pro-
cessfully engineered renal tissue absorbed by duced from a porcine model in 2013. These scaf-
the proximal tubule was shown to have the folds were then seeded with human amniotic
ability of metabolic and endocrine functions fluid-derived stem cells (hAFSCs) and porcine
[99]. Yu et al. [4] successfully reported that pancreatic islets. The scaffolds promoted cell
renal decellularized scaffolds induce the regen- proliferation and maintained cellular function
eration of injured kidney (Fig. 6.5). There is a [102]. Pancreatic acinar cells and β cells were
possibility that various cytokines in the scaf- used to construct decellularized pancreatic scaf-
fold play an important role in the recovery of folds in vitro and resulted in an increased insulin
postoperative renal function after partial level post-subcutaneous transplantation [103]. In
nephrectomy. recent studies, after implantation into mice in
136 T. Kwon and K.H. Moon

in vivo, constructs were shown that synthesized defects in the internal organs, before the clinical
pancreas using an artificial three-dimensional application of internal organ scaffolding. Bladder
material with β cells that regulate the blood glu- acellular scaffold was used to repair a defect in
cose level [104]. the bladder in rats in 1996 [109]. Because of the
similarity in anatomical simplicity, bladder acel-
lular scaffold has also been used in the
6.8.6 Spinal Cord and Brain ­reconstruction of other visceral organs, such as
the tympanic membrane [110], esophagus [111],
First, spinal scaffolds made the cellular structure, trachea [112], larynx [113], glottis [114], tho-
myelin, and nervous process of rats disappeared, racic wall [115], ventricular wall [116], small
whereas most ECM structural proteins were pre- intestine [117], and arteries [118]. The process
served [105]. the weakness of the immunogenic- based on the scaffolding of the internal organs, in
ity of the subcutaneous spinal cord scaffold addition to mobility, requires adequate blood
positioning, when implanted, and infiltration of supply to support the repair of the structure and
CD4+ and CD8+ cells were not obvious. Rat spi- components of the organ [119]. In the presence of
nal scaffolds were combined with mesenchymal a blood supply, i.e., scaffold can promote trans-
stem cells from human umbilical cord blood and planted stem cells to enhance recovery and the
then implanted into rat spinal cords. The results growth of cell function [120, 121]. In addition,
showed that nerve cells migrated into the scaf- modified scaffold can inhibit the inflammatory
fold, accompanied by the formation of new response for better integration into the recipient
myelinated axons, resulting in motor function site [122].
recovery [106]. Decellularized cerebral scaffolds,
derived from porcine brains, failed to maintain
the original structure, but the ECM, containing 6.8.8 Skin
GAGs, was successfully preserved [107].
The study suggests that ECM could be used The development of bioengineered products of
for cell culture because of nerve biocompatibil- different skin layers—including the tissue-­
ity. Nerve cells derived from human-iPSCs have engineered epidermis, dermis, and composite
grown and mature on the matrix. Brain matrix skin—has provided innovative tools for clinical
can also be processed into an injectable hydrogel applications. Cultured epithelial autograft
fiber structure. Porcine brain, spinal cord, and (CEA), an approach to obtaining epidermal
optic nerve were decellularized using a combina- grafts, has been used in the repair of major burns.
tion of freeze–thaw, trypsin digestion, and chem- Tissue-­engineered epithelial cells prepared by
ical detergents. The generated cross-linked culturing human self-epidermal keratinocytes
scaffolds preserved various growth factors, were in vitro have been transported to two patients to
cultured with pc12 cells, and demonstrated an repair burns [123].
ability to promote cell proliferation, migration, However, absence of dermis layer and
and differentiation. ECM from the central ner- wound contracture and may lead poor cells
vous system is likely to be more effective than adhesion and subsequent survival. In addition,
bladder ECM at promoting the proliferation and swelling and scar contracture have been
differentiation of neural cells [108]. reported in the later stages. Artificial skin,
developed through extensive experimentation,
comprising a layer of silastic (epidermis) and a
6.8.7 Visceral Organs porous bovine collagen–chondroitin 6-sulfate
(dermis), was used physiologically to repair
Decellularized scaffolds derived from the bladder extensive burn injuries constituting 50–95% of
and mucosal layer of the small intestine were the body surface area [124]. Compared with
widely adapted to be used for the treatment of engineered epidermis, scaffolds of skin
6 Decellularization 137

p­ romote the migration of fibroblasts and angio- 8. Kasimir MT, Rieder E, Seebacher G, et al.
Comparison of different decellularization proce-
genesis and have the ability to provide optimal
dures of porcine heart valves. Int J Artif Organs.
mechanical and physicochemical properties 2003;26:421–7.
necessary for healing. 9. Conklin BS, Richter ER, Kreutziger KL, Zhong DS,
Chen C. Development and evaluation of a novel
decellularized vascular xenograft. Med Eng Phys.
2002;24:173–83.
6.9 Concluding Remarks 10. Dahl SL, Koh J, Prabhakar V, Niklason LE.
Decellularized native and engineered arterial
The galloping field of decellularization holds scaffolds for transplantation. Cell Transplant.
2003;12:659–66.
great promise for the creation of biologic scaf-
11. Chen RN, Ho HO, Tsai YT, Sheu MT. Process devel-
folds for tissue engineering and regenerative med- opment of an acellular dermal matrix (ADM) for bio-
icine. It is unlikely that any combination of medical applications. Biomaterials. 2004;25:2679–86.
methods will eliminate all cell components from a 12. Hudson TW, Liu SY, Schmidt CE. Engineering an
improved acellular nerve graft via optimized chemi-
tissue or organ. However, it seems obvious that
cal processing. Tissue Eng. 2004;10:1346–58.
methods that can eliminate most or all the cellular 13. Borschel GH, Dennis RG, Kuzon WM Jr. Contractile
components result in biologic scaffolds that are skeletal muscle tissue-engineered on an acellular
safe for transplantation. The beneficial effects of scaffold. Plast Reconstr Surg. 2004;113:595–602.
discussion 3-4
biologic scaffolds in the field of tissue engineer-
14. Cartmell JS, Dunn MG. Effect of chemical treat-
ing and regenerative medicine can be achieved if ments on tendon cellularity and mechanical proper-
optimal methods of decellularization are applied. ties. J Biomed Mater Res. 2000;49:134–40.
Furthermore, these biologic scaffolds are clini- 15. Woods T, Gratzer PF. Effectiveness of three extrac-
tion techniques in the development of a decellular-
cally relevant and important in the continued
ized bone-anterior cruciate ligament-bone graft.
development of decellularization methods. Biomaterials. 2005;26:7339–49.
16. Wang ZQ, Watanabe Y, Toki A. Experimental
assessment of small intestinal submucosa as a
small bowel graft in a rat model. J Pediatr Surg.
References 2003;38:1596–601.
17. Kropp BP, Eppley BL, Prevel CD, et al. Experimental
1. Langer R, Vacanti JP. Tissue engineering. Science. assessment of small intestinal submucosa as a blad-
1993;260:920–6. der wall substitute. Urology. 1995;46:396–400.
2. Yu Y, Alkhawaji A, Ding Y, Mei J. Decellularized 18. Lin P, Chan WC, Badylak SF, Bhatia SN. Assessing
scaffolds in regenerative medicine. Oncotarget. porcine liver-derived biomatrix for hepatic tissue
2016;7:58671–83. engineering. Tissue Eng. 2004;10:1046–53.
3. Pariente JL, Kim BS, Atala A. In vitro biocompat- 19. Song JJ, Ott HC. Organ engineering based
ibility assessment of naturally derived and synthetic on decellularized matrix scaffolds. Trends
biomaterials using normal human urothelial cells. J Mol Med. 2011;17(8):424–32. doi:10.1016/j.
Biomed Mater Res. 2001;55:33–9. molmed.2011.03.005.
4. Yu YL, Shao YK, Ding YQ, et al. Decellularized 20. Lehr EJ, Rayat GR, Chiu B, et al. Decellularization
kidney scaffold-mediated renal regeneration. reduces immunogenicity of sheep pulmonary
Biomaterials. 2014;35:6822–8. artery vascular patches. J Thorac Cardiovasc Surg.
5. Exposito JY, D’Alessio M, Solursh M, Ramirez 2011;141:1056–62.
F. Sea urchin collagen evolutionarily homologous 21. Cortiella J, Niles J, Cantu A, et al. Influence of acel-
to vertebrate pro-alpha 2(I) collagen. J Biol Chem. lular natural lung matrix on murine embryonic stem
1992;267:15559–62. cell differentiation and tissue formation. Tissue Eng
6. Constantinou CD, Jimenez SA. Structure of cDNAs A. 2010;16:2565–80.
encoding the triple-helical domain of murine alpha 22. Patel N, Solanki E, Picciani R, et al. Strategies to
2 (VI) collagen chain and comparison to human and recover proteins from ocular tissues for proteomics.
chick homologues. Use of polymerase chain reaction Proteomics. 2008;8:1055–70.
and partially degenerate oligonucleotide for genera- 23. Hopkinson A, Shanmuganathan VA, Gray T, et al.
tion of novel cDNA clones. Matrix. 1991;11:1–9. Optimization of amniotic membrane (AM) denuding
7. Booth C, Korossis SA, Wilcox HE, et al. Tissue engi- for tissue engineering. Tissue Eng Part C Methods.
neering of cardiac valve prostheses I: development and 2008;14:371–81.
histological characterization of an acellular porcine 24. Jackson DW, Grood ES, Wilcox P, et al. The
scaffold. J Heart Valve Dis. 2002;11:457–62. effects of processing techniques on the mechanical
138 T. Kwon and K.H. Moon

p­ roperties of bone-anterior cruciate ligament-bone 39. Cole MB Jr. Alteration of cartilage matrix mor-
allografts. An experimental study in goats. Am J phology with histological processing. J Microsc.
Sports Med. 1988;16:101–5. 1984;133:129–40.
25. Sasaki S, Funamoto S, Hashimoto Y, et al. In vivo 40. Petersen TH, Calle EA, Zhao L, et al. Tissue-­
evaluation of a novel scaffold for artificial cor- engineered lungs for in vivo implantation. Science.
neas prepared by using ultrahigh hydrostatic pres- 2010;329:538–41.
sure to decellularize porcine corneas. Mol Vis. 41. Waldrop FS, Puchtler H, Meloan SN, Younker
2009;15:2022–8. TD. Histochemical investigations of different types
26. Funamoto S, Nam K, Kimura T, et al. The use of of collagen. Acta Histochem Suppl. 1980;21:23–31.
high-hydrostatic pressure treatment to decellularize 42. Yang M, Chen CZ, Wang XN, Zhu YB, Gu
blood vessels. Biomaterials. 2010;31:3590–5. YJ. Favorable effects of the detergent and enzyme
27. Gilbert TW, Wognum S, Joyce EM, et al. Collagen extraction method for preparing decellularized
fiber alignment and biaxial mechanical behavior of bovine pericardium scaffold for tissue engineered
porcine urinary bladder derived extracellular matrix. heart valves. J Biomed Mater Res B Appl Biomater.
Biomaterials. 2008;29:4775–82. 2009;91:354–61.
28. Hodde J, Janis A, Ernst D, et al. Effects of ster- 43. Rieder E, Kasimir MT, Silberhumer G, et al.
ilization on an extracellular matrix scaffold. Decellularization protocols of porcine heart valves
I. Composition and matrix architecture. J Mater Sci differ importantly in efficiency of cell removal and
Mater Med. 2007;18:537–43. susceptibility of the matrix to recellularization with
29. Prasertsung I, Kanokpanont S, Bunaprasert T, human vascular cells. J Thorac Cardiovasc Surg.
Thanakit V, Damrongsakkul S. Development of 2004;127:399–405.
acellular dermis from porcine skin using periodic 44. Gailit J, Ruoslahti E. Regulation of the fibronectin
pressurized technique. J Biomed Mater Res B Appl receptor affinity by divalent cations. J Biol Chem.
Biomater. 2008;85:210–9. 1988;263:12927–32.
30. Cox B, Emili A. Tissue subcellular fractionation 45. Maurer P, Hohenester E. Structural and functional
and protein extraction for use in mass-spectrometry-­ aspects of calcium binding in extracellular matrix
based proteomics. Nat Protoc. 2006;1:1872–8. proteins. Matrix Biol. 1997;15:569–80. discussion
31. Xu CC, Chan RW, Tirunagari N. A biodegrad- 81
able, acellular xenogeneic scaffold for regenera- 46. Wainwright JM, Czajka CA, Patel UB, et al.
tion of the vocal fold lamina propria. Tissue Eng. Preparation of cardiac extracellular matrix from an
2007;13:551–66. intact porcine heart. Tissue Eng Part C Methods.
32. Alhamdani MS, Schroder C, Werner J, et al. 2010;16:525–32.
Single-step procedure for the isolation of proteins 47. Gillies AR, Smith LR, Lieber RL, Varghese
at near-native conditions from mammalian tissue S. Method for decellularizing skeletal muscle with-
for proteomic analysis on antibody microarrays. J out detergents or proteolytic enzymes. Tissue Eng
Proteome Res. 2010;9:963–71. Part C Methods. 2011;17:383–9.
33. Cebotari S, Tudorache I, Jaekel T, et al. Detergent 48. Badylak S, Kokini K, Tullius B, Whitson B. Strength
decellularization of heart valves for tissue engi- over time of a resorbable bioscaffold for body wall
neering: toxicological effects of residual deter- repair in a dog model. J Surg Res. 2001;99:282–7.
gents on human endothelial cells. Artif Organs. 49. Badylak SF, Freytes DO, Gilbert TW. Extracellular
2010;34:206–10. matrix as a biological scaffold material: structure
34. Meyer SR, Chiu B, Churchill TA, et al. Comparison and function. Acta Biomater. 2009;5:1–13.
of aortic valve allograft decellularization techniques 50. Hodde JP, Badylak SF, Brightman AO, Voytik-­
in the rat. J Biomed Mater Res A. 2006;79:254–62. Harbin SL. Glycosaminoglycan content of small
35. Lumpkins SB, Pierre N, McFetridge PS. A mechani- intestinal submucosa: a bioscaffold for tissue
cal evaluation of three decellularization methods replacement. Tissue Eng. 1996;2:209–17.
in the design of a xenogeneic scaffold for tissue 51. Hodde JP, Record RD, Liang HA, Badylak
engineering the temporomandibular joint disc. Acta SF. Vascular endothelial growth factor in porcine-­
Biomater. 2008;4:808–16. derived extracellular matrix. Endothelium.
36. Levy RJ, Vyavahare N, Ogle M, et al. Inhibition of 2001;8:11–24.
cusp and aortic wall calcification in ethanol- and 52. Hodde J, Record R, Tullius R, Badylak
aluminum-­ treated bioprosthetic heart valves in S. Fibronectin peptides mediate HMEC adhesion to
sheep: background, mechanisms, and synergism. J porcine-derived extracellular matrix. Biomaterials.
Heart Valve Dis. 2003;12:209–16. discussion 16 2002;23:1841–8.
37. Brown BN, Freund JM, Han L, et al. Comparison of 53. McDevitt CA, Wildey GM, Cutrone RM.
three methods for the derivation of a biologic scaf- Transforming growth factor-beta1 in a sterilized tis-
fold composed of adipose tissue extracellular matrix. sue derived from the pig small intestine submucosa.
Tissue Eng Part C Methods. 2011;17:411–21. J Biomed Mater Res A. 2003;67:637–40.
38. Jamur MC, Oliver C. Cell fixatives for immunostain- 54. Badylak S, Kokini K, Tullius B, Simmons-Byrd
ing. Methods Mol Biol. 2010;588:55–61. A, Morff R. Morphologic study of small intestinal
6 Decellularization 139

submucosa as a body wall repair device. J Surg Res. 70. Qiu QQ, Leamy P, Brittingham J, et al. Inactivation
2002;103:190–202. of bacterial spores and viruses in biological material
55. Gilbert TW, Stewart-Akers AM, Simmons-Byrd A, using supercritical carbon dioxide with sterilant. J
Badylak SF. Degradation and remodeling of small Biomed Mater Res B Appl Biomater. 2009;91:572–8.
intestinal submucosa in canine Achilles tendon 71. Nagata S, Hanayama R, Kawane K. Autoimmunity
repair. J Bone Joint Surg Am. 2007;89:621–30. and the clearance of dead cells. Cell.
56. Badylak SF, Vorp DA, Spievack AR, et al. 2010;140:619–30.
Esophageal reconstruction with ECM and muscle 72. Manfredi AA, Capobianco A, Bianchi ME, Rovere-­
tissue in a dog model. J Surg Res. 2005;128:87–97. Querini P. Regulation of dendritic- and T-cell fate
57. Crapo PM, Gilbert TW, Badylak SF. An overview of by injury-associated endogenous signals. Crit Rev
tissue and whole organ decellularization processes. Immunol. 2009;29:69–86.
Biomaterials. 2011;32:3233–43. 73. Brown BN, Valentin JE, Stewart-Akers AM,
58. Hodde J, Hiles M. Virus safety of a porcine-derived McCabe GP, Badylak SF. Macrophage phenotype
medical device: evaluation of a viral inactivation and remodeling outcomes in response to biologic
method. Biotechnol Bioeng. 2002;79:211–6. scaffolds with and without a cellular component.
59. Gorschewsky O, Klakow A, Riechert K, Pitzl M, Biomaterials. 2009;30:1482–91.
Becker R. Clinical comparison of the Tutoplast 74. Grauss RW, Hazekamp MG, Oppenhuizen F, et al.
allograft and autologous patellar tendon (bone-­ Histological evaluation of decellularised porcine
patellar tendon-bone) for the reconstruction of the aortic valves: matrix changes due to different decel-
anterior cruciate ligament: 2- and 6-year results. Am lularisation methods. Eur J Cardio-Thoracic Surg.
J Sports Med. 2005;33:1202–9. 2005;27:566–71.
60. Gorschewsky O, Puetz A, Riechert K, Klakow A, 75. Schenke-Layland K, Vasilevski O, Opitz F, et al.
Becker R. Quantitative analysis of biochemical char- Impact of decellularization of xenogeneic tissue on
acteristics of bone-patellar tendon-bone allografts. extracellular matrix integrity for tissue engineering
Biomed Mater Eng. 2005;15:403–11. of heart valves. J Struct Biol. 2003;143:201–8.
61. Keane TJ, Swinehart IT, Badylak SF. Methods of 76. Lovekamp JJ, Simionescu DT, Mercuri JJ, et al.
tissue decellularization used for preparation of Stability and function of glycosaminoglycans in
biologic scaffolds and in vivo relevance. Methods. porcine bioprosthetic heart valves. Biomaterials.
2015;84:25–34. 2006;27:1507–18.
62. Freytes DO, Stoner RM, Badylak SF. Uniaxial and 77. Conconi MT, De Coppi P, Di Liddo R, et al. Tracheal
biaxial properties of terminally sterilized porcine matrices, obtained by a detergent-enzymatic
urinary bladder matrix scaffolds. J Biomed Mater method, support in vitro the adhesion of chondro-
Res B Appl Biomater. 2008;84:408–14. cytes and tracheal epithelial cells. Transplant Int.
63. Rosario DJ, Reilly GC, Ali Salah E, et al. 2005;18:727–34.
Decellularization and sterilization of porcine urinary 78. Jackson DW, Simon TM. Donor cell survival and
bladder matrix for tissue engineering in the lower repopulation after intraarticular transplantation of
urinary tract. Regen Med. 2008;3:145–56. tendon and ligament allografts. Microsc Res Tech.
64. Jackson DW, Windler GE, Simon TM. Intraarticular 2002;58:25–33.
reaction associated with the use of freeze-dried, ethyl- 79. Gilbert TW, Sellaro TL, Badylak
ene oxide-sterilized bone-patella tendon-bone allografts SF. Decellularization of tissues and organs.
in the reconstruction of the anterior cruciate ligament. Biomaterials. 2006;27:3675–83.
Am J Sports Med. 1990;18:1–10. discussion-1 80. Schmitt T, Fox PM, Woon CY, et al. Human
65. Sun WQ, Leung P. Calorimetric study of extracel- flexor tendon tissue engineering: in vivo effects of
lular tissue matrix degradation and instability after stem cell reseeding. Plastic Reconstructive Surg.
gamma irradiation. Acta Biomater. 2008;4:817–26. 2013;132:567e–76e.
66. Bailey AJ, Tromans WJ. Effects of ionizing radia- 81. Lin CH, Yang JR, Chiang NJ, Ma H, Tsay
tion on the ultrastructure of collagen fibrils. Radiat RY. Evaluation of decellularized extracellular matrix
Res. 1964;23:145–55. of skeletal muscle for tissue engineering. Int J Artif
67. Gouk SS, Lim TM, Teoh SH, Sun WQ. Alterations Organs. 2014;37:546–55.
of human acellular tissue matrix by gamma irradia- 82. Wang Q, Zhang C, Zhang L, et al. The prepara-
tion: histology, biomechanical property, stability, tion and comparison of decellularized nerve scaf-
in vitro cell repopulation, and remodeling. J Biomed fold of tissue engineering. J Biomed Mater Res A.
Mater Res B Appl Biomater. 2008;84:205–17. 2014;102:4301–8.
68. Moreau MF, Gallois Y, Basle MF, Chappard 83. Uygun BE, Soto-Gutierrez A, Yagi H, et al. Organ
D. Gamma irradiation of human bone allografts alters reengineering through development of a transplant-
medullary lipids and releases toxic compounds for able recellularized liver graft using decellularized
osteoblast-like cells. Biomaterials. 2000;21:369–76. liver matrix. Nat Med. 2010;16:814–20.
69. Sikin AM, Rizvi SS. Recent patents on the steriliza- 84. Shupe T, Williams M, Brown A, Willenberg B,
tion of food and biomaterials by supercritical fluids. Petersen BE. Method for the decellularization of
Recent Pat Food Nutr Agric. 2011;3:212–25. intact rat liver. Organogenesis. 2010;6:134–6.
140 T. Kwon and K.H. Moon

85. Barakat O, Abbasi S, Rodriguez G, et al. Use of 102. Mirmalek-Sani SH, Orlando G, McQuilling JP,
decellularized porcine liver for engineering human- et al. Porcine pancreas extracellular matrix as a
ized liver organ. J Surg Res. 2012;173:e11–25. platform for endocrine pancreas bioengineering.
86. Soto-Gutierrez A, Zhang L, Medberry C, et al. A Biomaterials. 2013;34:5488–95.
whole-organ regenerative medicine approach for 103. Goh SK, Bertera S, Olsen P, et al. Perfusion-­
liver replacement. Tissue Eng Part C Methods. decellularized pancreas as a natural 3D scaffold
2011;17:677–86. for pancreatic tissue and whole organ engineering.
87. Zhang H, Zhang Y, Ma F, Bie P, Bai L. Orthotopic Biomaterials. 2013;34:6760–72.
transplantation of decellularized liver scaffold in 104. Yuan X, Huang Y, Guo Y, et al. Controlling the blood
mice. Int J Clin Exp Med. 2015;8:598–606. glucose of type 1 diabetes mice by co-culturing
88. Zhou P, Lessa N, Estrada DC, et al. Decellularized MIN-6 beta cells on 3D scaffold. Pediatr Transplant.
liver matrix as a carrier for the transplantation of 2015;19:371–9.
human fetal and primary hepatocytes in mice. Liver 105. Guo SZ, Ren XJ, Wu B, Jiang T. Preparation of the
Transplant. 2011;17:418–27. acellular scaffold of the spinal cord and the study of
89. Bao J, Shi Y, Sun H, et al. Construction of a portal biocompatibility. Spinal Cord. 2010;48:576–81.
implantable functional tissue-engineered liver using 106. Liu J, Chen J, Liu B, et al. Acellular spinal cord scaf-
perfusion-decellularized matrix and hepatocytes in fold seeded with mesenchymal stem cells promotes
rats. Cell Transplant. 2011;20:753–66. long-distance axon regeneration and functional
90. Ott HC, Matthiesen TS, Goh SK, et al. Perfusion-­ recovery in spinal cord injured rats. J Neurol Sci.
decellularized matrix: using nature's platform to engi- 2013;325:127–36.
neer a bioartificial heart. Nat Med. 2008;14:213–21. 107. DeQuach JA, Yuan SH, Goldstein LS, Christman
91. Lu TY, Lin B, Kim J, et al. Repopulation of decellu- KL. Decellularized porcine brain matrix for cell cul-
larized mouse heart with human induced pluripotent ture and tissue engineering scaffolds. Tissue Eng A.
stem cell-derived cardiovascular progenitor cells. 2011;17:2583–92.
Nat Commun. 2013;4:2307. 108. Crapo PM, Medberry CJ, Reing JE, et al. Biologic
92. Venugopal JR, Prabhakaran MP, Mukherjee S, scaffolds composed of central nervous system extra-
et al. Biomaterial strategies for alleviation of myo- cellular matrix. Biomaterials. 2012;33:3539–47.
cardial infarction. J Roy Soc Interface Roy Soc. 109. Sutherland RS, Baskin LS, Hayward SW, Cunha
2012;9:1–19. GR. Regeneration of bladder urothelium, smooth
93. Wu YF, Zhang J, Gu YQ, et al. Reendothelialization muscle, blood vessels and nerves into an acellular
of tubular scaffolds by sedimentary and rota- tissue matrix. J Urol. 1996;156:571–7.
tive forces: a first step toward tissue-engineered 110. Parekh A, Mantle B, Banks J, et al. Repair of the
venous graft. Cardiovasc Revascularization Med. tympanic membrane with urinary bladder matrix.
2008;9:238–47. Laryngoscope. 2009;119:1206–13.
94. Cegielski M, Calkosinski I, Dziegiel P, Zabel M. The 111. Nieponice A, McGrath K, Qureshi I, et al. An extra-
search for stem cells of the epithelium in pulmonary cellular matrix scaffold for esophageal stricture
alveoli. Folia Morphol (Warsz). 2004;63:221–3. prevention after circumferential EMR. Gastrointest
95. Kim J, Yaszemski MJ, Lu L. Three-dimensional Endosc. 2009;69:289–96.
porous biodegradable polymeric scaffolds fabricated 112. Gilbert TW, Gilbert S, Madden M, Reynolds SD,
with biodegradable hydrogel porogens. Tissue Eng Badylak SF. Morphologic assessment of extracel-
Part C Methods. 2009;15:583–94. lular matrix scaffolds for patch tracheoplasty in a
96. White ES. Lung extracellular matrix and fibroblast func- canine model. Ann Thoracic Surg. 2008;86:967–74.
tion. Ann Am Thorac Soc. 2015;12(Suppl 1)):S30–3. discussion 74
97. Guan Y, Liu S, Liu Y, et al. Porcine kidneys as a 113. Huber JE, Spievack A, Simmons-Byrd A, Ringel
source of ECM scaffold for kidney regeneration. RL, Badylak S. Extracellular matrix as a scaffold for
Mater Sci Eng C Mater Biol Appl. 2015;56:451–6. laryngeal reconstruction. Ann Otol Rhinol Laryngol.
98. Orlando G, Farney AC, Iskandar SS, et al. 2003;112:428–33.
Production and implantation of renal extracellular 114. Kitamura M, Hirano S, Kanemaru SI, et al. Glottic
matrix scaffolds from porcine kidneys as a platform regeneration with a tissue-engineering technique,
for renal bioengineering investigations. Ann Surg. using acellular extracellular matrix scaffold in a canine
2012;256:363–70. model. J Tissue Eng Regen Med. 2014;10:825–32.
99. Song JH, Humes HD. The bioartificial kidney in the 115. Gilbert TW, Nieponice A, Spievack AR, et al. Repair
treatment of acute kidney injury. Curr Drug Targets. of the thoracic wall with an extracellular matrix scaf-
2009;10:1227–34. fold in a canine model. J Surg Res. 2008;147:61–7.

100. Coronel MM, Stabler CL. Engineering a local 116. Robinson KA, Li J, Mathison M, et al. Extracellular
microenvironment for pancreatic islet replacement. matrix scaffold for cardiac repair. Circulation.
Curr Opin Biotechnol. 2013;24:900–8. 2005;112:I135–43.
101. Hosseini-Tabatabaei A, Jalili RB, Hartwell R, et al. 117. Badylak S, Meurling S, Chen M, Spievack A, Simmons-
Embedding islet in a liquid scaffold increases islet via- Byrd A. Resorbable bioscaffold for esophageal repair
bility and function. Can J Diabetes. 2013;37:27–35. in a dog model. J Pediatr Surg. 2000;35:1097–103.
6 Decellularization 141

118. Kajitani M, Wadia Y, Hinds MT, et al. Successful 122. Zhou HY, Zhang J, Yan RL, et al. Improving the
repair of esophageal injury using an elastin based antibacterial property of porcine small intesti-
biomaterial patch. ASAIO J. 2001;47:342–5. nal submucosa by nano-silver supplementation:
119. Totonelli G, Maghsoudlou P, Fishman JM, et al. a promising biological material to address the
Esophageal tissue engineering: a new approach for need for contaminated defect repair. Ann Surg.
esophageal replacement. World J Gastroenterol. 2011;253:1033–41.
2012;18:6900–7. 123. Wainwright D, Madden M, Luterman A, et al.
120. Pokrywczynska M, Jundzill A, Bodnar M, et al. Clinical evaluation of an acellular allograft dermal
Do mesenchymal stem cells modulate the milieu matrix in full-thickness burns. J Burn Care Rehabil.
of reconstructed bladder wall? Arch Immunol Ther 1996;17:124–36.
Exp. 2013;61:483–93. 124. Burke JF, Yannas IV, Quinby WC Jr, Bondoc CC,
121. Hori Y, Nakamura T, Kimura D, et al. Functional Jung WK. Successful use of a physiologically
analysis of the tissue-engineered stomach wall. Artif acceptable artificial skin in the treatment of exten-
Organs. 2002;26:868–72. sive burn injury. Ann Surg. 1981;194:413–28.
Part III
Cell and Tissue Applications
Kidney
7
Bum Soo Kim and Hyun Tae Kim

7.1 Introduction which has demonstrated therapeutic effects on


improved renal functions and increased survival.
The kidney is a vital organ which plays various However, dialysis compromises patients’ quality
important roles in humans’ body, including of life and cannot substitute other renal functions,
excretion of waste products, production of eryth- such as erythropoietin production and activation
ropoietin, and maintenance of systemic blood of vitamin D, while it replaces function of filtra-
pressure through regulation of fluid volume and tion by removing toxic substances from the blood
electrolytes. The nephrons, which are the func- [4]. Kidney transplantation, which remains the
tional units of the kidney, maintain these renal only potential curative treatment, is another
functions. Specific conditions, such as hyperten- option, but critical shortage of organs is a limita-
sion, diabetes, glomerulonephritis, and nephro- tion. In addition, despite advances in renal trans-
toxic drugs, can cause the damage of nephrons, plant immunology, approximately 20% of
and if damages persist to progress, renal function recipients experience an episode of acute rejec-
gradually deteriorates, and the kidney finally tion within 5 years of kidney transplantation, and
enters the state of renal failure. about 40% of recipients lose graft function or die
Renal failure is a worldwide health issue, con- within 10 years [5, 6].
stituting about 8–16% of the adult population These limitations of current therapies for
suffering from chronic kidney disease, which is renal failure have encouraged researchers to
defined as a reduced glomerular filtration rate explore a better treatment for end-stage renal
and increased urinary albumin excretion [1]. disease that could improve, restore, or replace
Another type of renal failure, acute kidney injury, either partial or total renal function. Tissue
which is defined as a sudden increase in serum engineering and regenerative medicine repre-
creatinine concentration and decreased urine out- sent two of the promising and innovative alter-
put can often become permanent and progress to natives. The basis of tissue engineering is that
chronic kidney disease [2, 3]. There are two main cells can be expanded and differentiated in vitro,
therapeutic options for the current treatment of placed on a scaffold made of feasible biomate-
end-stage renal disease. One is lifelong dialysis, rial, and finally implanted into the host.
Moreover, xenoembryo transplantation
approach and blastocyst complementation
methods were suggested and showed promising
B.S. Kim (*) • H.T. Kim
results for potential kidney regeneration [7].
Department of Urology, School of Medicine,
Kyungpook National University, Daegu, South Korea In 1999, Chan et al. reported the first attempt
e-mail: urokbs@knu.ac.kr; ht-kim1212@hanmail.net to develop a whole functional renal unit using

© Springer Nature Singapore Pte Ltd. 2018 145


B.W. Kim (ed.), Clinical Regenerative Medicine in Urology, DOI 10.1007/978-981-10-2723-9_7
146 B.S. Kim and H.T. Kim

pronephros from animal caps in Xenopus [8]. amniotic fluid stem cells; and (4) reconstruction
Transplantation of this pronephros improved the of artificial kidney or renal components using
edema in bilaterally nephrectomized tadpoles, stem cells and biomaterial scaffold [11].
and they could survive for up to 1 month. Since emerging technologies in the field of
However, the pronephros, which was used in this kidney regeneration still have limitations, various
study, was too primitive for clinical application in approaches were attempted, and strategies can be
humans. Since this trial, many investigations broadly classified into several categories. Current
have been performed to regenerate whole kid- major strategies for kidney regeneration are as
neys de novo. Advances in stem cell research follows: (1) stem/progenitor cell-based therapy,
over the past decades accelerated significant (2) developmental biology, (3) bioartificial kid-
progress in the field of kidney regeneration. ney, and (4) whole-organ reconstruction
During the last decade, studies have shown that it (Fig.  7.1). In this chapter, all of the contents
is possible to seed cultured cells into a three-­ above related to kidney regeneration will be dis-
dimensional scaffold system in vitro and implant cussed in detail.
the cell-seeded scaffold in vivo to restore renal
functions, and other studies also have shown the
possibility of growing new kidneys in living hosts 7.2  echanisms of Kidney
M
or integrated new nephrons into immature kid- Regeneration
neys [4, 9].
However, kidney regeneration is more com- 7.2.1 C
 ells Involved in Kidney
plex than regeneration of other solid organs. Repair
Despite recent advancement of tissue engineer-
ing and stem cell therapy, we could not achieve Various types of cells are considered to be able
the appropriate functional kidney regeneration to either contribute directly to renal repair after
due to the complicated architecture and func- damage or substantially improve renal injury
tion of kidneys as well as the lack of precise without direct contributing to the renal epithe-
understanding of molecular mechanisms for lium. The proper regulation of these cells and
cell differentiation and development of kid- successful delivery are important for the devel-
neys. Although the regeneration of an entire opment of kidney regeneration. There are four
kidney may be difficult, functional recovery of possible main origins for cells contributing to
as little as 10% of renal filtration function can kidney repair: (1) interstitial cell transdifferenti-
allow for the withdrawal of dialysis in patients ation to epithelium [12], (2) recruitment of stem
with end-stage renal disease, thus improving cells from the bone marrow [13–17], (3) tubular
patients’ quality of life [10]. Therefore, research cell dedifferentiation and proliferation in
in the field of regeneration of the kidney is con- response to injury [12, 18, 19], and (4) repopula-
tinuing to make progress, with the aim of not tion of the renal tubules by a resident kidney
only making a whole functional kidney but also stem/progenitor cells [20–22]. The first two
improving partial renal function for renal options involve nonepithelial cell transdifferen-
failure. tiation into renal epithelium, whereas the other
There are four major steps for the research of two options involve epithelial cells within the
kidney regeneration: (1) identification of reno- renal epithelium itself. The theory that renal
tropic factors and signaling pathway, which repair involves cells within the renal epithelium
involve cell differentiation and growth; (2) iden- of the nephron was supported by lineage studies
tification of renal stem/progenitor cells in the showing no evidence of dilution of the cap mes-
adult or embryonic kidney; (3) cell therapies enchyme-derived (Six2+) tubular epithelium
using mesenchymal stem cells, such as bone with a nonepithelial cell source [23]. However,
marrow or adipose tissue-derived cells, and this study did not answer whether any cell within
7 Kidney 147

Scaffold

Renal stem/ Exogenous


progenitor cell stem cell

Kidney
Regeneration

Blastocyst Embryonic
complementation metanephros

Bioartificial
kidney

Fig. 7.1  Schematic representation of the current major strategies for kidney regeneration

the epithelium can contribute to repair or whether the proximal tubule [28], tubular epithelial cells
repair relied on a resident stem cell population [30], Bowman’s capsule [24], glomeruli [31],
within the tubules. renal papilla [32], and cortical stroma. These
renal progenitor cells possess high cloning effi-
7.2.1.1 Renal Progenitor Cells ciency, self-renewal potential, and differentiate
Although many studies have investigated the into specific renal cell lineages, suggesting a role
existence, location, and contribution of renal pro- in renal regeneration [24]. Several transcriptional
genitor cells to epithelial repair [21, 22, 24, 25], regulators, such as Six2 and Foxd+1, are essen-
existence and identification of renal stem or pro- tial for the persistence of these progenitor cell
genitor cells in the adult kidney tissues have been populations beyond development, while Pax2 is
somehow controversial [3]. Grobstein first dem- required for later differentiation [33, 34].
onstrated the presence of renal progenitor cells in Renal progenitor cells could be harvested and
the metanephric blastema [26, 27]. Discrete pop- isolated from adult human kidneys as well as
ulations of these renal progenitors have been human urine, expanded in vitro, maintain their
shown to persist into adulthood, and numerous renal phenotypes through several passages, and
studies have identified renal progenitor cells in reimplanted [20, 35, 36]. Bussolati et al. showed
148 B.S. Kim and H.T. Kim

that intravenously injected renal progenitor cells of inducing extrarenal stem cells to differentiate
migrated to the site of tubular injury and contrib- into renal progenitor cells has not been appropri-
uted to renal repair [20]. These results may sup- ately established [40, 41].
port that differentiated renal epithelial cells
isolated from kidney tissue have proliferation 7.2.1.2 Proliferation of Mature Renal
capacity through dedifferentiation of the surviv- Epithelial Cells
ing epithelia during cell culture. Lineage tracing studies in mice suggested that
In contrast to the stem cells, renal progenitor the proliferating populations after kidney injury
cells can only differentiate to specific cellular lin- were mature renal epithelial cells rather than a
eage and display limited self-renewal capacity specific progenitor cell population [42], whereas
[37]. Renal progenitor cells are identified by cell analysis of cell division using unbiased DNA
marker CD133 and CD24. CD133 is a marker of labeling also suggested randomness in the cells
several types of adult tissue stem cells, while reentering cell division within the tubule [43].
CD24 is a surface molecule which is expressed in Anatomical reanalysis of human tubular epithe-
human metanephric mesenchyme [38, 39]. Renal lium has showed a distinct subpopulation of cells
progenitor cells can differentiate into glomerular within the tubules referred to as scattered tubular
as well as tubular epithelial cells. Tubular pro- cells [44]. The first study of scattered tubular
genitor cells represent about 2–6% of all tubular cells performed by Berger et al provided the evi-
epithelial cells in healthy adult kidneys and dence that this cell population is not a defined
express CD133, CD24, vimentin, cytokeratins 7 intratubular progenitors of fixed morphology but
and 19, Pax2, and nestin that are not expressed by may arise from any tubular cell as a result of
differentiated tubular epithelial cells [20]. spontaneously dedifferentiation triggered by
Glomerular progenitor cells localize within the local injury [45]. Indeed, it has been known that
Bowman’s capsule and can differentiate toward cells within the proximal tubule can express mes-
podocyte phenotype. These glomerular progeni- enchymal cell markers (vimentin) after kidney
tor cells can express CD106, whereas tubular injury, suggesting a dedifferentiation process can
progenitor cells cannot express this surface occur in response to injury [19].
marker.
Renal progenitor cells have higher resistance 7.2.1.3 Mesenchymal Stem Cells
to injury compared to other differentiated cells of For decades, many studies demonstrated that
the kidney. When injected in severe combined cells from the bone marrow could migrate into
immunodeficient mice affected by the sites of injury around the body, including the
rhabdomyolysis-­ induced acute kidney injury, kidney [17]. Although bone marrow-derived cells
these cell populations displayed the capacity to were initially considered to have a capacity to
integrate into the tubules, generate new tubular generate new tissue even in remote injured sites,
epithelial cells, and improve renal function [22]. several studies have showed it may not occur in
It can be an attractive strategy if renal progenitor some cases [46, 47]. Mesenchymal stem cells, a
cells can be induced via lineage-instructive repro- population of cells within the bone marrow, are
gramming or differentiated from extrarenal stem clogenic, able to undergo proliferation, and to be
cells, such as mesenchymal, embryonic, and differentiated into various cell types, including
induced pluripotent stem cells. These regenerated bone, cartilage, and fat [48]. Although they were
exogenous renal progenitor cells may enhance originally investigated as a support population
renal repair combined with a small population of within the bone marrow, they have also been
endogenous renal progenitor cells. In comparison investigated for the therapeutic potential with
to stem cells, renal progenitor cells have several respect to mesenchymal tissue replacement.
advantages, such as no need of intermediate cell Recent several studies suggested that bone
culture conditions and direct transit from one marrow-­derived mesenchymal stem cells are able
phenotype to another. However, a reliable method to improve deteriorated renal function in response
7 Kidney 149

to kidney injury and to reduce allograft rejection regenerating tubular cells after ischemic renal
[49–52]. However, the mechanisms of how these injury [71–73].
mesenchymal stem cells can improve renal func- Recently, the role of these renotropic factors
tion were not well established. This is not in kidney injury has been demonstrated. Chen
regarded as a result of integration into the dam- et al. showed the importance of EGF receptor
aged epithelium or transdifferentiation into an activation in the recovery phase after acute kid-
epithelial cell type. These beneficial effects are ney injury through mice with a specific EGF
rather considered to be induced by paracrine receptor deletion in renal proximal tubules [74].
effects and immunomodulatory capacity of mes- Deletion of the BMP receptor activin-like kinase
enchymal stem cells [48, 53–59]. 3 in the tubular epithelium enhances epithelial
Although mesenchymal stem cells were origi- damage and fibrosis [75]. Conditional knockout
nally identified in the bone marrow, mesenchy- mice lacking the HGF receptor, c-met, specifi-
mal stem cell-like cells have been also identified cally in renal tubules demonstrated the antiapop-
to reside in many adult tissues, including the kid- totic of anti-inflammatory effect of c-met
ney. It may represent a perivascular cell popula- signaling in renal protection after acute kidney
tion involved in normal tissue repair [53, 54, 57]. injury [76].
Recently, a population of endogenous kidney A negative regulatory factor of kidney repair
mesenchymal stem cells was characterized from has also been identified. Several studies using
the adult murine kidney [60]. It showed the char- transgenic mice expressing truncated activin type
acteristic gene and protein expression profile, II receptor, an in vitro tubulogenesis model, the
clonogenicity, and potential to differentiate to Wolffian duct culture, and isolated rat embryonic
other cell types, such as bone, cartilage, and fat. kidney culture showed that activin A is an endog-
This cell population showed enrichment for enous inhibitor of renal organogenesis [77–83].
kidney-­specific genes, compared to bone marrow-­ Activin A is also a potent inhibitor of renal regen-
derived mesenchymal stem cells. eration after kidney injury [84].
Most of these renotropic factors can induce
tubular cell proliferation after kidney injury when
7.2.2 Renotropic Factors exogenously administered. However, it is still not
well known whether these factors are involved in
Soluble factors involved in kidney development cell maturation, modulation of renal blood flow,
have been identified by gene targeting tech- neutrophil infiltration, and restoration of polarity.
niques, in vitro tubulogenesis models, and organ It will be interesting to examine if these renotro-
culture systems. The regeneration process after pic factors promote renal regeneration via the
renal injury resembles the kidney organogenesis. activation of intrinsic renal stem cells.
Most of these factors have been proved to regu-
late renal recovery as potential renotropic factors
by using animal kidney injury models. These 7.2.3 Macrophage
renotropic factors are as follows: hepatocyte
growth factor (HGF) [61], epidermal growth fac- Generally, macrophages have a function to clear
tor (EGF) [62], heparin-binding EGF-like growth foreign pathogens in the body that can disrupt tis-
factor (HB-EGF) [63, 64], bone morphogenetic sue homeostasis [85] and also play an important
protein-7 (BMP-7) [65, 66], insulin-like growth role to mediate kidney repair. Although macro-
factor-I (IGF-I) [67, 68], platelet-derived growth phages are well known to promote the fibrotic
factor (PDGF) [69], and uterine sensitization-­ and inflammation reaction during renal disease,
associated gene-1 (USAG-1) [70]. In addition, they have been also demonstrated to have a pro-
several key regulatory molecules required for tective role in acute and chronic kidney diseases
renal organogenesis, such as Pax-2, leukemia [86]. Moreover, Wyburn et al. reported that mac-
inhibitory factor, and Wnt4, are reactivated in rophages can ameliorate allograft rejection [87].
150 B.S. Kim and H.T. Kim

Macrophages represent a heterogeneous pop- 7.2.4.1 PI3K/AKT/mTOR Pathway


ulation with distinct functions depending on It has been demonstrated that growth factors, such
resident tissue and phase of tissue injury or dis- as EGF, IGF-1, and HGF, can accelerate recovery
ease. In some cases, macrophages display classi- of renal function after acute kidney injury [107].
cal activation through the release of nitric oxide, These growth factors activate a lipid kinase
reactive oxygen species, and proinflammatory ­(phosphatidylinositol-3 kinase, PI3K) that phos-
cytokines, termed an M1 response or phenotype phorylates phosphatidylinositol 4,5-­bisphosphate
[88, 89]. In another stage of disease, macro- to yield phosphatidylinositol 3,4,5-trisphosphate.
phages can play antifibrotic and anti-­The phosphatidylinositol 3,4,5-trisphosphate
inflammatory roles, termed M2 phenotype [90, phosphorylates and activates Akt. After being
91]. These M2 macrophages have a reparative activated, Akt stimulates mammalian target of
function in ischemia-­ reperfusion injury and rapamycin (mTOR) by regulating the activity of
obstructive nephropathy, induced by unilateral intermediary kinases. The activation of mTOR
ureteric obstruction [92–95]. Many studies leads to phosphorylation of downstream sub-
reported that changes in the local microenviron- strates and then induces cell regeneration. It is
ment are important to trigger an M1 phenotype demonstrated that inhibition of mTOR by rapamy-
macrophages, such as macrophage colony-stim- cin delays recovery of renal function [108]. Akt
ulating factor, which can be released by tubular can activate antiapoptotic genes and might inacti-
epithelial cells to stimulate this process [96, 97]. vate proapoptotic factors, such as Bcl-2-associated
Based on these findings, macrophages were death promoter, forkhead family transcription fac-
directed toward an M2 phenotype via genetic tors, and ­procaspase-­9 [109]. Deletion of the EGF
manipulation or cytokine alteration, such as receptor in renal proximal tubular epithelial cells
overexpression of interleukin 4 (IL-4), IL-10, impairs phosphotidylinositol-3 kinase/Akt signal-
inducible nitric oxide synthase, or heme oxygen- ing and delays recovery from acute kidney
ase, resulting in improved renal function [98– injury [74].
106]. In terms of regenerative medicine,
macrophages are considered to have therapeutic 7.2.4.2 Wnt/GSK3/β-Catenin Pathway
potential and be one of the options for cellular or The Wnts are a family of secreted and glycosyl-
cytokine/growth factor-based therapies in kidney ated protein ligands. Wnt signals can inhibit
diseases. glycogen synthase kinase 3 (GSK3) by phos-
­
phorylation. When GSK3 is inhibited, β-catenin
is stabilized and translocates into the nucleus to
7.2.4 Signaling Pathways act as a transcriptional coactivator of the T-cell
in Mediating the Regenerative factor/lymphoid enhancer-binding factor family
Process or transcription factors and drive the expression
of its target genes [110]. This pathway is involved
Many cells, such as renal progenitor cells, endog- in the regulation of cell fate, cell mobility, prolif-
enous renal tubular epithelial cells, bone marrow-­ eration, survival, protein synthesis, and glycogen
derived stem cells, and macrophages, are involved metabolism [111]. Acute kidney injury induces
in kidney regeneration. However, the exact mech- activation of Wnt pathway, whereas genetic inac-
anism of cell to cell interaction and influencing tivation of Wnt signaling can deteriorate renal
factors for cell reaction involving regenerative function recovery and kidney regeneration [112].
process after kidney injury is still not exactly Among the increased Wnt ligands after kidney
known. Although several studies suggested pos- injury, macrophage-­derived Wnt7b can promote
sible pathways involved in kidney regeneration, tubular epithelial cell regeneration and kidney
further research to identify the factors in specific repair. In the downstream of Wnt signaling,
signaling pathways will be needed. GSK3 is inhibited by Wnt ligands, IGF, EGF, and
7 Kidney 151

fibroblast growth factors 16, 19, and 23 [113]. In N-terminal kinase (JNK), and p38MAPK [120,
acute kidney injury, GSK3 can promote the sys- 121]. ERK is mainly activated by mitogenic
temic inflammatory response and participate in a stimuli, such as growth factor, and ERK1/2 path-
number of apoptotic signaling pathways by pho- way has been widely studied in kidney regenera-
phorylating transcription factors that regulate tion. It was demonstrated that activation of ERK
apoptosis [114]. TDZD-8, a GSK3β inhibitor, pathway can enhance renal epithelial cell sur-
can inactivate ischemia-induced activation of vival during oxidative injury in vitro [122].
GSK3, caspase 3, and Bax, ameliorate tubular Activation of the signal transducer and activator
epithelial cell apoptosis, and protect renal func- of transcription-­3 (STAT3) during oxidative
tion [115]. Acute kidney injury can induce the stress can attenuate EGF receptor-mediated ERK
expression of β-catenin, and renal tubule-specific activation and renal tubular cell survival [123]. It
knockout of endogenous β-catenin aggravates was also reported that inhibition of ERK path-
kidney injury in mice [116]. It is also reported way reduces kidney regeneration in rats with
that inhibition of GSK3β can ameliorate nonste- myoglobinuric acute kidney injury in vivo [124].
roidal anti-­inflammatory drug-induced acute kid-
ney injury [117]. Consequently, it is considered
that activation of the Wnt/GSK3/β-catenin path- 7.3 I mproving Renal Function
way is beneficial for kidney disease and GSK3 After Kidney Injury
inhibitor can be a target of therapeutic agents in
the future. 7.3.1 Cell-Based Approach

7.2.4.3 JAK/STAT Pathway Early studies using stem cells for kidney regen-
When EGF binds to the EGF receptor, Janus-­ eration usually have shown that the predominant
activated kinase (JAK) is activated and phosphor- recovery mechanism of the acute kidney injury
ylates the intracellular domain of the receptor was mediated through the transdifferentiation of
and allows recruitment and phosphorylation of the infused mesenchymal stem cells into specific
STAT. Salahudeen et al. demonstrated that renal cell types, especially podocytes, renal tubu-
erythropoiesis-­
stimulating proteins suppress lar cells, glomerular cells, and mesangial cells
renal tubular cell apoptosis in vitro and enhance [13, 125–127]. More recent studies support the
renal recovery in the cisplatin-induced acute kid- concept that cytokines and/or growth factors
ney injury through the activation of JAK/STAT secreted from the injected mesenchymal stem
pathway [118]. In contrast, Ucero et al. reported cells stimulate endogenous cells to proliferate
that inhibition of the JAK/STAT pathway can and regenerate the infused renal tissues [128–
decrease tubular epithelial cell apoptosis and kid- 131]. Several studies suggested that the effect of
ney inflammation in murine acute kidney injury mesenchymal stem cells in acute kidney injury
model [119]. (Therefore, further researches will models is associated with paracrine mechanisms,
be needed to clarify the effect of JAK/STAT path- such as immune-related response and mitogenic,
way on renal repair.) antiapoptotic, and vasoprotective effect [132,
133]. It was evidenced by the results of several
7.2.4.4 MAPK/ERK Pathway studies that bone marrow-derived mesenchymal
Mitogen-activated protein kinases (MAPKs) are stem cell-cultured medium contains microvesi-
a family of kinases that have been commonly cles and growth factors that reduce inflammation
investigated on the kidney disease. There are and enhance renal repair through interactions
four different MAPK pathways: extracellular with renal progenitors [15, 125, 134, 135].
signal-­regulated kinase-1 and extracellular Bone marrow-derived mesenchymal stem
signal-­regulated kinase-2 (ERK1/2), extracellu- cells are multipotent, migrate across tissues, are
lar signal-regulated kinase-5 (ERK5), c-Jun easily harvested, are produced throughout life,
152 B.S. Kim and H.T. Kim

and contribute to the repair of organs [136]. The compared to amniotic fluid stem cells without
administration of bone marrow stem cells has treatment. The authors suggested that a combi-
been shown to promote neovascularization, nation of transdifferentiation of amniotic fluid
reduce inflammation, inhibit apoptosis, and stim- stem cells and paracrine signaling involved the
ulate differentiation and proliferation in multiple regenerative effect through this study. Hauser
systems [137]. Poulsom et al. first reported the et al. reported that intravenous infusion of amni-
effect of engrafted bone marrow stem cells into otic fluid stem cells induced more rapid recovery
the damaged kidney, improving repair of renal of renal function in an animal model of acute
tissues after acute kidney injury [17]. However, kidney injury, when compared to that of bone
despite these benefits of bone marrow stem cells, marrow stem cells. In this study, bone marrow
clinical application is still in question because stem cells showed higher potential for prolifera-
negative results were also reported. Bone marrow tion, whereas amniotic fluid stem cells showed
stem cells have been shown to promote intersti- better capacity for antiapoptosis and persistence
tial fibrosis in mouse models, and direct renal within the peritubular capillaries. In addition,
injection of bone marrow stem cells induced the they isolated different cytokines and growth fac-
development of angiomyeloproliferative lesions tors from bone marrow and amniotic fluid stem
of unknown neoplastic potential [138, 139]. cells, ­ suggesting different modes of action
Therefore, further safety studies are necessary between both cell sources.
before clinical applications to ensure safe and Induced pluripotent stem cells (iPSCs) have
successful outcomes for kidney regeneration gained increasing interest in the field of regenera-
using bone marrow stem cells. tive medicine, as they possess pluripotent capa-
Another promising cell source of mesenchy- bility and potentially provide an inexhaustible
mal stem cells is adipose-derived stem cells. source of patient- and tissue-specific stem cells.
Several studies reported the effectiveness of Recently, iPSCs have been successfully gener-
adipose-­derived stem cells to improve renal func- ated from human renal sources, such as kidney
tions in animal models, including mouse, rat, and mesangial cells, proximal tubular cells, podo-
pig [140–143]. Administration of adipose-­ cytes, and epithelial cells derived from urine
derived stem cells into an acute kidney injury [147–149]. IPSCs have several advantages com-
model reduced renal fibrosis at 6 weeks [144], pared with embryonic stem cell, such as absence
and intrarenal injection of adipose-derived stem of ethical issues related to cell sourcing, fewer
cells showed improved angiogenesis and pre- immune rejection, and less potential for abnor-
served the renal structure integrity, which restored mal tissue formation. Moreover, they also have
renal function at 14 days [145]. more capabilities to facilitate targeted, organ-­
Amniotic fluid-derived stem cells are easily specific differentiation due to retaining the epi-
harvested and cultured with lower risk of tumori- genetic pattern of the parent cell [150]. Recent
genicity and similar multipotency to bone mar- studies reported the improvement of renal func-
row stem cells. Rota et al. demonstrated the tions in an acute kidney injury rodent model after
therapeutic potential of human amniotic fluid administration of iPSCs [151, 152]. However,
stem cells [146]. In their study, engrafted amni- iPSCs have been shown to express abnormal
otic fluid stem cells localized primarily to the genes and induce T-cell-dependent immune reac-
peritubular region, limiting tubular damage, tion in syngeneic mice [153]. This unexpected
improving renal function, and prolonging sur- immune response is a big hurdle of iPSCs for
vival of the animals. They also reported that clinical application.
amniotic fluid stem cells that were precondi- Aforementioned renal progenitor cells are
tioned with glial cell line-derived neurotrophic also promising cell sources for the treatment of
factor were observed to promote better func- acute kidney injury. It has been demonstrated that
tional recovery by contributing to renal regener- CD24+/CD133+/CD106- renal progenitor cells
ation in acute kidney injury models when are resistant to apoptosis and promote recovery
7 Kidney 153

of tubular injury in the rhabdomyolysis-induced sought to create, implant, and maintain a renal
acute kidney injury rat model [22]. Neural cell structure that mimics the physical and physiolog-
adhesion molecule (NCAM)-positive cells that ical characteristics of the native kidney.
were identified from human kidney tissue showed The concept of early investigations in this
clonogenic and renal progenitor properties [154]. field endeavored to infuse new nephrons to devel-
After engraftment into the chick embryo, oping kidneys by implantation of embryonic
NCAM+ human nephron progenitor cells formed metanephric tissue into the renal cortex of neona-
a renal-like structure. Administration of human tal mice [155]. Metanephros is the embryological
nephron progenitor cells into the kidneys of acute precursor of the adult kidney, characterized by
kidney injury rat model induced inhibition of dis- mesenchyme and ureteric buds (Fig. 7.2) [156].
ease progression and improvement of renal func- Transplantation of primitive metanephric tissue
tion. Taken together, these results support the over fully developed kidney may have some
concept that renal progenitor cells have the advantages, such as reduced immunogenicity due
capacity to migrate and proliferate to improve to the absence of native vasculature and antigen-­
structural and functional damage after acute kid- presenting cells [156, 157]. Thus, metanephros
ney injury. can be maintained in vivo without host immuno-
suppression. Although transplanted metanephric
tissue was successfully differentiated and devel-
7.3.2 Developmental Biology oped into functional nephrons in the host kidney
of neonate, it was not initially reproduced in host
Although advancement of stem cell technology kidneys of adult due to lack of differentiation and
has showed promising results in the field of kid- acute graft rejection [158]. However, another
ney regeneration, stem cells themselves have study reported successful implantation and dif-
limitations to maintain the structure of the kid- ferentiation of metanephroi into adult hosts, with
ney. Thus, many researchers tried to develop the demonstrated glomerular filtration and plasma
cellular approach by incorporating principles of clearance after modifying the protocol [156].
developmental biology. This investigative field Encouraged by these results, further studies to

a b

Fig. 7.2 Isolated metanephros from embryo of E15 from metanephric mesenchyme under the vision of light
transgenic mouse in which a green fluorescent protein is microscope. (b) Ureter and branches of ureteric bud can
expressed in the ureteric bud under the control of the be seen with green color under the vision of
Hoxb7 promoter. (a) Only the ureter can be distinguished immunofluoro-microscope
154 B.S. Kim and H.T. Kim

investigate the possibility of xenotransplantation bioengineered scaffold was generated by Lanza


have been performed. More information related et al. in 2002 [167]. They used a nuclear trans-
to xenotransplantation will be discussed in plantation technique in which dermal fibroblasts
another section. isolated from an adult cow were transferred into
enucleated bovine oocytes and then transferred
into progestin-synchronized recipients.
7.4  e Novo Organ
D Metanephroi taken from embryos were dissoci-
Regeneration ated to single-cell suspension using collagenase,
and the cells were expanded in vitro. Then, the
The basic concept for regeneration of solid organ cells were seeded onto a specialized polymer
is to collect stem or progenitor cells from appro- tube as the artificial scaffold. The cell-seeded
priate cell sources and expand these cells in scaffold was implanted into the same cow from
culture with or without differentiation.
­ which the cells had been cloned and produced
Subsequently, cells are seeded into a scaffold and urine-like liquid. Histologic analysis showed that
placed into a bioreactor to promote cell acclima- it had well-differentiated kidney-like structure,
tion, attachment, and maturation. Then, finally including organized glomerulus-like, tubular-­
this cell-­seeded scaffold can be implanted in the like, and vascular components. This investigation
host [159, 160]. established the possibility of kidney regeneration
using bioengineered scaffolds.
Synthetic biodegradable and biocompatible
7.4.1 Artificial Scaffold polyesters, such as polyglycolic acid (PGA),
polylactic acid-polyglycolic acid (PLGA), and
Extracellular matrix is the naturally occurring polycaprolactone (PCL), also can be used as a
scaffold material secreted and manufactured by scaffold for kidney regeneration. Although these
the resident cells of each organ, and it plays a synthetic polymers are a US Food and Drug
critical role in development and repair of organ Administration (FDA)-approved biocompatible
[161, 162]. The extracellular matrix is a complex material, they have critical drawbacks, such as
three-dimensional framework of structural and producing acidic products during degradation
functional proteins in a state of dynamic equilib- and inducing an inflammatory response in the
rium within the surrounding tissues and provides host, which restricts the clinical applications
the means by which cells communicate with each [168–172]. To overcome these adverse effects of
other and the external environment [163]. The synthetic biomaterial, several investigations are
extracellular matrix contains growth factors and in progress.
bioinductive cytokines, which facilitate the
reparative process, cell attachment, and tissue 7.4.1.2 Decellularized Cadaveric
integration [164, 165]. Consequently, the roles of Scaffold
extracellular matrix for organogenesis and regen- Artificial scaffold also can be generated from
eration can be categorized by providing a three-­ organs of animal or human’s cadaver through
dimensional scaffold for the spatial organization decellularization [173–176]. The decellulariza-
of cells, secreting and storing growth factors and tion process can remove DNA, cellular material,
cytokines, and regulating signal transduction and cell surface antigens from the extracellular
[166]. matrix scaffold while preserving the structural
and functional characteristics of vascular chan-
7.4.1.1 Bioengineered Scaffold nels. The process of decellularization includes
Development of biomaterial engineering has pro- the repeated irrigation of cadaveric organs with
duced bioengineered scaffolds that facilitate acids or detergents through the innate vascula-
improved differentiation of transplanted cells. ture, although organs with higher fat component,
The first histocompatible functional kidney using such as pancreas, often require the addition of
7 Kidney 155

lipid solvents [177]. To generate cadaveric scaf- cadherin, and pan-­ cytokeratin. Through this
fold, complete decellularization must be achieved study, the authors suggested several points to
because residual cellular material may contain advance this research field that pretreatment of
antigenic epitopes that trigger inflammatory reac- the embryonic stem cell with prodifferentiation
tions and compromise subsequent recellulariza- agents, such as retinoic acid, activin-A, and
tion [178, 179]. Ethylene oxide or paracetic acid BMP-7, may promote implantation and prolifer-
can effectively sterilized the extracellular matrix ation by providing a more kidney-specific lin-
without denaturing the extracellular matrix pro- eage. Moreover, while injected cells through
teins or growth factors after decellularization innate vascular channels can be evenly distrib-
[180, 181]. uted in the renal cortex, the cells may not local-
Using this technique, a functional artificial rat ize in the collecting system. Thus, retrograde
heart using a cadaveric heart as a scaffold was seeding via ureter was attempted to resolve this
successfully generated [182]. For this, a whole-­ problem, but this method caused uneven cell dis-
heart scaffold with intact three-dimensional persion. Furthermore, murine scaffolds are not
geometry and vasculature was created via coro- feasible for adult human kidney regeneration in
nary perfusion with detergents into the cadaveric terms of size and structure. Therefore, organs of
heart, followed by repopulation with neonatal larger animals, such as pig, or primates had to be
cardiac cells or rat aortic endothelial cells and considered to be i­nvestigated using this tech-
cultured under physiological conditions to stimu- nique to meet human renal requirements.
late organ maturation. In this study, the injected However, application of this decellularization
neonatal cardiac cells formed a contractile myo- and recellularization technique to larger and
cardium, which performed the stroke function. complex organs is a great challenge, because
Decellularized cadaveric scaffolds have also been larger organs with greater parenchymal mass
used to generate other organs, such as the liver, require stronger detergents, higher perfusion
respiratory tract, and urinary bladder [183–185]. pressures, and prolonged detergent exposure time
Recent advancement of decellularization-­ that may damage the native vasculature and
recellularization technique enabled to generate extracellular matrix proteins [187]. The first trial
more complex organs, such as the kidney. Ross to decellularize primate kidney performed by
et al. first reported successful regeneration of an Nakayama et al. confirmed effective decellular-
entire kidney using the decellularized rat kidney ization with preservation of native extracellular
as a scaffold [186]. After fabricating intact scaf- matrix architecture and protein complement
fold, murine embryonic stem cells were injected [188]. After recellularization, immunohisto-
through the innate vasculature. In this study, chemical analysis showed effective local migra-
embryonic stem cells were used for seeding pop- tion and attachment of renal cells to the border of
ulation because of their high doubling capacity, scaffold, demonstrating the feasibility of primate
pluripotency, and potential to differentiate and kidneys for decellularization and recellulariza-
integrate into primordial kidney cultures. The tion technique.
kidney scaffold successfully supported the More recently, intact, whole-organ extracellu-
growth and migration of the seeded embryonic lar matrix scaffolds were generated from porcine
stem cells within glomerular, vascular, and tubu- kidneys [187, 189]. These studies reported suc-
lar structures while inducing differentiation cessful decellularization using detergent perfu-
down renal cell lines. Immunohistochemical sion through the innate vasculature and confirmed
analysis indicated that the seeded embryonic total cell clearance with preserving the architec-
stem cells had lost their embryonic appearance ture of scaffold, including glomeruli, tubules, and
and had shown gross morphological changes vasculatures. Despite these advances of generat-
consistent with mature kidney cells, as well as ing decellularized scaffold technique, regenera-
expression of immunohistochemical markers of tion of a whole functional kidney using this
renal differentiation, such as Pax-2, Ksp- method is still not achieved. Although several
156 B.S. Kim and H.T. Kim

attempts have been performed, effective seeding administered BrdU to rat and mouse neonates
population of cells was not established. Moreover, and found BrdU-retaining cells localized in the
it is unclear whether larger kidney scaffold can papilla, after chasing them for more than 2
support the retrograde ureteric perfusion to recel- months [32]. After the induction of ischemia,
lularize the collecting system. It is also not certain LRCs disappeared from papilla, whereas most of
whether extracellular matrix of the decellularized them expressed Ki67, which is a proliferation
kidney scaffold can support the proliferation and marker. Through this study, the authors demon-
differentiation of stem cells and whether recellu- strated that the renal papilla is a niche for adult
larization by perfusion through the innate vascu- kidney stem cells and that LRCs proliferate and
lature does not cause thrombogenic complications migrate following ischemic kidney injury. They
after reimplantation. Furthermore, even after reli- also showed similar results in another study using
able protocols of recellularization are established, H2B-GFP transgenic mice [190]. H2B-GFP is
various functions of regenerated kidney, such as more stable and easily detected than BrdU, and it
homeostasis, resorption, endocrine, metabolism, can label the cells in a cell cycle regardless of the
and immunomodulation, should be confirmed. stage, whereas BrdU can only label the cells in
the S phase [191]. In this study, H2B-GFP-­
positive cells proliferated and migrated to renal
7.4.2 Isolation of Cells tubules after kidney injury.
Maeshima et al. identified LRCs using BrdU
The kidney has approximately 20 different spe- predominantly in proximal tubules of the adult
cific cell types. For efficient kidney regeneration, rat kidney [30]. After ischemic injury, most of
all cells must be characterized for repopulation. the LRCs were found in tubules and were posi-
Although many attempts have been performed to tive for PCNA, indicating that LRCs proliferate
establish a reliable cell source and optimal cell after injury. The LRCs were successfully iso-
growth conditions to provide adequate enrich- lated and cultured, and they initially expressed
ment for achieving stable renal cell expansion vimentin, a mesenchymal stem cell marker, and
systems, it is still challenging. It is essential to eventually became positive for E-cadherin, an
isolate feasible stem or progenitor cells for cell epithelial cell marker, after multiple cell divi-
expansion and differentiation in vitro, and renal sions. In another study, they demonstrated that
stem/progenitor cells are one of the good candi- LRCs became positive for proximal tubule and
dates. There are five different methods which ureteric bud markers when transplanted into the
have been used to isolate renal stem/progenitor metanephric kidney of embryonic rat, indicating
cells from the adult kidney. that LRCs were integrated into epithelial compo-
nents of the nephrons in the process of kidney
7.4.2.1 Label-Retaining Cells development [192].
One of the characteristics that distinguish stem Although labeling with BrdU is an effective
cells from differentiated cells is their infrequent technique to detect stem cells, considerable
cell division. To conserve the proliferation capac- problems were also suggested. The BrdU label
ity for a lifetime and to prevent genetic injuries may be released from dying cells and taken up
during mitosis, the cycle of stem cells is very by adjacent dividing cells [193]. Moreover, the
slow. Renal stem/progenitor cells were isolated possibility that BrdU-retaining cells are not
using this property. When these cells are labeled stem/progenitor cells but fully differentiated
with markers such as 5-bromo-2-deoxyuridine cells was proposed [194]. Vogetseder et al.
(BrdU) and histone 2B-GFP (H2B-GFP), they reported that LRCs expressed differentiated
can retain the label for a long time. Then, these tubule markers, such as Na-K-ATPase, NaPi-IIa,
slow-cycling label-retaining cells (LRCs) can be PMP70, and megalin. Similarly, H2B-GFP label-
detected following a chase period. Oliver et al. ing has also drawbacks because the labeling is
7 Kidney 157

diluted after massive cell proliferation. To obtain 7.4.2.3 Cell Surface Markers
a better understanding of LRCs, a more accurate Another approach for isolating putative stem
purification and characterization technique are cells is utilizing stem cell markers, such as
required. CD133, CD24, and stem cell antigen-1 (Sca-1).
Although CD133 is not a specific marker for
7.4.2.2 Side Population Cells kidney stem cells, it is a universal marker for
Since stem cells extrude dyes, such as Rhodamine stem cells in other tissues, such as hematopoi-
123 and Hoechst 33342 via ATP-binding cassette etic stem cells, cancer stem cells, and vascular
transporters, they are located in a unique position endothelial progenitor cells [198, 199].
in fluorescent-activated cell sorting (FACS) anal- Bussolati et al. isolated CD133+ cells from nor-
ysis and are called side population (SP) cells mal adult human kidney [20]. These cells
[195]. SP cells are characterized as Hoechst low expressed Pax-2 (embryonic kidney marker),
cells and are isolated from several tissues, includ- but not CD34, CD45 (hematopoietic lineage
ing the heart, liver, and skeletal muscle. Several marker), CD 90, or c-Kit (stem cell markers),
groups isolated and characterized SP cells from indicating that they were originated from renal
the kidney. Iwatani et al. isolated SP cells from tissues. They were capable of self-expansion
adult rat kidney, but these SP cells were not and limited self-renewal and could be induced
involved in kidney repair following gentamicin-­ to differentiate into epithelial and endothelial
induced nephropathy [196]. Hishikawa et al. iso- cells in vitro and in vivo. When CD133+ cells
lated SP cells from adult mouse kidney [197]. were injected intravenously in a glycerol-
These cells expressed Musculin/MyoR, a tran- induced acute kidney injury model of severe
scription factor found in skeletal muscle precur- combined immunodeficiency mice, they
sors. Musculin/MyoR-positive cells reside in the migrated to the injured kidney and were incor-
renal interstitium and are decreased following porated predominantly into the proximal and
cisplatin-induced acute kidney injury. Systemic distal tubules. Sagrinati et al. isolated CD24+
injection of kidney SP cells into mice improved and CD133+ cells from parietal epithelial cells
renal function in cisplatin-induced acute tubular in the adult human kidney after culturing glom-
injury model. In addition, SP cells expressed a eruli [24]. These cells also expressed the stem
high level of leukemia inhibitory factor (LIF) and cell-­specific transcription factors, such as Oct-4
renoprotective factors, such as HGF, vascular and BmI-1. These cells had a high self-renewal
endothelial growth factor (VEGF), and BMP-7 in potential, and when cultured in appropriate con-
a cisplatin-induced acute kidney injury model. ditions, these cells could be differentiated into
Challen et al. also isolated SP cells from adult tubular epithelial cells, osteocytes, adipocytes,
mouse kidney, and these cells expressed genes and neuronal cells in vitro. These cells were
involved in Notch signaling. These SP cells are incorporated into regenerating tubules, and
located predominantly in the proximal tubules renal function was improved when they were
and integrated into the mesenchyme- and ureteric administered intravenously into severe com-
bud-derived structures when injected into the bined immunodeficiency mice with glycerol-
embryonic kidney, suggesting that they have mul- induced acute kidney injury. Dekel et al. isolated
tilineage differentiation potential. Administration Sca-1-positive cells from adult mouse kidney
of these cells improved renal function in adriamy- [200]. The Sca-1+ cells were located mainly in
cin-induced kidney injury model, but these cells the papilla and could differentiate into myo-
were barely incorporated into the renal tissues genic, osteogenic, adipogenic, and neural lin-
after infusion. Taken together, SP cells are consid- eages in vitro. When injected into the renal
ered to release renoprotective factors in paracrine parenchyma of ischemia-induced acute kidney
fashion, but the exact function or composition of injury model, some of these cells were inte-
SP cells is still unclear. grated into renal tubules.
158 B.S. Kim and H.T. Kim

7.4.2.4 Cell Culture only regenerate to podocytes. Finally, cells local-


Selective culture conditions are useful to isolate ized at the vascular pole did not express progeni-
stem cells [201]. A unique cell population can be tor markers but displayed phenotypic features of
isolated during the culture of dispersed cells differentiated podocytes (CD24-, CD133-, PDX+
derived from the adult kidney. Gupta et al. iso- cells). Moreover, the in vivo properties of the
lated progenitor-like cells from adult rat kidneys cells were also evaluated by Ronconi et al [204].
by using culture conditions similar to those used Administration of CD24+, CD133+, and PDX-
for bone marrow-derived cells [202]. These cells cells, but not CD24+, CD133+, PDX+ or CD24-,
were characterized by long-term self-renewal CD133-, and PDX+ cells, into mice with
without senescence and the expression of vimen- adriamycin-­induced kidney injury reduced pro-
tin, CD90, Pax-2, and Oct-4 without any markers teinuria and improved glomerular damage, sug-
of major histocompatibility complex (MHC) gesting that CD24+, CD133+, and PDX- cells are
class or other differentiated cell markers. These potential therapeutic targets for glomerular disor-
cells can also be induced to multiple lineages ders characterized by podocyte injury. Appel
in vitro, including hepatocytes, neurons, and et al. reported that the CD133+ parietal epithelial
endothelial cells. The cells could differentiate cells have the capacity to proliferate, migrate
into renal tubular cells when injected under the along the glomerular tuft, and differentiate to
capsule of the kidney or intra-arterially in unin- mature podocytes after podocyte injuries by
jured kidney or ischemia-reperfusion injury of using immunostaining, label retention, and trans-
the kidney. It is proposed that these progenitor genic mice approaches [205]. The cells at the
cells isolated by using selective culture condition base of the vascular pole adjacent to the podo-
participate in the regenerative response of the cytes were stained with both the parietal e­ pithelial
kidney to acute injury. However, possibility of cell marker (claudin-1) and podocyte-specific
contamination of differentiated kidney cells and markers (nestin, dipeptidyl peptidase IV, amino-
blood cells is a limitation of this method. peptidase A). The idea that parietal epithelial
cells migrate to become podocytes was also sup-
7.4.2.5 Parietal Epithelial Cells ported by BrdU staining of rat parietal epithelial
Mature podocytes are highly differentiated non- cells and genetic tagging of parietal epithelial
dividing cells, and it is not clear whether damage cells utilizing parietal epithelial cell-specific pro-
of podocyte in adulthood can be repaired. moter (hPODXL1). The researchers demon-
Glomerular epithelial stem cells may reside in strated that the cells at the urinary pole of the
the kidney and may be capable of regenerating Bowman’s capsule migrate down to the proximal
podocytes, and it was suggested that glomerular tubules and are involved in the turnover of tubu-
epithelial stem cells exist in the parietal epithelial lar epithelium. Finally, they concluded that the
cells [203]. It was reported that some of the pari- cells localized at the border of different compart-
etal epithelial cells localized in the Bowman’s ments have features of progenitor cells.
capsule of adult human kidney have the charac-
teristics of stem cells [24]. In this study, parietal
epithelial cells were divided into three subpopu- 7.4.3 P
 otential Cell Sources
lations according to the locations and characters. for Renal Regeneration
First, cells localized at the urinary pole express-
ing CD24 and CD133, but not podocalyxin Recent advances in stem cell biology and cell
(PDX), a differentiated podocyte marker (CD24+, culture techniques have facilitated the develop-
CD133+, PDX- cells), could regenerate both ment of cell therapy for clinical translation [206,
tubular cells and podocytes. Second, cells local- 207]. Compared with other approaches for kid-
ized between the urinary pole and the vascular ney regeneration, this method using cellular
pole expressing both progenitor and podocytes approach can be more practically applied to kid-
markers (CD24+, CD133+, PDX+ cells) could ney tissue regeneration due to the relatively
7 Kidney 159

simple cell manipulation process, easy access to Primary renal cells can be harvested from both
the target site in a less invasive manner, and effec- normal and diseased kidney tissues and expanded
tive integration of administered cells with the in vitro while maintaining the phenotype and
host tissues. Consequently, many studies have function which they are derived. Among these
been performed using this cellular approach to primary renal cells, proximal tubular cells play
treat renal failure [3, 41, 206]. In this section, cel- important roles in kidney functions [210, 211].
lular approach for kidney tissue regeneration, The main roles of proximal tubular cells are reab-
including various cell sources used as well recent sorption of proteins and electrolytes, production
advances made in preclinical and clinical studies, of erythropoietin, and hydrolase activity.
will be discussed. Proximal tubular cells occupy the highest per-
centage of renal cell population in normal kid-
7.4.3.1 Kidney Tissue-Derived Cells ney; thus, isolation and expansion of functional
Although multipotent stem cells have been primary proximal tubular cells from renal tissues
known to play important roles for various tissues can be considered as an attractive renal cell
and organs of humans, no definite evidence to source for cell-based kidney regeneration [212].
date establishes the existence of a pluripotent, Primary proximal tubular cell cultures have sev-
self-renewing cell population in the adult kidney. eral advantages and are more representative of
During kidney development, condensed mesen- normal physiology of proximal tubular cells than
chyme around the tips of the ureteric bud pos- immortalized cell lines, but primary renal cells,
sesses self-renewing cells capable of generating including proximal tubular cells, can lose expres-
all other elements of the nephrons, interstitium, sion of specific genes during in vitro culture and
and vasculature via a mesenchyme-epithelial are limited to only two to five passages [213].
transition [208]. The cells of condensed mesen- The optimal combination of high purity, well dif-
chyme are regarded as the renal stem cell popula- ferentiation, and consistent proliferation is
tion. These stem cells of condensed mesenchyme observed at passage 2–3 [214].
cease asymmetric division and self-renewal and Culture method of primary kidney cells from
then exhibit spontaneous commitment to human kidney tissues for cell expansion has been
mesenchyme-­ epithelial transition. However, established [35]. In histological analyses, the
these cells can be exhausted before the perinatal majority of the cultured cells retained a proximal
stage [209]. This phenomenon indicates that tubular phenotype, while distal tubular cells and
complete regeneration involving a complete podocytes were present in a lower percentage of
replacement of the lost nephron does not occur in the entire cell population. In addition, when the
the mammalian kidney. Nevertheless, researchers expanded cells were cultured under a three-­
have discovered the stem cell-like pluripotent dimensional environment, tubule-like structures
cells in adult kidney, which are called renal pro- with functional properties were formed from the
genitor cells, and there are two major theories to cultured cells. These results indicated that the
explain the origin of proliferating tubular epithe- established cell harvesting and culture method
lia after acute kidney injury. The first one is that may potentially be developed as an effective cell-­
stem or progenitor cells in renal tubule can gener- based approach for kidney regeneration.
ate new epithelia, and the other one is that fully Several protocols for other kidney cell types,
differentiated epithelia may dedifferentiate, reen- except proximal tubular cells, also have been
ter cell cycle, and generate new epithelial cells established. Presnell et al. established a culture
through self-duplication. method for primary cell cultures isolated from all
major compartments of the kidney, especially the
Primary Kidney Cells tubular cell-enriched subpopulation and the
Kidney tissue consists of more than 20 specialized erythropoietin-producing subpopulation that
cell types that are structurally organized into func- were reproducibly developed from both normal
tionally and anatomically distinct compartments. and diseased kidneys [215]. Recent advances of
160 B.S. Kim and H.T. Kim

immunomagnetic cell isolation technique enabled cells in the correct niche. Renal stem/progenitor
to isolate additional subtypes of kidney-derived cells derived from the S3 segment of adult rat
epithelial cells. Renal epithelial cells were suc- kidney expressed stem cell markers, such as Sca-­
cessfully separated from the ascending limb and 1, c-kit, Musashi-1, and nestin, as well as renal
the distal tubule of the human kidney using lineage markers, such as WT-1 and PAX-2.
glycoprotein-­coated magnetic beads [216]. This Although it was assumed that these cells were
result demonstrated that ascending limb of similar to metanephric mesenchymal cells, based
Henle’s loop and distal tubule can be a promising on marker protein expression, the cluster could
cell source for cell-based kidney regeneration differentiate into collecting duct-like cells or
and establishment of in vitro cell culture system mesangial-like cells, which are not considered to
for various cell types of the kidney can be be derived from metanephric mesenchyme.
realized. Resident mesenchymal stem cells have been
described in many organs as a multipotent popu-
Renal Stem/Progenitor Cells lation adjacent to endothelial cells in the micro-
Stem/progenitor cells isolated from many adult vasculature and characterized by expression of
organs demonstrate clonogenic, self-renewing PDGF, neuron glial antigen-2, CD146, and coex-
ability and can give rise to terminally differenti- pression of other mesenchymal markers [53].
ated cells of the original tissue. Renal stem/pro- Isolation of resident mesenchymal stem cells
genitor population disappears in the adult kidney, from the murine kidney was also reported [54].
possibly due to the loss of its niche [209, 217]. Subsequently, mesenchymal stem cells were iso-
However, renal stem/progenitor cells still exist in lated from human glomeruli and showed self-­
the adult kidney and are located in specific loca- renewal capability, clonogenicity, and
tions, such as renal papilla, tubular epithelial multipotency [219]. Furthermore, Wang et al.
cells, Bowman’s capsule, and the S3 segment of reported that renal mesenchymal stem cells iso-
the proximal tubules [24, 28–30, 32]. lated from adult murine kidney could differenti-
A recent study showed that renal stem/pro- ate into renin-producing cells [220]. The
genitor cells derived from the S3 segment of comparative study of gene expression in mesen-
adult rat kidney nephrons were able to reconsti- chymal stem cells between kidney and bone mar-
tute a three-dimensional kidney-like structure row origin identified selected patterns of genes in
in vitro [218]. In this study, kidney-like structures renal mesenchymal stem cells possibly related to
were formed when a cluster of renal stem/pro- a memory of tissue origin [60]. This result sug-
genitor cells was suspended in an extracellular gests that renal mesenchymal stem cells might
matrix gel and cultured in the presence of several display organ-specific regenerative capacities
growth factors, such as glial cell-derived neuro- that could overcome those of mesenchymal stem
trophic factor (GDNF), basic fibroblast growth cells from other organs, such as the bone marrow
factor (bFGF), HGF, EGF, and BMP-7. The clus- or adipose tissue, and that could be exploited for
ters from dissociated S3 segment-derived cells therapeutic applications.
were induced by the hanging drop method in Although adult renal stem/progenitor cells
three-dimensional culture, while two-­dimensional still remain poorly understood, they have been
culture conditions were not able to reconstruct reported to be nontumorigenic when injected into
kidney-like structures. The reconstructed kidney-­ a mouse model, whereas pluripotent stem cells,
like structures included most of renal substruc- such as embryonic stem cells or iPSCs, have a
tures, such as glomeruli, proximal and distal risk of potential tumorigenicity as a clinical
tubules, loop of Henle, and collecting ducts, but source [221, 222]. In this regard, the results of
not the vasculature. This study suggested that a aforementioned studies support the possibility
cluster of tissue stem/progenitor cells may have that adult renal stem/progenitor cells may repre-
the ability to reconstitute the minimum unit of its sent a safer clinical source than pluripotent stem
original organ by differentiating into specialized cells. Therefore, renal stem/progenitor cells may
7 Kidney 161

be a promising cell source for kidney regenera- or prevention of inflammatory cell influx. During
tion, if it is possible to establish renal stem/pro- the repair phase, bone marrow-derived stem cells
genitor cells with multipotency for kidney secrete several factors that promote tubular epi-
reconstruction. thelial cell dedifferentiation and proliferation
[231]. The transdifferentiation of bone marrow-­
7.4.3.2 Mesenchymal Stem Cells derived stem cells observed in early studies may
Mesenchymal stem cells are stromal cells that reflect cell fusion. If bone marrow-derived stem
can be found and isolated from various tissues. cells fuse with resident cells in the kidney, they
They also have the capacity to differentiate into a acquire a phenotype of resident cells. Therefore,
mesenchymal tissue types, including bone, carti- it could appear as if bone marrow-derived stem
lage, muscle, tendon, fat, and other connective cells were differentiated into resident cells.
tissues. Unlike pluripotent stem cells, such as Indeed, it was reported that bone marrow-derived
embryonic stem cells and iPSCs, mesenchymal stem cells could fuse with other cell types [232,
stem cells can be practically applied for cell-­ 233].
based therapies with fewer safety concerns and Togel et al. performed the first phase I clinical
ethical issues. These mesenchymal stem cells study to evaluate the efficacy and safety of allo-
have been studied in recent decades as a thera- geneic bone marrow-derived stem cell adminis-
peutic agent for various tissues and organs due to tration for the prevention of acute kidney injury
its relatively easy approach to harvest, multilin- after open heart surgery [234]. Sixteen patients
eage differentiation potential, ability to migrate who underwent cardiac surgery and who were at
to the site of injury, and no ethical concerns. a high risk of postoperative acute renal failure
due to underlying chronic kidney disease,
Bone Marrow-Derived Stem Cells advanced age, congestive heart failure, and dia-
Since 2000, bone marrow-derived stem cells betes mellitus were enrolled in this study.
have been used in experimental kidney disease Allogeneic bone marrow-derived stem cells were
models. Several studies have shown that treat- injected into the suprarenal aorta after surgery.
ment with bone marrow-derived stem cells can Preliminary data showed that renal function was
ameliorate some of injured renal tissues, such as well preserved after surgery for up to 16 months
tubular epithelial cells, podocytes, mesangial and none of the patients who received stem cell
cells, and endothelial cells [15–17, 125, 127, injection required hemodialysis, while 20% of
223–226]. Treatment using bone marrow-derived case controls developed acute renal failure. The
stem cells can contribute to the attenuation of length of hospital stay and readmission rates of
renal fibrosis during progression of chronic kid- stem cell group reduced by 40% compared to
ney disease [138]. Although early studies sug- control group, and no adverse events related to
gested that bone marrow-derived stem cells could this therapy occurred.
be differentiated into renal epithelial cells and Recently, two subsequent clinical trials were
mesangial cells [227–229], recent researches performed to evaluate the effect of bone marrow-­
have supported that migration of bone marrow-­ derived stem cells in patients who underwent kid-
derived stem cells into the kidney is very rare and ney transplantation [235, 236]. These clinical
their ability to direct differentiation is limited trials have shown that bone marrow-derived stem
[47]. Thus, the beneficial effects of bone marrow-­ cells were safe and offered an early recovery of
derived stem cells are considered to be mainly graft function. However, early posttransplant
mediated by paracrine action of injected cells stem cell infusion was found to be possibly asso-
that have antiapoptotic, immunomodulatory, and ciated with acute graft dysfunction due to engraft-
proangiogenic effects [50, 51, 230]. Taking isch- ment syndrome [235]. This adverse effect, which
emic renal injury, for example, bone marrow-­ was accompanied by neutrophil recruitment and
derived stem cells can initially ameliorate the complement C3 deposition, was possibly attrib-
injury either by direct inhibition of cell apoptosis utable to the inflammatory condition occurring
162 B.S. Kim and H.T. Kim

after kidney transplantation, leading to altered Adipose Tissue-Derived Stem Cells


functions of mesenchymal stem cells [237]. Adipose tissue-derived stem cells are another
More recently, a phase I clinical study using type of mesenchymal stem cells residing in sub-
autologous bone marrow-derived mesenchymal cutaneous adipose tissues. They are an attractive
stem cells for the treatment of allograft rejection source of cells with regenerative properties that
after kidney transplantation from living donors are similar to those of bone marrow-derived stem
was performed [238]. In this trial, bone marrow-­ cells. Moreover, adipose tissue-derived stem cells
derived stem cells were administered intrave- are promising stem cells for clinical use, because
nously at intervals of 7 days when a protocol of the subcutaneous adipose tissues are abundant in
renal biopsy at 1 or 6 months showed signs of humans and can be easily harvested using lipo-
rejection and/or an increase in interstitial fibrosis suction. In addition, these cells have no ethical
or tubular atrophy. Although only six patients issues regarding the cell source and less concern
were enrolled in this study, two renal biopsies about safety of allo- and xenografting.
after treatment with bone marrow-derived stem Furthermore, adipose tissue-derived stem cells
cells showed resolution of cell infiltrates, sug- have more potential of anti-inflammatory and
gesting a regenerative and anti-inflammatory immunomodulatory functions than bone marrow-­
effect of the cell therapy on the renal tissue. The derived stem cells. Adipose tissue-derived stem
engraftment syndrome, one of the negative cells have the capacity to differentiate into other
effects of mesenchymal stem cell administration, cell types, such as adipocytes, osteoblasts, neu-
was not observed in this study possibly due to a rons, and myocytes [145]. Several studies have
late timing of stem cell administration, beyond 6 demonstrated that cell therapy using adipose
months, after transplantation. tissue-­derived stem cells minimized kidney dam-
Despite these encouraging progresses of these age or improved renal dysfunction after renal
clinical trials, questions related to the establish- damages, such as ischemic injury, progressive
ment of the best way to apply mesenchymal stem renal fibrosis in a mouse model, cisplatin- or folic
cells to kidney disease still remain unsolved. In acid-induced acute kidney injury, atherosclerotic
addition, therapeutic mechanism is also still renal artery stenosis in swine models, and anti-
poorly understood. Mesenchymal stem cells do glomerular basement membrane disease in a rat
not engraft in the kidney when injected intra-­ model [140–143, 145, 240–242]. Adipose tissue-­
arterially or intravenously; rather, they can form derived stem cells also seem to improve renal
emboli in the lung [239]. Thus, these studies are function mainly via paracrine effects, such as
not cell transplantation but cell infusion that suppressing oxidative stress and inflammatory
exerts effects through paracrine mechanisms. response.
This implies that if we can identify the bioactive
substances secreted by bone marrow-derived Umbilical Cord Blood-Derived Stem Cells
stem cells, infusion of these substances without The umbilical cord blood has been known to con-
cells would be enough. Indeed, conditioned tain mesenchymal stem cells, and several studies
media from mesenchymal stem cells protected evaluated the efficacy of administration of umbil-
kidneys from acute injury as well as injection of ical cord blood-derived stem cells in the restora-
the cells did in preclinical study [49]. Incomplete tion of renal function in animal acute kidney
understanding of the mechanism of mesenchy- injury models [243, 244]. Injection of human
mal stem cells for cell therapy is not a reason not umbilical cord blood-derived mesenchymal stem
to pursue these clinical trials, but more basic cells to immunodeficient mice with cisplatin-­
research to investigate the biology of mesenchy- induced acute kidney injury was performed
mal stem cells for kidney protection and renal [243]. This study showed that umbilical cord
regeneration as well as further clinical trials blood-derived stem cells ameliorated tubular
devoted to testing efficacy of this therapy is injury, resulting in the recovery of renal function.
required. Coculturing of umbilical cord blood-derived
7 Kidney 163

stem cells with cisplatin-treated proximal tubular animal models of kidney disease delayed pro-
cells showed that the expression of HGF was par- gression of fibrosis [250]. More recent studies
ticularly induced and that of interleukin 1-β and have investigated whether amniotic fluid-derived
tumor necrosis factor-α was significantly stem cells can generate renal cell types in vitro,
decreased in the coculture system. These results with evidence for a capacity to differentiate into
indicated that the process of renal function podocyte-like cells [252]. In addition, it was pro-
improvement by umbilical cord blood-derived posed that these cells could be propagated in a
stem cells was also correlated to the modulation nephron progenitor state for many passages in
of paracrine factors in the kidney. Both umbilical order to provide an expandable source of cells to
cord blood- and bone marrow-derived stem cells give rise to this specific renal cell type.
expressed similar set of genes in the comparative Comparison of the characteristics between amni-
study of gene expression profile between these otic fluid- and bone marrow-derived stem cells
two kinds of stem cells, but bone marrow-derived was performed [248]. In this study, amniotic
stem cells predominantly expressed a set of genes fluid-derived stem cells had a more potent anti-
related to antimicrobial activity and osteogenesis, apoptotic activity against renal tubular cells but
whereas umbilical cord blood-derived stem cells lesser stimulatory activity for the proliferation of
predominantly expressed genes related to matrix renal tubular cells compared with bone marrow-­
remodeling and angiogenesis [245]. This result derived stem cells. It was also demonstrated that
suggested that umbilical cord blood- and bone amniotic fluid- and bone marrow-derived stem
marrow-derived stem cells may have distinct cells expressed different sets of paracrine factors,
activities in vivo. indicating that both cell populations may have
distinct activities in vivo. In another study,
Amniotic Fluid Stem Cells ­mammalian target of rapamycin was reported as
As multipotent stem cells at the fetal stage, amni- an important factor in renal differentiation of
otic fluid-derived stem cells have been consid- amniotic fluid-derived stem cells [253].
ered an attractive cell source for regenerative
medicine. Amniotic fluid-derived stem cells Endothelial Progenitor Cells
exhibit characteristics of both embryonic and Endothelial progenitor cells participate in the
adult stem cells, are easily harvested, and retain repair of tissues, including the kidney, under
high self-renewal potential and multiple differen- specific physiological and pathological condi-
tiation capacity without development of teratoma tions. Renal ischemia mobilized endothelial
formation compared to embryonic stem cells or progenitor cells, and transplantation of endothe-
iPSCs [246]. Perin et al. isolated amniotic fluid-­ lial progenitor cell-enriched cells from the
derived stem cells from human amniotic fluid medullopapillary parenchyma provided partial
obtained at 12–18 weeks gestation and showed renoprotection after ischemic injury of the kid-
that these cells expanded in vitro could survive, ney [254]. Acute elevation of uric acid acts as an
proliferated, and integrated into the embryonic endogenous mediator of mobilization and reno-
kidney and underwent organ development, dem- protection of endothelial progenitor cells [255].
onstrating a potential cell source for kidney Endothelial progenitor cells can be isolated
regeneration [247]. Therapeutic effects of human from human peripheral blood using CD34 as a
amniotic fluid-derived stem cells have been dem- marker for positive selection [256]. The CD34+
onstrated in kidney injury animal models, such as mononuclear blood cells obtained the character-
glycerol- or cisplatin-induced acute kidney istics of vascular endothelial cells when cul-
injury, a mouse model of unilateral ureteral tured on fibronectin-coated dishes. Endothelial
obstruction, and Alport syndrome [146, 248– progenitor cells were incorporated in ischemic
251]. Initial studies into the properties of amni- tissues in vivo and expressed vascular endothe-
otic fluid-derived stem cells in kidney injury lial cell markers, such as CD31, when intro-
demonstrated that the injection of such cells into duced into the circulation using a hindlimb
164 B.S. Kim and H.T. Kim

ischemia model. The efficacy of administration Recently, an efficient method has been devel-
of endothelial progenitor cells for the improve- oped to induce the differentiation of intermediate
ment of renal function was also reported in ani- mesodermal cells from pluripotent stem cells
mal models of acute kidney injury and renal using a combination of activin A and CHIR99021,
artery stenosis [254, 257, 258]. In a chronic followed by combined treatment with BMP-7
renal artery stenosis model, a single intrarenal and CHIR99021 [272, 274]. Odd-skipped related
injection of autologous endothelial progenitor 1 (OSR1)+ intermediate mesodermal cells were
cells preserved microvascular architecture and differentiated from human iPSCs using this pro-
decreased microvascular remodeling by pre- tocol with an efficiency greater than 90% [274].
serving hemodynamics [257]. However, it was Several studies also have endeavored to generate
reported that the function of endothelial progen- metanephric nephron progenitor cells by direct
itor cells was deteriorated in chronic kidney dis- differentiation from pluripotent stem cells.
ease patients, suggesting that the transplantation Takasato et al. induced a primitive streak from
of autologous endothelial progenitor cells may human embryonic stem cells using activin A (or
not be feasible for the treatment of chronic kid- CHIR99021) and BMP-4 [275]. The differenti-
ney disease [259]. ated human embryonic stem cells formed renal
vesicles combined with dissociated embryonic
7.4.3.3 Pluripotent Stem Cell mouse kidney cells, involving the integration of
Pluripotent stem cells have the potential to dif- human cells into mouse renal structures. In con-
ferentiate into any cell type in the body and self-­ trast, Taguchi et al. demonstrated more reason-
assemble into heterogeneous tissues or organs, able protocols to induce renal structures such as
and they have indeed been shown to generate nephrons and proximal tubules [277]. They pro-
mature cells in vitro [260–262]. Pluripotent posed the concept of “posteriorization” to induce
stem cells include both embryonic stem cells, nephron progenitor cells from pluripotent stem
which are derived from embryos and grown in cells in a mouse and human model, indicating
primary culture, and iPSCs, which are produced that nephron progenitor cells of the metanephric
from terminally differentiated cells using trans- mesenchyme could be derived from posteriorly
fection factors, such as c-myc, Oct4, Klf4, and located intermediate mesoderm.
Sox-2. Pluripotent stem cells have been success- The ureteric bud lineage cells were also
fully differentiated into various types of cells attempted to be differentiated from pluripotent
and tissues, including hematologic, neural, car- stem cells by stepwise treatment of iPSCs or
diac, hepatic, pancreatic, and intestinal lineages human embryonic stem cells with a combination
[263–271]. Recent progress toward generating of fibroblast growth factor 2 and BMP-4 for 2
renal cell types from embryonic stem cells or days, followed by combined treatment with
iPSCs has generated human nephron progenitor activin A and BMP-2 for another 2 days [276].
cells, including intermediate mesoderm and After the 4 days of treatment, intermediate
metanephric mesenchyme cells [272–277]. mesoderm-­ like cells expressing PAX2, OSR1,
However, the progress of kidney regeneration WT1, and LHX1 were produced, and the ureteric
using pluripotent stem cells is notably slow bud markers, such as HOXB7, RET, and GFRA1,
compared to other fields, and there have been were upregulated in these cells after additional 2
few efficient protocols in the kidney, whereas days of differentiation, indicating that ureteric
full protocols for the derivation of neurons, bud progenitor-like cells could be differentiated
hepatocytes, and cardiac myocytes were already from pluripotent cells.
established [278–280]. Although different stud-
ies have used different growth factor protocols Embryonic Stem Cell
in human pluripotent stem cells, the Wnt agonist Embryonic stem cells are pluripotent stem cells
CHIR99021 was commonly used to promote and were initially derived from the inner cell
mesoderm differentiation [273]. mass of the blastocyst of mouse embryos [281].
7 Kidney 165

These cells have the ability to differentiate into risk of uncontrolled growth as well as legal and
various cell types of the mesodermal, endoder- ethical problems associated with the use of
mal, and ectodermal lineages; thus, they have embryonic tissue is limitation of the embryonic
been considered to be used as an effective tool for stem cells. Although embryonic stem cells are a
kidney regeneration. Although research on valuable cell source for investigating the mecha-
human embryonic stem cells is limited due to nism of kidney regeneration, there are still sev-
ethical concerns, extensive research was con- eral hurdles for clinical applications of these
ducted using murine embryonic stem cells to cells.
establish the methods to efficiently induce differ-
entiation into renal cells [282]. Thomson et al. Induced Pluripotent Stem Cell
first established human embryonic stem cell line Another cell source that possesses pluripotency
in 1998 [283], and subsequently human embry- is the iPSC, which was first generated by
onic stem cell lines have been found to be capa- Takahashi et al. through reprogramming human
ble of differentiating in vitro into extraembryonic fibroblasts in 2006 [261]. These novel discover-
and somatic cell lineages [284]. When human ies have completely changed view of the devel-
embryonic stem cells were cultured with a mix- opment and cellular specialization. After then,
ture of growth factors, including bFGF, trans- iPSCs have been successfully established from
forming growth factor (TGF) β1, activin-A, several mammalian species, including rat, rabbit,
BMP-4, HGF, EGF, β-nerve growth factor, and pig, monkey, and humans [262, 291–296].
retinoic acid, they differentiated into cells Nevertheless, not all adult cells can be similarly
expressing WT-1 and renin [285]. Moreover, it reprogrammed, indicating that critical factors for
was shown that mouse embryonic stem cells sta- reprogramming are cell dependent. For example,
bly transfected with Wnt4 differentiated into reprogramming of mature B cells from adult
tubular-like structures that express aquaporin-2 spleen to iPSCs required an additional factor, C/
when cultured with combination of HGF and EBPα, as well as Oct4, Sox2, Klf4, and c-Myc
activin-A [286]. The mixture of LY294002, [20]. Recently, generation of iPSCs has been
CCG1423, and Janus-associated tyrosine kinase reported from human mesangial cells, tubular
inhibitor 1 was also shown to enhance the differ- cells, and epithelial cells derived from urine [147,
entiation of mouse embryonic stem cells into 149, 297]. The iPSCs derived from renal tissues
intermediate mesoderm and renal progenitor may have advantages for renal cell therapy or
cells [287]. In another study investigating an kidney regeneration, because a persistent
ex vivo culture system, embryonic stem cells genome-wide epigenetic memory of the somatic
were microinjected into the developing meta- cell of origin can be retained in iPSCs [298].
nephros, and this was cultured to analyze the dif- Indeed, when iPSCs derived from human mesan-
ferentiation capacity of embryonic stem cells into gial cells that were differentiated in podocyte
renal cells [288]. Tubule-like renal epithelial were engrafted in a metanephric kidney, they
structures were identified with an efficiency of were integrated in the developing glomeruli [147,
about 50%, and individual embryonic stem cells 148]. In addition, there are several advantages of
were occasionally observed in structures resem- iPSCs for kidney regeneration, such as no ethical
bling glomerular tufts. Furthermore, when issues and no immune rejection especially com-
embryonic stem cells treated with retinoic acid, pared to embryonic stem cells. However,
activin A, and BMP-7 were injected into a devel- increased risk of uncontrolled growth is one of
oping metanephros, they contributed to the tubu- the great concerns, because these cells are repro-
lar epithelia with almost 100% efficiency [289]. grammed using Klf4 and c-Myc, which are onco-
Despite these promising results, there is a con- genic factors [261, 299]. Hopefully, iPSCs can be
cern about this technique that embryonic stem established from human renal proximal tubular
cells can develop teratoma at 2 to 4 weeks after cells without transfection of Klf4 and c-Myc,
transplantation into mouse [290]. In addition, the suggesting that the oncogenic risk associated
166 B.S. Kim and H.T. Kim

with iPSC generation can be decreased by possibility of organogenesis for therapeutic


expressing only Oct4 and Sox2 [299]. Therefore, regeneration using this approach [301–304].
it is possible to prepare patient-specific pluripo- It was demonstrated that a single multipotent
tent cells without manipulating germ cells progenitor cell from embryonic mouse kidney
because iPSCs are pluripotent and can be gener- can express Sall-1 and differentiate into several
ated from adult somatic cells. types of renal cells, such as podocytes and tubu-
Recent study reported the therapeutic effect lar epithelial cells, and eventually reconstruct a
of iPSCs on renal ischemia [151]. In this study, three-dimensional kidney structure [305].
iPSCs were generated without transfection of Another study also reported that single-cell sus-
c-Myc and administered via intrarenal arterial pensions from embryonic kidney reaggregated
route into kidneys of ischemia/reperfusion-­ and formed organotypic renal structures [306].
induced acute kidney injury rat models. The Because intermediate mesoderm cells differenti-
administration of iPSCs reduced the expression ated into renal progenitor cells and then into
of oxidative substances, proinflammatory cyto- maturing renal cells, if pluripotent stem cells can
kines, and apoptotic factors and eventually be differentiated into intermediate mesoderm, all
improved survivals of rats. kinds of renal cells may be generated from
Nevertheless, there are still several hurdles embryonic stem cells or iPSCs. The protocols to
of iPSCs-based therapy, such as no established differentiate human iPSCs into intermediate
differentiation protocols for moving from plu- mesoderm cells were established using bacterial
ripotent state to functional renal cells, unde- artificial chromosome-based vectors and single-­
fined optimal final culture conditions for target nucleotide polymorphism array-based detection
cell, and multiple steps each requiring different [274]. These cells expressed intermediated meso-
factors to induce a stepwise differentiation derm marker genes and could mature into various
[41]. In addition, it was reported that some types of cells including those found in organs,
cells differentiated from iPSCs expressed such as the kidney, adrenal gland, and gonad.
abnormal gene and induced T-cell-dependent These results suggested that three-dimensional
immune response even in syngeneic recipients kidney structure could be constructed if pluripo-
[300]. Therefore, it is necessary to evaluate the tent stem cells can be made to generate renal pro-
immunogenicity of specific cells derived from genitor cells.
iPSCs before they can be used in a clinical
application.
7.4.5 Blastocyst Complementation

7.4.4 I n Vitro Kidney Regeneration Blastocyst is an early developmental stage of the


Without Scaffolds embryo that occurs 5 days after fertilization.
Injection of pluripotent stem cells into blasto-
There have been many efforts to generate mature cysts synchronizes the development of two line
cells using pluripotent stem cells in vitro. Several cells, and the combined blastocyst can generate a
studies reported successful generation of mature chimeric body. The chimera can also be produced
cells, which have characteristics of liver, pan- by injecting pluripotent stem cells into xenoblas-
creas, and intestine, from embryonic stem cells tocyst, which lacks potential to form any particu-
or iPSCs using stepwise protocols mimicking the lar cell lineage. The chimera is then transplanted
mechanism of embryonic development [263, into a foster uterus, and the deficient cells are
265, 267, 270, 271]. Furthermore, autonomous exclusively derived from injected pluripotent
formation of three-dimensional adenohypophy- stem cells. It was first reported that injection of
sis, polarized cortical tissues, optic cap, and ret- normal embryonic stem cells into the blastocysts
ina structures using three-dimensional culture of recombination-activating gene 2-deficient
systems of embryonic stem cells suggested the mice, which have no mature B or T lymphocytes,
7 Kidney 167

generated somatic chimeras with embryonic stem arcuate, and interlobular arterioles, was a chime-
cell-derived mature B and T cells [307]. Recent ric structure originating from both donor iPSCs
several studies demonstrated that several types of and host cells. Whether urine was produced by
tissues and organs, such as the thymic epithe- this regenerated kidney was not certain due to the
lium, yolk sac, germ cells, hepatocytes, heart, lack of precise urine analysis. In addition, gener-
pancreas, and kidney, could be reconstructed ation of rat kidneys in mice failed after injection
using this blastocyst complementation system of rat iPSCs into kidney-deficient mouse blasto-
[308–315]. Kobayashi et al. successfully gener- cysts. This result indicates that the key molecules
ated a functional pancreas using this approach in mice involved in interactions between the
[310]. In this study, iPSCs of rat were injected metanephric mesenchyme and ureteric bud do
into Pdx-/- (pancreatogenesis-disabled) blasto- not cross-react with those in rats. Therefore, the
cysts of mouse that produced newborn rat/mouse generation of xenogeneic organs using interspe-
chimeras with a pancreas which originated cific blastocysts requires a host animal strain
almost entirely from the injected iPSCs of rat. lacking all renal lineages.
The mouse and rat iPSC-derived pancreas pro- Although this strategy using blastocyst comple-
duced insulin, and the transplantation of this mentation has showed promising results for kidney
iPSC-derived pancreas islets improved hypergly- regeneration, there are several limitations for clini-
cemia in a diabetic rodent model [217]. This cal applications. It is difficult to generate interspe-
study suggested that iPSC-derived cellular prog- cific chimeras in livestock. In addition, tissue
eny could aggregate and differentiate into the rejection also should be overcome. Moreover, ethi-
deficient organ in a vacant developmental niche. cal issue is one of the most serious problems. There
Moreover, these results also demonstrated that has been always a concern about the possibility of
interspecific blastocyst complementation could generating interspecific ­chimeras containing brain
be used to generate organs derived from donor derived from injected pluripotent stem cells.
pluripotent stem cells in vivo using a xenogeneic
environment. This study group generated a Pdx-
/- pig and succeeded in generating bigger pan- 7.4.6 Embryonic Organ
creas using this technique, suggesting that Transplantation
human-scale organs could theoretically be gener-
ated [311]. Likewise, chimeric mice carrying a The embryonic metanephros (Fig. 7.3), which
liver derived from injected iPSCs of mouse were constitutes the primordial mammalian kidney, has
also generated from blastocysts with fumarylace- been used for the investigation of kidney regenera-
toacetate hydrolase deficiency [308]. Hepatocytes tion and showed promising results as a potential
derived from the injected iPSCs also had a prolif- source for the generation of a functional whole
erative characteristic of normal hepatocytes. kidney [156, 316–324]. If a metanephros is trans-
Kidney reconstruction using this blastocyst planted into the renal cortex of a host mouse, it can
complementation system has been attempted. continue to grow [155]. The transplanted meta-
Usui et al. injected normal mouse iPSCs into nephros contains vascularized glomeruli and
blastocysts from kidney-deficient mice lacking mature proximal tubules endowed with a capacity
the SAL-like 1 protein, which is essential for kid- for glomerular filtration. Collecting duct-like
ney development; eventually, the neonatal mice structures also appeared and extended from the
had kidneys derived from injected iPSCs [315]. transplant toward the papilla of the host. Although
However, the vascular and nervous systems were there is no direct evidence that these collecting
not constructed from iPSC-originated cells; thus, duct-like structures connect with the collecting
the kidney was not completely complemented. system of the host or that the transplanted meta-
Immunohistochemical analysis of the regener- nephros functions in a manner similar to that of
ated kidney showed that the vascular system, native kidney, these results offer a rationale for the
including renal segmental, lobar, interlobar, existence of renal stem cells in the metanephros
168 B.S. Kim and H.T. Kim

a b

Fig. 7.3 Metanephroi of E13 mouse embryo. (a) of the embryo (red arrow, both metanephroi). (b) Intact
Metanephroi, which are located just above aortic bifurca- metanephros can be isolated using microforceps in the
tion, can be observed in E13 mouse embryo after remov- vision of microscopy
ing lower half of the spinal cord and dorsal muscular layer

from embryos, which can be a potential source of were transplanted into either the omentum of mice
transplantable regenerated kidney. However, there in which costimulation was blocked or area under
are still several unsolved problems related to this the kidney capsules of immunodeficient mice and
technique, including questions as to the suitability developed to generate fully functional nephrons
of the renal capsule of patients with dialysis as a [316, 320]. The average levels of creatinine and
transplant site, given the significant disruptions to urea nitrogen were higher in the cystic fluid that
this area, such as the vasculature, and the fact that arose from the transplanted tissue than in the sera
space limitations beneath the renal capsule may of the transplanted host mice. These successful
inhibit the growth of transplanted metanephros. To xenotransplantation of metanephroi between dif-
overcome these concerns, the metanephroi were ferent species was based on previous studies show-
transplanted into a host omentum in another study ing minimal immunogenicity in tissues, including
[156]. The transplanted metanephroi into the the metanephros harvested at earlier gestational
omentum were not confined by a tight organ cap- ages [325]. In cases of allotransplantation, such as
sule or disturbed by dialysis. The metanephroi rat metanephros to rat omentum, transplants
transplanted into a host omentum also could dif- assumed a kidney-like shape in situ that was
ferentiate into mature vascularized glomeruli and approximately one-third the diameter of the native
proximal tubules and produce urine. Moreover, kidney, and the transplants contained well-­
after an intact ureteroureterostomy with the ureter differentiated kidney structure in histological anal-
of the native kidney that was removed, anephric ysis. In addition, the transplantation can be
host animals started to void and showed prolonged performed without immunosuppression. However,
survival. These metanephroi developed in host in xenotransplantation, although transplanted
animals exhibit metabolic renal function and pro- metanephros grew and differentiated into renal tis-
duce erythropoietin and renin leading to detectable sue in the host omentum, immunosuppressants
levels of these proteins in serum [319, 323, 324]. were required. Without these agents, the trans-
Furthermore, metanephroi from porcine embryos plants disappeared soon after transplantation.
7 Kidney 169

Transplantation of metanephros was also ablated by apoptosis, leaving the autologous


shown to maintain blood pressure in anephric rats erythropoietin-­producing tissues. In addition, it
with induced acute hypotension and reduce vas- was also confirmed that the metanephros estab-
cular calcification in rats with chronic renal fail- lished by this method no longer needed immuno-
ure, indicating that transplanted metanephroi have suppressants while maintaining erythropoietin
multiple renal functions as well as urine produc- production [318]. These results indicate that xeno-
tion [323, 324]. Although the omentum has been metanephroi per se could acquire some renal func-
preferred because a tight renal capsule does not tions in the host omentum as well as supply a niche
confine it and the host vessels can be preferen- for host stem cells to regenerate renal tissues and
tially integrated into the transplanted metaneph- then be rebuilt to constitute only host cell compo-
ros, the low hydrostatic pressure in the omentum nents. This technique can be helpful to reduce the
may influence the acquisition of some renal func- adverse effects of long-term administration of
tion in the developed metanephros. Indeed, it was immunosuppressants and to moderate the ethical
reported that the para-aortic area is better for the issues surrounding xenotransplantation.
establishment of renin-producing tissue [319].
Recently, it was reported that xenotransplanted
metanephros could supply endogenous mesenchy- 7.4.7 R
 enal Tissue Regeneration
mal stem cells with a niche for differentiation into Using Metanephric Progenitor
erythropoietin-producing renal tissues [318]. Cells
Polymerase chain reaction (PCR) using species-
specific primers and sequence analysis showed One of the most promising methods for kidney
that xenotransplanted metanephroi expressed host engineering is the generation of fetal or mature
animal-originated erythropoietin. This result renal tissues using intrinsic proficiency of cells
implied that the erythropoietin-producing cells to organize themselves into fully self-derived
were originated from the host animals and devel- sophisticated structures [301, 303]. Several early
oped to produce erythropoietin in the transplanted studies using this strategy demonstrated that
metanephros. It was also shown that the erythro- renal epithelial cell lines cultured in extracellular
poietin-producing cells were not originated from matrix gels could be induced to create branching
integrating vessels but from circulating host mes- tubules either by the whole embryonic kidney or
enchymal stem cells mobilized from the bone by the metanephric mesenchyme [326].
marrow. Advancement of this methodology showed that
Conventional transplantation of metanephros cells isolated from ureteric bud (Fig. 7.4) of mice
requires strong and persistent immunosuppression at embryonic day 11.5 (E11.5) and propagated
to avoid humoral rejection associated with the
xenogeneic barrier, which can cause several
adverse effects, such as carcinogenicity and severe
rejection. Thus, for safety, when the xenotransplant
is not needed anymore, it should be discarded by
introducing a cell fate-regulating system in which a
suicide gene is expressed on demand. To avoid the
xenogeneic barrier, metanephroi isolated from
transgenic ER-E2F1 mice that are expressing E2F1
under the control of tamoxifen can be used. E2F1 is
a transcription factor which regulates cell prolifera-
tion and ectopic expression of which induces apop-
tosis. The administration of tamoxifen in host rats
wherein metanephroi of ER-E2Fa mice were trans-
planted activated the suicide gene, and the xeno- Fig. 7.4  Mechanically isolated ureteric bud from the
compartments of transplanted metanephroi were metanephros of E11.5 mouse embryo
170 B.S. Kim and H.T. Kim

in vitro could undergo branching tubulogenesis nephros with functional properties still remained
when stimulated by a conditioned medium from unclear.
a metanephric mesenchyme-derived cell line Using simple suspensions of singe kidney
[327]. Similarly, a tubular epithelial cell line or cells, “organoids” that could carry out renal func-
mesenchymal stem cells generated glomerular tions when implanted into a living animal, such
and tubular structures in subcutaneous spaces as glomerular filtration, tubular reabsorption of
when stimulated by a media from vascular endo- macromolecules, and production of erythropoie-
thelial and tubular cells [328]. These results sug- tin, were finally constructed [322]. These organ-
gested that simple cultures of cells derived from oids were generated from single-cell suspensions
undifferentiated metanephric mesenchyme and derived from E11.5 mouse kidneys and then
unbranched ureteric bud could be used to create implanted under the renal capsule of an athymic
branching tubular structures similar to those that rat. The main procedure to obtain complete glo-
form in normal metanephros. merulogenesis consisted of soaking the organ-
Generation of three-dimensional renal epi- oids in a solution containing VEGF, and then
thelia which contain glomerular-like structures injection of VEGF into the recipient animals after
in vitro was attempted using suspensions of the organoids had been implanted in the ­kidney.
murine metanephric mesenchymal E11.5 pro- This method restituted the efficacy of the podo-
genitor cells selected for a high expression of cyte VEGF axis that is necessary for glomerular
Sall-1, which is essential during kidney devel- capillary endothelial induction and resulted in the
opment [305]. This cell preparation required formation of vascularized glomeruli with fully
coculture with an exogenous spinal cord cell differentiated glomerular capillary walls, includ-
layer, as has been performed in traditional ing endothelial fenestrae, slit diaphragms, and
developmental investigations [329]. Although foot processes. These organoids could develop
the functional capacity of these structures has glomeruli with normal-appearing slits in vivo,
not been investigated in vivo, this approach sug- wherein the proportion of diaphragms were com-
gested that dispersed cell suspensions can be parable to that found in the adult glomerulus.
used as starting material to obtain rudimental When the ultrafiltering function of the intragraft
three-dimensional kidney tissues. Further devel- nephrons was tested by injecting fluorescent dex-
opment of this method enabled the generation of trans of increasingly high molecular weight into
fetal kidney tissue from suspensions of E11.5 the host blood system, the proximal tubular cells
kidney cells without using any exogenous tissue concentrated dextrans only of lower molecular
[306]. While this method successfully repro- weights from the lumen, indicating efficient
duced the structures and differentiation states of ultrafiltration.
nephrons and stroma, collecting ducts devel- Approaches starting from the metanephros
oped as a multitude of very small collecting and dissociating down to the single-cell level
duct trees rather than a continuous one as would offer a valuable source for performing pharma-
happen in a normal kidney. Combination of cologic or genetic modifications in individual
reaggregated single-cell suspensions of ureteric cells prior to the in vitro aggregation step [322,
bud and metanephric mesenchyme allowed the 331]. The single-cell stage allows the use of
formation of nephrons arranged around a single genetic engineering approaches to humanize
collecting duct tree [330]. However, this cells, by introducing immunomodulatory genes
endeavor did not achieve development of glom- or by switching off others, to reduce the possibil-
eruli to any meaningful extent due to avascular ity of rejection and facilitate the xenotransplan-
environment in vitro. This limitation inhibits tation of animal renal tissues [331]. These
further progress of research related to matura- self-forming organoids can be exploited to gen-
tion potential of tissue obtained from a simple erate human renal tissue by constructing chime-
culture or suspension of precursor cells. ric kidneys that combine animal progenitor cells
Therefore, the capacity of these cells to generate and human stem cells, followed by the selective
7 Kidney 171

elimination of animal cells after transplantation. grown in a whole-embryo culture system [335].
Previous studies have shown that the organoids For the successful generation of the organ using
could incorporate cells of another source, such this technique, human mesenchymal stem cells
as human amniotic fluid stem cells or iPSC- must be placed in a specific embryonic niche to
derived OSR1-­positive cells, to build chimeric allow their exposure to the nephrogenic signals
tissue in vitro [253, 274]. Although several tech- before the metanephros begins to develop. This
nical problems remain to be overcome, these can be achieved by implanting human mesen-
systems are useful to examine the mechanisms chymal stem cells into the nephrogenic site of a
of renal progenitor differentiation and suggest developing embryo. Therefore, the researchers
the possibility of developing a whole kidney established a culture system in combination
from a single stem cell. with a whole-embryo culture system, followed
by metanephric organ culture. This relay culture
enabled the development of the metanephros
7.4.8 Kidney Regeneration Using from ­structures present before budding until the
Xenoembryos occurrence of complete organogenesis ex utero.
Using this system, embryos were isolated before
The idea of generating human organs in animals budding and were grown in a culture bottle until
was previously suggested by several studies [332, the formation of rudimentary kidneys so that it
333]. Regeneration of a whole functional kidney could be further developed by organ culture
that produced urine and renal hormones using a in vitro (Fig. 7.5). It was also observed that rudi-
developing heterozoic embryo as an organ fac- mentary kidneys also continued to grow with
tory has been endeavored [334–336]. This strat- fine tubulogenesis and ureteric bud branching
egy is based on the concept of “borrowing” the in vitro, demonstrating that the metanephros can
developing program of a growing xenoembryo continue developing ex utero even if the embryo
by applying stem cells at the niche of organogen- is dissected prior to sprouting of the ureteric bud
esis. During development of the metanephros, the (Fig. 7.6). In this study, before injection into the
metanephric mesenchyme initially forms from metanephros, the human mesenchymal stem
the caudal portion of the nephrogenic cord and cells were genetically engineered to express
secretes GDNF, which induces the nearby GDNF temporally using adenovirus and were
Wolffian duct to produce a ureteric bud [337]. labeled with the LacZ gene and dioctadecyl-
Then, the metanephric mesenchyme conse- 3 , 3 , 3 ′ , 3 ′ - t e t r a m e t h y l i n d o c a r b o cy a n i n e .
quently forms the glomerulus, proximal and dis- However, viral-­free manipulation can also be
tal tubule, loop of Henle, and the interstitium, as performed using a thermoreversible GDNF
a result of reciprocal epithelial-mesenchymal polymer during this process [339]. Consequently,
induction between the ureteric bud and meta- donor human mesenchymal stem cells were
nephric mesenchyme [338]. For this epithelial-­ integrated into the rudimentary metanephros
mesenchymal induction to occur, GDNF must and differentiated morphologically into tubular
interact with its receptor, c-ret, which is expressed and glomerular epithelial cells and interstitial
in the Wolffian duct. Thus, it can be hypothesized cells. Subsequently, they transplanted the devel-
that GDNF-expressing stem cells can differenti- oped metanephros into the omentum to allow
ate into kidney structures if they are placed at the vascular integration from the recipient to form a
budding site and stimulated by several factors functional nephron. Finally, a human mesenchy-
spatially and temporally identical to those found mal stem cell-derived neokidney was generated,
in the developmental milieu. which contained a human nephron associated
To investigate this hypothesis, Yokoo et al. with host vasculature [334]. The neokidney pro-
microinjected GDNF-expressing human mesen- duced urine with higher concentrations of cre-
chymal stem cells into the developing meta- atinine and urea nitrogen than those of sera of
nephros in vitro, and the recipient embryo was the recipient. This result suggested that the
172 B.S. Kim and H.T. Kim

a b

Fig. 7.5 Development of metanephros in vitro. culture (a), more branching of ureteric bud can be
Metanephros was isolated from E12 mouse and trans- observed in the metanephros cultured in vitro for 5 days
ferred into a culture dish. Then, it continued to grow. (b)
Compared to the morphology of metanephros at day 1 of

a b c

Fig. 7.6  Immunohistochemical staining of in vitro cul- (c) Merged image of ureteric bud and metanephric mesen-
ture metanephros (red, Six2; green, cytokeratin 8; blue, chyme. (d) Kidney-shaped whole metanephros consisting
E-cadherin). (a) Fine tubulogenesis and multiple branch- of metanephric mesenchyme and multiple branches of
ing of ureteric bud can be observed. (b) Six2 (nephron ureteric bud
progenitor marker) expressing metanephric mesenchyme.
7 Kidney 173

neokidney that developed in the host omentum such as the glomerulus and the tubule, to replace
possesses the ability to produce urine by filter- both excretory and metabolic functions of the
ing the host’s blood. In addition, the human kidney [341, 342].
mesenchymal stem cell-derived neokidney The development of bioartificial kidney repre-
secreted human erythropoietin, which was stim- sents the intersection of regenerative medicine
ulated by the induction of anemia in the host and renal replacement therapy. Bioartificial kid-
animal [336]. This finding demonstrated that ney consists of a hemofilter used in conventional
this system could preserve the normal hormonal dialysis with a bioreactor unit containing human
regulation of erythropoietin levels. However, primary renal proximal tubule cells, termed a
ureter or collecting ducts derived from ureteric renal tubule assist device [343]. The addition of
bud could not be reconstructed using this sys- the bioreactor is intended to provide homeostatic,
tem. Thus, another investigation to determine metabolic, endocrine, and immunomodulatory
whether mesenchymal stem cells can differenti- functions of the kidney that cannot be accom-
ate into the ureteric bud progenitor cells was plished by dialysis. Moreover, these units were
performed using chick embryo [340]. Human designed to be wearable or implantable to offer
mesenchymal stem cells expressing Pax2 were continuous renal support system with minimal
injected into the chicken ureteric bud progenitor deterioration of life quality. Since the bioartificial
region, and then the cells migrated caudally kidney was first proposed by Aebischer et al. in
with the elongating Wolffian duct and were inte- 1987, the technology has developed from con-
grated into the Wolffian duct epithelia and cept to clinical trial [344]. Recently, Humes et al.
expressed LIM1. This result indicated that reported the safe and efficacious use of bioartifi-
human mesenchymal stem cells can differenti- cial kidneys in patients with acute renal failure
ate into Wolffian duct cells under the influence [345]. The bioartificial kidney demonstrated
of local xenosignals. Taken together, a whole metabolic, endocrine, and immunomodulatory
kidney may be reconstructed by transplanting activities, including glutathione degradation,
human mesenchymal stem cells at an appropri- hydroxylation of calcifediol, and reduction of
ate time and location to regenerate derivatives of proinflammatory cytokines. More details of this
the metanephric mesenchyme and ureteric bud. progressive development of bioartificial kidney
are presented in the following sections.

7.5 Bioartificial Kidney


7.5.1 Renal Tubule Assist Device
Although the final goal of the research for kidney
regeneration is generation of a whole functional Replacement of the various tubular functions of
kidney, it still has many hurdles to be realized. the kidney cannot be achieved with inanimate
Instead, ex vivo renal-supporting device, such as membrane devices, as has been accomplished
bioartificial kidney using various biomaterials, with the renal ultrafiltration process, but requires
has been investigated. Current renal replacement the use of the naturally evolved biologic mem-
therapy with either hemodialysis or peritoneal branes of the renal tubular epithelium. In this
dialysis provides only filtration function and does regard, the tissue engineering of a bioartificial
not replace the homeostatic, metabolic, endo- renal tubule as a cell therapy device to replace
crine, and immunomodulatory functions of the these missing functions can be conceived as a
kidney. Thus, when designing a bioartificial kid- combination of living cells supported on syn-
ney for renal function replacement, these critical thetic scaffolds [346, 347]. A bioartificial tubule
elements of renal function must be replaced. The can be generated using progenitor cells of renal
functioning excretory unit of the kidney is com- tubule culture on semipermeable hollow fiber
posed of the filtering unit (glomerulus) and the membranes on which extracellular matrix has
regulatory or transport unit (tubule). Therefore, a been layered to enhance the attachment and
bioartificial kidney requires these two main units, growth of epithelial cells [348, 349]. These
174 B.S. Kim and H.T. Kim

h­ ollow fiber synthetic membranes provide immu- immediately following bacterial administration.
noprotection, which has been observed in the Acute kidney injury model was successfully gen-
long-term implantation of the bioartificial pan- erated in all animals within 2–4 h after bacterial
creas in a xenogeneic host, as well as the archi- administration. Renal tubule assist device-treated
tectural scaffold for these cells [350]. group showed better cardiovascular performance,
Subsequently, it was investigated whether the including cardiac output and renal blood flow, for
renal tubule assist device could maintain differ- longer periods than control group. Consequently,
entiated renal functional performance, similar to experimental group also survived longer than con-
that observed in vitro, and viability in an extra- trol groups. Treatment with renal tubule assist
corporeal hemoperfusion circuit in an acute ure- device was associated with significantly lower
mic animal model [351]. The renal tubule assist plasma circulating levels of IL-6, a proinflamma-
device was accordingly placed in series with a tory cytokine, and interferon-ɤ. These findings
standard hollow fiber hemofiltration cartridge. indicate that septic shock results in early acute kid-
The two cartridges had ultrafiltrate and post-­ ney injury and that treatment with renal tubule
filtered blood connections to duplicate the struc- assist device in a bioartificial kidney extracorpo-
tural anatomy of the nephron and to mimic the real circuit improves cardiovascular performance
functional relationship between the glomerulus associated with changes in cytokine profiles and
and tubule. In this study, fluid and electrolyte bal- confers a significant survival advantage.
ances in the animal were adequately controlled With these encouraging preclinical outcomes,
with the bioartificial kidney, and especially phases I and II clinical studies using renal tubule
plasma potassium and blood urea nitrogen levels assist device containing human cells were per-
were more easily controlled during the treatment formed in patients with acute kidney injury
using renal tubule assist device as compared with receiving continuous renal replacement therapy.
controls. In addition, active transport of potas- Human kidney cells were isolated from kidneys
sium, bicarbonate, and glucose and successful donated for cadaveric renal transplantation but
metabolic functions, such as ammoniagenesis, found unsuitable due to anatomic of fibrotic
were also observed. defects. The results of phase I study in the first
An animal study to investigate whether treat- ten patients treated with renal tubule assist
ment with the renal tubule assist device could alter device containing cells demonstrated that this
the course of acute renal failure with sepsis was treatment modality can be delivered safely under
performed [352]. Mongrel dogs with nephrec- study protocol guidelines for up to 24 h when
tomy-induced acute renal failure were treated with used in conjunction with continuous venovenous
continuous venovenous hemofiltrations and either hemofiltrations [345]. Cardiovascular stability
a renal tubule assist device containing cells or an of the patients was maintained with increased
identically prepared sham cartridge without cells. urine outputs. These results also demonstrated
After 4 hours of therapy, endotoxin was infused the viability, durability, and functionality of
intravenously to stimulate Gram-negative septic renal tubule assist device with cells in clinical
shock. Mean peak levels of an anti-inflammatory setting. Of these ten patients who had high risk
cytokine, IL-10, and mean arterial blood pressures of mortality, six survived past 28 days with renal
were significantly higher in the animals treated by function recovery. Plasma cytokine levels
renal tubule assist device containing cells. This implied that treatment with renal tubule assist
study group also performed similar experiments in device produces dynamic and individualized
a larger animal model [353]. E. coli was adminis- responses in patients, depending on their unique
tered intraperitoneally into pigs with normal renal pathophysiologic conditions. Several cytokines,
function to induce septic shock. Then, animals such as IL-6, IL-10, and granulocyte colony-
were treated using continuous venovenous hemo- stimulating factor, were significantly decreased
filtrations with either a renal tubule assist device after treatment with renal tubule assist device
containing cells or a sham cartridge without cells compared to baselines in patients who had
7 Kidney 175

e­ xcessive proinflammatory levels. These encour- spread adoption of renal cell therapy [357]. The
aging results led to perform randomized, con- bioartificial renal epithelial cell system is a model
trolled, open-label phase II trial, which was of a cell therapeutic device that maintains a high
completed at ten clinical sites to assess the safety density of renal cells for therapeutic application
and early efficacy of this cell-based renal tubule within an extracorporeal circuit. Compared to
assist device therapy. other cell therapies involving the direct applica-
In this phase II clinical trial, 58 patients with tion or injection of stem cells into the body, the
acute kidney injury requiring renal replacement extracorporeal approach of the bioartificial renal
therapy in the intensive care unit were enrolled epithelial cell system allows for cell therapeutic
and randomized to receive continuous renal application while maintaining immunoisolation
replacement therapy with renal tubule assist using a series of filters. Compared to renal tubule
device (n = 40) or renal replacement therapy assist device, bioartificial renal epithelial cell
alone (n = 18). Treatment with renal tubule assist system enables mass fabrication, cryopreserva-
device containing cells promoted a statistically tion for storage, and distribution to meet on-­
significant survival advantage over 180 days of demand clinical need.
follow-up having a 67% survival rate compared In summary, the bioartificial renal epithelial
to a 39% survival rate of the control group and cell system is a perfusion bioreactor that utilizes
demonstrated an acceptable safety profile. A fol- primary renal epithelial cells derived from the
low-­up phase IIb study to evaluate a commercial kidney, expanded from progenitor cells during
manufacturing process was not completed due to in vitro culture. Cells are seeded on porous
difficulties with the manufacturing process and niobium-­coated carbon disks, which are placed
clinical study design, which led to a suspension in a media flow path within the bioartificial
of the clinical development of this approach renal epithelial cell system. Renal progenitor
[354]. However, this suspended study has led to cells are directed toward a renal tubule cell fate
an innovative biomimetic membrane cell pro- and maintained in perfusion culture prior to
cessing device to treat systemic inflammatory therapeutic application. In vitro cell viability
response syndrome and has advanced to a pivotal and metabolic activity are confirmed in the bio-
clinical trial in patients with multiorgan failure reactor renal epithelial cell system by measur-
[355, 356]. ing lactate production and oxygen consumption.
Despite these successes of preclinical and Metrics of metabolism are consistent through-
clinical studies of the renal tubule assist device out the duration of perfusion culture, with an
containing cells in renal replacement therapy for estimated total cell number of approximately
patients with acute kidney injury, there are sev- 108 cells [357]. Renal epithelial cells in the bio-
eral limitations for successful commercialization artificial renal epithelial cell system maintain
of cell-based therapeutic devices, such as the renal differentiated phenotypic characteristics
need for a reliable and consistent source of cells over time in perfusion culture. Cryopreservation
to manufacture thousands of these cell devices and thawing of renal epithelial cell in bio-
and the requirement to develop a cost-effective artificial renal epithelial cell system were
manufacturing, storage, and distribution process accomplished using a commercially available
for these devices. cryopreservation media. Post-­cryopreservation
cell retention was optimized in combination
with a controlled rate freezer, resulting in aver-
7.5.2 B
 ioartificial Renal Epithelial age cell retention of more than 80% and viabil-
Cell System ity of more than 90% [357]. This system offers
an all-in-one system for cell culture, cryostor-
The bioartificial renal epithelial cell system aims age, and distribution of cell therapy for on-
to address the important issues of cell cryopreser- demand use for acute indications, such as acute
vation, storage, and distribution for the wide- kidney injury and sepsis.
176 B.S. Kim and H.T. Kim

7.5.3 W
 earable or Implantable nutrient stream has been avoided to prevent risks
Bioartificial Kidney of clotting and infection. Similar to acute appli-
cations of cell-based therapies, the compact,
The ideal bioartificial kidney suitable for long-­ cryopreservable design of bioartificial renal epi-
term use in patients with end-stage renal disease thelial cell system and the enhanced propagation
would be capable of performing continuously to methods used to isolate and expand the renal
reduce risks from fluctuations in volume status, cells in the bioartificial renal epithelial cell sys-
electrolytes, and solute concentrations and to tem are enabling advancements toward the devel-
maintain regulation of acid-base and uremic opment of a wearable bioartificial kidney for
toxin. Such treatment requires the design and chronic applications which integrates a wearable
manufacture of a compact wearable or implant- sorbent dialysis system and bioartificial renal
able dialysis unit and the development of com- epithelial cell system with recycling peritoneal
pact renal tubular cell devices with long fluid. Further investigation and refinement
durability. Therefore, next-generation renal according to this wearable bioartificial kidney are
tubule assist device should be like that of an still going on.
implantable device similar to the pacemaker. For better patients’ quality of life, there have
Recently, a wearable bioartificial kidney has been attempts to create a miniaturized version
been suggested and evaluated in preclinical study of the renal tubule assist device that would be
utilizing either hemodialysis or peritoneal dialy- suitable for implantation and offer the benefits
sis as the therapeutic circuit delivery route. The of a transplanted kidney. An implantable bioar-
wearable bioartificial kidney is comprised of the tificial kidney has the potential to overcome
use of sorbent-based technologies to replace the both shortage of kidney donors and the burden
excretory function of the kidney and the compact of therapy of intensive maintenance dialysis.
bioartificial renal epithelial cell system to replace Microelectromechanical system technology has
the metabolic function of the kidney. The first been utilized to develop and implantable renal
trial to develop wearable dialysis system gener- tubule assist device. The silicon nanopore mem-
ated a lightweight, wearable, continuous ambula- branes have been used for both hemofilter and
tory ultrafiltration device consisting of a hollow an immunoisolation scaffold for a cellular bio-
fiber hemofilter, a battery-operated pulsatile reactor [342, 361–364]. The ultrathin silicon
pump, and two micropumps to control heparin nanopore membranes have a uniform slit pore
infusion and ultrafiltration [358]. This device design giving higher hydraulic permeability and
regenerates dialysate with activated carbon, selectivity compared to the roughly circular-
immobilized urease, zirconium hydroxide, and shaped pores in standard hollow fiber mem-
zirconium phosphate. However, this approach branes [365]. Since the silicon nanopore
requires continuous blood access to allow ade- membranes can have pore sizes as small as 5 nm
quate ultrafiltration. with less than 1% variability, these membranes
Another approach to utilize continuous regen- can function like the glomerulus of the native
eration of peritoneal dialytic fluid relying on kidney by selectively filtering solutes based on
continuous-flow peritoneal dialysis systems was molecular weight cutoffs. The selectivity of sili-
also investigated [359, 360]. With the develop- con nanopore membranes has been shown
ment of a sorbent-based wearable, continuous-­ in vitro using β2-microglobulin and several
flow recycling peritoneal dialysis, the integration globular proteins. Silicon readily forms a thin
of renal epithelial cell therapy in the peritoneal oxide coating upon exposure to atmospheric
dialysis circuit to treat patients with end-stage oxygen. This silica film is negatively charged at
renal disease is also being evaluated in large ani- physiologic pH, and then it absorbs plasma pro-
mal models with uremia. Although a cell therapy teins and activates the coagulation cascade. It is
device requires a continuous support of nutrients necessary to modify the silicon surface with a
and oxygen, the use of blood circuits for this highly hydrated polymer to reduce adverse
7 Kidney 177

blood-material interactions. The successful out- technique, blastocyst complementation and


comes of grafting several organic polymers on embryonic organ, and bioartificial kidney.
silicon surfaces demonstrated the feasibility of Although there are many interesting approaches
using silicon nanopore membranes coated with and new frontiers are being explored in the field
antifouling films for hemofiltration, allowing of kidney regeneration, many unsolved problems
for progress toward an implantable bioartificial still remain. In addition, determination of the
kidney [366]. optimal cell sources is also important for appro-
Despite these advancements in biocompatibil- priate regeneration of kidney tissue. The use of
ity of silicon membranes, vascular perfusion of renal stem/progenitor cells in kidney regenera-
the bioartificial kidney will be needed to maintain tion is still somehow limited due to their low
filtration and cell viability with the use of long-­ prevalence and growth restriction. Mesenchymal
term anticoagulation. In addition, there are still stem cells are easily accessible and usually do not
limitations to miniaturize and implant the hemo- require technical manipulations, but mesenchy-
dialysis circuit, such as the size and pump require- mal stem cells from patients with chronic kidney
ments of modern dialyzers and the water volume disease may not be suitable for regenerative ther-
required for dialytic therapy. Hollow fiber poly- apy, because uremia induces functional incompe-
mer membranes have been immensely successful tence of mesenchymal stem cells. Although
in treating renal failure with extracorporeal thera- embryonic stem cells are pluripotent, there are
pies but require super-physiologic driving pres- ethical issues associated with manipulation of
sures for blood circulation through the cartridge. germ cells in producing embryonic stem cells.
The long cylindrical hollow fibers present a high Whereas, iPSCs are pluripotent and free from
resistance to blood flow, which rises further in the ethical issues, but the use of retroviral transduc-
distal portion of the fiber as ultrafiltration tion and our limited understanding of its effects
increases hematocrit and viscosity, necessitating still inhibit clinical applications of iPSCs.
energy requiring roller pumps to circulate blood However, new insights into the processes
through the device. The pores in hollow fiber dia- controlling endogenous renal repair and the
lyzers are irregular, cylindrical, and multiform in capacity to isolate progenitor cells from the adult
size, and this polydispersity compromises the renal epithelium are providing possible options
trade-off between permeability and selectivity. for the treatment of acute kidney injury. Major
The majority of the membrane’s pores should be advances in the directed differentiation of stem
maintained smaller than the desired cutoff target cells to a large range of kidney cell types have
of the membrane to prevent albumin leakage also showed significant possibility of kidney
through the largest pores in the membrane. regeneration. Moreover, cell-scaffold technol-
Another limitation is the volume of dialytic water, ogy gave us more chance to reconstruct a three-
which is most readily demonstrated by the esti- dimensional whole functional kidney similar to
mated volume of water required during a 4-h dial- the native human kidney. Furthermore, recent
ysis with a dialysate flow of 600 ml/min, efforts to regenerate de novo a whole functional
consuming 144 l of dialysate, being as much as kidney using xenoembryo have suggested
additional 200 l of tap water [361, 365]. another potential strategy.
In the future, we should characterize gene
expression profiles of regeneration associated
7.6 Future Perspectives cells and elucidate corresponding signaling mol-
ecules more clearly. Based on the knowledge of
In this chapter, we have reviewed recent advances renal development, a more reliable technique
in renal regenerative therapy including renal should be established to manipulate stem/pro-
stem/progenitor cells and exogenous stem cells genitor cells. Enhanced understanding of the
to treat damaged renal tissue, de novo whole mechanism of currently available stem cells with
functional organ regeneration using cell-scaffold the capacity of renoprotection can also help us to
178 B.S. Kim and H.T. Kim

find the novel pathway for kidney regeneration, marrow-derived cells to differentiate to glomerular
mesangial cells. J Am Soc Nephrol. 2001;12:1401–9.
and more specific cell or gene therapy should be
15. Ito T, Suzuki A, Imai E, Okabe M, Hori M. Bone
discovered for different kidney diseases. Finally, marrow is a reservoir of repopulating mesangial cells
continued collaborative efforts are required to during glomerular remodeling. J Am Soc Nephrol.
achieve the generation of a bioengineered kidney 2001;12:2625–35.
16. Ito T, Suzuki A, Okabe M, Imai E, Hori M. Application
that is capable of restoring renal function in
of bone marrow-derived stem cells in experimental
patients with end-stage renal disease. nephrology. Exp Nephrol. 2001;9:444–50.
17. Poulsom R, Forbes SJ, Hodivala-Dilke K, Ryan E,
Wyles S, Navaratnarasah S, et al. Bone marrow con-
tributes to renal parenchymal turnover and regenera-
References tion. J Pathol. 2001;195:229–35.
18. Witzgall R, Brown D, Schwarz C, Bonventre
1. Jha V, Garcia-Garcia G. Global kidney disease – JV. Localization of proliferating cell nuclear anti-
authors’ reply. Lancet. 2013;382:1244. gen, vimentin, c-Fos, and clusterin in the postisch-
2. McCampbell KK, Wingert RA. Renal stem cells: fact emic kidney. Evidence for a heterogenous genetic
or science fiction? Biochem J. 2012;444:153–68. response among nephron segments, and a large pool
3. Li Y, Wingert RA. Regenerative medicine for the of mitotically active and dedifferentiated cells. J Clin
kidney: stem cell prospects & challenges. Clin Invest. 1994;93:2175–88.
Transl Med. 2013;2:11. 19. Bonventre JV. Dedifferentiation and proliferation of
4. Chung HC, Ko IK, Atala A, Yoo JJ. Cell-based ther- surviving epithelial cells in acute renal failure. J Am
apy for kidney disease. Korean journal of urology. Soc Nephrol. 2003;14(Suppl 1):S55–61.
2015;56:412–21. 20. Bussolati B, Bruno S, Grange C, Buttiglieri S,
5. Wolfe RA, Ashby VB, Milford EL, Ojo AO, Ettenger Deregibus MC, Cantino D, et al. Isolation of renal
RE, Agodoa LY, et al. Comparison of mortality in progenitor cells from adult human kidney. Am J
all patients on dialysis, patients on dialysis awaiting Pathol. 2005;166:545–55.
transplantation, and recipients of a first cadaveric 21. Lazzeri E, Crescioli C, Ronconi E, Mazzinghi B,
transplant. N Engl J Med. 1999;341:1725–30. Sagrinati C, Netti GS, et al. Regenerative potential
6. Katari R, Peloso A, Zambon JP, Soker S, Stratta of embryonic renal multipotent progenitors in acute
RJ, Atala A, et al. Renal bioengineering with scaf- renal failure. J Am Soc Nephrol. 2007;18:3128–38.
folds generated from human kidneys. Nephron Exp 22. Angelotti ML, Ronconi E, Ballerini L, Peired A,
Nephrol. 2014;126:119. Mazzinghi B, Sagrinati C, et al. Characterization
7. Fukui A, Yokoo T. Kidney regeneration using devel- of renal progenitors committed toward tubular lin-
oping xenoembryo. Curr Opin Organ Transplant. eage and their regenerative potential in renal tubular
2015;20:160–4. injury. Stem Cells. 2012;30:1714–25.
8. Chan TC, Ariizumi T, Asashima M. A model sys- 23. Humphreys BD, Valerius MT, Kobayashi A,
tem for organ engineering: transplantation of in vitro Mugford JW, Soeung S, Duffield JS, et al. Intrinsic
induced embryonic kidney. Naturwissenschaften. epithelial cells repair the kidney after injury. Cell
1999;86:224–7. Stem Cell. 2008;2:284–91.
9. Xinaris C, Yokoo T. Reforming the kidney start- 24. Sagrinati C, Netti GS, Mazzinghi B, Lazzeri E,
ing from a single-cell suspension. Nephron Exp Liotta F, Frosali F, et al. Isolation and character-
Nephrol. 2014;126:107–12. ization of multipotent progenitor cells from the
10. Locatelli F, Buoncristiani U, Canaud B, Kohler Bowman's capsule of adult human kidneys. J Am
H, Petitclerc T, Zucchelli P. Dialysis dose and fre- Soc Nephrol. 2006;17:2443–56.
quency. Nephrol Dial Transplant. 2005;20:285–96. 25. Romagnani P, Remuzzi G. Renal progenitors in
11. Maeshima A, Nakasatomi M, Nojima non-diabetic and diabetic nephropathies. Trends
Y. Regenerative medicine for the kidney: renotropic Endocrinol Metab. 2013;24:13–20.
factors, renal stem/progenitor cells, and stem cell 26. Grobstein C. Trans-filter induction of tubules in
therapy. Biomed Res Int. 2014;2014:1–10. mouse metanephrogenic mesenchyme. Exp Cell
12. Strutz F, Okada H, Lo CW, Danoff T, Carone RL, Res. 1956;10:424–40.
Tomaszewski JE, et al. Identification and charac- 27. Grobstein C. Inductive tissue interaction in develop-
terization of a fibroblast marker: FSP1. J Cell Biol. ment. Adv Cancer Res. 1956;4:187–236.
1995;130:393–405. 28. Kitamura S, Yamasaki Y, Kinomura M, Sugaya T,
13. Cornacchia F, Fornoni A, Plati AR, Thomas A, Sugiyama H, Maeshima Y, et al. Establishment and
Wang Y, Inverardi L, et al. Glomerulosclerosis is characterization of renal progenitor like cells from
transmitted by bone marrow-derived mesangial cell S3 segment of nephron in rat adult kidney. FASEB J.
progenitors. J Clin Invest. 2001;108:1649–56. 2005;19:1789–97.
14. Imasawa T, Utsunomiya Y, Kawamura T, Zhong Y, 29. Lindgren D, Bostrom AK, Nilsson K, Hansson J,
Nagasawa R, Okabe M, et al. The potential of bone Sjolund J, Moller C, et al. Isolation and characteriza-
7 Kidney 179

tion of progenitor-like cells from human renal proximal 45. Berger K, Bangen JM, Hammerich L, Liedtke C,
tubules. Am J Pathol. 2011;178:828–37. Floege J, Smeets B, et al. Origin of regenerating
30. Maeshima A, Yamashita S, Nojima Y. Identification tubular cells after acute kidney injury. Proc Natl
of renal progenitor-like tubular cells that participate Acad Sci U S A. 2014;111:1533–8.
in the regeneration processes of the kidney. J Am 46. Dekel B, Shezen E, Even-Tov-Friedman S, Katchman
Soc Nephrol. 2003;14:3138–46. H, Margalit R, Nagler A, et al. Transplantation of
31. Bruno S, Camussi G. Isolation and characterization human hematopoietic stem cells into ischemic and
of resident mesenchymal stem cells in human glom- growing kidneys suggests a role in vasculogenesis
eruli. Methods Mol Biol. 2012;879:367–80. but not tubulogenesis. Stem Cells. 2006;24:1185–93.
32. Oliver JA, Maarouf O, Cheema FH, Martens TP, 47. Duffield JS, Park KM, Hsiao LL, Kelley VR,
Al-Awqati Q. The renal papilla is a niche for adult Scadden DT, Ichimura T, et al. Restoration of tubular
kidney stem cells. J Clin Invest. 2004;114:795–804. epithelial cells during repair of the postischemic kid-
33. Kobayashi A, Valerius MT, Mugford JW, Carroll TJ, ney occurs independently of bone marrow-derived
Self M, Oliver G, et al. Six2 defines and regulates a stem cells. J Clin Invest. 2005;115:1743–55.
multipotent self-renewing nephron progenitor popu- 48. Friedenstein AJ, Latzinik NW, Grosheva AG,
lation throughout mammalian kidney development. Gorskaya UF. Marrow microenvironment transfer
Cell Stem Cell. 2008;3:169–81. by heterotopic transplantation of freshly isolated
34. Self M, Lagutin OV, Bowling B, Hendrix J, Cai Y, and cultured cells in porous sponges. Exp Hematol.
Dressler GR, et al. Six2 is required for suppres- 1982;10:217–27.
sion of nephrogenesis and progenitor renewal in the 49. Bi B, Schmitt R, Israilova M, Nishio H, Cantley
developing kidney. EMBO J. 2006;25:5214–28. LG. Stromal cells protect against acute tubular
35. Guimaraes-Souza NK, Yamaleyeva LM, injury via an endocrine effect. J Am Soc Nephrol.
AbouShwareb T, Atala A, Yoo JJ. In vitro recon- 2007;18:2486–96.
stitution of human kidney structures for renal cell 50. Imberti B, Morigi M, Tomasoni S, Rota C, Corna
therapy. Nephrol Dial Transplant. 2012;27:3082–90. D, Longaretti L, et al. Insulin-like growth factor-1
36. Lazzeri E, Ronconi E, Angelotti ML, Peired A, sustains stem cell mediated renal repair. J Am Soc
Mazzinghi B, Becherucci F, et al. Human urine-­ Nephrol. 2007;18:2921–8.
derived renal progenitors for personalized model- 51. Togel F, Hu Z, Weiss K, Isaac J, Lange C,
ing of genetic kidney disorders. J Am Soc Nephrol. Westenfelder C. Administered mesenchymal stem
2015;26:1961–74. cells protect against ischemic acute renal failure
37. Weissman IL, Anderson DJ, Gage F. Stem and pro- through differentiation-independent mechanisms.
genitor cells: origins, phenotypes, lineage commit- Am J Physiol Renal Physiol. 2005;289:F31–42.
ments, and transdifferentiations. Annu Rev Cell Dev 52. Togel F, Cohen A, Zhang P, Yang Y, Hu Z,
Biol. 2001;17:387–403. Westenfelder C. Autologous and allogeneic marrow
38. Coskun V, Wu H, Blanchi B, Tsao S, Kim K, Zhao J, stromal cells are safe and effective for the treatment of
et al. CD133+ neural stem cells in the ependyma of acute kidney injury. Stem Cells Dev. 2009;18:475–85.
mammalian postnatal forebrain. Proc Natl Acad Sci 53. Crisan M, Yap S, Casteilla L, Chen CW, Corselli M,
U S A. 2008;105:1026–31. Park TS, et al. A perivascular origin for mesenchy-
39. Ivanova L, Hiatt MJ, Yoder MC, Tarantal AF, mal stem cells in multiple human organs. Cell Stem
Matsell DG. Ontogeny of CD24 in the human kid- Cell. 2008;3:301–13.
ney. Kidney Int. 2010;77:1123–31. 54. da Silva ML, Chagastelles PC, Nardi NB. Mesenchymal
40. Pleniceanu O, Harari-Steinberg O, Dekel B. Concise stem cells reside in virtually all post-natal organs and
review: kidney stem/progenitor cells: differentiate, tissues. J Cell Sci. 2006;119:2204–13.
sort out, or reprogram? Stem Cells. 2010;28:1649–60. 55. Dominici M, Le Blanc K, Mueller I, Slaper-­
41. Hendry CE, Little MH. Reprogramming the kid- Cortenbach I, Marini F, Krause D, et al. Minimal cri-
ney: a novel approach for regeneration. Kidney Int. teria for defining multipotent mesenchymal stromal
2012;82:138–46. cells. The International Society for Cellular Therapy
42. Kusaba T, Lalli M, Kramann R, Kobayashi A, position statement. Cytotherapy. 2006;8:315–7.
Humphreys BD. Differentiated kidney epithelial 56. Horwitz EM, Le Blanc K, Dominici M, Mueller I,
cells repair injured proximal tubule. Proc Natl Acad Slaper-Cortenbach I, Marini FC, et al. Clarification
Sci U S A. 2014;111:1527–32. of the nomenclature for MSC: The International
43. Humphreys BD, Czerniak S, DiRocco DP, Hasnain Society for Cellular Therapy position statement.
W, Cheema R, Bonventre JV. Repair of injured prox- Cytotherapy. 2005;7:393–5.
imal tubule does not involve specialized progenitors. 57. Nombela-Arrieta C, Ritz J, Silberstein LE. The elu-
Proc Natl Acad Sci U S A. 2011;108:9226–31. sive nature and function of mesenchymal stem cells.
44. Smeets B, Boor P, Dijkman H, Sharma SV, Jirak Nat Rev Mol Cell Biol. 2011;12:126–31.
P, Mooren F, et al. Proximal tubular cells contain 58. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK,
a phenotypically distinct, scattered cell popula- Douglas R, Mosca JD, et al. Multilineage potential
tion involved in tubular regeneration. J Pathol. of adult human mesenchymal stem cells. Science.
2013;229:645–59. 1999;284:143–7.
180 B.S. Kim and H.T. Kim

59. Salem HK, Thiemermann C. Mesenchymal stromal 72. Yoshino J, Monkawa T, Tsuji M, Hayashi M, Saruta
cells: current understanding and clinical status. Stem T. Leukemia inhibitory factor is involved in tubular
Cells. 2010;28:585–96. regeneration after experimental acute renal failure. J
60. Pelekanos RA, Li J, Gongora M, Chandrakanthan V, Am Soc Nephrol. 2003;14:3090–101.
Scown J, Suhaimi N, et al. Comprehensive transcrip- 73. Terada Y, Tanaka H, Okado T, Shimamura H,
tome and immunophenotype analysis of renal and Inoshita S, Kuwahara M, et al. Expression and func-
cardiac MSC-like populations supports strong con- tion of the developmental gene Wnt-4 during experi-
gruence with bone marrow MSC despite maintenance mental acute renal failure in rats. J Am Soc Nephrol.
of distinct identities. Stem Cell Res. 2012;8:58–73. 2003;14:1223–33.
61. Matsumoto K, Mizuno S, Nakamura T. Hepatocyte 74. Chen J, Chen JK, Harris RC. Deletion of the epider-
growth factor in renal regeneration, renal disease mal growth factor receptor in renal proximal tubule
and potential therapeutics. Curr Opin Nephrol epithelial cells delays recovery from acute kidney
Hypertens. 2000;9:395–402. injury. Kidney Int. 2012;82:45–52.
62. Humes HD, Cieslinski DA, Coimbra TM, Messana 75. Sugimoto H, LeBleu VS, Bosukonda D, Keck P,
JM, Galvao C. Epidermal growth factor enhances Taduri G, Bechtel W, et al. Activin-like kinase 3 is
renal tubule cell regeneration and repair and acceler- important for kidney regeneration and reversal of
ates the recovery of renal function in postischemic fibrosis. Nat Med. 2012;18:396–404.
acute renal failure. J Clin Invest. 1989;84:1757–61. 76. Zhou D, Tan RJ, Lin L, Zhou L, Liu Y. Activation
63. Sakai M, Zhang M, Homma T, Garrick B, Abraham of hepatocyte growth factor receptor, c-met, in renal
JA, McKanna JA, et al. Production of heparin bind- tubules is required for renoprotection after acute kid-
ing epidermal growth factor-like growth factor in the ney injury. Kidney Int. 2013;84:509–20.
early phase of regeneration after acute renal injury. 77. Maeshima A, Miya M, Mishima K, Yamashita S,
Isolation and localization of bioactive molecules. J Kojima I, Nojima Y. Activin A: autocrine regula-
Clin Invest. 1997;99:2128–38. tor of kidney development and repair. Endocr J.
64. Homma T, Sakai M, Cheng HF, Yasuda T, Coffey RJ 2008;55:1–9.
Jr, Harris RC. Induction of heparin-binding epidermal 78. Maeshima A, Nojima Y, Kojima I. The role of the
growth factor-like growth factor mRNA in rat kidney activin-follistatin system in the developmental and
after acute injury. J Clin Invest. 1995;96:1018–25. regeneration processes of the kidney. Cytokine
65. Zeisberg M, Hanai J, Sugimoto H, Mammoto T, Growth Factor Rev. 2001;12:289–98.
Charytan D, Strutz F, et al. BMP-7 counteracts 79. Maeshima A, Sakurai H, Choi Y, Kitamura S, Vaughn
TGF-beta1-induced epithelial-to-mesenchymal tran- DA, Tee JB, et al. Glial cell-derived neurotrophic
sition and reverses chronic renal injury. Nat Med. factor independent ureteric bud outgrowth from the
2003;9:964–8. Wolffian duct. J Am Soc Nephrol. 2007;18:3147–55.
66. Vukicevic S, Basic V, Rogic D, Basic N, Shih MS, 80. Maeshima A, Vaughn DA, Choi Y, Nigam SK. Activin
Shepard A, et al. Osteogenic protein-1 (bone mor- A is an endogenous inhibitor of ureteric bud outgrowth
phogenetic protein-7) reduces severity of injury from the Wolffian duct. Dev Biol. 2006;295:473–85.
after ischemic acute renal failure in rat. J Clin Invest. 81. Maeshima A, Yamashita S, Maeshima K, Kojima I,
1998;102:202–14. Nojima Y. Activin a produced by ureteric bud is a
67. Ding H, Kopple JD, Cohen A, Hirschberg differentiation factor for metanephric mesenchyme.
R. Recombinant human insulin-like growth factor- J Am Soc Nephrol. 2003;14:1523–34.
I accelerates recovery and reduces catabolism in
­ 82. Maeshima A, Zhang YQ, Furukawa M, Naruse
rats with ischemic acute renal failure. J Clin Invest. T, Kojima I. Hepatocyte growth factor induces
1993;91:2281–7. branching tubulogenesis in MDCK cells by modu-
68. Miller SB, Martin DR, Kissane J, Hammerman lating the activin-follistatin system. Kidney Int.
MR. Insulin-like growth factor I accelerates recov- 2000;58:1511–22.
ery from ischemic acute tubular necrosis in the rat. 83. Ritvos O, Tuuri T, Eramaa M, Sainio K, Hilden K,
Proc Natl Acad Sci U S A. 1992;89:11876–80. Saxen L, et al. Activin disrupts epithelial branching
69. Nakagawa T, Sasahara M, Haneda M, Kataoka H, morphogenesis in developing glandular organs of
Nakagawa H, Yagi M, et al. Role of PDGF B-chain the mouse. Mech Dev. 1995;50:229–45.
and PDGF receptors in rat tubular regeneration after 84. Maeshima A, Zhang YQ, Nojima Y, Naruse T,
acute injury. Am J Pathol. 1999;155:1689–99. Kojima I. Involvement of the activin-follistatin sys-
70. Yanagita M, Okuda T, Endo S, Tanaka M, Takahashi tem in tubular regeneration after renal ischemia in
K, Sugiyama F, et al. Uterine sensitization-­ rats. J Am Soc Nephrol. 2001;12:1685–95.
associated gene-1 (USAG-1), a novel BMP antag- 85. Gordon S, Taylor PR. Monocyte and macrophage
onist expressed in the kidney, accelerates tubular heterogeneity. Nat Rev Immunol. 2005;5:953–64.
injury. J Clin Invest. 2006;116:70–9. 86. Ricardo SD, van Goor H, Eddy AA. Macrophage
71. Imgrund M, Grone E, Grone HJ, Kretzler M, diversity in renal injury and repair. J Clin Invest.
Holzman L, Schlondorff D, et al. Re-expression of 2008;118:3522–30.
the developmental gene Pax-2 during experimen- 87. Wyburn KR, Jose MD, Wu H, Atkins RC, Chadban
tal acute tubular necrosis in mice 1. Kidney Int. SJ. The role of macrophages in allograft rejection.
1999;56:1423–31. Transplantation. 2005;80:1641–7.
7 Kidney 181

88. Kluth DC, Erwig LP, Rees AJ. Multiple fac- 103. Wilson HM, Stewart KN, Brown PA, Anegon I,
ets of macrophages in renal injury. Kidney Int. Chettibi S, Rees AJ, et al. Bone-marrow-derived
2004;66:542–57. macrophages genetically modified to produce IL-10
89. Sean Eardley K, Cockwell P. Macrophages and reduce injury in experimental glomerulonephritis.
progressive tubulointerstitial disease. Kidney Int. Mol Ther. 2002;6:710–7.
2005;68:437–55. 104. Yamagishi H, Yokoo T, Imasawa T, Mitarai T,
90. Gordon S. Alternative activation of macrophages. Kawamura T, Utsunomiya Y. Genetically modified
Nat Rev Immunol. 2003;3:23–35. bone marrow-derived vehicle cells site specifically
91. Sica A, Mantovani A. Macrophage plasticity deliver an anti-inflammatory cytokine to inflamed
and polarization: in vivo veritas. J Clin Invest. interstitium of obstructive nephropathy. J Immunol.
2012;122:787–95. 2001;166:609–16.
92. Ikezumi Y, Atkins RC, Nikolic-Paterson DJ. 105. Yokoo T, Ohashi T, Utsunomiya Y, Kojima H,
Interferon-gamma augments acute macrophage-­ Imasawa T, Kogure T, et al. Prophylaxis of antibody-­
mediated renal injury via a glucocorticoid-sensitive induced acute glomerulonephritis with genetically
mechanism. J Am Soc Nephrol. 2003;14:888–98. modified bone marrow-derived vehicle cells. Hum
93. Lee S, Huen S, Nishio H, Nishio S, Lee HK, Choi Gene Ther. 1999;10:2673–8.
BS, et al. Distinct macrophage phenotypes contrib- 106. Ferenbach DA, Ramdas V, Spencer N, Marson L,
ute to kidney injury and repair. J Am Soc Nephrol. Anegon I, Hughes J, et al. Macrophages express-
2011;22:317–26. ing heme oxygenase-1 improve renal function
94. Nishida M, Fujinaka H, Matsusaka T, Price J, Kon in ischemia/reperfusion injury. Mol Ther. 2010;
V, Fogo AB, et al. Absence of angiotensin II type 1 18:1706–13.
receptor in bone marrow-derived cells is detrimen- 107. Nigam eS, Lieberthal W. Acute renal failure. III. The
tal in the evolution of renal fibrosis. J Clin Invest. role of growth factors in the process of renal regen-
2002;110:1859–68. eration and repair. Am J Physiol Renal Physiol.
95. Lopez-Guisa JM, Cai X, Collins SJ, Yamaguchi I, 2000;279:F3–F11.
Okamura DM, Bugge TH, et al. Mannose recep- 108. Lieberthal W, Fuhro R, Andry CC, Rennke H,
tor 2 attenuates renal fibrosis. J Am Soc Nephrol. Abernathy VE, Koh JS, et al. Rapamycin impairs
2012;23:236–51. recovery from acute renal failure: role of cell-cycle
96. Menke J, Iwata Y, Rabacal WA, Basu R, Yeung YG, arrest and apoptosis of tubular cells. Am J Physiol
Humphreys BD, et al. CSF-1 signals directly to renal Renal Physiol. 2001;281:F693–706.
tubular epithelial cells to mediate repair in mice. J 109. Fresno Vara JA, Casado E, de Castro J, Cejas P,
Clin Invest. 2009;119:2330–42. Belda-Iniesta C, Gonzalez-Baron M. PI3K/Akt
97. Alikhan MA, Jones CV, Williams TM, Beckhouse signalling pathway and cancer. Cancer Treat Rev.
AG, Fletcher AL, Kett MM, et al. Colony-­ 2004;30:193–204.
stimulating factor-1 promotes kidney growth and 110. Angers S, Moon RT. Proximal events in Wnt sig-
repair via alteration of macrophage responses. Am J nal transduction. Nat Rev Mol Cell Biol. 2009;
Pathol. 2011;179:1243–56. 10:468–77.
98. Cao Q, Wang Y, Zheng D, Sun Y, Lee VW, Zheng G, 111. Grimes CA, Jope RS. The multifaceted roles of gly-
et al. IL-10/TGF-beta-modified macrophages induce cogen synthase kinase 3beta in cellular signaling.
regulatory T cells and protect against adriamycin Prog Neurobiol. 2001;65:391–426.
nephrosis. J Am Soc Nephrol. 2010;21:933–42. 112. Lin SL, Li B, Rao S, Yeo EJ, Hudson TE, Nowlin
99. Kluth DC, Ainslie CV, Pearce WP, Finlay S, Clarke BT, et al. Macrophage Wnt7b is critical for kidney
D, Anegon I, et al. Macrophages transfected with repair and regeneration. Proc Natl Acad Sci U S A.
adenovirus to express IL-4 reduce inflammation 2010;107:4194–9.
in experimental glomerulonephritis. J Immunol. 113. Doble BW, Woodgett JR. GSK-3: tricks of the
2001;166:4728–36. trade for a multi-tasking kinase. J Cell Sci.
100. Lu J, Cao Q, Zheng D, Sun Y, Wang C, Yu X, et al. 2003;116:1175–86.
Discrete functions of M2a and M2c macrophage 114. Jope RS, Johnson GV. The glamour and gloom of
subsets determine their relative efficacy in treating glycogen synthase kinase-3. Trends Biochem Sci.
chronic kidney disease. Kidney Int. 2013;84:745–55. 2004;29:95–102.
101. Riquelme P, Tomiuk S, Kammler A, Fandrich F, 115. Wang Z, Havasi A, Gall J, Bonegio R, Li Z, Mao H,
Schlitt HJ, Geissler EK, et al. IFN-gamma-induced et al. GSK3beta promotes apoptosis after renal isch-
iNOS expression in mouse regulatory macrophages emic injury. J Am Soc Nephrol. 2010;21:284–94.
prolongs allograft survival in fully immunocompe- 116. Zhou D, Li Y, Lin L, Zhou L, Igarashi P, Liu
tent recipients. Mol Ther. 2013;21:409–22. Y. Tubule-specific ablation of endogenous beta-­
102. Wilson HM, Chettibi S, Jobin C, Walbaum D, Rees catenin aggravates acute kidney injury in mice.
AJ, Kluth DC. Inhibition of macrophage nuclear Kidney Int. 2012;82:537–47.
factor-­kappaB leads to a dominant anti-­inflammatory 117. Bao H, Ge Y, Zhuang S, Dworkin LD, Liu Z, Gong
phenotype that attenuates glomerular inflammation R. Inhibition of glycogen synthase kinase-3beta pre-
in vivo. Am J Pathol. 2005;167:27–37. vents NSAID-induced acute kidney injury. Kidney
Int. 2012;81:662–73.
182 B.S. Kim and H.T. Kim

118. Salahudeen AK, Haider N, Jenkins J, Joshi M, and promote tissue repair in an injury specific fash-
Patel H, Huang H, et al. Antiapoptotic properties ion. Cell Transplant. 2013;22:423–36.
of erythropoiesis-stimulating proteins in models of 133. Duffy MM, Ritter T, Ceredig R, Griffin
cisplatin-induced acute kidney injury. Am J Physiol MD. Mesenchymal stem cell effects on T-cell effec-
Renal Physiol. 2008;294:F1354–65. tor pathways. Stem Cell Res Ther. 2011;2:34.
119. Ucero AC, Berzal S, Ocana-Salceda C, Sancho M, 134. Camussi G, Deregibus MC, Tetta C. Paracrine/
Orzaez M, Messeguer A, et al. A polymeric nano- endocrine mechanism of stem cells on kidney
medicine diminishes inflammatory events in renal repair: role of microvesicle-mediated transfer of
tubular cells. PLoS One. 2013;8:e51992. genetic information. Curr Opin Nephrol Hypertens.
120. Lewis TS, Shapiro PS, Ahn NG. Signal transduction 2010;19:7–12.
through MAP kinase cascades. Adv Cancer Res. 135. Ke YH, He JW, Fu WZ, Zhang ZL. Identification
1998;74:49–139. of two novel mutations in the OCRL1 gene in two
121. Kyriakis JM, Avruch J. Mammalian mitogen-­ Chinese families with Lowe syndrome. Nephrology
activated protein kinase signal transduction path- (Carlton). 2012;17:20–5.
ways activated by stress and inflammation. Physiol 136. Phinney DG, Prockop DJ. Concise review: mesen-
Rev. 2001;81:807–69. chymal stem/multipotent stromal cells: the state of
122. di Mari JF, Davis R, Safirstein RL. MAPK activa- transdifferentiation and modes of tissue repair--cur-
tion determines renal epithelial cell survival during rent views. Stem Cells. 2007;25:2896–902.
oxidative injury. Am J Physiol. 1999;277:F195–203. 137. Burdon TJ, Paul A, Noiseux N, Prakash S, Shum-­
123. Arany I, Megyesi JK, Nelkin BD, Safirstein Tim D. Bone marrow stem cell derived paracrine
RL. STAT3 attenuates EGFR-mediated ERK activa- factors for regenerative medicine: current perspec-
tion and cell survival during oxidant stress in mouse tives and therapeutic potential. Bone Marrow Res.
proximal tubular cells. Kidney Int. 2006;70:669–74. 2011;2011:207326.
124. Ishizuka S, Yano T, Hagiwara K, Sone M, Nihei H, 138. Li J, Deane JA, Campanale NV, Bertram JF, Ricardo
Ozasa H, et al. Extracellular signal-regulated kinase SD. The contribution of bone marrow-derived cells
mediates renal regeneration in rats with myoglo- to the development of renal interstitial fibrosis. Stem
binuric acute renal injury. Biochem Biophys Res Cells. 2007;25:697–706.
Commun. 1999;254:88–92. 139. Thirabanjasak D, Tantiwongse K, Thorner PS.
125. Ikarashi K, Li B, Suwa M, Kawamura K, Morioka T, Angiomyeloproliferative lesions following autol-
Yao J, et al. Bone marrow cells contribute to regen- ogous stem cell therapy. J Am Soc Nephrol.
eration of damaged glomerular endothelial cells. 2010;21:1218–22.
Kidney Int. 2005;67:1925–33. 140. Donizetti-Oliveira C, Semedo P, Burgos-Silva M,
126. Lin F, Moran A, Igarashi P. Intrarenal cells, not Cenedeze MA, Malheiros DM, Reis MA, et al.
bone marrow-derived cells, are the major source for Adipose tissue-derived stem cell treatment pre-
regeneration in postischemic kidney. J Clin Invest. vents renal disease progression. Cell Transplant.
2005;115:1756–64. 2012;21:1727–41.
127. Prodromidi EI, Poulsom R, Jeffery R, Roufosse CA, 141. Eirin A, Zhu XY, Krier JD, Tang H, Jordan KL,
Pollard PJ, Pusey CD, et al. Bone marrow-derived Grande JP, et al. Adipose tissue-derived mesenchy-
cells contribute to podocyte regeneration and ame- mal stem cells improve revascularization outcomes
lioration of renal disease in a mouse model of Alport to restore renal function in swine atherosclerotic
syndrome. Stem Cells. 2006;24:2448–55. renal artery stenosis. Stem Cells. 2012;30:1030–41.
128. Bruno S, Grange C, Deregibus MC, Calogero RA, 142. Kim JH, Park DJ, Yun JC, Jung MH, Yeo HD, Kim
Saviozzi S, Collino F, et al. Mesenchymal stem cell-­ HJ, et al. Human adipose tissue-derived mesen-
derived microvesicles protect against acute tubular chymal stem cells protect kidneys from cisplatin
injury. J Am Soc Nephrol. 2009;20:1053–67. nephrotoxicity in rats. Am J Physiol Renal Physiol.
129. Caplan AI, Dennis JE. Mesenchymal stem cells as tro- 2012;302:F1141–50.
phic mediators. J Cell Biochem. 2006;98:1076–84. 143. Zhu XY, Urbieta-Caceres V, Krier JD, Textor SC,
130. Tomasoni S, Longaretti L, Rota C, Morigi M, Conti Lerman A, Lerman LO. Mesenchymal stem cells and
S, Gotti E, et al. Transfer of growth factor recep- endothelial progenitor cells decrease renal injury in
tor mRNA via exosomes unravels the regenerative experimental swine renal artery stenosis through dif-
effect of mesenchymal stem cells. Stem Cells Dev. ferent mechanisms. Stem Cells. 2013;31:117–25.
2013;22:772–80. 144. de Almeida DC, Donizetti-Oliveira C, Barbosa-­
131. Wang Y, He J, Pei X, Zhao W. Systematic review and Costa P, Origassa CS, Camara NO. In search of
meta-analysis of mesenchymal stem/stromal cells mechanisms associated with mesenchymal stem
therapy for impaired renal function in small animal cell-based therapies for acute kidney injury. Clin
models. Nephrology (Carlton). 2013;18:201–8. Biochem Rev. 2013;34:131–44.
132. Xinaris C, Morigi M, Benedetti V, Imberti B, 145. Chen YT, Sun CK, Lin YC, Chang LT, Chen YL,
Fabricio AS, Squarcina E, et al. A novel strategy to Tsai TH, et al. Adipose-derived mesenchymal stem
enhance mesenchymal stem cell migration capacity cell protects kidneys against ischemia-reperfusion
7 Kidney 183

injury through suppressing oxidative stress and 160. Basu J, Ludlow JW. Developmental engineering
inflammatory reaction. J Transl Med. 2011;9:51. the kidney: leveraging principles of morphogenesis
146. Rota C, Imberti B, Pozzobon M, Piccoli M, De for renal regeneration. Birth Defects Res C Embryo
Coppi P, Atala A, et al. Human amniotic fluid stem Today. 2012;96:30–8.
cell preconditioning improves their regenerative 161. Cuppage FE, Tate A. Repair of the nephron fol-
potential. Stem Cells Dev. 2012;21:1911–23. lowing injury with mercuric chloride. Am J Pathol.
147. Song B, Niclis JC, Alikhan MA, Sakkal S, Sylvain 1967;51:405–29.
A, Kerr PG, et al. Generation of induced pluripotent 162. Oliver J. Correlations of structure and function and
stem cells from human kidney mesangial cells. J Am mechanisms of recovery in acute tubular necrosis.
Soc Nephrol. 2011;22:1213–20. Am J Med. 1953;15:535–57.
148. Song B, Smink AM, Jones CV, Callaghan JM, Firth 163. Bissell MJ, Hall HG, Parry G. How does the extra-
SD, Bernard CA, et al. The directed differentiation cellular matrix direct gene expression? J Theor Biol.
of human iPS cells into kidney podocytes. PLoS 1982;99:31–68.
One. 2012;7:e46453. 164. Vorotnikova E, McIntosh D, Dewilde A, Zhang
149. Zhou T, Benda C, Duzinger S, Huang Y, Li X, Li Y, J, Reing JE, Zhang L, et al. Extracellular matrix-­
et al. Generation of induced pluripotent stem cells derived products modulate endothelial and pro-
from urine. J Am Soc Nephrol. 2011;22:1221–8. genitor cell migration and proliferation in vitro and
150. Polo JM, Liu S, Figueroa ME, Kulalert W, Eminli stimulate regenerative healing in vivo. Matrix Biol.
S, Tan KY, et al. Cell type of origin influences 2010;29:690–700.
the molecular and functional properties of mouse 165. Bornstein P, Sage EH. Matricellular proteins: extra-
induced pluripotent stem cells. Nat Biotechnol. cellular modulators of cell function. Curr Opin Cell
2010;28:848–55. Biol. 2002;14:608–16.
151. Lee PY, Chien Y, Chiou GY, Lin CH, Chiou CH, 166. Lelongt B, Ronco P. Role of extracellular matrix in
Tarng DC. Induced pluripotent stem cells without kidney development and repair. Pediatr Nephrol.
c-Myc attenuate acute kidney injury via downregu- 2003;18:731–42.
lating the signaling of oxidative stress and inflam- 167. Lanza RP, Chung HY, Yoo JJ, Wettstein PJ,
mation in ischemia-reperfusion rats. Cell Transplant. Blackwell C, Borson N, et al. Generation of histo-
2012;21:2569–85. compatible tissues using nuclear transplantation. Nat
152. Imberti B, Tomasoni S, Ciampi O, Pezzotta A, Biotechnol. 2002;20:689–96.
Derosas M, Xinaris C, et al. Renal progenitors 168. Bergsma EJ, Rozema FR, Bos RR, de Bruijn
derived from human iPSCs engraft and restore func- WC. Foreign body reactions to resorbable poly(L-­
tion in a mouse model of acute kidney injury. Sci lactide) bone plates and screws used for the fixation
Rep. 2015;5:8826. of unstable zygomatic fractures. J Oral Maxillofac
153. Zhao T, Zhang ZN, Rong Z, Xu Y. Immunogenicity Surg. 1993;51:666–70.
of induced pluripotent stem cells. Nature. 169. Lo H, Kadiyala S, Guggino SE, Leong KW. Poly(L-­
2011;474:212–5. lactic acid) foams with cell seeding and
154. Harari-Steinberg O, Metsuyanim S, Omer D, Gnatek controlled-­release capacity. J Biomed Mater Res.
Y, Gershon R, Pri-Chen S, et al. Identification of 1996;30:475–84.
human nephron progenitors capable of generation 170. Martin C, Winet H, Bao JY. Acidity near erod-
of kidney structures and functional repair of chronic ing polylactide-polyglycolide in vitro and in vivo
renal disease. EMBO Mol Med. 2013;5:1556–68. in rabbit tibial bone chambers. Biomaterials.
155. Woolf AS, Palmer SJ, Snow ML, Fine LG. Creation 1996;17:2373–80.
of a functioning chimeric mammalian kidney. 171. Zhang R, Ma PX. Poly(alpha-hydroxyl acids)/
Kidney Int. 1990;38:991–7. hydroxyapatite porous composites for bone-tissue
156. Rogers SA, Lowell JA, Hammerman NA, Hammerman engineering. I. Preparation and morphology. J
MR. Transplantation of developing metanephroi into Biomed Mater Res. 1999;44:446–55.
adult rats. Kidney Int. 1998;54:27–37. 172. Nam YS, Yoon JJ, Park TG. A novel fabrication
157. Statter MB, Fahrner KJ, Barksdale EM Jr, Parks DE, method of macroporous biodegradable polymer
Flavell RA, Donahoe PK. Correlation of fetal kidney scaffolds using gas foaming salt as a porogen addi-
and testis congenic graft survival with reduced major tive. J Biomed Mater Res. 2000;53:1–7.
histocompatibility complex burden. Transplantation. 173. Orlando G, Baptista P, Birchall M, De Coppi P,
1989;47:651–60. Farney A, Guimaraes-Souza NK, et al. Regenerative
158. Abrahamson DR, St John PL, Pillion DJ, Tucker medicine as applied to solid organ transplantation:
DC. Glomerular development in intraocular and current status and future challenges. Transpl Int.
intrarenal grafts of fetal kidneys. Lab Invest. 2011;24:223–32.
1991;64:629–39. 174. Orlando G, Wood KJ, De Coppi P, Baptista PM,
159. Mikos AG, Herring SW, Ochareon P, Elisseeff J, Lu Binder KW, Bitar KN, et al. Regenerative medi-
HH, Kandel R, et al. Engineering complex tissues. cine as applied to general surgery. Ann Surg.
Tissue Eng. 2006;12:3307–39. 2012;255:867–80.
184 B.S. Kim and H.T. Kim

175. Orlando G, Wood KJ, Stratta RJ, Yoo JJ, Atala A, Decellularization methods of porcine kidneys for
Soker S. Regenerative medicine and organ trans- whole organ engineering using a high-throughput
plantation: past, present, and future. Transplanta- system. Biomaterials. 2012;33:7756–64.
tion. 2011;91:1310–7. 190. Oliver JA, Klinakis A, Cheema FH, Friedlander J,
176. Badylak SF, Taylor D, Uygun K. Whole-organ tissue Sampogna RV, Martens TP, et al. Proliferation and
engineering: decellularization and recellularization migration of label-retaining cells of the kidney
of three-dimensional matrix scaffolds. Annu Rev papilla. J Am Soc Nephrol. 2009;20:2315–27.
Biomed Eng. 2011;13:27–53. 191. Reule S, Gupta S. Kidney regeneration and resident
177. Crapo PM, Medberry CJ, Reing JE, Tottey S, van der stem cells. Organogenesis. 2011;7:135–9.
Merwe Y, Jones KE, et al. Biologic scaffolds com- 192. Maeshima A, Sakurai H, Nigam SK. Adult kidney
posed of central nervous system extracellular matrix. tubular cell population showing phenotypic plas-
Biomaterials. 2012;33:3539–47. ticity, tubulogenic capacity, and integration capa-
178. Badylak SF, Gilbert TW. Immune response to biologic bility into developing kidney. J Am Soc Nephrol.
scaffold materials. Semin Immunol. 2008;20:109–16. 2006;17:188–98.
179. Brown BN, Valentin JE, Stewart-Akers AM, 193. Humphreys BD, Bonventre JV. The contribution
McCabe GP, Badylak SF. Macrophage phenotype of adult stem cells to renal repair. Nephrol Ther.
and remodeling outcomes in response to biologic 2007;3:3–10.
scaffolds with and without a cellular component. 194. Vogetseder A, Karadeniz A, Kaissling B, Le Hir
Biomaterials. 2009;30:1482–91. M. Tubular cell proliferation in the healthy rat kid-
180. Brown B, Lindberg K, Reing J, Stolz DB, Badylak ney. Histochem Cell Biol. 2005;124:97–104.
SF. The basement membrane component of biologic 195. Challen GA, Little MH. A side order of stem cells:
scaffolds derived from extracellular matrix. Tissue the SP phenotype. Stem Cells. 2006;24:3–12.
Eng. 2006;12:519–26. 196. Iwatani H, Ito T, Imai E, Matsuzaki Y, Suzuki A,
181. Reing JE, Brown BN, Daly KA, Freund JM, Gilbert Yamato M, et al. Hematopoietic and nonhemato-
TW, Hsiong SX, et al. The effects of processing poietic potentials of Hoechst(low)/side population
methods upon mechanical and biologic properties cells isolated from adult rat kidney. Kidney Int.
of porcine dermal extracellular matrix scaffolds. 2004;65:1604–14.
Biomaterials. 2010;31:8626–33. 197. Hishikawa K, Marumo T, Miura S, Nakanishi A,
182. Ott HC, Matthiesen TS, Goh SK, Black LD, Kren Matsuzaki Y, Shibata K, et al. Musculin/MyoR is
SM, Netoff TI, et al. Perfusion-decellularized expressed in kidney side population cells and can
matrix: using nature's platform to engineer a bioarti- regulate their function. J Cell Biol. 2005;169:921–8.
ficial heart. Nat Med. 2008;14:213–21. 198. Hristov M, Erl W, Weber PC. Endothelial progeni-
183. Baptista PM, Siddiqui MM, Lozier G, Rodriguez tor cells: mobilization, differentiation, and homing.
SR, Atala A, Soker S. The use of whole organ Arterioscler Thromb Vasc Biol. 2003;23:1185–9.
­decellularization for the generation of a vascularized 199. Bidlingmaier S, Zhu X, Liu B. The utility and
liver organoid. Hepatology. 2011;53:604–17. limitations of glycosylated human CD133 epitopes
184. Song JJ, Kim SS, Liu Z, Madsen JC, Mathisen in defining cancer stem cells. J Mol Med (Berl).
DJ, Vacanti JP, et al. Enhanced in vivo function of 2008;86:1025–32.
bioartificial lungs in rats. J Biomed Mater Res A. 200. Dekel B, Zangi L, Shezen E, Reich-Zeliger S,
2011;92:998–1005. discussion-6 Eventov-Friedman S, Katchman H, et al. Isolation
185. Loai Y, Yeger H, Coz C, Antoon R, Islam SS, and characterization of nontubular sca-1+lin- mul-
Moore K, et al. Bladder tissue engineering: tissue tipotent stem/progenitor cells from adult mouse kid-
regeneration and neovascularization of HA-VEGF-­ ney. J Am Soc Nephrol. 2006;17:3300–14.
incorporated bladder acellular constructs in mouse 201. Bertoncello I, Williams B. Hematopoietic stem cell
and porcine animal models. J Biomed Mater Res A. characterization by Hoechst 33342 and rhodamine
2010;94:1205–15. 123 staining. Methods Mol Biol. 2004;263:181–200.
186. Ross EA, Williams MJ, Hamazaki T, Terada N, 202. Gupta S, Verfaillie C, Chmielewski D, Kren S,
Clapp WL, Adin C, et al. Embryonic stem cells pro- Eidman K, Connaire J, et al. Isolation and charac-
liferate and differentiate when seeded into kidney terization of kidney-derived stem cells. J Am Soc
scaffolds. J Am Soc Nephrol. 2009;20:2338–47. Nephrol. 2006;17:3028–40.
187. Orlando G, Farney AC, Iskandar SS, Mirmalek-Sani 203. Lasagni L, Romagnani P. Glomerular epithelial stem
SH, Sullivan DC, Moran E, et al. Production and cells: the good, the bad, and the ugly. J Am Soc
implantation of renal extracellular matrix scaffolds Nephrol. 2010;21:1612–9.
from porcine kidneys as a platform for renal bioengi- 204. Ronconi E, Sagrinati C, Angelotti ML, Lazzeri E,
neering investigations. Ann Surg. 2012;256:363–70. Mazzinghi B, Ballerini L, et al. Regeneration of
188. Nakayama KH, Batchelder CA, Lee CI, Tarantal glomerular podocytes by human renal progenitors. J
AF. Decellularized rhesus monkey kidney as a three-­ Am Soc Nephrol. 2009;20:322–32.
dimensional scaffold for renal tissue engineering. 205. Appel D, Kershaw DB, Smeets B, Yuan G, Fuss
Tissue Eng Part A. 2010;16:2207–16. A, Frye B, et al. Recruitment of podocytes from
189. Sullivan DC, Mirmalek-Sani SH, Deegan DB, glomerular parietal epithelial cells. J Am Soc
Baptista PM, Aboushwareb T, Atala A, et al. Nephrol. 2009;20:333–43.
7 Kidney 185

206. Eirin A, Lerman LO. Mesenchymal stem cell treat- 221. Simerman AA, Dumesic DA, Chazenbalk
ment for chronic renal failure. Stem Cell Res Ther. GD. Pluripotent muse cells derived from human adi-
2014;5:83. pose tissue: a new perspective on regenerative medi-
207. Rosenberg ME. Cell-based therapies in kidney dis- cine and cell therapy. Clin Transl Med. 2014;3:12.
ease. Kidney Int Suppl. 2011;3:364–7. 222. Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE,
208. Herzlinger D, Koseki C, Mikawa T, al-Awqati Keene CD, Ortiz-Gonzalez XR, et al. Pluripotency
Q. Metanephric mesenchyme contains multipotent of mesenchymal stem cells derived from adult mar-
stem cells whose fate is restricted after induction. row. Nature. 2002;418:41–9.
Development. 1992;114:565–72. 223. Hayakawa M, Ishizaki M, Hayakawa J, Migita M,
209. Hartman HA, Lai HL, Patterson LT. Cessation of renal Murakami M, Shimada T, et al. Role of bone marrow
morphogenesis in mice. Dev Biol. 2007;310:379–87. cells in the healing process of mouse experimental
210. Strutz F, Zeisberg M, Renziehausen A, Raschke B, glomerulonephritis. Pediatr Res. 2005;58:323–8.
Becker V, van Kooten C, et al. TGF-beta 1 induces 224. Kale S, Karihaloo A, Clark PR, Kashgarian M,
proliferation in human renal fibroblasts via induction Krause DS, Cantley LG. Bone marrow stem cells
of basic fibroblast growth factor (FGF-2). Kidney contribute to repair of the ischemically injured renal
Int. 2001;59:579–92. tubule. J Clin Invest. 2003;112:42–9.
211. Phillips AO, Steadman R. Diabetic nephropathy: 225. Rookmaaker MB, Smits AM, Tolboom H, Van’t
the central role of renal proximal tubular cells Wout K, Martens AC, Goldschmeding R, et al.
in tubulointerstitial injury. Histol Histopathol. Bone-marrow-derived cells contribute to glomerular
2002;17:247–52. endothelial repair in experimental glomerulonephri-
212. Helbert MJ, Dauwe S, De Broe ME. Flow cytomet- tis. Am J Pathol. 2003;163:553–62.
ric immunodissection of the human nephron in vivo 226. Sugimoto H, Mundel TM, Sund M, Xie L, Cosgrove
and in vitro. Exp Nephrol. 1999;7:360–76. D, Kalluri R. Bone-marrow-derived stem cells repair
213. Cummings BS, Lasker JM, Lash LH. Expression basement membrane collagen defects and reverse
of glutathione-dependent enzymes and cytochrome genetic kidney disease. Proc Natl Acad Sci U S A.
P450s in freshly isolated and primary cultures 2006;103:7321–6.
of proximal tubular cells from human kidney. J 227. Herrera MB, Bussolati B, Bruno S, Fonsato V,
Pharmacol Exp Ther. 2000;293:677–85. Romanazzi GM, Camussi G. Mesenchymal stem
214. Qi W, Johnson DW, Vesey DA, Pollock CA, Chen cells contribute to the renal repair of acute tubular
X. Isolation, propagation and characterization of epithelial injury. Int J Mol Med. 2004;14:1035–41.
primary tubule cell culture from human kidney. 228. Morigi M, Imberti B, Zoja C, Corna D, Tomasoni S,
Nephrology (Carlton). 2007;12:155–9. Abbate M, et al. Mesenchymal stem cells are reno-
215. Presnell SC, Bruce AT, Wallace SM, Choudhury S, tropic, helping to repair the kidney and improve
Genheimer CW, Cox B, et al. Isolation, character- function in acute renal failure. J Am Soc Nephrol.
ization, and expansion methods for defined primary 2004;15:1794–804.
renal cell populations from rodent, canine, and 229. Wong CY, Cheong SK, Mok PL, Leong
human normal and diseased kidneys. Tissue Eng CF. Differentiation of human mesenchymal stem
Part C Methods. 2011;17:261–73. cells into mesangial cells in post-glomerular injury
216. Baer PC, Geiger H. Human renal cells from the thick murine model. Pathology. 2008;40:52–7.
ascending limb and early distal tubule: characteriza- 230. Togel F, Weiss K, Yang Y, Hu Z, Zhang P,
tion of primary isolated and cultured cells by reverse Westenfelder C. Vasculotropic, paracrine actions of
transcription polymerase chain reaction. Nephrology infused mesenchymal stem cells are important to
(Carlton). 2008;13:316–21. the recovery from acute kidney injury. Am J Physiol
217. Aggarwal S, Moggio A, Bussolati B. Concise Renal Physiol. 2007;292:F1626–35.
review: stem/progenitor cells for renal tissue repair: 231. Chou YH, Pan SY, Yang CH, Lin SL. Stem cells
current knowledge and perspectives. Stem Cells and kidney regeneration. J Formos Med Assoc.
Transl Med. 2013;2:1011–9. 2014;113:201–9.
218. Kitamura S, Sakurai H, Makino H. Single adult 232. Terada N, Hamazaki T, Oka M, Hoki M, Mastalerz
kidney stem/progenitor cells reconstitute three-­ DM, Nakano Y, et al. Bone marrow cells adopt the
dimensional nephron structures in vitro. Stem Cells. phenotype of other cells by spontaneous cell fusion.
2014;33:774–84. Nature. 2002;416:542–5.
219. Bruno S, Bussolati B, Grange C, Collino F,
233. Alvarez-Dolado M, Pardal R, Garcia-Verdugo JM,
di Cantogno LV, Herrera MB, et al. Isolation Fike JR, Lee HO, Pfeffer K, et al. Fusion of bone-mar-
and characterization of resident mesenchymal row-derived cells with Purkinje neurons, cardiomyo-
stem cells in human glomeruli. Stem Cells Dev. cytes and hepatocytes. Nature. 2003;425:968–73.
2009;18:867–80. 234. Togel FE, Westenfelder C. Mesenchymal stem cells:
220. Wang H, Gomez JA, Klein S, Zhang Z, Seidler B, a new therapeutic tool for AKI. Nat Rev Nephrol.
Yang Y, et al. Adult renal mesenchymal stem cell-­like 2010;6:179–83.
cells contribute to juxtaglomerular cell recruitment. J 235. Perico N, Casiraghi F, Introna M, Gotti E, Todeschini
Am Soc Nephrol. 2013;24:1263–73. M, Cavinato RA, et al. Autologous mesenchymal
186 B.S. Kim and H.T. Kim

stromal cells and kidney transplantation: a pilot 248. Hauser PV, De Fazio R, Bruno S, Sdei S, Grange C,
study of safety and clinical feasibility. Clin J Am Soc Bussolati B, et al. Stem cells derived from human
Nephrol. 2011;6:412–22. amniotic fluid contribute to acute kidney injury
236. Tan J, Wu W, Xu X, Liao L, Zheng F, Messinger recovery. Am J Pathol. 2010;177:2011–21.
S, et al. Induction therapy with autologous mesen- 249. Perin L, Sedrakyan S, Giuliani S, Da Sacco S,
chymal stem cells in living-related kidney trans- Carraro G, Shiri L, et al. Protective effect of human
plants: a randomized controlled trial. JAMA. amniotic fluid stem cells in an immunodeficient
2012;307:1169–77. mouse model of acute tubular necrosis. PLoS One.
237. Casiraghi F, Perico N, Remuzzi G. Mesenchymal 2010;5:e9357.
stromal cells to promote solid organ transplan- 250. Sedrakyan S, Da Sacco S, Milanesi A, Shiri L,
tation tolerance. Curr Opin Organ Transplant. Petrosyan A, Varimezova R, et al. Injection of amni-
2013;18:51–8. otic fluid stem cells delays progression of renal
238. Reinders ME, de Fijter JW, Roelofs H, Bajema IM, fibrosis. J Am Soc Nephrol. 2012;23:661–73.
de Vries DK, Schaapherder AF, et al. Autologous 251. Sun D, Bu L, Liu C, Yin Z, Zhou X, Li X, et al.
bone marrow-derived mesenchymal stromal cells Therapeutic effects of human amniotic fluid-derived
for the treatment of allograft rejection after renal stem cells on renal interstitial fibrosis in a murine
transplantation: results of a phase I study. Stem Cells model of unilateral ureteral obstruction. PLoS One.
Transl Med. 2013;2:107–11. 2013;8:e65042.
239. Lee RH, Pulin AA, Seo MJ, Kota DJ, Ylostalo J, 252. Da Sacco S, Lemley KV, Sedrakyan S, Zanusso I,
Larson BL, et al. Intravenous hMSCs improve myo- Petrosyan A, Peti-Peterdi J, et al. A novel source of
cardial infarction in mice because cells embolized in cultured podocytes. PLoS One. 2013;8:e81812.
lung are activated to secrete the anti-inflammatory 253. Siegel N, Rosner M, Unbekandt M, Fuchs C, Slabina
protein TSG-6. Cell Stem Cell. 2009;5:54–63. N, Dolznig H, et al. Contribution of human amniotic
240. Ebrahimi B, Eirin A, Li Z, Zhu XY, Zhang X, Lerman fluid stem cells to renal tissue formation depends on
A, et al. Mesenchymal stem cells improve medullary mTOR. Hum Mol Genet. 2010;19:3320–31.
inflammation and fibrosis after r­evascularization of 254. Patschan D, Krupincza K, Patschan S, Zhang Z,
swine atherosclerotic renal artery stenosis. PLoS Hamby C, Goligorsky MS. Dynamics of mobilization
One. 2013;8:e67474. and homing of endothelial progenitor cells after acute
241. Katsuno T, Ozaki T, Saka Y, Furuhashi K, Kim H, renal ischemia: modulation by ischemic precondition-
Yasuda K, et al. Low serum cultured adipose tissue-­ ing. Am J Physiol Renal Physiol. 2006;291:F176–85.
derived stromal cells ameliorate acute kidney injury 255. Patschan D, Patschan S, Gobe GG, Chintala S,
in rats. Cell Transplant. 2013;22:287–97. Goligorsky MS. Uric acid heralds ischemic tissue
242. Furuhashi K, Tsuboi N, Shimizu A, Katsuno T, Kim injury to mobilize endothelial progenitor cells. J Am
H, Saka Y, et al. Serum-starved adipose-derived stro- Soc Nephrol. 2007;18:1516–24.
mal cells ameliorate crescentic GN by promoting 256. Asahara T, Murohara T, Sullivan A, Silver M, van
immunoregulatory macrophages. J Am Soc Nephrol. der Zee R, Li T, et al. Isolation of putative pro-
2013;24:587–603. genitor endothelial cells for angiogenesis. Science.
243. Morigi M, Rota C, Montemurro T, Montelatici E, 1997;275:964–7.
Lo Cicero V, Imberti B, et al. Life-sparing effect 257. Chade AR, Zhu X, Lavi R, Krier JD, Pislaru S,
of human cord blood-mesenchymal stem cells Simari RD, et al. Endothelial progenitor cells restore
in experimental acute kidney injury. Stem Cells. renal function in chronic experimental renovascular
2010;28:513–22. disease. Circulation. 2009;119:547–57.
244. Shalaby RH, Rashed LA, Ismaail AE, Madkour 258. Chade AR, Zhu XY, Krier JD, Jordan KL, Textor
NK, Elwakeel SH. Hematopoietic stem cells derived SC, Grande JP, et al. Endothelial progenitor cells
from human umbilical cord ameliorate cisplatin-­ homing and renal repair in experimental renovascu-
induced acute renal failure in rats. Am J Stem Cells. lar disease. Stem Cells. 2010;28:1039–47.
2014;3:83–96. 259. Jie KE, Zaikova MA, Bergevoet MW, Westerweel
245. Panepucci RA, Siufi JL, Silva WA Jr, Proto-Siquiera PE, Rastmanesh M, Blankestijn PJ, et al. Progenitor
R, Neder L, Orellana M, et al. Comparison of gene cells and vascular function are impaired in
expression of umbilical cord vein and bone marrow-­ patients with chronic kidney disease. Nephrol Dial
derived mesenchymal stem cells. Stem Cells. Transplant. 2010;25:1875–82.
2004;22:1263–78. 260. Evans MJ, Kaufman MH. Establishment in culture
246. De Coppi P, Bartsch G Jr, Siddiqui MM, Xu T, of pluripotential cells from mouse embryos. Nature.
Santos CC, Perin L, et al. Isolation of amniotic stem 1981;292:154–6.
cell lines with potential for therapy. Nat Biotechnol. 261. Takahashi K, Yamanaka S. Induction of pluripotent
2007;25:100–6. stem cells from mouse embryonic and adult fibroblast
247. Perin L, Giuliani S, Jin D, Sedrakyan S, Carraro G, cultures by defined factors. Cell. 2006;126:663–76.
Habibian R, et al. Renal differentiation of amniotic 262. Takahashi K, Tanabe K, Ohnuki M, Narita M,
fluid stem cells. Cell Prolif. 2007;40:936–48. Ichisaka T, Tomoda K, et al. Induction of pluripotent
7 Kidney 187

stem cells from adult human fibroblasts by defined embryonic stem cell differentiation towards a renal
factors. Cell. 2007;131:861–72. lineage generates a self-organizing kidney. Nat Cell
263. Basma H, Soto-Gutierrez A, Yannam GR, Liu L, Ito Biol. 2014;16:118–26.
R, Yamamoto T, et al. Differentiation and transplan- 276. Xia Y, Nivet E, Sancho-Martinez I, Gallegos T,
tation of human embryonic stem cell-derived hepa- Suzuki K, Okamura D, et al. Directed differ-
tocytes. Gastroenterology. 2009;136:990–9. entiation of human pluripotent cells to ureteric
264. Chambers SM, Fasano CA, Papapetrou EP, bud kidney progenitor-­ like cells. Nat Cell Biol.
Tomishima M, Sadelain M, Studer L. Highly effi- 2013;15:1507–15.
cient neural conversion of human ES and iPS cells by 277. Taguchi A, Kaku Y, Ohmori T, Sharmin S, Ogawa
dual inhibition of SMAD signaling. Nat Biotechnol. M, Sasaki H, et al. Redefining the in vivo origin of
2009;27:275–80. metanephric nephron progenitors enables generation
265. D'Amour KA, Bang AG, Eliazer S, Kelly OG, of complex kidney structures from pluripotent stem
Agulnick AD, Smart NG, et al. Production of pan- cells. Cell Stem Cell. 2014;14:53–67.
creatic hormone-expressing endocrine cells from 278. Cai J, Zhao Y, Liu Y, Ye F, Song Z, Qin H, et al.
human embryonic stem cells. Nat Biotechnol. Directed differentiation of human embryonic stem
2006;24:1392–401. cells into functional hepatic cells. Hepatology.
266. Dhara SK, Stice SL. Neural differentiation of 2007;45:1229–39.
human embryonic stem cells. J Cell Biochem. 2008; 279. Wichterle H, Lieberam I, Porter JA, Jessell
105:633–40. TM. Directed differentiation of embryonic stem
267. Hay DC, Zhao D, Fletcher J, Hewitt ZA, McLean D, cells into motor neurons. Cell. 2002;110:385–97.
Urruticoechea-Uriguen A, et al. Efficient differen- 280. Laflamme MA, Chen KY, Naumova AV, Muskheli
tiation of hepatocytes from human embryonic stem V, Fugate JA, Dupras SK, et al. Cardiomyocytes
cells exhibiting markers recapitulating liver develop- derived from human embryonic stem cells in pro-­
ment in vivo. Stem Cells. 2008;26:894–902. survival factors enhance function of infarcted rat
268. Ledran MH, Krassowska A, Armstrong L, Dimmick hearts. Nat Biotechnol. 2007;25:1015–24.
I, Renstrom J, Lang R, et al. Efficient hematopoi- 281. Martin GR. Isolation of a pluripotent cell line from
etic differentiation of human embryonic stem cells early mouse embryos cultured in medium condi-
on stromal cells derived from hematopoietic niches. tioned by teratocarcinoma stem cells. Proc Natl
Cell Stem Cell. 2008;3:85–98. Acad Sci U S A. 1981;78:7634–8.
269. Takahashi T, Lord B, Schulze PC, Fryer RM, Sarang 282. Osafune K. In vitro regeneration of kidney from plu-
SS, Gullans SR, et al. Ascorbic acid enhances differ- ripotent stem cells. Exp Cell Res. 2010;316:2571–7.
entiation of embryonic stem cells into cardiac myo- 283. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz
cytes. Circulation. 2003;107:1912–6. MA, Swiergiel JJ, Marshall VS, et al. Embryonic
270. Zhang D, Jiang W, Liu M, Sui X, Yin X, Chen S, stem cell lines derived from human blastocysts.
et al. Highly efficient differentiation of human ES Science. 1998;282:1145–7.
cells and iPS cells into mature pancreatic insulin-­ 284. Reubinoff BE, Pera MF, Fong CY, Trounson A,
producing cells. Cell Res. 2009;19:429–38. Bongso A. Embryonic stem cell lines from human
271. Spence JR, Mayhew CN, Rankin SA, Kuhar MF, blastocysts: somatic differentiation in vitro. Nat
Vallance JE, Tolle K, et al. Directed differentiation Biotechnol. 2000;18:399–404.
of human pluripotent stem cells into intestinal tissue 285. Schuldiner M, Yanuka O, Itskovitz-Eldor J, Melton
in vitro. Nature. 2011;470:105–9. DA, Benvenisty N. Effects of eight growth factors
272. Gadue P, Huber TL, Paddison PJ, Keller GM. Wnt on the differentiation of cells derived from human
and TGF-beta signaling are required for the induc- embryonic stem cells. Proc Natl Acad Sci U S A.
tion of an in vitro model of primitive streak forma- 2000;97:11307–12.
tion using embryonic stem cells. Proc Natl Acad Sci 286. Kobayashi T, Tanaka H, Kuwana H, Inoshita S,
U S A. 2006;103:16806–11. Teraoka H, Sasaki S, et al. Wnt4-transformed
273. Lam AQ, Freedman BS, Morizane R, Lerou PH, mouse embryonic stem cells differentiate into renal
Valerius MT, Bonventre JV. Rapid and efficient dif- tubular cells. Biochem Biophys Res Commun.
ferentiation of human pluripotent stem cells into 2005;336:585–95.
intermediate mesoderm that forms tubules express- 287. Mae S, Shirasawa S, Yoshie S, Sato F, Kanoh Y,
ing kidney proximal tubular markers. J Am Soc Ichikawa H, et al. Combination of small molecules
Nephrol. 2014;25:1211–25. enhances differentiation of mouse embryonic stem
274. Mae S, Shono A, Shiota F, Yasuno T, Kajiwara M, cells into intermediate mesoderm through BMP7-­
Gotoda-Nishimura N, et al. Monitoring and robust positive cells. Biochem Biophys Res Commun.
induction of nephrogenic intermediate mesoderm 2010;393:877–82.
from human pluripotent stem cells. Nat Commun. 288. Steenhard BM, Isom KS, Cazcarro P, Dunmore JH,
2013;4:1367. Godwin AR, St John PL, et al. Integration of embry-
275. Takasato M, Er PX, Becroft M, Vanslambrouck JM, onic stem cells in metanephric kidney organ culture.
Stanley EG, Elefanty AG, et al. Directing human J Am Soc Nephrol. 2005;16:1623–31.
188 B.S. Kim and H.T. Kim

289. Kim D, Dressler GR. Nephrogenic factors promote 304. Nakano T, Ando S, Takata N, Kawada M, Muguruma
differentiation of mouse embryonic stem cells into K, Sekiguchi K, et al. Self-formation of optic cups
renal epithelia. J Am Soc Nephrol. 2005;16:3527–34. and storable stratified neural retina from human
290. Yamamoto M, Cui L, Johkura K, Asanuma K, ESCs. Cell Stem Cell. 2012;10:771–85.
Okouchi Y, Ogiwara N, et al. Branching ducts simi- 305. Osafune K, Takasato M, Kispert A, Asashima
lar to mesonephric ducts or ureteric buds in terato- M, Nishinakamura R. Identification of multipo-
mas originating from mouse embryonic stem cells. tent progenitors in the embryonic mouse kidney
Am J Physiol Renal Physiol. 2006;290:F52–60. by a novel colony-forming assay. Development.
291. Esteban MA, Xu J, Yang J, Peng M, Qin D, Li W, 2006;133:151–61.
et al. Generation of induced pluripotent stem cell 306. Unbekandt M, Davies JA. Dissociation of embryonic
lines from Tibetan miniature pig. J Biol Chem. kidneys followed by reaggregation allows the forma-
2009;284:17634–40. tion of renal tissues. Kidney Int. 2010;77:407–16.
292. Ezashi T, Telugu BP, Alexenko AP, Sachdev S, Sinha 307. Chen J, Lansford R, Stewart V, Young F, Alt
S, Roberts RM. Derivation of induced pluripotent FW. RAG-2-deficient blastocyst complementation:
stem cells from pig somatic cells. Proc Natl Acad an assay of gene function in lymphocyte develop-
Sci U S A. 2009;106:10993–8. ment. Proc Natl Acad Sci U S A. 1993;90:4528–32.
293. Honda A, Hirose M, Hatori M, Matoba S, Miyoshi 308. Espejel S, Roll GR, McLaughlin KJ, Lee AY, Zhang
H, Inoue K, et al. Generation of induced pluripotent JY, Laird DJ, et al. Induced pluripotent stem cell-­
stem cells in rabbits: potential experimental mod- derived hepatocytes have the functional and prolif-
els for human regenerative medicine. J Biol Chem. erative capabilities needed for liver regeneration in
2010;285:31362–9. mice. J Clin Invest. 2010;120:3120–6.
294. Li W, Wei W, Zhu S, Zhu J, Shi Y, Lin T, et al. 309. Fraidenraich D, Stillwell E, Romero E, Wilkes D,
Generation of rat and human induced pluripotent Manova K, Basson CT, et al. Rescue of cardiac
stem cells by combining genetic reprogramming and defects in id knockout embryos by injection of
chemical inhibitors. Cell Stem Cell. 2009;4:16–9. embryonic stem cells. Science. 2004;306:247–52.
295. Liao J, Cui C, Chen S, Ren J, Chen J, Gao Y, et al. 310. Kobayashi T, Yamaguchi T, Hamanaka S, Kato-Itoh
Generation of induced pluripotent stem cell lines M, Yamazaki Y, Ibata M, et al. Generation of rat pan-
from adult rat cells. Cell Stem Cell. 2009;4:11–5. creas in mouse by interspecific blastocyst injection
296. Liu H, Zhu F, Yong J, Zhang P, Hou P, Li H, et al. of pluripotent stem cells. Cell. 2010;142:787–99.
Generation of induced pluripotent stem cells from 311. Matsunari H, Nagashima H, Watanabe M, Umeyama
adult rhesus monkey fibroblasts. Cell Stem Cell. K, Nakano K, Nagaya M, et al. Blastocyst comple-
2008;3:587–90. mentation generates exogenic pancreas in vivo in
297. Montserrat N, Ramirez-Bajo MJ, Xia Y, Sancho-­ apancreatic cloned pigs. Proc Natl Acad Sci U S A.
Martinez I, Moya-Rull D, Miquel-Serra L, et al. 2013;110:4557–62.
Generation of induced pluripotent stem cells from 312. Muller SM, Terszowski G, Blum C, Haller C, Anquez
human renal proximal tubular cells with only two V, Kuschert S, et al. Gene targeting of VEGF-A in thy-
transcription factors, OCT4 and SOX2. J Biol Chem. mus epithelium disrupts thymus blood vessel architec-
2012;287:24131–8. ture. Proc Natl Acad Sci U S A. 2005;102:10587–92.
298. Kim K, Doi A, Wen B, Ng K, Zhao R, Cahan P, et al. 313. Ueno H, Turnbull BB, Weissman IL. Two-step oli-
Epigenetic memory in induced pluripotent stem goclonal development of male germ cells. Proc Natl
cells. Nature. 2010;467:285–90. Acad Sci U S A. 2009;106:175–80.
299. Zhou T, Benda C, Dunzinger S, Huang Y, Ho JC, Yang 314. Ueno H, Weissman IL. Clonal analysis of mouse
J, et al. Generation of human induced pluripotent stem development reveals a polyclonal origin for yolk sac
cells from urine samples. Nat Protoc. 2012;7:2080–9. blood islands. Dev Cell. 2006;11:519–33.
300. Okita K, Nagata N, Yamanaka S. Immunogenicity 315. Usui J, Kobayashi T, Yamaguchi T, Knisely AS,
of induced pluripotent stem cells. Circ Res. Nishinakamura R, Nakauchi H. Generation of kid-
2011;109:720–1. ney from pluripotent stem cells via blastocyst com-
301. Eiraku M, Takata N, Ishibashi H, Kawada M, plementation. Am J Pathol. 2012;180:2417–26.
Sakakura E, Okuda S, et al. Self-organizing optic-­ 316. Dekel B, Burakova T, Arditti FD, Reich-Zeliger S,
cup morphogenesis in three-dimensional culture. Milstein O, Aviel-Ronen S, et al. Human and porcine
Nature. 2011;472:51–6. early kidney precursors as a new source for trans-
302. Eiraku M, Watanabe K, Matsuo-Takasaki M, plantation. Nat Med. 2003;9:53–60.
Kawada M, Yonemura S, Matsumura M, et al. Self-­ 317. Hammerman MR. Renal organogenesis from trans-
organized formation of polarized cortical tissues planted metanephric primordia. J Am Soc Nephrol.
from ESCs and its active manipulation by extrinsic 2004;15:1126–32.
signals. Cell Stem Cell. 2008;3:519–32. 318. Matsumoto K, Yokoo T, Matsunari H, Iwai S, Yokote
303. Suga H, Kadoshima T, Minaguchi M, Ohgushi M, S, Teratani T, et al. Xenotransplanted embryonic
Soen M, Nakano T, et al. Self-formation of func- kidney provides a niche for endogenous mesenchy-
tional adenohypophysis in three-dimensional cul- mal stem cell differentiation into erythropoietin-­
ture. Nature. 2011;480:57–62. producing tissue. Stem Cells. 2012;30:1228–35.
7 Kidney 189

319. Matsumoto K, Yokoo T, Yokote S, Utsunomiya Y, 334. Yokoo T, Fukui A, Ohashi T, Miyazaki Y,
Ohashi T, Hosoya T. Functional development of a Utsunomiya Y, Kawamura T, et al. Xenobiotic kid-
transplanted embryonic kidney: effect of transplan- ney organogenesis from human mesenchymal stem
tation site. J Nephrol. 2012;25:50–5. cells using a growing rodent embryo. J Am Soc
320. Rogers SA, Talcott M, Hammerman Nephrol. 2006;17:1026–34.
MR. Transplantation of pig metanephroi. ASAIO J. 335. Yokoo T, Ohashi T, Shen JS, Sakurai K, Miyazaki
2003;49:48–52. Y, Utsunomiya Y, et al. Human mesenchymal stem
321. Sariola H, Ekblom P, Lehtonen E, Saxen cells in rodent whole-embryo culture are repro-
L. Differentiation and vascularization of the meta- grammed to contribute to kidney tissues. Proc Natl
nephric kidney grafted on the chorioallantoic mem- Acad Sci U S A. 2005;102:3296–300.
brane. Dev Biol. 1983;96:427–35. 336. Yokoo T, Fukui A, Matsumoto K, Ohashi T, Sado
322. Xinaris C, Benedetti V, Rizzo P, Abbate M, Corna D, Y, Suzuki H, et al. Generation of a transplantable
Azzollini N, et al. In vivo maturation of functional erythropoietin-­producer derived from human mesen-
renal organoids formed from embryonic cell suspen- chymal stem cells. Transplantation. 2008;85:1654–8.
sions. J Am Soc Nephrol. 2012;23:1857–68. 337. Davies JA, Fisher CE. Genes and proteins in renal
323. Yokote S, Yokoo T, Matsumoto K, Ohkido I, development. Exp Nephrol. 2002;10:102–13.
Utsunomiya Y, Kawamura T, et al. Metanephros 338. Lipschutz JH. Molecular development of the kidney:
transplantation inhibits the progression of vascular a review of the results of gene disruption studies. Am
calcification in rats with adenine-induced renal fail- J Kidney Dis. 1998;31:383–97.
ure. Nephron Exp Nephrol. 2012;120:e32–40. 339. Gheisari Y, Yokoo T, Matsumoto K, Fukui A,
324. Yokote S, Yokoo T, Matsumoto K, Utsunomiya Y, Sugimoto N, Ohashi T, et al. A thermoreversible
Kawamura T, Hosoya T. The effect of metaneph- polymer mediates controlled release of glial cell
ros transplantation on blood pressure in anephric line-derived neurotrophic factor to enhance kidney
rats with induced acute hypotension. Nephrol Dial regeneration. Artif Organs. 2010;34:642–7.
Transplant. 2012;27:3449–55. 340. Fukui A, Yokoo T, Matsumoto K, Kawamura T,
325. Dekel B, Marcus H, Herzel BH, Bocher WO, Passwell Hosoya T, Okabe M. Integration of human mes-
JH, Reisner Y. In vivo modulation of the allogeneic enchymal stem cells into the Wolffian duct in
immune response by human fetal kidneys: the role of chicken embryos. Biochem Biophys Res Commun.
cytokines, chemokines, and cytolytic effector mol- 2009;385:330–5.
ecules. Transplantation. 2000;69:1470–8. 341. Pino CJ, Humes HD. Stem cell technology for the
326. Steer DL, Nigam SK. Developmental approaches treatment of acute and chronic renal failure. Transl
to kidney tissue engineering. Am J Physiol Renal Res. 2010;156:161–8.
Physiol. 2004;286:F1–7. 342. Fissell WH, Fleischman AJ, Humes HD, Roy S.
327. Sakurai H, Barros EJ, Tsukamoto T, Barasch J, Development of continuous implantable renal replace-
Nigam SK. An in vitro tubulogenesis system using ment: past and future. Transl Res. 2007;150:327–36.
cell lines derived from the embryonic kidney shows 343. Tasnim F, Deng R, Hu M, Liour S, Li Y, Ni M,
dependence on multiple soluble growth factors. Proc et al. Achievements and challenges in bioartificial
Natl Acad Sci U S A. 1997;94:6279–84. kidney development. Fibrogenesis Tissue Repair.
328. Machiguchi T, Nakamura T. Cellular interactions 2010;3:14.
via conditioned media induce in vivo nephron gen- 344. Aebischer P, Ip TK, Panol G, Galletti PM. The bio-
eration from tubular epithelial cells or mesenchy- artificial kidney: progress towards an ultrafiltration
mal stem cells. Biochem Biophys Res Commun. device with renal epithelial cells processing. Life
2013;435:327–33. Support Syst. 1987;5:159–68.
329. Auerbach R, Grobstein C. Inductive interaction of 345. Humes HD, Weitzel WF, Bartlett RH, Swaniker FC,
embryonic tissues after dissociation and reaggrega- Paganini EP, Luderer JR, et al. Initial clinical results
tion. Exp Cell Res. 1958;15:384–97. of the bioartificial kidney containing human cells in
330. Ganeva V, Unbekandt M, Davies JA. An improved ICU patients with acute renal failure. Kidney Int.
kidney dissociation and reaggregation culture system 2004;66:1578–88.
results in nephrons arranged organotypically around 346. MacKay SM, Funke AJ, Buffington DA, Humes
a single collecting duct system. Organogenesis. HD. Tissue engineering of a bioartificial renal
2011;7:83–7. tubule. ASAIO J. 1998;44:179–83.
331. D’Agati VD. Growing new kidneys from embry- 347. Nikolovski J, Gulari E, Humes HD. Design engi-
onic cell suspensions: fantasy or reality? J Am Soc neering of a bioartificial renal tubule cell therapy
Nephrol. 2012;23:1763–6. device. Cell Transplant. 1999;8:351–64.
332. Ogle B, Cascalho M, Platt JL. Fusion of approaches 348. Humes HD, Cieslinski DA. Interaction between
to the treatment of organ failure. Am J Transplant. growth factors and retinoic acid in the induction of
2004;4(Suppl 6):74–7. kidney tubulogenesis in tissue culture. Exp Cell Res.
333. Cascalho M, Ogle BM, Platt JL. Xenotransplantation 1992;201:8–15.
and the future of renal replacement. J Am Soc 349. Humes HD, Krauss JC, Cieslinski DA, Funke
Nephrol. 2004;15:1106–12. AJ. Tubulogenesis from isolated single cells of adult
190 B.S. Kim and H.T. Kim

mammalian kidney: clonal analysis with a recombi- cell system (BRECS): a compact, cryopreservable
nant retrovirus. Am J Physiol. 1996;271:F42–9. extracorporeal renal replacement device. Cell Med.
350. O’Neil JJ, Stegemann JP, Nicholson DT, Mullon CJ, 2012;4:33–43.
Maki T, Monaco AP, et al. Immunoprotection pro- 358. Gura V, Beizai M, Ezon C, Polaschegg
vided by the bioartificial pancreas in a xenogeneic HD. Continuous renal replacement therapy for end-­
host. Transplant Proc. 1997;29:2116–7. stage renal disease. The wearable artificial kidney
351. Humes HD, Buffington DA, MacKay SM, Funke (WAK). Contrib Nephrol. 2005;149:325–33.
AJ, Weitzel WF. Replacement of renal function in 359. Roberts M, Ash SR, Lee DB. Innovative perito-
uremic animals with a tissue-engineered kidney. Nat neal dialysis: flow-thru and dialysate regeneration.
Biotechnol. 1999;17:451–5. ASAIO J. 1999;45:372–8.
352. Fissell WH, Dyke DB, Weitzel WF, Buffington DA, 360. Ash SR, Janle EM. Continuous flow-through peri-
Westover AJ, MacKay SM, et al. Bioartificial kid- toneal dialysis (CFPD): comparison of efficiency to
ney alters cytokine response and hemodynamics in IPD, TPD, and CAPD in an animal model. Perit Dial
endotoxin-challenged uremic animals. Blood Purif. Int. 1997;17:365–72.
2002;20:55–60. 361. Fissell WH, Dubnisheva A, Eldridge AN, Fleischman
353. Humes HD, Buffington DA, Lou L, Abrishami S, AJ, Zydney AL, Roy S. High-performance silicon
Wang M, Xia J, et al. Cell therapy with a tissue-­ nanopore hemofiltration membranes. J Memb Sci.
engineered kidney reduces the multiple-organ 2009;326:58–63.
consequences of septic shock. Crit Care Med. 362. Fissell WH, Roy S. The implantable artificial kidney.
2003;31:2421–8. Semin Dial. 2009;22:665–70.
354. Humes HD, Sobota JT, Ding F, Song JH. A selective 363. Roy S, Goldman K, Marchant R, Zydney A, Brown
cytopheretic inhibitory device to treat the immuno- D, Fleischman A, et al. Implanted renal replace-
logical dysregulation of acute and chronic renal fail- ment for end-stage renal disease. Panminerva Med.
ure. Blood Purif. 2010;29:183–90. 2011;53:155–66.
355. Tumlin JA, Chawla L, Tolwani AJ, Mehta R, Dillon 364. Conlisk AT, Datta S, Fissell WH, Roy
J, Finkel KW, et al. The effect of the selective cyto- S. Biomolecular transport through hemofiltration
pheretic device on acute kidney injury outcomes membranes. Ann Biomed Eng. 2009;37:722–36.
in the intensive care unit: a multicenter pilot study. 365. Kanani DM, Fissell WH, Roy S, Dubnisheva A,
Semin Dial. 2012;26:616–23. Fleischman A, Zydney AL. Permeability – selectiv-
356. Pino CJ, Yevzlin AS, Lee K, Westover AJ, Smith PL, ity analysis for ultrafiltration: effect of pore geom-
Buffington DA, et al. Cell-based approaches for the etry. J Memb Sci. 2010;349:405.
treatment of systemic inflammation. Nephrol Dial 366. Muthusubramaniam L, Lowe R, Fissell WH, Li L,
Transplant. 2013;28:296–302. Marchant RE, Desai TA, et al. Hemocompatibility
357. Buffington DA, Pino CJ, Chen L, Westover AJ, of silicon-based substrates for biomedical implant
Hageman G, Humes HD. Bioartificial renal epithelial applications. Ann Biomed Eng. 2011;39:1296–305.
Bladder
8
Yun-Sok Ha and Tae Gyun Kwon

8.1 Introduction fistulae, bowel obstruction, and prolonged epi-


sodes of ileus, intestinal failure, or malignancy,
The bladder is susceptible to a variety of con- as well as significant morbidity and functional
genital anomalies, injuries, and disorders, such alterations [4]. To overcome these problems,
as cancer, trauma, infection, and chronic numerous alternative reconstructive procedures
inflammation [1]. In cases of muscle-invasive have been introduced including autoaugmenta-
and refractory non-muscle-invasive bladder tion and ureterocystoplasty [5–7], as well as tis-
cancer and end-stage (congenital) bladder dis- sue expansion [8, 9]. However, almost all of them
ease, the current standard treatment is radical resulted in failure due to technical difficulty or
cystectomy in combination with urinary diver- complication. Tissue engineering techniques,
sion or bladder augmentation, which serves to transplantation of biomaterials seeded with cells,
restore bladder capacity and compliance and have been developed as a promising alternative
prevent vesicoureteral reflux and renal damage that circumvent many of the limitations and com-
[2, 3]. The method of diversion depends in part plications associated with enterocystoplasty;
on the nature of defect and the patient’s needs therefore, it is considered a potentially promising
and wishes. Bladder reconstruction is one of option for bladder reconstruction.
the greatest surgical challenges in the field of Regenerative medicine aims to regenerate tis-
urology. To repair or replace the bladder, gas- sues and organs by creating biological equiva-
trointestinal segments are commonly used. lents through supplementation of scaffolding
However, gastrointestinal tissues are designed materials with bioactive components, cells, or a
to absorb specific solutes, whereas bladder combination thereof [10]. Various attempts have
tissues are designed to excrete solutes.
­ been made to reconstruct the bladder in both ani-
Conventional bladder reconstruction using gas- mal and human studies using regenerative medi-
trointestinal tissue (enterocystoplasty) is asso- cine techniques [11]. In 2006, the first clinical
ciated with numerous complications, including application of regenerative medicine for bladder
mucus production, bacterial colonization, elec- reconstruction was published by Atala et al. [12]
trolyte imbalance, anastomotic leakage, enteric In this study, they implanted a collagen/polygly-
colic acid composite scaffold with urothelial and
smooth muscle cells (SMCs) into the bladder
dome area after partial cystectomy.
Y.-S. Ha (*) • T.G. Kwon The fields of tissue engineering and regen-
Department of Urology, School of Medicine, erative medicine have achieved substantial
Kyungpook National University, Daegu, South Korea
e-mail: yunsokha@gmail.com; tgkwon@knu.ac.kr progression over the previous two decades. It

© Springer Nature Singapore Pte Ltd. 2018 191


B.W. Kim (ed.), Clinical Regenerative Medicine in Urology, DOI 10.1007/978-981-10-2723-9_8
192 Y.-S. Ha and T.G. Kwon

encompasses the cell biology, transplantation, types are being investigated for this purpose,
material science, and biomedical engineering, including bone marrow stem cells, skeletal myo-
toward identifying alternatives that can rees- blasts, and adipose progenitor cells [21–24].
tablish and preserve the regular function of In the 1950s, efforts to find alternatives to
damaged tissues and organs [13]. Studies for intestinal cystoplasty included the use of allo-
bladder tissue engineering strategies have plastic materials [25–31]. Although these early
been orientated in two directions: firstly, to research efforts met with very limited success
identify the most appropriate type of cells for owing to foreign body complications, they pro-
regeneration and to proficiently incorporate it vided the foundation for regenerative medicine
into bladder cells and, secondly, to determine and led to a focus on biodegradable, collagen-­
the most appropriate biomaterial and tech- rich tissues for use as a scaffold with and without
nique of embedding cells into engineered cell seeding [32–38]. Atala et al. [12] reported
grafts [14, 15]. The selected grafts must the first data regarding the use of a seeded biode-
exhibit all the qualities of the native tissue, gradable construct for augmentation of the neu-
acting ultimately as microenvironments for rogenic cell population. This work led to an
the implanted cells to proliferate and differen- industry-supported, prospective, multi-institution
tiate. Numerous studies have been tried to investigation using a neo-bladder construct.
identify ideal materials for bladder regenera- Unfortunately, compliance improved in only four
tion including acellular tissue matrices and patients after 12 months and in five after 36
synthetic matrices [16]. For example, bladder months but in none to a clinically or statistically
submucosa matrix (BSM), obtained by decel- significant degree. Capacity also did not improve
lularization of bladder submucosa, has been [39]. Using autologous tissue from a neuropathic
shown to be an effective material for clinical source may lower the yield of usable cells or
applications because it maintains the extracel- could potentiate the growth of abnormal tissue
lular matrix of the bladder as well as the bio- [40]. An alternate cell source for regenerative tis-
logical activity of growth factors [17]. sue, such as stem cells, might circumvent these
On the other hand, efforts have been exerted to concerns. However, the use of stem cells carries
find ideal cell source for bladder regeneration. its own set of obstacles [41–44]. Further investi-
Autologous bladder cells are the gold standard gation into the clinical applicability of regenera-
source for bladder regeneration in cell-based tis- tive tissue engineering is required, but this
sue engineering. To provide functional cells for technology remains poised to alter reconstructive
tissue reconstruction and remodeling, cultured bladder surgery.
cells at early passage (before passage 5) provide This chapter is focused on current status of
optimal results. In addition, cells should be cul- regenerative medicine including achievements
tured on the scaffold for a short time (less than 2 and perspectives on the use of biomaterials and
weeks) since SMCs lose their phenotype after stem cells in the field of bladder reconstruction.
long-term static culture [18]. Mechanical precon-
ditioning of muscle cells might be an alternative
culture method that would maintain the pheno- 8.2  he Use of Matrices
T
type and functional characteristics of cells before for Bladder Regeneration
implantation [19]. Immortalized muscle cell lines
are attractive since they may generate a large The bladder is considered as a good candidate for
number of functional cells. However, these types regenerative medicine-based approach because it
of cell lines have limited clinical application, as is one of the simple structured organs (not so
immortalized cells carry the risk of tumor forma- many different kinds of cells, minimal metabolic
tion [20]. For patients with bladder cancer, normal activity, and less interaction with adjacent
bladder cells are not available; thus, an alternative organs). Therefore, many researchers have stud-
cell source needs to be found, and ­several cell ied bladder reconstruction with various methods.
8 Bladder 193

One of the approaches is implantation of bio- i­ nkjet technology, it has been possible to use bio-­
materials into the bladder. In this approach, printing to create a naturally derived 3-D
selection of the appropriate bioscaffold for large construct, with a precise arrangement of growth
bladder tissue replacement is a critical compo- factors and other cellular components, into a
nent. There are distinct benefits to using biocom- patient-specific scaffold [16].
patible material in regenerative medicine for the
purpose of cell delivery vehicles and for bearing
the physical maintenance required for tissue 8.2.2 Synthetic Matrices
replacement [45]. Scaffolds are constructs that
are designed to direct tissue development and the Synthetic polymers are promising materials
growth of cells during the process of healing because of their reproducibility, large-scale pro-
[46]. Bladder replacements should therefore pro- duction, mechanical properties, ease of fabrica-
vide provisional mechanical support, adequate tion, and manipulable strength, degradation rate,
to endure forces exerted from neighboring struc- and microstructure [49]. The typical synthetic
tures, while maintaining a potential zone for tis- biomaterials utilized for urinary tissue regenera-
sue development. Biomaterials used for bladder tion are polyglycolic acid (PGA) and polylactic-­
replacements should possess the ability to be co-­glycolic acid (PLGA), which are
easily manipulated into a hollow, spherical con- biocompatible, biodegradable, and FDA-­
figuration. Furthermore, the biomaterials should approved for human applications [1]. However,
possess the ability to biodegrade for complete the brittleness of these synthetic polymers and
tissue development, without causing inflamma- the acidic by-products produced during their deg-
tion. An ideal scaffold for bladder tissue recon- radation have been linked to inflammation and
struction should have an adjustable 3-D porous immune dysfunction [50–54]. Furthermore, stud-
structure, high mechanical strength, uniformity, ies have shown that stiff synthetic materials can
and appropriate degradation rate, and it should cause mechanical failure and urinary stone for-
be easily fabricated. Over the last few decades, mation, and over time, the degradation products
several bladder wall substitutes, composed of cause fibroblast deposition, scarring, and graft
both synthetic and organic materials, have been contracture and reduce reservoir volume [25, 55,
investigated extensively. Biocompatible syn- 56]. To avoid these side effects, interest in poly-
thetic materials are considered potential substi- caprolactone (PCL) has increased because it is
tutes for bladder tissue. Biomaterials can be flexible, biocompatible, stable, and resistant to
divided into four main categories: (1) naturally resorption; its degradation products show low
derived matrices, including collagen; (2) syn- toxicity; and it could help protect the urothelium
thetic matrices, including polylactic-co-glycolic [57, 58]. However, it is well known that the use of
acid (PLGA); (3) acellular tissue matrices, scaffolds with low hydrophilicity (hydrophobic),
including bladder submucosa; and (4) hybrid or like PCL, can lead to low initial cell seeding effi-
composite scaffold [47]. cacy and heterogeneous/slow cell growth due to
inadequate diffusion of the cell culture medium
into the scaffold and the lack of specific interac-
8.2.1 Naturally Derived Matrices tion sites with cells [59]. Besides abovemen-
tioned biomaterials, numerous synthetic materials
Collagen, a naturally derived matrix, is consid- including polyvinyl sponge, Teflon, collagen
ered to be the most ubiquitous protein in the matrices, Vicryl PGA matrices, and silicone have
human body, and it is often used alongside algi- been studied in both experimental and clinical
nate as a natural matrix. It is useful in tissue engi- settings. However, most of these materials failed
neering, as it possesses the ability to be easily to show possibilities of clinical application due to
manipulated and does not provoke an immune mechanical, structural, functional, or biocompat-
response [48]. Through the use of innovative ibility problems. Permanent synthetic materials
194 Y.-S. Ha and T.G. Kwon

often succumb to mechanical failure and urinary it was difficult to delineate the junction between
stone formation, while degradable materials lead the host bladder and BAMG. Neural regeneration
to fibroblast deposition, scarring, graft contrac- continued to improve, and normal bladder capac-
ture, and a reduced reservoir volume [60, 61]. ity was maintained throughout the study. They
concluded that the BAMG appeared to serve,
without rejection, as a collagen and elastin frame-
8.2.3 Acellular Tissue Matrices work for the ingrowth of all bladder wall
components.
Decellularized matrices are the most commonly Small intestinal submucosa (SIS), a biode-
used naturally derived urological matrices. They gradable, acellular, xenogeneic collagen-based
are usually harvested from autologous, allogenic, tissue matrix graft, was first described in the early
or xenogeneic tissue [55, 62]. Chemical or 1960s as an acellular matrix for tissue replace-
mechanical processing decellularizes the matrix, ment in the vascular field [65]. The matrix is
removing all cellular components and leaving a derived from pig small intestine in which the
natural platform for tissue development [63]. The mucosa is mechanically removed from the inner
most common origin of decellularized matrices is surface and the serosa and muscular layer are
the tissue harvested from the bladder or small removed from the outer surface. Animal studies
intestinal mucosa. have shown that, when used for bladder augmen-
Bladder submucosa matrix (BSM), obtained tation, non-seeded SIS matrix is able to regener-
by decellularization of bladder submucosa, has ate [66]. In this study, 15-month small intestinal
been shown to be an effective material for clini- submucosa-regenerated canine bladder strips
cal applications because it maintains the extra- in vitro were analyzed by muscle bath compli-
cellular matrix of the bladder as well as the ance, contractility testing, and immunohisto-
biological activity of growth factors [17]. Non- chemical staining, and the results showed that
seeded allogeneic acellular bladder matrices small intestinal submucosa-regenerated canine
have been used as scaffolds for the ingrowth of bladder. Compliance studies demonstrated no
host bladder wall components. These matrices significant difference between small intestinal
are prepared by mechanically and chemically submucosa-regenerated and control bladders,
removing all cellular components from bladder which were 30-fold more compliant than a native
tissue. Probst et al. reported a method of bladder small intestinal submucosal graft. Contractility
augmentation that avoided the complications studies demonstrated that the contractile
encountered with the use of bowel segments responses and innervation were similar to those
using a newly developed acellular biomaterial, of normal canine bladder. Afferent nerves were
the bladder acellular matrix graft (BAMG), as a observed by immunohistochemical techniques.
homologous graft [64]. Thirty-four rats under- Histologically, the transitional layer was the
went a partial cystectomy (40–50%) and grafting same as that of the native bladder tissue; how-
with a BAMG of equal size. After initial bladder ever, as with other non-seeded collagen matrices,
enlargement, the graft was progressively infil- the muscle layer was not fully developed. In vitro
trated by host vessels and SMCs, and the muco- contractility studies performed on SIS-­
sal lining was complete within 10 days. After 4 regenerated dog bladders showed a 50% decrease
weeks, histological analysis showed that all in maximal contractile response compared to that
bladder wall components were present in the of normal bladder tissues.
graft. Ingrowth was complete after 8 weeks, Bladder augmentation with porcine acellular
except for neural regeneration, which was only bowel tissue matrix, human placental mem-
partial. At 12 weeks, the bladder wall muscle branes, or porcine SIS was performed using lap-
structure in the graft was so well developed that aroscopic techniques in minipigs. At 12 weeks
8 Bladder 195

post-op, the grafts had contracted to 60% of their showed an obvious effect on cell proliferation.
original size, and histologically, the grafts These findings demonstrate that the growth fac-
showed predominantly mucosal regeneration tors and extracellular matrix components in the
[67]. The human placental membranes, acellular BSM maintain biological activity even after
tissue matrices, and SIS grafts persisted as decellularization and extraction, supporting the
well-­
­ vascularized fibrous bands, without evi- wide applicability of BSM in tissue regeneration.
dence of significant inflammatory responses. The identification and characterization of the
These results suggest that a laparoscopic tech- growth factors and extracellular matrix compo-
nique for partial bladder wall replacement using nents in BSM are a prerequisite for understanding
a cell-free graft is feasible. At 1 year, the hemi- tissue regeneration using this scaffold. A study by
cystectomy and bladder replacement with SIS Kikuno et al. showed that grafts of acellular col-
group showed muscle tissue at the graft periph- lagen matrices can be enhanced by adding growth
ery and center; however, it consisted of small factors to improve bladder regeneration [69].
fused bundles with significant fibrosis. Compared They evaluated the combined effects of nerve
with primary bladder closure after hemicystec- growth factor (NGF) and vascular endothelial
tomy, there was no improvement in bladder growth factor (VEGF) on regeneration of the
capacity or compliance [68]. In this study, 12 BAMG in spinal cord injury (SCI)-mediated neu-
minipigs underwent laparoscopic hemicystec- rogenic bladder in rats. Bladder capacity and
tomy. Then, six pigs underwent bladder recon- compliance were significantly increased in all
struction with SIS and ipsilateral ureteral BAMG groups 8 weeks after surgery compared
reimplantation. In the SIS group, four out of five with that before bladder replacement surgery.
surviving pigs had unobstructed reimplanted ure- However, bladder capacity and compliance were
ters without evidence of hydroureteronephrosis, much higher in the VEGF and NGF group than in
while one had a high-­grade obstruction at the the control, NGF alone, and VEGF alone groups.
reimplantation site. Histopathologic studies 1 NGF had a significant synergistic effect on the
year after implantation revealed muscle at the development, differentiation, and functional res-
graft periphery and center; however, it consisted toration of the BAMG when administered with
of small fused bundles with significant fibrosis. VEGF in neurogenic bladders. Therefore, NGF
Although nerves were present at the graft periph- may be a useful cytokine for enhancing the regen-
ery and center, they were decreased in number. eration of a functional bladder following acellular
Compared to primary bladder closure, no increase matrix grafting in a neurogenic rat model.
in bladder capacity or compliance was observed.
Studies of acellular matrices that may provide
the necessary environment to promote cell migra- 8.2.4 Hybrid or Composite Scaffolds
tion, growth, and differentiation have been con-
ducted. Chin et al. identified and characterized the While it is difficult to find a single material that
bioactive factors in decellularized BSM by using satisfies the numerous requirements for bladder
ELISA, Western blotting, and immunohistochem- regeneration, composite materials might have
istry for its effective utilization in regenerative the necessary characteristics for use as scaffolds
medicine [17]. At least ten growth factors, includ- [70]. Tensile strength and elastic modulus are
ing VEGF, BMP4, PDGF-BB, KGF, TGF-­beta 1, important properties for urine storage and
IGF, bFGF, EGF, and TGF-alpha, were detected expulsion, and changes in the viscoelastic prop-
in the decellularized BSM. The presence of erties of the bladder wall can lead to voiding
­collagen (types 1, 2, 3, 4), laminin, and elastin dysfunction [71]. Therefore, composite scaf-
within the matrix was also demonstrated. folds which have many benefits, including
Supplementation with soluble BSM extracts reproducibility, large-­ scale production, and
196 Y.-S. Ha and T.G. Kwon

BSM 3 wt%BSM/PCL/F127
× 400

Fig. 8.1  Field emission scanning electron microscopy urine stem cells were used. The BSM scaffold was used as
images of cell morphology on a 3 wt% BSM composite the control. PCL polycaprolactone, F127 Pluronic F127
scaffold. For the analyses, upper urinary tract-derived BSM bladder submucosa matrix

appropriate mechanical properties, as well as BSM scaffold was used as the control. The pri-
adjustable strength, degradation rate, and micro- mary upper urinary tract-derived urine stem
structure, may be useful synthetic polymers, cells (uUSCs) on the 3 wt% BSM/F127 com-
because the desirable configuration is easily posite scaffold were flattened, with cell pro-
achieved. Pluronic F127 (F127) does not alter cesses that extended to the surface of the
the properties of the scaffold, but it improves scaffold. A similar morphology was seen in the
hydrophilicity and the microenvironment for control. FE-SEM analysis showed that the
cell attachment [72]. PCL was investigated as a uUSCs were tightly attached to the surface of
potential substitute for b­ladder tissue recon- the scaffold and had extended into the pores on
struction because of its flexibility, biocompati- day 8, indicating that the BSM/PCL/F127 com-
bility, stability, and resistance to resorption [57]. posite scaffold provided a suitable microenvi-
Jang et al. [1] fabricated a blend of PCL with ronment. Evaluation of the in vivo
F127 and BSM, which has been shown to be a tumorigenicity of the scaffold with or without
nonimmunogenic, noncytotoxic, collagen-rich uUSCs 8 weeks after implantation showed that
membrane that can be rapidly replaced by native the 3 wt% BSM composite scaffold did not
tissues. In their study, the artificial polymer cause teratomas. The tumorigenicity of the
PCL/F127 was chosen for its natural elastic 3 wt% BSM/F127 composite scaffolds was
characteristics and physiochemical properties, evaluated by implanting the scaffold into the
which enable fabrication of a reservoir of suffi- subcapsular space of the kidneys in ICR mice
cient volume, and naturally occurring BSM was (Fig.  8.2). BSM was used as the control.
used to improve its biocompatibility by increas- Scaffolds seeded with uUSCs were also tested.
ing the proliferation of the urothelium and After 8 weeks, H&E staining showed no signs
SMCs [1]. Testing for the proper PCL/F127/ of tumor formation in any scaffold, with or
BSM ratio for the scaffold showed that a PCL/ without cells. Evaluation of the in vivo tumori-
F127/3 wt% BSM composite scaffold exhibited genicity of the scaffold with or without uUSCs,
significantly enhanced hydrophilicity, the sur- 8 weeks after implantation, showed that the
face was easily immobilized, and there was no 3 wt% BSM composite scaffold did not cause
evidence of teratoma formation in vivo. The cell teratomas. With respect to tumorigenicity, this
morphology on the 3 wt% BSM composite scaf- result supports the safety of the scaffold with or
fold was assessed by field emission scanning without cells for therapeutic applications.
electron microscopy (FE-SEM; Fig. 8.1). The
8 Bladder 197

Control

× 10

3 wt% BSM/PCL/F127 BSM

uUSCs
3 wt% BSM/PCL/F127 BSM

Fig. 8.2  Tumorigenicity analysis of the 3 wt% BSM composite scaffold. For this analysis, uUSCs were used. The BSM
scaffold and a sham-operated kidney were used as controls. Control, non-scaffold-treated kidney
198 Y.-S. Ha and T.G. Kwon

8.3  ladder Regeneration Using


B Amniotic fluid- and bone marrow-derived stem
Cell Transplantation cells can also be used in an autologous manner
and have the potential to differentiate into blad-
Regenerative medicine with selective cell trans- der muscle and urothelium [74–76]. Embryonic
plantation may provide a means to create new stem cells and uUSCs also have the potential to
functional bladder segments. The success of cell differentiate into bladder tissue [77, 78].
transplantation strategies for bladder reconstruc-
tion depends on the ability to efficiently use
donor tissue and provide the right conditions for 8.3.1 Mesenchymal Stem Cell
long-term survival, differentiation, and growth.
Various cell sources have been explored for blad- De Coppi et al. [74] set up a model of bladder
der regeneration. Cilento et al. [73] showed that acute necrotizing injury to test the efficacy of
native cells are preferable because they can be adult rat bone marrow mesenchymal stem cell
used without rejection. A simple method for har- (MSC) vs. fetal rat amniotic fluid MSC trans-
vesting bladder cell types from surgical ­specimens plantation for the treatment of the impaired detru-
was used to generate normal human urothelial sor muscle contractility that develops when a
cell lines that could be reproducibly cultivated, cryoinjury is applied to the bladder wall. Impaired
passaged, and extensively expanded in serum- detrusor muscle contractility is part of the wound-­
free medium. Immunostaining of the bladder epi- healing process, which is characterized by SMC
thelial cells with broadly reacting anti-­cytokeratin depletion, followed by SMC hyperplasia and tis-
antibodies and an anti-cytokeratin antibody spe- sue remodeling in the surviving bladder. At 30
cific to cytokeratin 7, a transitional cell marker, days after transplantation, only a few fetal or
indicated that they expressed a stable epithelial adult MSCs gave rise to enteric or vascular
phenotype during serial passaging. Low levels of SMCs, whereas most MSCs appeared incapable
immunostaining for E-cadherin and E-cadherin of specific differentiation. In vitro coculture
messenger ribonucleic acid by Northern blot experiments using SMCs with fetal or adult
analysis and strongly positive immunostaining MSCs selectively labeled with distinct fluoro-
for vimentin indicated that the uroepithelial cells chromes showed the presence of hybrid cells,
express a non-barrier-forming phenotype under suggesting that some MSCs can undergo cell
these culture conditions. However, when the uro- fusion. Surprisingly, the major effect of rat bone
thelial cells were implanted subcutaneously into marrow or amniotic fluid MSC transplantation
athymic mice on biodegradable synthetic poly- seemed to be preventing the cryoinjury-induced
mers, they formed multilayered structures, sug- hypertrophy of surviving SMCs. In this model,
gesting that they retain the capability to stem cell transplantation had a limited effect on
differentiate in a living host. The urothelial cells SMC regeneration. Instead, it regulated post-­
proliferated in an epidermal growth factor-inde- injury bladder remodeling, possibly via a para-
pendent manner and expressed high levels of crine mechanism.
transforming growth factor-alpha and amphireg- The potential risk of using diseased tissue as a
ulin messenger ribonucleic acids, suggesting source of cells for tissue engineering necessitates
autocrine regulation of growth by epidermal the investigation of other possible cell sources.
growth factor-like factors. Cytogenetic analysis One promising source is the bone marrow, which
indicated that urothelial cells cultured for six pas- is a known source of stem cells for hematopoietic
sages possessed a normal chromosomal comple- and mesenchymal lineages. Bone marrow-­
ment. These results demonstrate that primary derived MSCs mobilize from the marrow in
cultures of autologous human bladder epithelial response to tissue damage and contribute to the
cells can be extensively expanded in vitro and, normal regeneration process, and MSCs have
consequently, might be useful in cell transplanta- been shown to localize in urinary bladders under-
tion strategies for genitourinary reconstruction. going tissue regeneration after acellular bladder
8 Bladder 199

augmentation in rats [79]. Shukla et al. [67] char- microscopic architecture and expression of pro-
acterized bone marrow-derived MSCs from pigs teins confirming functional characteristics.
and demonstrated their ability to differentiate Specifically, the induced urothelium expressed
into SMCs and utility for autologous augmenta- uroplakin, a marker of urothelial differentiation.
tion cystoplasty. MSCs were isolated from pigs These differentiated bladder structures also
and analyzed for common markers of MSCs by showed appropriate alpha-smooth muscle actin
flow cytometry, and SMC differentiation was staining. Finally, Hoechst staining of the xeno-
assessed by immunoblotting. MSCs were iso- grafts revealed a nuclear architecture consistent
lated, genetically labeled, expanded in vitro, with a mouse mesenchymal stem cell origin in
seeded onto SIS, and used for autologous bladder the urothelium, supporting the differentiation of
augmentation. Porcine MSCs are morphologi- these cells. In the appropriate signaling environ-
cally and immunophenotypically similar to ment, bone marrow-derived MSCs can undergo
human MSCs. Culturing MSCs at low density directed differentiation toward endodermal-­
enhances their proliferation rates, whereas main- derived urothelium and develop into mature blad-
tenance at confluence consistently induces der tissue within a tissue recombination model.
­differentiation into mature SMCs. Labeled MSCs This model serves as an important tool for the
grew on SIS for 1 week in vitro and survived a study of bladder development with a long-term
2-week implantation as an autologous bladder goal of cell replacement therapy applications.
augment in vivo. Some SMC label-positive cells
with typical SMC morphology were detected;
however, most cells were SMC label negative. 8.3.2 Embryonic Stem Cell
Notably, many cells with a urothelial morphol-
ogy stained positively for SMC markers. Porcine In 1981, embryonic stem (ES) cells were isolated
MSCs have properties similar to those of MSCs from mice for the first time [80]. This major
from other species and consistently undergo dif- breakthrough revolutionized the field of develop-
ferentiation into mature SMCs in vitro under spe- mental biology. ES cells are capable of prolonged
cific culture conditions. The addition of MSCs to self-renewal and differentiation, providing a tool
SIS may enhance tissue regeneration in augmen- to investigate the molecular mechanisms occur-
tation cystoplasty; however, they may not be sig- ring during differentiation from the embryo to
nificantly incorporated into smooth muscle adult. ES cells are considered to be pluripotent
bundles. and can differentiate into almost all cell types
Anumanthan et al. [76] reported directed dif- that arise from the three embryonic germ layers
ferentiation of bone marrow-derived MSCs into [81]. In vitro, these cells can differentiate into
bladder urothelium for use as a source of pluripo- multiple embryonic and adult cell types but rarely
tent or multipotent progenitor cells. The epithe- cells of endodermal lineage [82]. In contrast, dif-
lium was separated from the mesenchymal shells ferentiation of ES cells in an in vivo environment
of embryonic day 14 rat bladders. MSCs were shows their full developmental potential. Whether
isolated from mouse femoral and tibial bone mar- stem and/or progenitor cells exist within the blad-
row, and heterospecific recombinant xenografts der is unknown, but hypothetically, they should
were created by combining embryonic rat blad- exist. Identification of stem cells within the vast
der mesenchymal shells with the MSCs and population of cells in the bladder would be chal-
grafting them into the renal subcapsular space of lenging, especially without bladder-specific
athymic nude mice. Grafts were harvested at time stem/progenitor cell markers. Oottamasathien
points of up to 42 days and stained for urothelial et al. [77] determined the specific mesenchymal
and stromal differentiation. Histological exami- to ES cell ratios necessary to promote organ-­
nation of xenografts comprising mouse MSCs specific differentiation while completely sup-
and rat embryonic rat bladder mesenchyma pressing teratomatous growth. The embryonic
yielded mature bladder structures with normal mesenchyme is well established as an inductive
200 Y.-S. Ha and T.G. Kwon

tissue that dictates organ-specific programming lected from three healthy men. The UTCs were
of epithelial tissues, and this study showed that able to form colonies, had a greater proliferation
embryonic bladder mesenchyme can also drive rate than the VUCs, and had a normal karyotype.
ES cell differentiation toward endodermal-­ The UTCs possessed stem cell characteristics
derived urothelium. These approaches allow us to (expression of CD44+, CD73+, CD90+,
capture specific stages of stem cell differentiation CD105+, and SSEA4+) and expressed several
and better define stem cell hierarchies. markers of the urothelial, smooth muscle, and
endothelial cell lineages. The UTCs did not form
teratomas when implanted into the subcapsular
8.3.3 Urine-Derived Stem Cell space of a mouse kidney. Since the UTCs pos-
sessed stem cell characteristics, they could
Urine-derived stem cells (USCs) consistently potentially be an alternative cell source for uro-
expressed MSC/pericyte markers and some key logic tissue regeneration in patients with bladder
cell surface markers but no hematopoietic stem cancer.
cell markers (except for MHC-1), endothelial
markers (CD31), or human leukocyte antigen
(locus) DR (HLA-DR) [83]. Compared to other 8.3.4 Induced Pluripotent Stem Cell
MSCs, USCs have several advantages: (1) they
can be collected using a simple, low-cost, safe, Induced pluripotent stem cells (iPSCs) are natu-
noninvasive procedure; (2) they display telomer- rally programmed to divide continuously and
ase activity, and, thus, they are able to generate remain undifferentiated. Although these cells
more cells; and (3) they can efficiently differen- can give rise to ectodermal, mesodermal, or
tiate into SMCs, UCs, and endothelial cells. endodermal cell lineages, a significant risk of
Chun and Kim et al. [70] investigated whether teratoma exists. Any undifferentiated iPSCs
cells isolated from the upper urinary tract (UTCs) placed in the body might continue to divide in an
possess stem cell characteristics and could be uncontrolled manner, forming tumors. In addi-
used as an alternative cell source for patients tion, it takes a long time (4 months) to derive and
with bladder cancer. Current tissue engineering characterize iPSCs from an individual.
approaches for urologic tissue regeneration Furthermore, the low efficiency of differentia-
require invasive tissue biopsies to obtain autolo- tion, genetic abnormalities, and high cost pro-
gous cells, and these procedures are associated hibit their clinical applicability. Despite this, a
with various potential complications, such as few studies of ESCs or iPSCs for bladder tissue
donor site morbidity. Recently, cells isolated engineering have been reported. Frimberger
from voided urine (VUCs) have been proposed et al. [76] reported that human embryoid body-
as an alternative cell source for urologic tissue derived stem cells showed improved migration
engineering. However, VUCs should not be used in the presence of mature human bladder SMCs
in patients with bladder cancer, because the and urothelial cells. In addition, Moad et al. [77]
voided urine sample could contain malignant reported the generation of human iPSCs derived
cells. In the study, urine samples were collected from normal and aging human urinary tract tis-
from the upper urinary tract of four male patients sue. These iPSCs underwent bladder differentia-
with bladder cancer using a ureteral catheter. tion more efficiently than skin-derived iPSCs, as
The samples were centrifuged, and the cell pel- shown by the expression of urothelial-specific
lets were plated for primary culture. The cells markers (uroplakins, claudins, and cytokeratin)
were analyzed for the number of colony-forming and stromal smooth muscle markers (alpha-
units, proliferation rate, cytogenetics, stem cell smooth muscle actin, calponin, and desmin),
characteristics, and tumorigenicity, and the indicating the importance of organ-specific
results were compared to those of VUCs col- iPSCs for tissue-specific studies. Immobilized
8 Bladder 201

cell lines are not suitable for bladder regenera- easily accessible and exhibit high reprogram-
tion due to safety concerns. Therefore, multipo- ming efficiency, offering several advantages over
tent adult stem cells are currently used in bladder other cell types used for iPSC generation. Using
repair and reconstruction. Of particular interest the approach described in this study, the authors
is the paper published by Xue et al. [84], in generated 93 iPSC lines from 20 donors with
which they describe a practical method to gener- diverse genetic backgrounds. The nonviral iPSC
ate human iPSCs from USCs under feeder-free, bank containing these cell lines is a valuable
virus-free, serum-free conditions without the resource for iPSC research, facilitating future
c-MYC oncogene. The authors showed that this applications of human iPSCs. Table 8.1 shows a
approach could be applied in a large population comparison of the various stem cell types used in
with different genetic backgrounds. USCs are bladder repair studies.

Table 8.1  Comparison of the various stem cell types used for bladder repair
Bladder SMCs
Cell type/parameter BMSCs ASCs USCs ESC/iPSCs and UCs
Self-renewal and Limited, PD High, PD 60–70 Very high, PD Limited, PD
expansion capability ~30 >200 <30
Multi-lineage Multipotent but Similar to Multipotent Pluripotent None
differentiation capability mainly limited BMSCs differentiation (can generate
to mesodermal potential all lineages)
cell lineages
Urothelial and endothelial Low (<10%) Low (10%) High (60–85%) Low
differentiation capability
Telomerase activity (TA)/ Cannot be Cannot be Up to 75% of USC Possess TA None
telomere length detected detected clones possess TA and long
and relatively long telomeres
telomeres
Harvesting approach Invasive Invasive Noninvasive, Invasive to Invasive
simple, low cost, harvest
safe somatic cells
to generate
iPSCs
Pure stem cell isolation Difficult Difficult Very easy Easy None
Number of stem cells MSC/104 bone 100–140 USC Unknown
harvested marrow stromal clones/24 h urine
cells in from adults
newborns,
1MSC/106
Angiogenic trophic Yes Yes Yes Unknown Moderate
factors
Immunomodulatory Yes Yes Yes Unknown Unknown
properties
Rejection after No rejection as allogenous or xenogenous cells Likely to be No rejection
implantation (e.g., human BMSCs or USCs) when implanted in rejected as autogenous
rodent, rabbit, or canine models cells
Oncogenic potential No No No Yes None
Clinical trial utility Potential Potential Potential Safety concern Yes
ASC adipose-derived stem cell, BMSC bone marrow-derived mesenchymal stromal cell, ESC embryonic stem cell, iPSC
induced pluripotent stem cell, MSC mesenchymal stem cell, PD population doubling, SMC smooth muscle cell UC
urothelial cell, USC urine-derived stem cell
202 Y.-S. Ha and T.G. Kwon

8.4 Tissue Engineering 8.4.1 Animal Models


Approach for Bladder
Regeneration Jayo et al. compared the in situ cellular responses
to two biopolymer implants, a polylactic-co-­
Even in multiple studies, implantation of bioma- glycolic acid-based biodegradable mesh scaffold
terials without cells into the bladder has shown with autologous urothelial cells and SMCs (con-
some promising results, especially the urothelial struct) and a PLGA-based biodegradable mesh
layer which was able to regenerate normally; scaffold without cells (scaffold), in a canine
however the regeneration of muscle layer was model of augmentation cystoplasty [87]. Healing
not fully developed [55, 62, 64, 66, 85]. events were correlated with urodynamic assess-
Therefore, many investigators preferred tissue ments. Construct implants regenerated baseline
engineering approach (grafting biomaterials urodynamics as early as 4 months after implanta-
seeded with cells) for bladder tissue regenera- tion. In contrast, following scaffold implantation,
tion. In the early stage of investigation, synthetic urodynamics failed to return to baseline by study
polymer seeded with autologous cells was the termination (9 months). The functional differ-
most commonly used approach. The autologous ences elicited by the construct and scaffold
urothelial and muscle cells can be expanded implants were correlated with structural differ-
in vitro, seeded onto polymer scaffolds, and ences in the neo-tissues. The construct stroma
allowed to attach and form sheets of cells. The had greater vascularization, with gently folded,
resulting cell-polymer scaffold can then be interwoven connective tissue elements.
implanted in vivo. Histological analysis showed Conversely, the scaffold stroma was dense, with
that viable cells were able to self-assemble back haphazardly organized connective tissue. The
into their respective tissue types and retain their urothelium was regenerated in response to both
native phenotypes [86]. Synthetic polymer fibers construct and scaffold implantation. However,
of polyglycolic acid can serve as both a scaffold only the construct urothelium had normal stroma,
and delivery vehicle for the implantation of rab- well-developed detrusor, and abundant alpha-­
bit uroepithelial cells into athymic host animals. smooth muscle actin cell staining at early time
The polymers, which slowly degrade in vivo, points, leading to a structurally and functionally
allow the urothelial cells to survive at the implant complete bladder wall at 9 months. They con-
site. In the abovementioned study [79], the cluded that early cellular and stromal events dis-
authors demonstrated that polyglycolic acid tinguished the healing processes that led to
polymers support the proliferation of rabbit uro- bladder wall regeneration or repair. Construct
thelial cells in situ and can serve as a malleable implants containing cells elicited early healing
substrate for the creation of new urological processes that culminated with the regeneration
structures that replace the degrading polymer of complete mucosal and muscular components.
fibers. They also showed that when implanted on In contrast, the response to scaffold implantation
polyglycolic acid fibers, human urothelial and was consistent with reparative healing, i.e.,
bladder muscle cells form new urological struc- mucosal growth, but incomplete tissue layer
tures composed of both cell types. Human cell- development. These independent studies demon-
polymer xenografts can be recovered from host strate that cells are necessary to improve bladder
animals at extended time points after implanta- function when a large bladder tissue implant is
tion. These data suggest the feasibility of using required.
polyglycolic acid polymers as substrates for the The utility of allogenic bladder submucosa
creation of human urothelial and muscle grafts seeded with cells as a biomaterial for bladder
for genitourinary reconstruction. These experi- augmentation was investigated by Yoo et al. in
ments demonstrated, for the first time, that com- 1998 [62]. Partial cystectomies were performed
posite-layered tissue-­ engineered structures in ten beagle dogs. Both urothelial and SMCs
could be created de novo. were harvested from five animals and expanded
8 Bladder 203

separately. Allogenic bladder submucosa lialization and differentiation. In addition, TGF-­


obtained from sacrificed dogs was seeded with beta type 2 receptor protein expression was
muscle cells on one side and urothelial cells on enhanced in the urothelium. Platelet-derived
the opposite side. All beagles underwent cruciate growth factor-A (PDGF-A) RNA was constitu-
cystotomies on the bladder dome. Augmentation tively expressed in the mucosa, but expression
cystoplasty was performed with the cell-seeded decreased in the reepithelialization phase. These
allogenic bladder submucosa in five animals and data are consistent with the notion that urothelial
unseeded allogenic bladder submucosa in five regeneration can be achieved through paracrine
animals. The augmented bladders were retrieved or autocrine mechanisms via urothelium-derived
2 and 3 months after augmentation. Bladders growth factors. The observation of analogous
augmented with the cell-seeded allogenic bladder growth factor RNA expression patterns in regen-
submucosa showed a 99% increase in capacity erating skin epidermis suggests a more general
compared to bladders augmented with the cell-­ growth factor-regulated mechanism for epithelial
free allogenic bladder submucosa, which showed regeneration.
only a 30% increase in capacity. All dogs showed Bladder muscle tissue is less likely to regener-
normal bladder compliance, as evidenced by uro- ate normally. Both urothelial and muscle ingrowth
dynamic studies. Histologically, all retrieved are believed to be initiated from the edges of the
bladders contained a normal cellular organization normal bladder toward the region of the graft
consisting of a urothelial-lined lumen surrounded [89]. Regeneration of smooth muscle appears to
by submucosal tissue and smooth muscle. take place within the fibrous tissue characteristi-
Immunocytochemical analyses confirmed the cally found when biodegradable collagen/Vicryl
urothelial and muscle cell phenotypes and prosthesis is used to repair full-thickness defects
showed the presence of nerve fibers. In summary, in the rabbit urinary bladder. The question of
these matrices can function as vehicles for partial whether the central smooth muscle was gener-
bladder regeneration, and no relevant antigenic- ated via myoblastic differentiation within the
ity is evident. fibrous tissue or arose from healthy preexisting
It has been known for decades that the bladder detrusor muscle was addressed by serial section-
can regenerate extensively over free grafts, as the ing and specific staining. Only in situ transmuta-
urothelium has a high reparative capacity [88]. In tion, or differentiation, explains the observed
their study, de Boer et al. investigated the spatio- morphology, and the results strongly suggest that
temporal changes in the RNA and protein expres- the central smooth muscle was regenerated from
sion of growth factors and their receptors by in within the repair area.
situ hybridization and immunocytochemistry However, contracture or resorption of the graft
during regeneration after acute injury of mouse is usually evident. The inflammatory response to
urothelium. These expression data were well cor- the matrix may contribute to resorption of the
related with the changes in cell morphology and free graft. It was hypothesized that building 3-D
proliferation. Except for enhanced muscular constructs in vitro prior to implantation might
transforming growth factor-beta 1 (TGF-beta 1) facilitate the eventual terminal differentiation of
and TGF-beta type 2 receptor expression, the the cells after implantation while minimizing the
changes in the expression patterns of growth fac- inflammatory response toward the matrix, thus
tors or receptors were confined to the urothelium. avoiding graft contracture and shrinkage. A study
Increased mucosal RNA expression of insulin-­ in dogs demonstrated a major difference between
like growth factor-2 (IGF-2) and particularly type matrices with autologous cells (tissue-engineered
1 IGF receptor, as well as fibroblast growth fac- matrices) and those without cells [62]. Matrices
tor-­1 (FGF-1) but not FGF-2, coincided with implanted with cells retained most of their
reepithelialization and urothelial proliferation. implanted diameter, whereas matrices implanted
High levels of urothelial TGF-beta 1 RNA and without cells showed graft contraction and
protein expression were associated with reepithe- shrinkage. The histomorphology demonstrated a
204 Y.-S. Ha and T.G. Kwon

marked paucity of muscle cells and a more Furthermore, the construct C/BW demonstrated
aggressive inflammatory reaction in the matrices the ability of the regenerated bladder to respond
implanted without cells. to growth regulation. However, not all scaffolds
To better address the functional parameters perform well when used to replace a large por-
of tissue-engineered bladders, a canine animal tion of the bladder. In a study using SIS for sub-
model was designed that required a subtotal total bladder replacement in dogs, both the
cystectomy and subsequent replacement with a unseeded and cell-seeded experimental groups
tissue-­engineered organ [90]. Cystectomy-only showed graft shrinkage and poor results [91]. In
and non-seeded controls maintained average the study, 22 male dogs had a 90% partial cys-
capacities of 22% and 46% of the preoperative tectomy and were divided into three groups. At
values, respectively. In contrast, an average 1 month after cystectomy, dogs in the unseeded
bladder capacity of 95% of the original precys- (n = 6) and seeded (n = 6) groups received a
tectomy volume was achieved in the cell-seeded bladder augmentation with a corresponding SIS
tissue-engineered bladder replacements. These graft. The dogs in the surgical control group
findings were confirmed radiographically. (n = 10) received no further surgery. All dogs
Histologically, the non-seeded scaffold bladders were evaluated before and after surgery with
presented a pattern of normal urothelial cells blood chemistry, urine culture, intravenous
with a thickened fibrotic submucosa and a thin urography, cystogram, and cystometrogram.
layer of muscle fibers. The retrieved After surgery (at 1, 5, and 9 months), the blad-
tissue-­
­ engineered bladders showed a normal ders were examined by routine histology and
cellular organization, consisting of a trilayer of immunohistochemistry. All 22 dogs survived
urothelium, submucosa, and muscle. These the subtotal cystectomy, and 18 survived to the
studies, performed with PGA-based scaffolds, end of their intended survival period. One dog in
have been repeated by other investigators, and the seeded group died 1 month after augmenta-
similar results in long-term studies of large tion due to a bladder perforation caused by a
numbers of animals have been reported [85, 87]. large piece of incompletely absorbed SIS. Three
Jayo et al. [85] evaluated bladder regeneration other dogs (two in the unseeded group and one
following 80% cystectomy and augmentation in the seeded group) died within 2 months after
using a synthetic biopolymer with autologous augmentation due to bladder obstruction by
urothelial and SMCs (autologous neo-bladder stones. Unseeded and seeded SIS grafts showed
augmentation construct [construct]) or auto- moderate to heavy adhesion and graft shrinkage,
transplantation of native bladder (reimplanted and some had bone and calcification at the graft
native urinary bladder [reimplant]) in canines. site. In both groups, histology showed limited
Voiding function, urodynamic assessment, and bladder regeneration. Interestingly, dogs in the
neo-organ capacity-­ to-­
body-weight ratio (C/ control group at 1 month after cystectomy
BW) were assessed longitudinally for 24 months (when the seeded and unseeded groups received
following trigone-sparing augmentation cysto- their augmentations) had severely shrunken
plasty in juvenile canines. Within 30 days post- bladders and histologically showed severe
implantation, hematology and urinalysis inflammation, fibroblast infiltration, and muscle
returned to baseline. Both the constructs and hypertrophy. These results verify the subtotal
reimplants yielded neo-organs with statistically cystectomy model. The use of seeded or
equivalent urodynamics and histology. Linear unseeded SIS in a subtotal cystectomy model
regression analysis of C/BW showed that the does not yield the same quality and quantity of
constructs regained baseline slope and contin- bladder regeneration observed in the 40% non-
ued to adapt with animal growth. Constructs and inflammatory cystectomy model. This study
reimplants regained and maintained native blad- provides important insights into the process of
der histology by 3 months, capacity at 3–6 regeneration in a severely damaged bladder.
months, and compliance by 12–24 months. These results led us to reevaluate the critical
8 Bladder 205

elements required for complete bladder replace- more physiological environment, with the use of
ment using tissue engineering. a bioreactor that mimics the dynamics of bladder
The type of scaffold used is critical for the filling and emptying, to acquire the proper physi-
success of tissue engineering-based bladder ological properties. In our model, fibroblasts and
replacement. The use of bioreactors, wherein urothelial cells were evolved in a 3-D culture to
mechanical stimulation is initiated at organ pro- obtain a reconstructed VE. This was then cul-
duction, has also been proposed as an important tured in our bioreactor, which delivers a cyclic
parameter for success [92, 93]. Farhat and Yegar pressure increase up to 15 cm H2O, followed by a
[92] reported that mechanical stimulation may rapid decrease, to achieve a dynamically cultured
have a role in urinary bladder tissue engineering. VE (dcVE). The dcVE was characterized by his-
Currently, tissue engineering of the urinary blad- tology and immunofluorescence and compared to
der relies on biocompatible scaffolds that deliver the characteristics of statically cultured
biological and physical functionality with negli- VE. Mechanical resistance was evaluated by uni-
gible immunogenic or tumorigenic risks, and axial tensile tests, and permeability was mea-
recent research suggests that autologous cells sured with 14C-urea. Compared to our static
propagated in culture and seeded on scaffolds model, the dynamic model led to a urothelium
prior to implantation improve clinical outcomes. profile similar to that of native bladder.
In addition, as normal urinary bladder develop- Permeability analysis showed a profile compara-
ment in utero requires regular filling and empty- ble to that of native bladder, coinciding with the
ing, current research suggests that bladders basal cell organization in the dcVE, and appro-
constructed in vitro may also benefit from regular priate resistance for suturing and handling was
mechanical stimulation. Such stimulation appears also shown. This new alternative method offers a
to induce favorable cellular changes, prolifera- promising avenue for regenerative medicine. It is
tion, and the production of structurally suitable distinguished by its autologous character and
extracellular matrix (ECM) components that are efficiency as a urea barrier. These properties
essential for the normal function of hollow could significantly reduce inflammation, necro-
dynamic organs. To mimic in vivo urinary blad- sis, and possibly rejection.
der dynamics, tissue bioreactors that imitate the
filling and emptying of a normal bladder have
been devised. A “urinary bladder tissue bioreac- 8.4.2 Human Models
tor” that is able to recapitulate these dynamics
while providing a cellular environment that facil- Clinical trials of engineered bladder tissue for
itates the normal cell-cell and cell-matrix interac- cystoplasty reconstruction began in 1998. The
tions may be necessary to successfully engineer first was a small pilot study of seven patients
bladder tissue. Validation of a urinary bladder tis- using either a cell-seeded collagen scaffold (with
sue bioreactor that permits careful control of or without omentum coverage) or a combined
physiological conditions will generate broad PGA-collagen cell-seeded scaffold with omental
interest from researchers in urinary bladder phys- coverage. The patients who underwent recon-
iology and tissue engineering. A similar study struction with the engineered bladder tissue cre-
was conducted by Bouhout et al. [86], showing a ated with the PGA-collagen cell-seeded scaffolds
bladder substitute that was reconstructed in a with omental coverage showed increased compli-
physiological pressure environment. Bladder ance, decreased end-filling pressure, increased
reconstruction by enterocystoplasty or with bio- capacity, and longer dry periods over time [12].
engineered substitutes is still associated with In this study, the outcomes were measured by
complications, which led us to develop an autolo- serial urodynamics, cystograms, ultrasounds,
gous vesical equivalent (VE). This model has bladder biopsies, and serum analyses, and the
already proven its structural conformity. The cur- average follow-up was 46 months (range, 22–61
rent challenge is to reconstruct our model in a months). Postoperatively, the mean bladder leak
206 Y.-S. Ha and T.G. Kwon

point pressure decreases at capacity, and the bladder is often successful from an oncological
greatest increase in volume and compliance was perspective, there is significant morbidity associ-
observed in the composite engineered bladders ated with enteric interposition within the genito-
with an omental wrap (56%, 1.58-fold and 2.79-­ urinary tract. Therefore, there is a great
fold, respectively). Bowel function returned opportunity to decrease the morbidity associated
promptly after surgery. No metabolic conse- with the current surgical management of bladder
quences were noted, urinary calculi did not form, cancer by utilizing novel technologies to create a
mucus production was normal, and renal function urinary diversion without the intestine. Clinical
was preserved. The engineered bladder biopsies trials using neo-urinary conduits (NUC) seeded
showed an adequate structural architecture and with autologous SMCs are currently in progress
phenotype. Based on these results, engineered and may offer a significant surgical advance by
bladder tissues created with autologous cells eliminating the complications associated with the
seeded on collagen-polyglycolic acid scaffolds use of gastrointestinal segments in urinary recon-
and wrapped in omentum after implantation can struction, simplifying the surgical procedure, and
be used in patients who require cystoplasty. greatly facilitating recovery from cystectomy. A
Although these results are promising since they conduit from the ureters to the skin surface
show that engineered tissues can be implanted addresses the current standard of care while sim-
safely, it is just a start in terms of accomplishing plifying the surgical procedure, and it may also
the goal of engineering fully functional blad- improve patient outcomes. The NUC created by
ders. This was a limited clinical experience, and Tengion serves as a template to catalyze the
the technology is not yet ready for wide dissem- regeneration of native-like urinary tissue that can
ination; further experimental and clinical studies connect the ureters to the skin surface. To ensure
are required, and phase 2 studies have been native urinary tissue regeneration, a biocompati-
completed. ble, biodegradable scaffold with an extended his-
tory of safety and clinical utility is necessary. The
broadly used PLGA scaffold can enhance tissue
8.4.3 Neo-urinary Conduits regeneration and promote neo-tissue integration
when properly seeded with SMCs. The NUC
From the aforementioned and the urethral stud- construct has two principal components. The first
ies, it is evident that the use of cell-seeded matri- is the biomaterials. The NUC scaffold is com-
ces is superior to non-seeded matrices for the posed of a PGA polymer mesh fashioned into the
creation of engineered bladder tissues. Although required tubular shape and coated with a 50/50
advances have been made in bladder tissue engi- blend of PLGA copolymer. The specific struc-
neering, many challenges remain. Much of the tural parameters of the construct can be modified
current research is aimed at the development of during the surgical procedure according to the
biologically active, “smart” biomaterials that patient’s needs. The choice of well-established,
may improve tissue regeneration. Similar engi- synthetic, degradable biopolymers reflects the
neering techniques are now being used in patients same requirements for reliability and reproduc-
with bladder cancer who are having engineered ibility inherent in the choice of these polymers
urinary conduits implanted after cystectomy [94]. for applications in other bladder-related neo-­
Muscle-invasive and recurrent non-muscle-­ organs. The second component is the cells.
invasive bladder cancers have been traditionally Autologous SMCs sourced from bladder or non-­
treated with a radical cystectomy and urinary bladder tissue may be applied for NUC construc-
diversion. The urinary diversion is generally tion. Based on the successful outcomes in a
accomplished through the creation of an inconti- porcine cystectomy model, Tengion has initiated
nent ileal conduit, continent catheterizable reser- phase I clinical trials of NUC constructs in human
voir, or orthotopic neo-bladder utilizing the small patients requiring urinary diversion. This phase I
or large intestine. While radical extirpation of the study, “Incontinent Urinary Diversion Using an
8 Bladder 207

Autologous Neo-Urinary Conduit” (http://www. from scaffolds seeded with autologous adipose-
clinicaltrials.gov/ct2/show/NCT01087697), is or peripheral blood-derived SMCs will greatly
currently recruiting patients, with the objective of facilitate the translation of urologic tissue engi-
implanting up to ten patients by the end of 2012. neering technologies into clinical practice.
The objective of the study is to evaluate if NUC
constructs made using autologous adipose-­
derived SMCs in combination with defined 8.4.4 Kyungpook National
degradable biomaterial scaffolds can form a University Experiences
functional conduit to safely facilitate passage of
urine from the kidneys subsequent to radical cys- In a very important study, Lee et al. [96] investi-
tectomy. Primary outcome indices over a gated the synergistic effect of human USCs and a
12-month postimplantation period include struc- surface-modified composite scaffold for bladder
tural integrity and conduit patency. CT scans will reconstruction in a rat model. The composite
be used to demonstrate that urine flows safely scaffold (PCL/F127/3 wt% BSM) was fabricated
through the NUC construct. Additional measures using an immersion precipitation method, and
of primary outcomes up to 12-month postimplan- heparin was immobilized on the surface via cova-
tation include evaluation of any product- or lent conjugation. A PCL pellet/Pluronic F127
procedure-­related adverse events. Similarly, sec- powder mixture (95/5 [w/w]) was dissolved in
ondary outcome indices will include analysis of tetraglycol (12 wt%), and BSM powder was
NUC structural integrity and patency over a evenly mixed with the polymer solution. The
12–60-month postimplantation period. CT scan mixed solution was poured into a polytetrafluoro-
and renal ultrasound will be applied to demon- ethylene mold (70 × 70 × 0.4 μL) and then
strate that urine flows safely through the NUC immersed in water for 1 h at room temperature.
construct up to 60 months after implantation. After additional washing and vacuum drying, the
Procedural- and product-related adverse events PCL/F127/BSM composite scaffold was steril-
will also be monitored up to 60 months after ized with ethanol. The PCL/F127/3 wt% BSM
implantation. Finally, the overall safety of the composite scaffold exhibited significantly
NUC construct will be assessed by evaluation of enhanced hydrophilicity, the surface was easily
nonproduct-/procedural-related adverse events immobilized, and there was no evidence of tera-
and patient vital signs. toma formation in vivo [1]. Basic fibroblast
Stem cells derived from fat can be differenti- growth factor (bFGF) was loaded onto the
ated into smooth muscle for use in the conduits, heparin-­immobilized scaffold by a simple dip-
thus avoiding the use of bladder cells from blad- ping method. bFGF has been shown to stimulate
der cancer patients [95]. Basu et al. [88] described the proliferation and survival of both SMCs and
the isolation and characterization of SMCs from urothelial cells [97], suggesting the benefit of
porcine adipose tissue and peripheral blood that bFGF-loaded scaffolds for urological tissue engi-
are phenotypically and functionally indistin- neering applications. To fabricate a scaffold for
guishable from bladder-derived SMCs. In a pre- bFGF delivery, the authors covalently conjugated
clinical Good Laboratory Practice study, we heparin to the surface of a scaffold to form a
demonstrated that autologous adipose- and heparin-­ immobilized scaffold, which was then
peripheral blood-derived SMCs can be used to loaded with bFGF (scaffoldheparin-bFGF) [98]. Urine
seed synthetic, biodegradable, tubular scaffold samples from the upper urinary tract were
structures and that implantation of these seeded obtained from a 52-year-old female patient. The
scaffolds into a porcine cystectomy model leads urine samples (100 mL each) were centrifuged,
to successful de novo regeneration of a tubular and the cell pellets were washed with PBS. The
neo-organ composed of urinary-like neo-tissue cells were cultured in a mixture of keratinocyte
that is histologically identical to native bladder. serum-free medium and progenitor cell medium
The ability to create urologic structures de novo (Gibco-Invitrogen, Grand Island, NY, USA) in a
208 Y.-S. Ha and T.G. Kwon

USC USC USC USC USC


bFGF bFGF bFGF bFGF bFGF
bFGF
bFGF bFGF bFGF bFGF bFGF bFGF
bFGF bFGF bFGF

USC USC USC


USC USC

USCs-scaffoldheparin-bFGF

Fig. 8.3  Schematic diagram of the USC-seeded composite scaffoldheparin-bFGF graft. USC urine-derived stem cell, bFGF
basic fibroblast growth factor

1:1 ratio. USCs have been proposed as an alterna- 35.62 ± 6.69 μL/cm H2O, scaffold group; and
tive stem cell source for urological tissue recon- 1.92 ± 0.29 mL and 40.74 ± 7.88 μL/cm H2O,
struction since they have mesenchymal stem cell scaffoldheparin-bFGF group; Table 8.2). In the histo-
characteristics and have the capacity to differen- logical and immunohistochemical analyses, the
tiate into a variety of urological cell lineages USC-scaffoldheparin-bFGF group showed pro-
[78]. Figure 8.3 shows a schematic diagram of nounced, well-differentiated, and organized
the USC-seeded composite scaffoldheparin-bFGF smooth muscle bundle formation, a multilayered
graft. Twenty-five rats were divided into five and pan-cytokeratin-positive urothelium, and
groups: (1) control (sham operated), (2) partial high condensation of the submucosal area. The
cystectomy group (an ~40% defect was created implanted scaffolds were not visible in the tissue
in the dome of the bladder wall), (3) scaffold (an sections because they were dissolved by xylene
unmodified scaffold was attached after partial during specimen processing. The histological fea-
cystectomy), (4) scaffoldheparin-bFGF (the heparin-­ tures of the implanted grafts showed that the
immobilized bFGF-loaded scaffold was attached regenerated portions of the bladders in the USC-­
after partial cystectomy), and (5) USC-­ scaffoldheparin-­bFGF group exhibited pronounced
scaffoldheparin-­bFGF (scaffoldheparin-bFGF combined smooth muscle bundles, a multilayered urothe-
with 1 × 104 USCs was attached after partial cys- lium, condensed submucosa layer formation, and
tectomy) groups. The single-layer scaffold (disk restored bladder volume. These anatomical recon-
shaped, 6 mm diameter) was sutured as a patch structions are essential for functional compliance.
onto the bladder defect with 7-0 Vicryl sutures. However, the other scaffold groups showed only
The omentum was loosely wrapped over the graft weak SMC bundles, a thin urothelium, and loose
and fixed with fibrin glue (Greenplast; Green submucosa regeneration at the graft. These results
Cross, Seoul, Korea) (Fig. 8.4). In maximal blad- suggest that the seeded USCs contributed to tissue
der capacity and compliance analyses at 8 weeks regeneration. While the seeded cells were only
postoperation, the USC-scaffoldheparin-bFGF group expected to survive for 2 weeks in vivo [99], these
showed significant functional improvement exogenous cells were more effective for bladder
(2.34 ± 0.25 mL and 55.09 ± 11.81 μL/cm H2O) regeneration than the cells recruited from the sur-
compared to the other groups (2.60 ± 0.23 mL rounding host tissues or circulating blood flow.
and 56.14 ± 9.00 μL/cm H2O, control group; Although the USC regenerative mechanism was
1.46 ± 0.18 mL and 34.27 ± 4.42 μL/cm H2O, not determined in this study, based on previous
partial cystectomy group; 1.76 ± 0.22 mL and reports, regeneration is presumed to result from
8 Bladder 209

a b c d

e f g

Fig. 8.4 Bladder reconstruction procedure using the (e) The single-layer scaffold (disk form; diameter, 6 mm)
USC-scaffoldheparin-bFGF. (a) Rats were anesthetized with an was sutured as a patch onto the defect with 7-0 Vicryl
intraperitoneal injection of ketamine (40 mg/kg) and sutures. (f) Representative picture of the scaffold and
Rompun (10 mg/kg), with periodic supplementation as USC-scaffoldheparin-bFGF. (g) A urodynamic study (filling
needed. (b) The rats were placed on the operating board in cystometry). The bladder was filled with PBS, and maxi-
a supine position in laminar flow. (c) After shaving and mal capacity was defined as the minimum infusion vol-
sterilization with povidone iodine scrub over the suprapu- ume that triggered urine leakage. Compliance was defined
bic region, the abdomen was opened through a 4-cm lon- as the maximal capacity/(pressure that triggered leak-
gitudinal midline incision, and the urinary bladder was age)—(baseline pressure). USC urine-derived stem cell,
exposed. (d) The upper half of the bladder (dome and bFGF basic fibroblast growth factor
upper portion) was transected and removed with scissors.

Table 8.2 Measurement of cross-sectional area and urodynamics, including maximal bladder capacity and
compliance
USC-scaffoldheparin-­
Control Partial cystectomy Scaffold Scaffoldheparin-bFGF bFGF

Cross-sectional 16.85 ± 1.21 7.87 ± 1.37 9.67 ± 0.87 11.19 ± 0.87 15.71 ± 1.34


area, mm2
(mean ± SD)
Maximal bladder 2.60 ± 0.23 1.46 ± 0.18 1.76 ± 0.22 1.92 ± 0.29 2.34 ± 0.25
capacity, mL
(mean ± SD)
Compliance, μL/ 56.14 ± 9.00 34.27 ± 4.42 35.62 ± 6.69 40.74 ± 7.88 55.09 ± 11.81
cm H2O
(mean ± SD)
USC urine-derived stem cell, bFGF basic fibroblast growth factor

the paracrine effects of trophic factors secreted increased bladder capacity, compliance, regen-
from the USCs [99]. Thus, the transplanted stem eration of smooth muscle tissue, multilayered
cells influenced the surrounding host cells, urothelium, and condensed submucosa layers in
resulting in improved cell migration and differ- the study. Based on these results, the USC-
entiation into the target cell type. The USC- scaffoldheparin-bFGF would be ideal for bladder
seeded scaffoldheparin-bFGF exhibited significantly reconstruction.
210 Y.-S. Ha and T.G. Kwon

8.4.5 Whole Bladder Reconstruction regenerative medicine and tissue engineering.


Various types of acellular matrices, naturally
An interesting study was published by derived materials, and synthetic polymers have
Hoogenkamp et al [100]. They showed that scaf- been used for either unseeded (cell-free) or autol-
folds made from molecularly defined biomateri- ogous cell-seeded tissue engineering scaffolds.
als are instrumental in the regeneration of tissues Different categories of cell sources, from autolo-
but are generally confined to small flat patches gous differentiated urothelial cells and SMCs to
and do not comprise the whole organ. In this natural or laboratory-derived stem cells, have
study, a simple, one-step casting method was been tested to obtain suitable “cell-seeded” tem-
developed to produce a seamless, large, hollow, plates. The current clinically validated bladder
collagen-based scaffold, mimicking the shape of tissue engineering approaches essentially consist
the whole bladder with integrated anastomotic of augmentation cystoplasty in patients suffering
sites for ureters and the urethra. This hollow from poorly compliant neuropathic bladder.
bladder scaffold is highly standardized with uni- There have been no clinical applications of whole
form wall thickness and a unidirectional pore tissue-engineered neo-bladder for radical-­
structure to facilitate cell infiltration in vivo. reconstructive surgical treatment of bladder
Human and porcine bladder urothelial cells and malignancies or chronic inflammation due to
SMCs were able to attach to the scaffold and vesical coarctation. The reasons why bladder tis-
maintained their phenotypes in vitro. The closed sue engineering has not yet been clinically
luminal side and the porous outside of the scaf- applied include the risk of graft ischemia and
fold facilitated the formation of a urothelial lin- subsequent fibrous contraction and perforation.
ing and infiltration of SMCs, respectively. The The generation of a graft vascular network (vas-
cells aligned according to the scaffold template. culogenesis), together with the promotion of sur-
The technology used is highly adjustable in terms rounding vessel sprouting (angiogenesis), could
of shape, size, and materials and could be used as allow an effective graft blood supply and avoid
a starting point for research aimed at an off-the-­ serious, ischemia-related complications.
shelf medical device for neo-bladders. Current research suggests that the use of
Tissue engineering of the bladder may become biomaterial-­ based, bladder-shaped scaffolds
a reality in the future. It is important to design a seeded with autologous urothelial cells and
preclinical study using the best predictive model SMCs is the best option for bladder tissue engi-
with an adequate disease background to mimic the neering. Further studies to develop novel bioma-
clinical situation. For bladder tissue engineering, terials and identify cell sources, as well as the
large animals with diseased bladders appear to rep- information gained from research in develop-
resent the best experimental model, which should mental biology, signal transduction, and wound
aid in the development of clinically applicable tis- healing, would be beneficial.
sue-engineered bladder augmentation or replace-
ment with satisfactory long-term outcomes.
References
8.5 Conclusions 1. Jang YJ, Chun SY, Kim GN, Kim JR, Oh SH, Lee
and Perspectives JH, et al. Characterization of a novel composite
scaffold consisting of acellular bladder submucosa
matrix, polycaprolactone and Pluronic F127 as a
To prevent the problematic outcomes of bowel-­ substance for bladder reconstruction. Acta Biomater.
based bladder reconstructive surgery, such as 2014;10:3117–25.
prosthetic tumors and systemic metabolic com- 2. Hautmann RE, Hautmann SH, Hautmann O.
Complications associated with urinary diversion. Nat
plications, the research focus has switched from
Rev Urol. 2011;8:667–77.
regenerating and strengthening the failing organ 3. Nieuwenhuijzen JA, de Vries RR, Bex A, van der Poel
or building an organ replacement in the 1990s to HG, Meinhardt W, Antonini N, et al. Urinary diversions
8 Bladder 211

after cystectomy: the association of clinical factors, 21. Chung SY, Krivorov NP, Rausei V, Thomas L,
complications and functional results of four different Frantzen M, Landsittel D, et al. Bladder reconstitu-
diversions. Eur Urol. 2008;53:834–42. discussion 42-4 tion with bone marrow derived stem cells seeded on
4. Jednak R. The evolution of bladder augmentation: small intestinal submucosa improves morphological
from creating a reservoir to reconstituting an organ. and molecular composition. J Urol. 2005;174:353–9.
Front Pediatr. 2014;2:10. 22. Zhang Y, Lin HK, Frimberger D, Epstein RB, Kropp
5. Cartwright PC, Snow BW. Bladder autoaugmenta- BP. Growth of bone marrow stromal cells on small
tion: early clinical experience. J Urol. 1989;142:505– intestinal submucosa: an alternative cell source for
8. discussion 20-1 tissue engineered bladder. BJU Int. 2005;96:1120–5.
6. Cartwright PC, Snow BW. Bladder autoaugmenta- 23. Lu SH, Sacks MS, Chung SY, Gloeckner DC,
tion: partial detrusor excision to augment the bladder Pruchnic R, Huard J, et al. Biaxial mechanical prop-
without use of bowel. J Urol. 1989;142:1050–3. erties of muscle-derived cell seeded small intes-
7. Adams MC, Brock JW 3rd, Pope JC, Rink tinal submucosa for bladder wall reconstitution.
RC. Ureterocystoplasty: is it necessary to detubular- Biomaterials. 2005;26:443–9.
ize the distal ureter? J Urol. 1998;160:851–3. 24. Jack GS, Almeida FG, Zhang R, Alfonso ZC, Zuk PA,
8. Lailas NG, Cilento B, Atala A. Progressive ureteral Rodriguez LV. Processed lipoaspirate cells for tissue
dilation for subsequent ureterocystoplasty. J Urol. engineering of the lower urinary tract: implications
1996;156:1151–3. for the treatment of stress urinary incontinence and
9. Satar N, Yoo JJ, Atala A. Progressive dilation for bladder reconstruction. J Urol. 2005;174:2041–5.
bladder tissue expansion. J Urol. 1999;162:829–31. 25. Gleeson MJ, Griffith DP. The use of alloplas-
10. Langer R, Vacanti JP. Tissue engineering. Science. tic biomaterials in bladder substitution. J Urol.
1993;260:920–6. 1992;148:1377–82.
11. Sloff M, Simaioforidis V, de Vries R, Oosterwijk 26. Kropp BP, Eppley BL, Prevel CD, Rippy MK,
E, Feitz W. Tissue engineering of the bladder– Harruff RC, Badylak SF, et al. Experimental assess-
reality or myth? A systematic review. J Urol. ment of small intestinal submucosa as a bladder wall
2014;192:1035–42. substitute. Urology. 1995;46:396–400.
12. Atala A, Bauer SB, Soker S, Yoo JJ, Retik AB. Tissue-­ 27. Kropp BP, Cheng EY, Lin HK, Zhang Y. Reliable
engineered autologous bladders for patients needing and reproducible bladder regeneration using
cystoplasty. Lancet. 2006;367:1241–6. unseeded distal small intestinal submucosa. J Urol.
13. Aboushwareb T, McKenzie P, Wezel F, Southgate J, 2004;172:1710–3.
Badlani G. Is tissue engineering and biomaterials the 28. Kanematsu A, Yamamoto S, Ogawa O. Changing
future for lower urinary tract dysfunction (LUTD)/ concepts of bladder regeneration. Int J Urol.
pelvic organ prolapse (POP)? Neurourol Urodyn. 2007;14:673–8.
2011;30:775–82. 29. Lewis JM, Cheng EY. Non-traditional manage-
14. Petrovic V, Stankovic J, Stefanovic V. Tissue engi- ment of the neurogenic bladder: tissue engineer-
neering of the urinary bladder: current concepts ing and neuromodulation. ScientificWorldJournal.
and future perspectives. ScientificWorldJournal. 2007;7:1230–41.
2011;11:1479–88. 30. Yamzon J, Perin L, Koh CJ. Current status of tissue
15. Shokeir AA, Harraz AM, El-Din AB. Tissue engi- engineering in pediatric urology. Curr Opin Urol.
neering and stem cells: basic principles and applica- 2008;18:404–7.
tions in urology. Int J Urol. 2010;17:964–73. 31. Roth CC, Mondalek FG, Kibar Y, Ashley RA, Bell
16. El-Taji OM, Khattak AQ, Hussain SA. Bladder CH, Califano JA, et al. Bladder regeneration in a
reconstruction: The past, present and future. Oncol canine model using hyaluronic acid-poly(lactic-co-­
Lett. 2015;10:3–10. glycolic-acid) nanoparticle modified porcine small
17. Chun SY, Lim GJ, Kwon TG, Kwak EK, Kim BW, intestinal submucosa. BJU Int. 2011;108:148–55.
Atala A, et al. Identification and characterization 32. Kelami A. Lyophilized human dura as a bladder wall
of bioactive factors in bladder submucosa matrix. substitute: experimental and clinical results. J Urol.
Biomaterials. 2007;28:4251–6. 1971;105:518–22.
18. Chamley-Campbell J, Campbell GR, Ross R. The 33. Fishman IJ, Flores FN, Scott FB, Spjut HJ, Morrow
smooth muscle cell in culture. Physiol Rev. B. Use of fresh placental membranes for bladder
1979;59:1–61. reconstruction. J Urol. 1987;138:1291–4.
19. Akhyari P, Fedak PW, Weisel RD, Lee TY, Verma 34. Atala A, Vacanti JP, Peters CA, Mandell J, Retik
S, Mickle DA, et al. Mechanical stretch regi- AB, Freeman MR. Formation of urothelial struc-
men enhances the formation of bioengineered tures in vivo from dissociated cells attached to
autologous cardiac muscle grafts. Circulation. biodegradable polymer scaffolds in vitro. J Urol.
2002;106:I137–42. 1992;148:658–62.
20. Koh GY, Klug MG, Soonpaa MH, Field 35. Kambic H, Kay R, Chen JF, Matsushita M, Harasaki
LJ. Differentiation and long-term survival of H, Zilber S. Biodegradable pericardial implants for
C2C12 myoblast grafts in heart. J Clin Invest. bladder augmentation: a 2.5-year study in dogs. J
1993;92:1548–54. Urol. 1992;148:539–43.
212 Y.-S. Ha and T.G. Kwon

36. Kropp BP, Badylak S, Thor KB. Regenerative blad- 53. Lo H, Kadiyala S, Guggino SE, Leong KW. Poly(L-­
der augmentation: a review of the initial preclinical lactic acid) foams with cell seeding and controlled-­
studies with porcine small intestinal submucosa. release capacity. J Biomed Mater Res. 1996;30:475–84.
Adv Exp Med Biol. 1995;385:229–35. 54. Nam YS, Yoon JJ, Park TG. A novel fabrication
37. Zhang Y, Kropp BP, Lin HK, Cowan R, Cheng method of macroporous biodegradable polymer
EY. Bladder regeneration with cell-seeded small scaffolds using gas foaming salt as a porogen addi-
intestinal submucosa. Tissue Eng. 2004;10:181–7. tive. J Biomed Mater Res. 2000;53:1–7.
38. Harrington DA, Sharma AK, Erickson BA, Cheng 55. Sutherland RS, Baskin LS, Hayward SW, Cunha
EY. Bladder tissue engineering through nanotech- GR. Regeneration of bladder urothelium, smooth
nology. World J Urol. 2008;26:315–22. muscle, blood vessels and nerves into an acellular
39. Joseph DB, Borer JG, De Filippo RE, Hodges SJ, tissue matrix. J Urol. 1996;156:571–7.
McLorie GA. Autologous cell seeded biodegradable 56. Monsour MJ, Mohammed R, Gorham SD, French
scaffold for augmentation cystoplasty: phase II study DA, Scott R. An assessment of a collagen/vicryl
in children and adolescents with spina bifida. J Urol. composite membrane to repair defects of the urinary
2014;191:1389–95. bladder in rabbits. Urol Res. 1987;15:235–8.
40. Subramaniam R, Hinley J, Stahlschmidt J, Southgate 57. Yu DS, Lee CF, Chen HI, Chang SY. Bladder wall
J. Tissue engineering potential of urothelial cells grafting in rats using salt-modified and collagen-­
from diseased bladders. J Urol. 2011;186:2014–20. coated polycaprolactone scaffolds: preliminary
41. Aboushwareb T, Atala A. Stem cells in urology. Nat report. Int J Urol. 2007;14:939–44.
Clin Pract Urol. 2008;5:621–31. 58. Beiser IH, Kanat IO. Biodegradable internal fixa-
42. Aitken KJ, Bagli DJ. The bladder extracellular tion. A literature review. J Am Podiatr Med Assoc.
matrix. Part II: regenerative applications. Nat Rev 1990;80:72–5.
Urol. 2009;6:612–21. 59. Oh SH, Lee JH. Hydrophilization of synthetic biode-
43. Soler R, Fullhase C, Atala A. Regenerative medi- gradable polymer scaffolds for improved cell/tissue
cine strategies for treatment of neurogenic bladder. compatibility. Biomed Mater. 2013;8:014101.
Therapy. 2009;6:177–84. 60. Atala A. This month in investigative urology:
44. Chen BS, Xie H, Zhang SL, Geng HQ, Zhou JM, Commentary on the replacement of urologic associ-
Pan J, et al. Tissue engineering of bladder using ated mucosa. J Urol. 1996;156:338–9.
vascular endothelial growth factor gene-modified 61. Atala A. Autologous cell transplantation for urologic
endothelial progenitor cells. Int J Artif Organs. reconstruction. J Urol. 1998;159:2–3.
2011;34:1137–46. 62. Yoo JJ, Meng J, Oberpenning F, Atala A. Bladder
45. Furth ME, Atala A, Van Dyke ME. Smart biomate- augmentation using allogenic bladder submucosa
rials design for tissue engineering and regenerative seeded with cells. Urology. 1998;51:221–5.
medicine. Biomaterials. 2007;28:5068–73. 63. Crapo PM, Gilbert TW, Badylak SF. An overview of
46. Chan BP, Leong KW. Scaffolding in tissue engineer- tissue and whole organ decellularization processes.
ing: general approaches and tissue-specific consider- Biomaterials. 2011;32:3233–43.
ations. Eur Spine J. 2008;17(Suppl 4):467–79. 64. Probst M, Dahiya R, Carrier S, Tanagho
47. Atala A. Tissue engineering of human bladder. Br EA. Reproduction of functional smooth muscle tis-
Med Bull. 2011;97:81–104. sue and partial bladder replacement. Br J Urol.
48. Dahms SE, Piechota HJ, Dahiya R, Lue TF, Tanagho 1997;79:505–15.
EA. Composition and biomechanical properties of 65. Rotthoff G, Braun B, Bilgin I, Mahjoub M,
the bladder acellular matrix graft: comparative anal- Zimmermann U, Zinganell K. Aortic replacement
ysis in rat, pig and human. Br J Urol. 1998;82:411–9. with multi-layer submucosa prostheses made from
49. Shakhssalim N, Dehghan MM, Moghadasali R, heterologous small intestine. J Cardiovasc Surg
Soltani MH, Shabani I, Soleimani M. Bladder tissue (Torino). 1969;10:31–5.
engineering using biocompatible nanofibrous elec- 66. Kropp BP, Sawyer BD, Shannon HE, Rippy MK,
trospun constructs: feasibility and safety investiga- Badylak SF, Adams MC, et al. Characterization
tion. Urol J. 2012;9:410–9. of small intestinal submucosa regenerated canine
50. Bergsma EJ, Rozema FR, Bos RR, de Bruijn detrusor: assessment of reinnervation, in vitro com-
WC. Foreign body reactions to resorbable poly(L-­ pliance and contractility. J Urol. 1996;156:599–607.
lactide) bone plates and screws used for the fixation 67. Portis AJ, Elbahnasy AM, Shalhav AL, Brewer A,
of unstable zygomatic fractures. J Oral Maxillofac Humphrey P, McDougall EM, et al. Laparoscopic
Surg. 1993;51:666–70. augmentation cystoplasty with different biodegradable
51. Martin C, Winet H, Bao JY. Acidity near eroding grafts in an animal model. J Urol. 2000;164:1405–11.
polylactide-polyglycolide in vitro and in vivo in rabbit 68. Landman J, Olweny E, Sundaram CP, Andreoni C,
tibial bone chambers. Biomaterials. 1996;17:2373–80. Collyer WC, Rehman J, et al. Laparoscopic mid
52. Zhang R, Ma PX. Poly(alpha-hydroxyl acids)/ sagittal hemicystectomy and bladder reconstruction
hydroxyapatite porous composites for bone-tissue with small intestinal submucosa and reimplantation
engineering. I. Preparation and morphology. J of ureter into small intestinal submucosa: 1-year fol-
Biomed Mater Res. 1999;44:446–55. lowup. J Urol. 2004;171:2450–5.
8 Bladder 213

69. Kikuno N, Kawamoto K, Hirata H, Vejdani K, 84. Xue Y, Cai X, Wang L, Liao B, Zhang H, Shan Y,
Kawakami K, Fandel T, et al. Nerve growth factor et al. Generating a non-integrating human induced
combined with vascular endothelial growth factor pluripotent stem cell bank from urine-derived cells.
enhances regeneration of bladder acellular matrix PLoS One. 2013;8:e70573.
graft in spinal cord injury-induced neurogenic rat 85. Jayo MJ, Jain D, Ludlow JW, Payne R, Wagner BJ,
bladder. BJU Int. 2009;103:1424–8. McLorie G, et al. Long-term durability, tissue regen-
70. Wang C, Meng G, Zhang L, Xiong Z, Liu eration and neo-organ growth during skeletal matu-
J. Physical properties and biocompatibility of a ration with a neo-bladder augmentation construct.
core-sheath structure composite scaffold for bone Regen Med. 2008;3:671–82.
tissue engineering in vitro. J Biomed Biotechnol. 86. Atala A, Freeman MR, Vacanti JP, Shepard J,
2012;2012:579141. Retik AB. Implantation in vivo and retrieval of
71. Ghoniem GM. The recalcitrant overactive bladder artificial structures consisting of rabbit and human
patient. Geriatrics. 2002;57(Suppl 1):23–9. urothelium and human bladder muscle. J Urol.
72. Khattak SF, Bhatia SR, Roberts SC. Pluronic F127 as a 1993;150:608–12.
cell encapsulation material: utilization of membrane- 87. Jayo MJ, Jain D, Wagner BJ, Bertram TA. Early cel-
stabilizing agents. Tissue Eng. 2005;11:974–83. lular and stromal responses in regeneration versus
73. Cilento BG, Freeman MR, Schneck FX, Retik AB, repair of a mammalian bladder using autologous
Atala A. Phenotypic and cytogenetic characteriza- cell and biodegradable scaffold technologies. J Urol.
tion of human bladder urothelia expanded in vitro. J 2008;180:392–7.
Urol. 1994;152:665–70. 88. de Boer WI, Schuller AG, Vermey M, van der
74. De Coppi P, Callegari A, Chiavegato A, Gasparotto Kwast TH. Expression of growth factors and recep-
L, Piccoli M, Taiani J, et al. Amniotic fluid and tors during specific phases in regenerating uro-
bone marrow derived mesenchymal stem cells can thelium after acute injury in vivo. Am J Pathol.
be converted to smooth muscle cells in the cryo-­ 1994;145:1199–207.
injured rat bladder and prevent compensatory hyper- 89. Gorham SD, French DA, Shivas AA, Scott R. Some
trophy of surviving smooth muscle cells. J Urol. observations on the regeneration of smooth muscle
2007;177:369–76. in the repaired urinary bladder of the rabbit. Eur
75. Shukla D, Box GN, Edwards RA, Tyson DR. Bone Urol. 1989;16:440–3.
marrow stem cells for urologic tissue engineering. 90. Oberpenning F, Meng J, Yoo JJ, Atala A. De novo
World J Urol. 2008;26:341–9. reconstitution of a functional mammalian urinary
76. Anumanthan G, Makari JH, Honea L, Thomas JC, bladder by tissue engineering. Nat Biotechnol.
Wills ML, Bhowmick NA, et al. Directed differentia- 1999;17:149–55.
tion of bone marrow derived mesenchymal stem cells 91. Zhang Y, Frimberger D, Cheng EY, Lin HK, Kropp
into bladder urothelium. J Urol. 2008;180:1778–83. BP. Challenges in a larger bladder replacement with
77. Oottamasathien S, Wang Y, Williams K, Franco OE, cell-seeded and unseeded small intestinal submu-
Wills ML, Thomas JC, et al. Directed differentia- cosa grafts in a subtotal cystectomy model. BJU Int.
tion of embryonic stem cells into bladder tissue. Dev 2006;98:1100–5.
Biol. 2007;304:556–66. 92. Farhat WA, Yeger H. Does mechanical stimulation
78. Chun SY, Kim HT, Lee JS, Kim MJ, Kim BS, Kim have any role in urinary bladder tissue engineering?
BW, et al. Characterization of urine-derived cells World J Urol. 2008;26:301–5.
from upper urinary tract in patients with bladder 93. Bouhout S, Gauvin R, Gibot L, Aube D, Bolduc
cancer. Urology. 2012;79:1186 e1-7. S. Bladder substitute reconstructed in a physiological
79. Kanematsu A, Yamamoto S, Iwai-Kanai E, Kanatani pressure environment. J Pediatr Urol. 2011;7:276–82.
I, Imamura M, Adam RM, et al. Induction of smooth 94. Hyndman ME, Kaye D, Field NC, Lawson KA,
muscle cell-like phenotype in marrow-derived cells Smith ND, Steinberg GD, et al. The use of regenera-
among regenerating urinary bladder smooth muscle tive medicine in the management of invasive bladder
cells. Am J Pathol. 2005;166:565–73. cancer. Adv Urol. 2012;2012:653652.
80. Martin GR. Isolation of a pluripotent cell line from 95. Basu J, Jayo MJ, Ilagan RM, Guthrie KI, Sangha N,
early mouse embryos cultured in medium condi- Genheimer CW, et al. Regeneration of native-like
tioned by teratocarcinoma stem cells. Proc Natl neo-urinary tissue from nonbladder cell sources.
Acad Sci U S A. 1981;78:7634–8. Tissue Eng Part A. 2012;18:1025–34.
81. Alison MR, Poulsom R, Forbes S, Wright NA. An 96. Lee JN, Chun SY, Lee HJ, Jang YJ, Choi SH, Kim
introduction to stem cells. J Pathol. 2002;197:419–23. DH, et al. Human Urine-derived Stem Cells Seeded
82. Trounson A. The production and directed differen- Surface Modified Composite Scaffold Grafts for
tiation of human embryonic stem cells. Endocr Rev. Bladder Reconstruction in a Rat Model. J Korean
2006;27:208–19. Med Sci. 2015;30:1754–63.
83. Qin D, Long T, Deng J, Zhang Y. Urine-derived stem 97. Lee M, Wu BM, Stelzner M, Reichardt HM, Dunn
cells for potential use in bladder repair. Stem Cell Res JC. Intestinal smooth muscle cell maintenance by
Ther. 2014;5:69. basic fibroblast growth factor. Tissue Eng Part A.
2008;14:1395–402.
214 Y.-S. Ha and T.G. Kwon

98. Yoon JJ, Chung HJ, Lee HJ, Park TG. Heparin-­ 100.
Hoogenkamp HR, Pot MW, Hafmans TG,
immobilized biodegradable scaffolds for local and Tiemessen DM, Sun Y, Oosterwijk E, et al.
sustained release of angiogenic growth factor. J Scaffolds for whole organ tissue engineer-
Biomed Mater Res A. 2006;79:934–42. ing: Construction and in vitro evaluation of a
99. Kim BS, Chun SY, Lee JK, Lim HJ, Bae JS, Chung seamless, spherical and hollow collagen blad-
HY, et al. Human amniotic fluid stem cell injection der construct with appendices. Acta Biomater.
therapy for urethral sphincter regeneration in an ani- 2016;43:112–21.
mal model. BMC Med. 2012;10:94.
Urethra
9
Yun-Sok Ha and Tae-Hwan Kim

9.1 Introduction can occur at any portion of the urethra between


the meatus and bladder neck [10]. In the con-
A variety of congenital and acquired urethral text of urethral stricture description, the term
pathologies including hypospadias, strictures, “urethra” typically refers to the anterior ure-
fistulae, and straddle injuries can severely thra including the bulbar and penile urethra
impair normal urethral function, necessitating [11]. However, posterior urethral strictures
surgical reconstruction [1–4]. No universally including those of the membranous and pros-
recognized algorithms for the surgical manage- tatic urethrae are not included in this defini-
ment of these conditions are in place, although tion; instead, these are typically termed
some progress in this field has been made [5– “urethral contractures” or “urethral stenoses”
7]. A urethral stricture is a narrowing of the [11]. It should be noted that urethroplasties are
urethral lumen, arising from iatrogenic (33%), performed not only in patients with urethral
idiopathic (33%), traumatic (19%), and inflam- strictures, but also in those with hypospadias
matory (15%) causes, that may lead to lower and other urethral defects (e.g., urethral fistu-
urinary tract obstruction [3, 8]. Such obstruc- lae). In particular, repair of the anterior urethra
tion may significantly impair patients’ quality is one of the most demanding surgical prob-
of life by causing voiding difficulties; these, in lems in urology. A multitude of techniques has
turn, may negatively affect the upper urinary been used to treat anterior urethral strictures
tract, resulting in deterioration of renal func- using buccal mucosa or a penile skin flap
tion [9]. The mechanism of urethral stricture for reconstruction of the long urethral defect
development involves the processes of fibrosis [12–14]. However, in addition to potential
and cicatrix formation in the urethral mucosa donor site morbidity, there is an inadequate
and surrounding connective tissues. Strictures amount of graft in many cases. Urothelial cell
based tissue engineering has shown promise as
an alternative to urethral substitution [15–20].
In this chapter, we have evaluated currently
available studies regarding the potential of tis-
sue engineering in urethral reconstruction, in
particular those describing the use of both
acellular and recellularized tissue-­engineered
Y.-S. Ha (*) • T.-H. Kim
constructs in animal and human models.
Department of Urology, School of Medicine,
Kyungpook National University, Daegu, South Korea Possible future developments in this field are
e-mail: yunsokha@gmail.com; doctork@knu.ac.kr also discussed.

© Springer Nature Singapore Pte Ltd. 2018 215


B.W. Kim (ed.), Clinical Regenerative Medicine in Urology, DOI 10.1007/978-981-10-2723-9_9
216 Y.-S. Ha and T.-H. Kim

9.2 Tissue Engineering 9.2.1 Natural Materials


for Urethroplasty to Treat
Urethral Lesions Natural polymers may be divided into natural
polymers and acellular matrices [32, 33]. In the
To reconstruct a new tissue, a triad of compo- quest for an ideal urethral substitute, acellular
nents is needed: cells, biomaterials to be used scaffolds are promising, as they have demon-
as supports or scaffolds, and growth factors strated the ability to induce tissue regeneration
[21–23]. Various strategies have been proposed layer by layer. Following several experimental
over the years for the regeneration of urethral studies, the use of acellular matrices for urethral
tissue. A woven mesh of polyglycolic acid reconstruction has become a clinical reality over
(PGA) both with and without cells was used to the last decade. Such biomaterials are prepared
regenerate urethras in various animal models via a process leading to the removal of all cells
[24–26]. Naturally derived collagen-based and their components, i.e. decellularization. The
materials such as bladder-derived acellular remaining product is an acellular matrix, mostly
submucosa, acellular urethral submucosa, and containing collagen that (theoretically) should be
collagen gels have also been tried experimen- non-immunogenic and should cause no allergic
tally in various animal models for urethral reactions. All the above-mentioned matrices are
reconstruction [27–29]. Urethral reconstruc- biodegradable and are gradually substituted by
tion involves the use of different “patches” the host’s own intercellular matrix.
(which may be called matrices, scaffolds, or The bladder submucosal matrix proved to be
substrates) that have to be transplanted into the a suitable graft for repair of urethral defects in
urethral wall, thus providing a patent lumen. rabbits [27]. The neourethras exhibited a normal
Conventional substitution urethroplasty uti- urothelial luminal lining and organized muscle
lizes different tissues from the patient. Tissue bundles. These results were confirmed clinically
engineered urethroplasty procedures may be in a series of patients with a history of failed
performed with both naturally derived and syn- hypospadias reconstruction wherein the urethral
thetic materials [30, 31]. defects were repaired with human bladder acel-
Many pediatric and adult patients with ure- lular collagen matrices [34]. The neourethras
thral disease have been successfully treated in an were created by anastomosing the matrix in an
onlay manner using collagen-based matrices. onlay fashion to the urethral plate. The size of
One of the advantages of this method over non- the created neourethra ranged from 5 to 15 cm.
genital tissue grafts used for urethroplasty is that After a 3-year follow-up, three of the four
the material is “off the shelf.” This eliminates the patients had a successful outcome with regard to
necessity of additional surgical procedures for cosmetic appearance and function. The acellular
graft harvesting, which may decrease operative collagen-­based matrix eliminated the necessity
time as well as eliminate morbidity from the har- for performing additional surgical procedures
vest procedure. for graft harvesting, and both operative time and
One of the fundamental tasks when develop- the potential morbidity from the harvest proce-
ing tissue-engineered “patches” is forming a dure were decreased. Similar results were
support matrix that can serve as a substrate for obtained in pediatric and adult patients with pri-
cells (cell-adhesion substrate) and control the mary urethral stricture disease using the same
configuration of the tissue-engineered structure collagen matrices [35]. Another study by
and the direction of tissue regeneration. Each of El-Kassaby et al. of 30 patients with recurrent
the naturally derived and synthetic biocompati- stricture disease showed that a healthy urethral
ble materials utilized for tissue engineering has bed (two or fewer prior urethral surgeries) was
its advantages and disadvantages. needed for successful urethral reconstruction
9 Urethra 217

using acellular collagen-­ based grafts [36]. In however, the smooth muscle in the matrix was
their study, the length of the strictures ranged less extensive than in the normal rabbit urethra
from 2 to 18 cm (average 6.9 cm). All grafts and was not well oriented.
were implanted as ventral onlay patches. The The value of homologous dermal acellular
patients were followed for a mean period of matrix grafts for urethral reconstruction in
25 months. Where the patients had one or no pre- humans was investigated by Lin et al. in 2005
vious interventions, the success rate between [42]. The dermal acellular matrix graft was
both groups was similar (100% for buccal sutured to a tubular graft and it replaced the
mucosa vs. 89% for acellular matrix). defect in the urethra. Following the operation,
Feng et al. compared the mechanical proper- urethrography revealed the excellent caliber of
ties and biocompatibility of biomaterials, includ- the reconstructed urethra. Urethroscopic exami-
ing bladder submucosa, small intestinal nation showed that the graft was covered by epi-
submucosa (SIS), acellular corpus spongiosum thelial tissue and grew into the native tissue. They
matrix, and PGA, to identify the optimal scaffold suggested that the homologous dermal acellular
for urethral tissue engineering [37]. Cytotoxicity, matrix graft may serve as an ideal replacement
tensile mechanical properties, and pore size were material for complex urethral strictures or
evaluated. Smooth muscle cells were seeded onto defects, without the risk of rejection.
the biomaterials to evaluate differences in cell Fossum et al. surgically treated six boys aged
infiltration. They concluded that the acellular 14–44 months with severe hypospadias with
corpus spongiosum matrix had better overall per- autologous urothelial cell constructs [43]. A two-­
formance, indicating that an organ specific matrix staged procedure starting with repair of the chor-
may be the most suitable type of scaffold for ure- dee was performed in all cases. Urothelial cells
thral reconstruction. were harvested via bladder lavage during the first
A clinical trial using tubularized nonseeded operation and were seeded onto acellular dermis.
SIS for endoscopic urethroplasty was performed The seeded scaffold was implanted in a second
in eight evaluable patients [38]. Two patients operation in an onlay fashion. These children
with short inflammatory strictures maintained were followed up for 3.5–5 years. One patient
urethral patency. Stricture recurrence developed developed a partial stricture, which was treated
in the six patients within 3 months of surgery. conservatively, and another developed an obstruc-
Authors concluded that endoscopic urethroplasty tion in the proximal anastomosis that was man-
using an unseeded SIS graft was unsuccessful. aged successfully with internal urethrotomy. Two
On the other hand, various authors usually other children developed fistulae requiring surgi-
reported favorable results from SIS urethroplas- cal correction.
ties in humans. Palminteri et al. observed an 85%
success rate with short-term follow-up (mean
21 months) [39]. A dorsal inlay graft was per- 9.2.2 Synthetic Polymers
formed in 14 cases, ventral onlay graft in 1, and
dorsal inlay plus ventral onlay in 5. The three Synthetic polymers of naturally occurring
failures were penile repairs with long strictures α-hydroxy acids are widely used in the field of
(over 5 cm). This group updated their series in regenerative medicine. Some of these have been
2012, publishing long-term results (71 months) approved by the US Federal Drug Administration
with an overall patency rate of 76% [40]. (FDA) for use in a number of applications,
Sievert et al. performed single-stage urethral including sutures [44]. Of interest for urethral tis-
reconstruction with homologous acellular ure- sue engineering are biodegradable matrices (both
thral matrix grafts [41]. All tissue components synthetic and natural). The degradation products
were observed in the grafted matrix after of synthetic biodegradable polymers based on
3 months, with further improvement over time; α-hydroxy acids are carbon dioxide and water.
218 Y.-S. Ha and T.-H. Kim

As these polymers are thermoplastics, they can The fabrication technique of electrospinning
easily be shaped into a three-dimensional (3D) allows the fast production of high-porosity scaf-
scaffold with the required porosity and size. folds with defined shapes and architectures. Han
Synthetic polymer scaffolds designed for cell and Gouma developed electrospun bioscaffolds
transplantation were reproducibly made on a that mimic the topology of extracellular matrix
large scale and studied with respect to biocom- (ECM) [48]. ECM is a natural scaffold for cell,
patibility, structure, and biodegradation rate [45]. tissue, and organ growth. Its topology plays an
The scaffold induced chondrocyte differentiation important role in cell differentiation. The design
with respect to morphology and phenotype and challenge is to fabricate biomaterials that mimic
represented a model cell culture substrate that the ECM’s three-dimensional (3D) structures
may be useful for a variety of tissue engineering with defined shapes and complex porous archi-
applications [45]. Micos et al. suggested that bio- tecture. The urinary bladder matrix (UBM) is
degradable foams of hydrophobic polymers can used in this work as the model system of the
be efficiently wet by a two-step immersion in ECM architecture. Cellulose acetate (CA) is the
ethanol and water, which overcomes the hindered biomaterial of choice for building UBM-­
entry of water into air-filled pores [46]. In their mimicking scaffolds. Electrospinning is the fab-
study, ethanol readily enters into the porous poly- rication method used to form complex, porous,
mer, after which it is diluted and replaced by 3D structures with specific designs in a single-­
water. This method was evaluated for porous step process. Lee et al. used thermal treatments
disks of poly(l-lactic acid) (PLLA) and poly(dl-­ to enhance the mechanical properties of electro-
lactic-­co-glycolic acid) (PLGA) foams of copo- spun poly-(epsilon-caprolactone) (PCL) scaf-
lymer ratios 85:15 and 50:50. Furthermore, water folds [49]. The biomechanical properties of the
entry even after 1 h was very close to the plateau thermally bonded electrospun PCL scaffolds
value for all pre-wet polymers tested. were significantly increased without any gross
Harris et al. established open pore biodegrad- observable and ultrastructural changes when
able matrices, formed using a gas foaming compared to untreated PCL scaffolds. They sug-
method developed for fabricating matrices with- gested that the introduction of thermal fiber
out the use of organic solvents and/or elevated bonding to electrospun PCL scaffolds improved
temperatures [47]. Disks comprised of polymer the biomechanical properties of these scaffolds,
and NaCl particles were compression molded at making them more suitable for tissue engineering
room temperature and subsequently allowed to applications.
equilibrate with high pressure CO2 gas (800 psi). PGA polymers were used as tissue-engineered
Creation of a thermodynamic instability led to autologous urethras for patients who required
the nucleation and growth of gas pores in the reconstruction [50]. In this study, epithelial cells
polymer particles, resulting in the expansion of were expanded and seeded onto tubularized poly-
the polymer particles. The polymer particles glycolic acid:poly(lactide-co-glycolide acid)
fused to form a continuous matrix with entrapped scaffolds. Patients then underwent urethral recon-
salt particles. The NaCl particles subsequently struction with the tissue-engineered tubularized
were leached to yield macropores within the urethras. Urethral biopsies showed that the engi-
polymer matrix. The overall porosity and extent neered grafts had developed apparently normal
of pore connectivity were regulated by the ratio architecture by 3 months after implantation.
of polymer/salt particles and the size of the salt Tubularized urethras can be engineered and can
particles. The utility of these matrices was dem- remain functional in a clinical setting for up to
onstrated by engineering smooth muscle tissue 6 years. Kanatami et al. reported fabrication of an
in vitro. This process, a combination of high-­ optimal urethral graft using collagen-sponge
pressure gas foaming and particulate leaching tubes reinforced with Copoly(l-lactide/epsilon-­
techniques, allows the fabrication of matrices caprolactone) [P(LA/CL)] fabric [51]. The tubes
with well-controlled porosity and pore structure. were dipped in aqueous collagen solution and
9 Urethra 219

lyophilyzed to prepare the P(LA/CL)-collagen cells and culture method used, this process may
sponge graft. The grafts were applied to a 1.5 cm take 4–12 days to 3–6 weeks [50, 54, 55]. As
rabbit urethral defect (n = 14 for each condition), soon as the required number of cells is obtained,
and tissue repair was evaluated using urethro- they are seeded onto a matrix and, 1–7 days later,
graphical, urethroscopical, and histological are implanted into a patient or animal. Frequently
examination 1, 3, and 6 months after surgery. For used autologous cells are as follows: urothelial
the mesh style graft, all urethras were patent, cells from the bladder, urethra, or ureter; buccal
without fistulae or stenoses, over the entire obser- mucosa epitheliocytes; keratinocytes; fibro-
vation period. Histologically, urethral structure blasts; and smooth muscle cells [50, 54, 56–58].
was disorganized for the stent style graft, whereas De Filippo et al. showed that acellular colla-
the urethral tissue on the mesh style graft was gen matrices derived from bladder submucosa
slightly fibrotic but completely epithelialized and seeded with cells from normal urethral tissue can
supported by a regenerated smooth muscle layer be used for tubularized replacement [56]. They
at 6 months. These findings suggest that creation suggested that the collagen matrices seeded with
of a scaffold suitable for urethral tissue regenera- cells may offer a useful alternative in the future
tion depends not only on biomaterial composi- for patients requiring a tubularized urethral seg-
tion, but also on the fabrication technique. ment replacement. Li et al. investigated the fea-
sibility of urethral reconstruction using oral
keratinocyte (OK)-seeded bladder acellular
9.2.3 Hybrid or Composite Scaffolds matrix grafts [57]. In their study, OKs had good
biocompatibility with bladder acellular matrix
Hybrid or composite scaffolds are constructed grafts, and urethral reconstruction with these
from a synthetic polymer combined with a cor- grafts was better than that with bladder acellular
responding natural matrix [52, 53]. Such scaf- matrix grafts alone. Fu et al. evaluated altera-
folds were developed by researchers in the quest tions in foreskin epidermal cells, which were
for an ideal tissue-engineered material to be used used to reconstruct male rabbit anterior urethras
in urology. Hybrid scaffolds are constructed by in combination with acellular collagen matrices.
electrospinning PLGA microfibers onto the ablu- They concluded that epidermal cells seeded onto
minal surface of a bladder acellular matrix [53]. acellular collagen matrices can be successfully
There are two types of matrix used in tissue-­ used to reconstruct urethras that have defects,
engineered urethral substitution procedures: and are transformed to transitional epithelial
acellular matrices and autologous cell–seeded cells [58]. Autologous tissue-engineered buccal
matrices. Acellular matrices have been utilized mucosa was confirmed as a clinically useful
for a long period of time. An acellular matrix autologous urethral replacement tissue in a
transfer procedure will be successful if: (a) the group of patients with lichen sclerosis urethral
entire abluminal surface is infiltrated by host stricture. However, it was not without complica-
epithelial cells; (b) the defect being substituted is tions, namely fibrosis and contraction [54].
short; and (c) there is a good vascular urethral Raya-Rivera et al. constructed engineered ure-
bed. Therefore, procedures using acellular matri- thras with autologous cells and implanted them
ces are expected to fail in patients with recurring into patients with urethral defects. A tissue
strictures, marked spongiofibrosis, or long stric- biopsy was taken from each patient, and the
tures. An autologous cell-seeded matrix is a muscle and epithelial cells were expanded and
tissue-­engineered construct containing an acel- seeded onto tubularized polyglycolic acid:poly
lular matrix populated (ex vivo) with autologous (lactide-co-glycolide acid) scaffolds. Patients then
cells. A biopsy is obtained from the patient. In underwent urethral reconstruction with the tissue-
specialized sterile laboratories, cells of the engineered tubularized urethras. Tubularized ure-
desired type are harvested from the biopsy and thras can be engineered and remain functional in
grown in a culture. Depending on the type of a clinical setting for up to 6 years. These engi-
220 Y.-S. Ha and T.-H. Kim

Natural polymers
Naturally
derived
Decellularized tissues

Synthetic
Origin
(Materials)
Hybrid

Other types

Matrices used for tissue


engineering urethral Acellular
reconstruction matrices
Presence of
autologous cells Monoculture
Matrices with
autologous cells
Co-culture

Patch

Shape
Tubularized

Fig. 9.1  Matrices and cells used for tissue-engineered urethral reconstruction

neered urethras can be used in patients who need and cells used for tissue-engineered urethral
complex urethral reconstruction [50]. reconstruction.
Tissue-engineered autologous cells can be
divided into two types: tissue-engineered con- 9.2.3.1 Acellular Matrices Utilized
structs developed using a monoculture; and those in Animal Models
using co-culture. To develop tissue-engineered Kropp et al. suggested using SIS as an acellular
constructs using a monoculture, autologous cells urethroplasty matrix [60]. In one of the control
are seeded onto one of the surfaces of the support groups, full thickness preputial skin from the host
matrix and, as a rule, a certain type of epithelio- rabbit was used. The success rates were the same
cyte is used [55]. Constructs using a co-culture in both groups (100%), yet preputial graft proce-
are more complex, as the abluminal surface is dures resulted in the formation of urethral diver-
seeded with epitheliocytes, while the opposite ticulum in all eight animals.
side is seeded with either fibroblasts or smooth Chen et al. utilized BAMG (obtained and pro-
muscle cells [50, 59]. cessed from porcine bladder submucosa) for ven-
According to macroscopic parameters, tissue-­ tral onlay urethroplasties in 10 rabbits [27].
engineered constructs can be divided into patch Complete graft epithelialization was achieved
constructs and tubularized constructs. The stric- after 2 months; the migration of organized mus-
ture length to be reconstructed with patch con- cle bundles was detected 6 months after implan-
structs grafts is usually limited, as it is in cases of tation. No strictures or complications were
conventional substitution procedures. Tubularized observed.
grafts are usually developed using a co-culture In a study conducted by Nuininga et al., rab-
and may be utilized to reconstruct entire ure- bits were divided into four groups, with six ani-
thras [50]. Figure 9.1 overviews matrices mals in each, according to the type of biomatrix
used: in group 1, the rabbits underwent partial
9 Urethra 221

urethral replacement, which was replaced with defects of varying length (0.5, 1.0, 2.0, and
one layer of SIS; in group 2, four-layer SIS patch 3.0 cm) were created. At week 4, only the 0.5 cm
grafts were utilized; group 3 was treated with group had a normal layer of epithelium sur-
collagen-based matrices produced from bovine rounded by a layer of smooth muscle within the
tendons; and in group 4, the animals underwent a urethral lumen. In all groups with longer defects,
sham control operation (urethras were incised strictures developed by 4 weeks. Therefore, this
ventrally and then sutured) [61]. In groups 1 and study proved that acellular matrices in tubular-
3, complete epithelialization was observed at ized grafts may be used successfully only to
1 month after implantation, whereas in group 2, it repair defects limited in size.
was achieved at 3 months. One rabbit from group Villoldo et al. assessed the efficacy of onlay
3 developed a stricture and one rabbit from group urethroplasty with SIS in rabbits [65]. A total of
2 developed a fistula near the operation site. 30 animals included in the study had 15 mm of
Yang et al. suggested using an acellular corpus the ventral wall of the penile urethra excised. One
spongiosum matrix substitute as a patch [62]. month later, the rabbits were divided into two
These kinds of matrices were obtained from rab- groups: group 1 was the control group; in group
bit urethras and transplanted to repair urethral 2, where the created defect was patched with an
defects of 10–15 mm in length. Complete epithe- SIS onlay graft, epithelialization was achieved
lialization of the extracellular matrix was after 15 days and smooth muscle bundles were
achieved at 3 weeks post operation. Well-formed detected at 6 months follow-up. The fact that ure-
smooth muscle cells were observed after 6 weeks. thral strictures were created 1 month before ure-
No strictures or complications were reported. throplasties were performed brings this study a
Huang et al. explored the potential of SIS for step closer to clinical reality.
urethral reconstruction in rabbits [63]. Tubular Chung et al. studied the potential of using
SIS grafts were applied in group 1 (n = 6), while acellular matrices derived from silk fibroin for
a ventral onlay graft was used as a patch in group urethral repair [66]. The results of urethroplasty
2 (n = 6). The epithelium covered the grafts fully with silk fibroin matrices (SFMs) were compared
after 6 weeks. to those with SIS. The follow-up period was
An interesting study was published by 3 months, with no intermediate time points. The
Kanatani et al., wherein two types of tubularized success rates of SFM and SIS were found to be
graft based on type I collagen sponge, reinforced the same. No strictures or complications were
with co-poly(Lactide/ε-caprolactone) (CLLC) reported. However, the silk fibroin scaffolds
fabric, were created [51]. In group 1, the P(LA/ showed reduced immunogenicity.
CL) fibers were knitted into a vascular stent Kajbafzadeh et al. explored the role of prepu-
style; in group 2, the P(LA/CL) fibers were tial acellular matrix (PAM) for urethral recon-
weaved into a mesh style. Each of the grafts was struction [67]. Prepuces were obtained from
applied to a 15-mm urethral defect. Numerous circumcised boys. The prepuces were decellular-
complications (stenoses, fistulae, or stone for- ized and the produced matrices were used as
mation) were observed in group 1, whereas no patches in ventral onlay urethroplasty proce-
complications were seen in group 2. The authors dures: PAM in group 1; and PAM plus fibrin
concluded that creation of a scaffold suitable for sealant in group 2. The effectiveness of repair
urethral tissue regeneration depends not only on was evaluated in both groups at a 9 month fol-
the biomaterial composition, but also on the fab- low-up. The authors noted that satisfactory vas-
rication technique. cularity and smooth muscle layer formation
In a very important study, Dorin et al. deter- were more significant in group 2 (PAM + fibrin
mined the maximum potential distance of normal sealant).
native tissue regeneration when using tubularized In all the above-mentioned studies, only
unseeded matrices [64]. Twelve rabbits were male rabbits were used as animal models. In
divided into four groups. In each group, urethral most studies, a longitudinal urethral defect was
222 Y.-S. Ha and T.-H. Kim

created, varying in length (10–20 mm); in some and hypospadias. No episodes of rejection were
studies it was a tubular defect 5–30 mm long. observed. During the 46 month follow-up period,
SIS was used as a urethroplasty matrix in four only four patients needed periodical urethral
of ten studies, with BAMG being the second dilatation.
most ­ frequently utilized type of scaffold Le Roux et al. evaluated SIS as a substitute in
(applied in three of ten studies). One should endoscopic urethroplasty [38]. Nine patients
note that no episodes of graft rejection were were enrolled. Optical urethrotomy was per-
reported. Acellular matrices were found to be formed prior to the SIS graft implantation.
highly effective in animal models, yet their Subsequently, a tubularized SIS graft was
application range was limited by the size of implanted into the stricture site. Only two patients
urethral defect. Complete formation of the uro- had patent lumen at 1 and 2 years follow-up,
thelial layer on the inner surface of the graft respectively; all the other patients developed
was achieved 4–12 weeks after implantation; recurring strictures. The authors concluded that
either single smooth muscle cells or a regener- endoscopic urethroplasty with unseeded SIS
ated smooth muscle layer was observed grafts is not to be recommended.
2–12 months after implantation. Donkov et al. assessed the success rates of SIS
grafts used for dorsal onlay substitution urethro-
9.2.3.2 Acellular Matrices Utilized plasty [69]. The graft was fixed using a modified
in Human Models Barbagli technique. The success rate was 89%
The feasibility of applying a bladder submucosal, (eight of nine patients) at 18 months follow-up.
collagen-based inert matrix as a free graft substi- While Hauser et al. also chose SIS for dorsal
tute for urethral repair in patients with hypospa- onlay substitution urethroplasty, the success rate
dias was explored [34]. All four patients had had was much lower: only one of five patients did not
a history of hypospadia repair procedures and have stricture recurrence at a 12 month follow-up
required yet another repair. The neourethras were [70]. The complications observed postopera-
created from BAMGs in the size range 5–15 cm. tively were extravasation, severe urethritis, and
Postoperatively, only one patient developed a fis- urinary tract infection.
tula. Histological evaluation showed typical ure- Palminteri et al. evaluated the role of SIS
thral stratified epithelium at the site of surgery. grafts for substitution urethroplasties [39]. Three
Mantovani et al. were the first to utilize an SIS techniques were employed: a dorsal inlay graft
graft for substitution urethroplasty in a 72 year-­ was performed in 14 patients, ventral onlay in
old patient with a long stricture of the penile and one patient, and dorsal onlay plus ventral onlay in
bulbar urethra [68]. The dorsal onlay technique five patients; 21 month follow-up data demon-
was used. At a 16 month follow-up, the maxi- strated an average success rate of 85%. The aver-
mum urine flow rate was 14 ml/s. No complica- age stricture length was 3 cm. No complications
tions were observed intraoperatively or were noted. The three failures were in penile and
postoperatively. In the same year, El-Kassaby penile–bulbar urethral strictures with an average
et al. used a BAMG matrix for urethral stricture length of 5.7 cm.
repair [35]. A total of 28 patients with urethral Fiala et al. used SIS grafts for urethroplasties
strictures of varied length (1.5–16 cm) underwent in 50 patients [71]. The success rate averaged
reconstructive surgery wherein the ventral onlay 80%, with a mean follow-up time of 31 months.
technique was used. The success rate was 86%, Recurring strictures developed in one of 10 bul-
as one patient developed a urethrocutaneous fis- bar, five of 31 bulbopenile, and four of nine penile
tula that closed spontaneously 1 year after repair. strictures; these occurred during the first 6 months
Lin et al. suggested using an acellular dermal postoperatively. Farahat et al. placed SIS grafts
matrix (ADM) graft for urethral reconstruction endoscopically [72]. Only two cases of 10 exhib-
[42]. Homologous ADM was applied as a tubu- ited stricture recurrence at a mean follow-­up time
larized graft in 16 patients with urethral strictures of 14 months; there were no complications.
9 Urethra 223

El-Kassaby et al. conducted a comparative length in the former study was 2.2 cm, as
study between BAMGs and buccal mucosal opposed to 1.47 cm in the latter study.
grafts in anterior urethral strictures [36]. The Apparently, the difference in the success rates
patients were followed for 25 months on average. can be attributed to these two factors.
The success rate for BAMG was 53%, and that of In addition, Donkov et al. and Hauser et al.
the buccal mucosal graft was 100%. The authors both applied SIS in a similar technique (dorsal
divided patients from the two groups into sub- onlay substitution), but the success rates were
groups: (a) those with a healthy urethral bed (less completely different (89% vs 20%, respectively)
than two prior surgeries), where BAMG surgeries [69, 70]. The difference can be explained by the
were successful in 89% of cases and buccal difference in the way SIS was secured: in the
mucosa surgeries in 100%; and (b) those with an study by Donkov et al. [69], the patch was spread-­
unhealthy urethral bed (more than two prior sur- fixed onto the tunica albuginea, while the mucosa
geries), where the success rates were 33% was sutured to the graft margins; in the study by
(BAMG) and 100% (buccal mucosa). Hauser et al. [70], SIS was anastomosed only to
Palminteri et al. reported the longest (as of the incised urethra, without being fixed onto the
today) mean follow-up period (71 months), hav- tunica albuginea. Moreover, the strictures in the
ing worked with patients who had bulbar urethral former study were shorter than those in the latter
strictures and underwent urethroplasties using (4–6 cm vs 3.5–10 cm).
SIS [40]. The success rate was 76%. It should be
noted that the failure rate was 14% for strictures 9.2.3.3 Cellularized Matrices Utilized
<4 cm and 100% for strictures >4 cm. in Animal Models
In the studies mentioned above, the most fre- (Monoculture)
quently used type of matrix was SIS, which De Filippo et al. were the first to use a tubularized
was, in most cases, used as a material for patch tissue-engineered construct derived from bladder
grafts. Tubularized grafts were applied twice: submucosa and seeded with autologous urothe-
in endoscopic urethroplasties (with a success lial bladder cells for urethroplasty in rabbits [56].
rate of 22%) [38] and in conventional urethro- The cells were obtained via an open biopsy.
plasties (with a success rate of 88%); however, Urethroplasties were performed to repair 10 mm-­
when describing the cases of conventional ure- long defects. In the control group, similar defects
throplastic surgery, the authors did not mention were repaired with unseeded tubularized
the lengths of the urethral stricture [42]. No BAMGs. After 6 months of follow-up, there were
episodes of graft rejection were reported. no strictures in the former group, whereas the
Biopsies taken in five studies out of 13 showed control group exhibited strictures at the site of
normal urethral tissue characteristics at the site surgery.
of surgery. Almost all studies using acellular A similar study was conducted by Li et al.,
matrices reported success rates of 75% or more. although instead of urothelial bladder cells they
Failures were more common in patients with used OKs [57]. The ventral onlay graft procedure
long urethral strictures (>4 cm), in those who was used to repair 20 mm-long defects. Rabbits
had had previous urethroplasties (unhealthy in the experimental group (BAMG plus OKs)
urethral bed, unsatisfactory vascularity), and in developed no strictures or complications; in the
those with penile or penile–bulbar strictures. control group, two rabbits died of infection, two
The two studies that used endoscopic had fistulae, and the remaining eight developed
approaches exhibited different success rates: strictures.
22% vs 80% [38, 72]. The key differences Fu et al. used tubularized BAMGs seeded with
among the groups were in the way the SIS autologous preputial keratinocytes to repair
grafts were secured: Le Roux et al. used tubu- 15 mm-long defects in three rabbits; there was no
larized grafts [38], while Farahat et al. chose control group in the study [58]. At a 2 month fol-
patches [72]. Additionally, the average stricture low-up, urethrography showed a wide urethral
224 Y.-S. Ha and T.-H. Kim

caliber in all three animals, and histological evalu- derived stem cells [75]. BAMGs were labelled
ation showed complete epithelialization at the site with 5-bromo-2′-deoxyuridine. Thirty-six rabbits
of surgery. were divided into three groups (12 animals/
Of particular interest is the paper published by group); 20 mm ventral urethral defects were cre-
Gu et al. [73] They implanted silastic tubes into ated. In group A, defects were substituted only
the peritoneal cavities of nine rabbits. After with BAMGs; in group B with BAMGs plus
2 weeks, the authors harvested the tubes covered TGFβ1 siRNA-transfected fibroblasts (Und-­
by tissue, histological analysis of which revealed rASCs); and in group C with BAMGs plus
myofibroblasts embedded in collagen bundles epithelial-­differentiated rabbit adipose-derived
covered by an outer layer of mesothelium. The stem cells (Epith-rASCs). Only one stricture was
tissue was everted and used as a tubularized graft observed in group C, whereas almost all animals
to repair 15 mm-long urethral defects. At 1 month developed strictures in the first two groups.
post operation, histological and immunohisto- Complete epithelialization was achieved only in
chemical analyses showed normal urethral group C.
architecture. Wang et al. used a denuded human amniotic
In their study, Micol et al. used high-density scaffold (dHAS); 5 × 10 mm defects were cre-
collagen gel tubes as grafts that were seeded ated on the ventral wall of the urethra and were
with autologous bladder smooth muscle cells repaired with either dHAS alone (group 1,
[29]. It took the authors approximately 24 h to n = 6) or dHAS + urethral urothelial cells (group
produce each tubularized graft. They did not 2, n = 6). In group 1, one animal developed an
place catheters postoperatively. Seven of six- infection and one had a fistula; group 2 exhib-
teen rabbits developed fistulae. In both groups ited no complications or strictures (the efficacy
(with eight animals in each), equal numbers of rate was 100%). Mild inflammatory infiltration
smooth muscle cells were observed after was observed in cell-seeded dHAS grafts, as
1 month. revealed by less pronounced accumulation of
Gu et al. isolated mesothelial cells via omen- CD4+ and CD8+ cells (or neutrophils or other
tum biopsy and seeded them onto BAMGs [74]. immune cells). Histopathological analysis iden-
Fifteen mm-long defects were substituted by tified a mild immune response in the cell-seeded
tubularized matrices seeded with cells (in nine dHAS group (p < 0.05). Urethral defects in
rabbits) and by those without cells (in nine rab- group 2 were completely resolved in 1 month.
bits from the control group). In the latter, all ani- At 3 months after surgery, the formation of a
mals developed strictures, while no stricture smooth muscular layer and rich blood vessels
formation was observed in the experimental was apparent [76].
group. At 6 months after implantation, the neo-­ In a very recently published Korean study by
urethras could not be histologically distinguished Chun and Kim et al., they evaluated the com-
from the native urethras. bined effect of acellular bladder submucosa
Xie et al. prepared electrospun silk fibroin matrix (BSM) and autologous urethral tissue for
matrices and seeded them with urothelial blad- the treatment of long segment urethral stricture
der cells that had been isolated and expanded in a rabbit model [20]. To prepare the BSM, por-
[18]. Dorsal urethral defects 30 mm long were cine bladder submucosa was processed, decel-
created in female dogs. At 6 months after lularized, configured into a sheet-like shape, and
implantation, the neo-urethras could not be his- sterilized (Fig. 9.2). Twenty rabbits were ran-
tologically distinguished from the native ure- domly divided into groups: normal control, ure-
thras. It should be noted that the artificial defects thral stricture, urethroplasty using BSM only, or
involved only the mucosa, not the smooth mus- BSM/autologous urethral tissue (n = 5 per
cle layer. group). Brief urethroplasty methods are rep-
Li et al. seeded BAMGs with either epithelial resented in Fig. 9.3. The width of the penile
differentiated or undifferentiated rabbit adipose-­ urethra was measured by postoperative
9 Urethra 225

a b

c d

Fig. 9.2 Preparation of bladder submucosa matrix ized by a thin sheet structure, acellular composition, mul-
(BSM). (a) Bladder extraction from pig. (b) Separation of tidirectional tensile strength, and lack of chemical
submucosa. (c) Gross image of acellular BSM, character- cross-links. (d) Lyophilized acellular BSM sheet

a b c d

Fig. 9.3  Process of nontransected ventral urethral stric- normal urethra. (c) Embedded urethral tissue on the BSM
ture model generation and urethroplasty. (a) Urethral with fibrin glue (shown below the magnified figure).
defect created by urethrectomy and suturing. (b) Excision Prepared combined graft with BSM and autologous ure-
of healthy urethral muscle and endothelial tissue from thral tissue. (d) Completed onlay graft. Scale bar, 10 μm

u­ rethrograms at 4, 8, and 12 weeks. The control autologous urethral tissue groups at week 12
urethrogram showed a wide urethral caliber. was 10.3 ± 0.80, 3.8 ± 1.35, 8.8 ± 0.84, and
Both graft groups showed a similar width to 9.1 ± 1.14 mm, respectively. Although the dif-
those of the normal group, and the stricture ference in the width of the BSM and BSM/tissue
group revealed stenosis. The mean urethral graft urethroplasty groups was not statistically
width of the control, stricture, BSM, and ­BSM/ significant, the BSM/tissue graft group diameter
226 Y.-S. Ha and T.-H. Kim

Normal- Urethral Stricture BSM only BSM +


Urethragram Urethral Tissue

Urethral diameter
b 14
** **
** Normal
12 * *
** ** ** ** ** ** Urethral Stricture

10 BSM only
BSM+Urethral Tissue
8
mm

0
4 8 12
Weeks

Fig. 9.4  Width of the penile urethra. (a) Retrograde ure- at P < 0.05. P values were obtained with analysis of vari-
thrography (representative images taken at week 12) ance and Tukey’s test. BSM, urethrotomy, and onlay ure-
shows complete tubularization of both grafted urethras throplasty with an acellular BSM scaffold graft; BSM/
similar to the control group throughout the study period. tissue, urethrotomy and onlay urethroplasty with a graft
The stricture group shows stenosis. (b) Urethral diameter composed of autologous urethral tissue and an acellular
measured at 4, 8, and 12 weeks post operation. The sym- BSM scaffold
bols on the top of the bars indicate significant differences

was approximately 0.567 mm wider (Fig. 9.4). activators for incorporation, promoting ingrowth
The histopathology study revealed that the of surrounding urethral tissue into the scaffold.
­BSM/autologous urethral tissue graft had a nor- This indicates that autologous urethral tissue at
mal urethral lumen area, compact muscular lay- the time of implantation is effective for urethral
ers, complete epithelialization, and progressive reconstruction. A BSM graft embedded with
infiltration by vessels in the regenerated urethra. autologous urethral tissue can be applied for
In contrast, the BSM grafts revealed keratinized clinical treatment of strictures requiring par-
epithelium, abundant collagenized fibrous con- tially resected urethra replacements.
nective tissue, and were devoid of bundles of Male rabbits were used in ten of the eleven
circular smooth muscle (Fig. 9.5). According to studies reviewed above. Female dogs were used
these results, the BSM/tissue graft had well- as animal models in one study. Despite the fact
organized integration with the recipient tissues, that preclinical and clinical studies demon-
and the cells within embedded autologous tissue strated better success rates with SIS, six of the
and BSM can synchronistically act as biological eleven reviewed studies utilized BAMGs. SIS
9 Urethra 227

After 12 weeks.
* Masson’s Trichrome
a b c d
Red : Muscle, keratin
Blue : Collagen

* IHC
Red : Pan-cytokeratin
(Epithelium stain)
Blue : DAPI
(Nucleus stain)

e f g h
H&E

i j k l
Masson’s-
Trichrome

m n o p
IHC

Scale bar 10 µm
BSM+
Normal Urethral Stricture BSM only
Urethral Tissue

Fig. 9.5  Histological, Masson’s trichrome, and immuno- cle, similar to controls. BSM grafts show fibrosis-like
histochemical analysis of harvested urethra. (a–d) Low-­ morphology and a simple smooth muscle layer as
magnification images of H&E-stained sections (40×); observed in the stricture group. (i–l) BSM/tissue grafts
both graft groups show thick circular smooth muscle and reveal scattered, variably sized circular bundles of smooth
a similar area of the urethral lumen as observed in the con- muscle. BSM grafts show few muscle fibers, extensive
trol group. The stricture group shows loose muscular lay- collagen deposition, and keratinized squamous cell epi-
ers and narrow lumen. (e–h) High-magnification images thelium. (m–p) BSM/tissue grafts express stratified
of H&E-stained sections (200×); the BSM/tissue grafts columnar urothelium and stratified squamous cells. BSM
show compressed columnar epithelium, newly formed grafts have a thin and irregular urothelial layer (400×)
capillaries, and abundant circular bundles of smooth mus-

grafts were not applied at all. In most studies, smooth muscle cells at the site of surgery were
matrices were seeded with epithelial cells. observed at a mean follow-up time of 4 weeks.
Also, ­mesothelial cells, bladder smooth muscle
cells, bone marrow mesenchymal stem cells, 9.2.3.4 Cellularized Matrices Utilized
and rabbit adipose-derived stem cells were in Human Models
used. The authors did not mention any prob- (Monoculture)
lems with expanding the cells or seeding them Fossum et al. obtained urothelial cells via bladder
onto different matrices; 50% of the surgeries washing [55]. The cells were cultured and seeded
were performed with tubularized constructs; onto decellularized dermal matrices that were
and 8 of 11 studies had control groups, where used to surgically treat six boys with scrotal or
similar matrices without cells were used. All 11 perineal hypospadias. The mean follow-up time
studies showed significantly better results was 87 months. Only one boy had to undergo an
with cellularized matrices. Epithelial cells and optical urethrotomy to improve voiding.
228 Y.-S. Ha and T.-H. Kim

9.2.3.5 Recellularized Matrices Utilized those in groups (b) and (c) had none. However, in
in Animal Models (Co-Culture) group (c) the formation of capillaries in the epi-
Feng et al. seeded two types of autologous cells thelial lower layer was observed at 6 months after
(lingual keratinocytes and corporal smooth mus- implantation.
cle cells) onto both sides of porcine acellular cor- Xie et al. seeded autologous OKs and fibro-
pus spongiosum matrices (ACSMs) [59]. The blasts onto silk fibroin matrices; 5 cm urethral
best results were achieved in the group in which mucosal defects were repaired using matrices
urethral defects were repaired with matrices con- alone and matrices with two cell types (in five
taining both types of cells. Animals in the other female dogs in each case) [80]. No strictures or
two groups (ACSMs alone, and ACSMs plus complications developed in the group with cell-­
OKs) developed strictures. seeded matrices.
De Filippo et al. substituted 30 mm defects Tissue-engineered constructs with different
with either tubularized BAMGs alone, or BAMGs cell types in a co-culture system are structurally
seeded with two types of cells (BAMGs plus more similar to the native urethra than other con-
BUCs plus BSMCs) [77]. The success rate in the structs. In three of the six studies reviewed above,
group wherein cell-seeded matrices were utilized BAMGs were utilized. The inner surface of the
was 100%. matrix was seeded with OKs (in four of six stud-
Mikami et al. formed tubularized tissue engi- ies) or bladder urothelial cells (in two of six stud-
neered urethras comprised of collagen mesh ies), while the outer surface of the matrix was
matrices and autologous cells of two types— seeded with smooth muscle cells or fibroblasts
keratinocytes and buccal mucosa smooth muscle (in two of six studies). Each study included a
cells [78]. These grafts were used to repair ure- control group in which matrices without cells
thral defects in the experimental group (10 dogs) were used. The authors did not mention any prob-
and were not applied to defects in the control lems regarding culturing autologous cells or cre-
group (10 dogs). The authors reported statisti- ating tissue-engineered constructs with two types
cally significant differences in the number of of cells. The animal models were male rabbits (in
complications between the two groups. The con- three of six studies), male dogs (in two of six
trol dogs developed fistulae and strictures. studies), and female dogs (in one study).
Orabi et al. used the same types of matrix in Tubularized constructs were applied in 50% of
male dogs; 6 cm perineal urethra segments were studies. The authors used tissue-engineered con-
removed and substituted with either tubularized structs containing different cell types with 100%
matrices with two types of cells (BAMGs plus success in five of six studies and with 70% suc-
BUCs and BSMCs) or BAMGs alone (control cess in one study.
group) [19]. In the control group, 100% of ani-
mals developed strictures and fistulae, whereas in 9.2.3.6 Recellularized Matrices Utilized
the experimental group there were no strictures at in Human Models (Co-Culture)
the 12 month follow-up. Bhargava et al. were the first to use a tissue-­
Li et al. hypothesized that TGFβ1 plays a very engineered construct for urethral repair in clini-
important role in the formation of fibrosis. Using cal patients [54]. Sterilized donor de-epidermized
OK- and TGFβ1 siRNA transfected fibroblast-­ dermis was utilized as a matrix, onto which buc-
seeded BAMGs, the authors were able to mini- cal mucosa keratinocytes and fibroblasts were
mize the activity of TGFβ1 and avoid the seeded. Five patients with urethral stricture sec-
formation of fibrosis [79]. In three groups of rab- ondary to lichen sclerosis participated in the
bits (nine animals/group) the urethral defects study. Three patients underwent a two-stage
were repaired with: (a) BAMGs alone; (b) procedure and two patients a one-stage proce-
BAMGs plus OKs; and (c) BAMGs plus OKs dure. Postoperatively, one patient required com-
plus TGF-β1-siRNA transfected fibroblasts. plete graft excision because of marked fibrosis,
Rabbits in group (a) developed strictures, while and one had partial graft excision due to
9 Urethra 229

h­yperproliferation of tissue. Three patients tissue-engineered constructs?” At present, clini-


required some form of instrumentation because cians are successfully utilizing oral (buccal)
of recurring strictures. At present, the mean fol- mucosa to repair complex urethral strictures and
low-up time is 111.8 months [81]. All patients hypospadias. Its use has yielded good results, not
who underwent urethroplasties are able to void only in patients with primary urethral strictures
independently. All four patients with tissue- but also in those with recurring strictures and
engineered buccal mucosa (TEBM) void spon- lichen sclerosis, with most authors reporting low
taneously. Three of five patients used intermittent donor-site morbidity (sometimes extremely low,
self-calibration. especially when mucosa is harvested from the
Raya-Rivera et al. used a PGA matrix, of cheek or the ventral surface of the tongue).
which the inner surface was seeded with expanded Moreover, the procedure of harvesting is simple,
bladder epithelial cells and the outer surface with reproducible, and well tolerated by the majority
bladder smooth muscle cells [50]. All patients of patients. The view that oral mucosa may not
had post-traumatic obliteration of membranous provide enough tissue for urethral repair seems to
urethra. They had scar tissues excised and be groundless. Mucosa from both cheeks as well
tissue-­engineered urethras implanted. The suc- as from the ventral surface of the tongue may
cess rate was 100% at a 71 month follow-up. provide enough material to create a 16 cm graft.
Biopsies showed that the grafts had developed a While buccal mucosa possesses many advan-
normal-­appearing architecture by 3 months after tages, its use is associated with a number of prob-
implantation. lems that have to be addressed. First, any graft
To sum up, the authors applied tissue-­ harvesting procedure entails some degree of
engineered constructs with two types of cells to donor site morbidity, which, no matter how mini-
manage very complicated cases of urethral stric- mal, still has a negative impact on the patients.
ture disease: lichen sclerosis in Bhargava et al. The larger the amount of tissue removed from the
and post-traumatic membranous urethral obliter- donor site, the higher the negative impact.
ation in Raya-Rivera et al. [50, 54]. In the former, Second, long strictures may require harvesting of
additional interventions were needed, while in long grafts from several donor sites. This
the latter, the use of tubularized synthetic matri- increases surgical duration as well as intraopera-
ces seeded with urothelial and smooth muscle tive and postoperative complication rates [82].
cells obtained from bladder demonstrated 100% Third, in cases of recurrent strictures where a
success at long-term follow-up. Histological secondary urethroplasty is needed, the lack of
evaluation showed that the engineered grafts had oral mucosa may drive the need to find other sub-
developed apparently normal architecture by stitution tissues. A similar problem may arise
3 months after implantation. All adverse fibrotic during the first stage of a two-stage reconstruc-
reactions were observed within the first year after tion that often requires additional epithelializa-
implantation. The results have been durable for tion of the urethral bed. Thus, selecting and
three out of five patients for 9 years, although all designing an ideal substitution material remains a
of them required re-interventions [81]. great challenge. Tissue-engineering options may
help address this challenge. The development of
a perfect tissue-engineered construct (matrix +
9.3 Current Challenges cells obtained using a non-invasive technique,
and Future Perspectives possibly from urine) will eliminate the problems
of donor site morbidity and limited amount of
Urethral tissue engineering provides fertile available substitution tissue [83, 84], even in
ground for further research, with many problems patients with long strictures and in those who
remaining unresolved. One of the questions to be have had previous urethroplasties. It will also
answered is, “What are the potential indications decrease operative time and the risks of intraop-
for substitution urethroplasty procedures using erative and postoperative complications.
230 Y.-S. Ha and T.-H. Kim

Among the problems our analysis of the mostly collagen and were devoid of any native
reviewed preclinical studies brings to light is the cellular components, thus having reduced immu-
problem of developing animal models for ure- nogenicity and bioactivity. The success of acel-
thral repair. Only one preclinical study reviewed lular matrices is known to be contingent on the
above created a model of urethral stricture: its vascularization of the graft and regeneration of
authors excised a segment of the urethra 1 month native mucosa at the implantation site. Moreover,
before urethroplasties were performed, thus sim- an ideal matrix must be biodegradable, its break-
ulating the conditions under which grafts nor- down products must be non-toxic, and it must be
mally take place [65]. In most of the reviewed substituted by the host’s own intercellular matrix
preclinical trials, a urethral defect was created [86, 87]. Also, an ideal matrix should provide an
intraoperatively, to be immediately repaired with environment in which adhesion, proliferation,
a matrix. However, the process of graft take in an migration, and differentiation of cells can occur
injured urethra may substantially differ from that to enable functional tissue formation [30, 31].
in a normal urethral bed. What is more, only four Therefore, when an acellular matrix is used for
out of twenty-eight preclinical studies used large urethral reconstruction, the following processes
animals (dogs) [18, 19, 78, 80], and only two of should take place: (a) the matrix becomes cov-
them used male dogs [19, 78]. While we are ered with the native urothelium; (b) it completely
aware that, anatomically, the female and male degrades; and (c) it becomes substituted by the
urethras are different, and that spongiofibrosis, host’s extracellular matrix (ideally, with smooth
being the major cause of strictures in males, muscle fibers being formed).
never occurs in females, we still decided to Another issue to be touched upon is the effi-
include the studies conducted on female animals cacy of acellular matrices in both preclinical and
in our review. The reason for doing so was that clinical studies. As was shown by Dorin et al.
the authors of the studies introduced a novel tech- [64], urothelial regeneration has some limita-
nique to prepare matrices by electrospinning, and tions, which is why acellular matrices are less
suggested using silk fibroin matrices [18, 80]. As successful in patients with long urethral strictures
for developing animal models for urethral repair, and in those who have had previous repairs
Sievert et al. introduced three methods of stric- (unsatisfactory vascularity). Hence, it is not
ture induction in large animals (minipigs): ure- advisable to use acellular matrices to repair stric-
throtomy, ligation, and thermocoagulation [85]. tures longer than 2 cm. As for short strictures
Stricture severity was found to be higher in the (<2 cm), which, in theory, can be reconstructed
model that used thermocoagulation. From these with acellular matrices, they are successfully
results, we may assume that it is best to conduct managed using either primary anastomosis or a
experiments on large animals and develop a non-transecting anastomotic technique [7, 88]. In
model of urethral stricture to mimic the condi- cases of longer strictures, where urethroplasty is
tions of graft take in humans. indicated [6], the use of acellular matrices does
It should be noted that SIS grafts were the not seem to be expedient. Patients with an
most frequently applied matrices in the preclini- unhealthy urethral bed (severe spongiofibrosis,
cal and clinical studies reviewed above. They recurring strictures, or previous urethroplasties)
were used in five of eleven preclinical studies and have higher risks of poor graft vascularization,
in nine of thirteen clinical studies. Synthetic graft contraction, and stricture recurrence, as was
matrices were not used at all, with the exception shown in several clinical trials [21, 39, 40, 70].
of one study that utilized a hybrid matrix [CLLC To sum up, the applications for acellular matrices
plus collagen sponge tubes (CST)]. All the matri- in the treatment of urethral strictures are limited.
ces took well and caused no immunogenic and/or Problems with acellular matrices made
proinflammatory reactions. All of them (except researchers look for other options. This is why
CLLC plus CST, and SFM) were homogeneous they turned to designing tissue-engineered con-
or heterogeneous acellular grafts that contained structs, containing not only a matrix but also
9 Urethra 231

autologous cells. Interestingly, the cells seeded in such cases, the inner surface is seeded with
onto the surface of a matrix (when cultured epithelial cells, while the outer surface is seeded
in vitro and immediately after implantation onto with smooth muscle cells. Preclinical studies
a patient) survive by molecular diffusion. Later, mostly applied BAMGs, whereas two clinical
the seeded cells enter a hypoxic state, thus stimu- studies used ADMs and PGA matrices (thus, in
lating the endogenous release of angiogenic the reviewed studies, a synthetic matrix was used
growth factors [89, 90]. If adequate angiogenesis only once). The inner surface was seeded either
is achieved, the cells expand and new tissue is with bladder epithelial cells or with buccal
formed. In the absence of adequate angiogenesis, mucosa cells. The use of bladder urothelial cells
the cells die. seems to be preferable; however, the main draw-
It is noteworthy that in the experimental stud- back is that they are obtained via an open bladder
ies that compared the efficacy of acellular and biopsy. Therefore, in this respect, buccal mucosa
cellularized tissue engineered constructs, failure cells appear to be a better choice, as the technique
rates when using acellular matrices were signifi- employed to obtain them is less invasive.
cantly higher than were those in the studies that Another important question regards graft
utilized only acellular matrices. This may be placement. Ventral graft placement is preferred in
explained either by some degree of bias or by dif- patients with urethral strictures located in the
ferences in study designs—the lengths of the ure- proximal bulbar urethra [14, 92, 93]. This is
thral defects in the studies presented in the animal because, in the proximal bulbar urethra, the
models using acellular matrices were 5–20 mm, lumen is dorsally eccentric, and the ventral aspect
whereas the lengths of the urethral defects in the of the surrounding tissues of the corpus spongio-
studies presented in the animal models using cel- sum is rather thick. This thick portion of the bul-
lularized matrices varied from 10 to 30 mm. bar urethra has sufficient blood supply to
While SIS has been successfully utilized as a vascularize the graft. Moving distally, the ure-
material for grafts in earlier trials, it was not used thral lumen becomes more centrally placed and
in the preclinical and clinical studies reviewed the corpus spongiosum becomes thinner, which
here. Most preclinical studies applied BAMGs, is why the use of ventral graft location may be
while two clinical studies used MukoCell® associated with higher risks of inadequate vascu-
matrices and ADMs. Furthermore, the authors larization and stricture recurrence. Thus, in
seeded matrices with different types of autolo- patients with strictures located in the distal bulbar
gous epithelial cells, smooth muscle cells, and urethra, one should opt for dorsal (onlay or inlay)
stem cells. Preclinical studies showed better suc- graft placement. This is why we believe dorsal
cess with cellularized matrices than with acellu- graft placement should be preferred in clinical
lar ones. Fossum et al. obtained urothelial cells studies assessing the use of tissue-engineered
via bladder washing [55]. Urethroplasties with constructs.
this type of cell were rather successful (83%) at a Overall, the evidence presented in the
mean follow-up time of 87 months. We believe reviewed studies appears to support the view
bladder washing is a promising technique, as it is that tissue-engineered constructs with different
minimally invasive and simple. Recently, some cell types should be used as grafts of choice in
authors have reported that stem cells can also be substitution urethroplasty procedures. Tissue-­
obtained from urine [83, 84, 91], which makes engineered constructs should be utilized in the
this approach even more attractive. following groups of patients: in those with
Since the native urethra is comprised of the long strictures (>2 cm); in those with multiple
inner epithelial lining and corpus spongiosum strictures; in those with subtotal and total stric-
that contain endothelial and smooth muscle cells, tures; in those with an unhealthy urethral bed
the next step taken by some researchers was to (severe spongiofibrosis, recurring strictures
create a tissue-engineered construct that would after multiple urethrotomies and dilations, pre-
contain a matrix and two types of cell. As a rule, vious urethroplasties); and in those with soft
232 Y.-S. Ha and T.-H. Kim

tissue deficit at the site of surgery (hypospa- References


dias, failed hypospadia repair). The use of
“off-the-shelf” tissue engineered constructs 1. Bhatnagar A, Upadhyaya VD, Kumar B. Congenital
urethrocutaneous fistula: case report with review of
can minimize (or, in cases where non-invasive literature. Indian J Plast Surg. 2012;45:563–5.
techniques to obtain autologous cells are 2. Elgammal MA. Straddle injuries to the bulbar urethra:
employed, even eliminate) donor site morbid- management and outcome in 53 patients. Int Braz J
ity, decrease operative time, and reduce the Urol. 2009;35:450–8.
3. Kim BS, Kwon TG. Urethral reconstruction using
risks of intraoperative and postoperative com- autologous vein grafts for the management of urethral
plications. However, further investigations are strictures. Curr Urol Rep. 2015;16:467.
needed to prove this. 4. Macedo A Jr, Rondon A, Ortiz V. Hypospadias. Curr
Interestingly, despite the fact that there has Opin Urol. 2012;22:447–52.
5. Chapple C. Anterior urethral surgery: current con-
been much international debate regarding natu- cepts and future directions. Eur Urol. 2010;58:42–5.
rally derived vs synthetic scaffolds, almost all 6. Chapple C, Andrich D, Atala A, Barbagli G,
the studies reviewed here utilized natural matri- Cavalcanti A, Kulkarni S, et al. SIU/ICUD consulta-
ces. Only two studies applied a hybrid matrix tion on urethral strictures: the management of anterior
urethral stricture disease using substitution urethro-
and a synthetic one. Therefore, more attention plasty. Urology. 2014;83:S31–47.
should be paid to synthetic matrices and their 7. Morey AF, Watkin N, Shenfeld O, Eltahawy E,
possible applications in substitution urethroplasty Giudice C. SIU/ICUD consultation on urethral stric-
procedures. tures: anterior urethra--primary anastomosis. Urology.
2014;83:S23–6.
Urethral tissue engineering is, without any 8. Fenton AS, Morey AF, Aviles R, Garcia CR. Anterior
doubt, a promising field of regenerative medi- urethral strictures: etiology and characteristics.
cine. Yet creating a tissue-engineered construct Urology. 2005;65:1055–8.
with different types of autologous cells is a 9. Tritschler S, Roosen A, Fullhase C, Stief CG, Rubben
H. Urethral stricture: etiology, investigation and treat-
complex biotechnological process, requiring ments. Dtsch Arztebl Int. 2013;110:220–6.
clean room laboratories and highly specialized 10. Lumen N, Hoebeke P, Willemsen P, De Troyer B,
personnel. A lack of the mentioned facilities Pieters R, Oosterlinck W. Etiology of urethral stricture
and specialists, as well as a lack of clinical disease in the 21st century. J Urol. 2009;182:983–7.
11.
Chapple C, Barbagli G, Jordan G, Mundy
studies, limit the wide application of tissue AR, Rodrigues-Netto N, Pansadoro V, et al.
engineering to replace urethras. Urethral tissue Consensus statement on urethral trauma. BJU Int.
engineering provides effective treatment 2004;93:1195–202.
options for patients with strictures and congeni- 12. Bhandari M, Dubey D, Verma BS. Dorsal or ventral
placement of the preputial/penile skin onlay flap for
tal anomalies. However, with the development anterior urethral strictures:does it make a difference?
of biotechnologies in this field, we will harness BJU Int. 2001;88:39–43.
new research results that will enable us to tackle 13.
Kozinn SI, Harty NJ, Zinman L, Buckley
other problems as well, such as various lesions JC. Management of complex anterior urethral stric-
tures with multistage buccal mucosa graft reconstruc-
of the bladder and ureter, chordee, or erectile tion. Urology. 2013;82:718–22.
dysfunction. 14. Barbagli G, Lazzeri M. Penile urethral stricture recon-
struction--flap or graft? Graft. J Urol. 2011;186:375–6.
Conclusions 15. Venn SN, Mundy AR. Urethroplasty for balanitis

xerotica obliterans. Br J Urol. 1998;81:735–7.
The studies reviewed in this chapter have 16. Xu YM, Xu QK, Fu Q, Sa YL, Zhang J, Song LJ,
described different approaches to the develop- et al. Oral complications after lingual mucosal graft
ment of matrices, harvesting and expanding harvesting for urethroplasty in 110 cases. BJU Int.
autologous cells, and producing tissue-­ 2011;108:140–5.
17. Cattan V, Bernard G, Rousseau A, Bouhout S,

engineered constructs of various complexities Chabaud S, Auger FA, et al. Mechanical stimuli-­
(patch grafts and tubularized grafts). More induced urothelial differentiation in a human tissue-­
clinical studies are needed to explore these engineered tubular genitourinary graft. Eur Urol.
and other problems on a larger scale. 2011;60:1291–8.
9 Urethra 233

18. Xie M, Song L, Wang J, Fan S, Zhang Y, Xu


34. Atala A, Guzman L, Retik AB. A novel inert col-
Y. Evaluation of stretched electrospun silk fibroin lagen matrix for hypospadias repair. J Urol.
matrices seeded with urothelial cells for urethra 1999;162:1148–51.
reconstruction. J Surg Res. 2013;184:774–81. 35. El-Kassaby AW, Retik AB, Yoo JJ, Atala A. Urethral
19. Orabi H, AbouShwareb T, Zhang Y, Yoo JJ, Atala stricture repair with an off-the-shelf collagen matrix.
A. Cell-seeded tubularized scaffolds for reconstruc- J Urol. 2003;169:170–3. discussion 3
tion of long urethral defects: a preclinical study. Eur 36. el-Kassaby A, AbouShwareb T, Atala A. Randomized
Urol. 2013;63:531–8. comparative study between buccal mucosal and acel-
20. Chun SY, Kim BS, Kwon SY, Park SI, Song PH, Yoo lular bladder matrix grafts in complex anterior ure-
ES, et al. Urethroplasty using autologous urethral thral strictures. J Urol. 2008;179:1432–6.
tissue-­embedded acellular porcine bladder submucosa 37. Feng C, Xu YM, Fu Q, Zhu WD, Cui L, Chen

matrix grafts for the management of long-segment J. Evaluation of the biocompatibility and mechanical
urethral stricture in a rabbit model. J Korean Med Sci. properties of naturally derived and synthetic scaffolds
2015;30:301–7. for urethral reconstruction. J Biomed Mater Res A.
21. Ahn HH, Kim KS, Lee JH, Lee MS, Song IB, Cho 2010;94:317–25.
MH, et al. Porcine small intestinal submucosa sheets 38. le Roux PJ. Endoscopic urethroplasty with unseeded
as a scaffold for human bone marrow stem cells. Int J small intestinal submucosa collagen matrix grafts: a
Biol Macromol. 2007;41:590–6. pilot study. J Urol. 2005;173:140–3.
22. Narayanan K, Leck KJ, Gao S, Wan AC. Three-­
39. Palminteri E, Berdondini E, Colombo F, Austoni

dimensional reconstituted extracellular matrix E. Small intestinal submucosa (SIS) graft urethro-
scaffolds for tissue engineering. Biomaterials. plasty: short-term results. Eur Urol. 2007;51:1695–
2009;30:4309–17. 701. discussion 701
23. Ng SL, Narayanan K, Gao S, Wan AC. Lineage
40. Palminteri E, Berdondini E, Fusco F, De Nunzio

restricted progenitors for the repopulation of decel- C, Salonia A. Long-term results of small intestinal
lularized heart. Biomaterials. 2011;32:7571–80. submucosa graft in bulbar urethral reconstruction.
24. Bazeed MA, Thuroff JW, Schmidt RA, Tanagho
Urology. 2012;79:695–701.
EA. New treatment for urethral strictures. Urology. 41. Sievert KD, Bakircioglu ME, Nunes L, Tu R, Dahiya
1983;21:53–7. R, Tanagho EA. Homologous acellular matrix graft
25. Olsen L, Bowald S, Busch C, Carlsten J, Eriksson I. for urethral reconstruction in the rabbit: histological
Urethral reconstruction with a new synthetic absorb- and functional evaluation. J Urol. 2000;163:1958–65.
able device. An experimental study. Scand J Urol 42. Lin J, Hao JR, Jin J, Deng SM, Hu J, Na

Nephrol. 1992;26:323–6. YQ. Homologous dermal acellular matrix graft for
26. Atala A, Vacanti JP, Peters CA, Mandell J, Retik urethral reconstruction in man (report of 16 cases).
AB, Freeman MR. Formation of urothelial struc- Zhonghua Yi Xue Za Zhi. 2005;85:1057–9.
tures in vivo from dissociated cells attached to 43. Fossum M, Svensson J, Kratz G, Nordenskjold

biodegradable polymer scaffolds in vitro. J Urol. A. Autologous in vitro cultured urothelium in hypo-
1992;148:658–62. spadias repair. J Pediatr Urol. 2007;3:10–8.
27. Chen F, Yoo JJ, Atala A. Acellular collagen matrix 44. Williams DF. Biomaterials and biocompatibility. Med
as a possible "off the shelf" biomaterial for urethral Prog Technol. 1976;4:31–42.
repair. Urology. 1999;54:407–10. 45. Freed LE, Vunjak-Novakovic G, Biron RJ, Eagles DB,
28. Sievert KD, Tanagho EA. Organ-specific acellular
Lesnoy DC, Barlow SK, et al. Biodegradable polymer
matrix for reconstruction of the urinary tract. World J scaffolds for tissue engineering. Biotechnology (N
Urol. 2000;18:19–25. Y). 1994;12:689–93.
29. Micol LA, Arenas da Silva LF, Geutjes PJ, Oosterwijk 46. Mikos AG, Lyman MD, Freed LE, Langer R. Wetting
E, Hubbell JA, Feitz WF, et al. In-vivo perfor- of poly(L-lactic acid) and poly(DL-lactic-co-­
mance of high-density collagen gel tubes for ure- glycolic acid) foams for tissue culture. Biomaterials.
thral regeneration in a rabbit model. Biomaterials. 1994;15:55–8.
2012;33:7447–55. 47. Harris LD, Kim BS, Mooney DJ. Open pore biode-
30. Atala A. Regenerative medicine strategies. J Pediatr gradable matrices formed with gas foaming. J Biomed
Surg. 2012;47:17–28. Mater Res. 1998;42:396–402.
31. Atala A. Tissue engineering of reproductive tissues 48.
Han D, Gouma PI. Electrospun bioscaffolds
and organs. Fertil Steril. 2012;98:21–9. that mimic the topology of extracellular matrix.
32. Micol LA, Ananta M, Engelhardt EM, Mudera VC, Nanomedicine. 2006;2:37–41.
Brown RA, Hubbell JA, et al. High-density colla- 49. Lee SJ, Oh SH, Liu J, Soker S, Atala A, Yoo JJ. The
gen gel tubes as a matrix for primary human bladder use of thermal treatments to enhance the mechanical
smooth muscle cells. Biomaterials. 2011;32:1543–8. properties of electrospun poly(epsilon-caprolactone)
33. Ribeiro-Filho LA, Sievert KD. Acellular matrix
scaffolds. Biomaterials. 2008;29:1422–30.
in urethral reconstruction. Adv Drug Deliv Rev. 50. Raya-Rivera A, Esquiliano DR, Yoo JJ, Lopez-­

2015;82-83:38–46. Bayghen E, Soker S, Atala A. Tissue-engineered
234 Y.-S. Ha and T.-H. Kim

a­utologous urethras for patients who need recon- 65. Villoldo GM, Loresi M, Giudice C, Damia O, Moldes
struction: an observational study. Lancet. 2011; JM, DeBadiola F, et al. Histologic changes after ure-
377:1175–82. throplasty using small intestinal submucosa unseeded
51. Kanatani I, Kanematsu A, Inatsugu Y, Imamura M, with cells in rabbits with injured urethra. Urology.
Negoro H, Ito N, et al. Fabrication of an optimal ure- 2013;81:1380 e1-5.
thral graft using collagen-sponge tubes reinforced 66. Chung YG, Tu D, Franck D, Gil ES, Algarrahi K,
with Copoly(L-lactide/epsilon-caprolactone) fabric. Adam RM, et al. Acellular bi-layer silk fibroin scaf-
Tissue Eng. 2007;13:2933–40. folds support tissue regeneration in a rabbit model of
52. Fu WJ, Xu YD, Wang ZX, Li G, Shi JG, Cui FZ, et al. onlay urethroplasty. PLoS One. 2014;9:e91592.
New ureteral scaffold constructed with composite 67. Kajbafzadeh AM, Sabetkish S, Tourchi A, Amirizadeh
poly(L-lactic acid)-collagen and urothelial cells by N, Afshar K, Abolghasemi H, et al. The application of
new centrifugal seeding system. J Biomed Mater Res tissue-engineered preputial matrix and fibrin sealant
A. 2012;100:1725–33. for urethral reconstruction in rabbit model. Int Urol
53. Horst M, Madduri S, Milleret V, Sulser T, Gobet R, Nephrol. 2014;46:1573–80.
Eberli D. A bilayered hybrid microfibrous PLGA-- 68. Mantovani F, Trinchieri A, Castelnuovo C, Romano
acellular matrix scaffold for hollow organ tissue engi- AL, Pisani E. Reconstructive urethroplasty using por-
neering. Biomaterials. 2013;34:1537–45. cine acellular matrix. Eur Urol. 2003;44:600–2.
54. Bhargava S, Patterson JM, Inman RD, MacNeil
69.
Donkov II, Bashir A, Elenkov CH, Panchev
S, Chapple CR. Tissue-engineered buccal mucosa PK. Dorsal onlay augmentation urethroplasty with
urethroplasty-clinical outcomes. Eur Urol. 2008;53: small intestinal submucosa: modified Barbagli tech-
1263–9. nique for strictures of the bulbar urethra. Int J Urol.
55. Fossum M, Skikuniene J, Orrego A, Nordenskjold 2006;13:1415–7.
A. Prepubertal follow-up after hypospadias repair 70. Hauser S, Bastian PJ, Fechner G, Muller SC. Small
with autologous in vitro cultured urothelial cells. Acta intestine submucosa in urethral stricture repair in a
Paediatr. 2012;101:755–60. consecutive series. Urology. 2006;68:263–6.
56. De Filippo RE, Yoo JJ, Atala A. Urethral replacement 71. Fiala R, Vidlar A, Vrtal R, Belej K, Student V. Porcine
using cell seeded tubularized collagen matrices. J small intestinal submucosa graft for repair of anterior
Urol. 2002;168:1789–92. discussion 92-3 urethral strictures. Eur Urol. 2007;51:1702–8. discus-
57. Li C, Xu YM, Song LJ, Fu Q, Cui L, Yin
sion 8
S. Urethral reconstruction using oral keratinocyte 72. Farahat YA, Elbahnasy AM, El-Gamal OM, Ramadan
seeded bladder acellular matrix grafts. J Urol. AR, El-Abd SA, Taha MR. Endoscopic urethroplasty
2008;180:1538–42. using small intestinal submucosal patch in cases of
58. Fu Q, Deng CL, Song XF, Xu YM. Long-term study recurrent urethral stricture: a preliminary study. J
of male rabbit urethral mucosa reconstruction using Endourol. 2009;23:2001–5.
epidermal cell. Asian J Androl. 2008;10:719–22. 73. Gu GL, Zhu YJ, Xia SJ, Zhang J, Jiang JT, Hong Y,
59. Feng C, Xu YM, Fu Q, Zhu WD, Cui L. Reconstruction et al. Peritoneal cavity as bioreactor to grow autolo-
of three-dimensional neourethra using lingual kera- gous tubular urethral grafts in a rabbit model. World J
tinocytes and corporal smooth muscle cells seeded Urol. 2010;28:227–32.
acellular corporal spongiosum. Tissue Eng Part A. 74. Gu GL, Xia SJ, Zhang J, Liu GH, Yan L, Xu ZH,
2011;17:3011–9. et al. Tubularized urethral replacement using tissue-­
60. Kropp BP, Ludlow JK, Spicer D, Rippy MK, Badylak engineered peritoneum-like tissue in a rabbit model.
SF, Adams MC, et al. Rabbit urethral regenera- Urol Int. 2012;89:358–64.
tion using small intestinal submucosa onlay grafts. 75. Li H, Xu Y, Xie H, Li C, Song L, Feng C, et al.
Urology. 1998;52:138–42. Epithelial-differentiated adipose-derived stem cells
61. Nuininga JE, van Moerkerk H, Hanssen A, Hulsbergen seeded bladder acellular matrix grafts for urethral
CA, Oosterwijk-Wakka J, Oosterwijk E, et al. Rabbit reconstruction: an animal model. Tissue Eng Part A.
urethra replacement with a defined biomatrix or small 2014;20:774–84.
intestinal submucosa. Eur Urol. 2003;44:266–71. 76. Wang F, Liu T, Yang L, Zhang G, Liu H, Yi X, et al.
62. Yang SX, Yao Y, Hu YF, Song C, Wang LL, Jin Urethral reconstruction with tissue-engineered human
HM. Reconstruction of rabbit urethra using urethral amniotic scaffold in rabbit urethral injury models.
extracellular matrix. Chin Med J. 2004;117:1786–90. Med Sci Monit. 2014;20:2430–8.
63. Huang X, Luo J, Liao Y, Qu Y, Yang Z. Study on 77. De Filippo RE, Kornitzer BS, Yoo JJ, Atala A. Penile
small intestinal submucosa as repair materials in ure- urethra replacement with autologous cell-seeded
thral reconstruction. Zhongguo Xiu Fu Chong Jian tubularized collagen matrices. J Tissue Eng Regen
Wai Ke Za Zhi. 2006;20:206–9. Med. 2015;9:257–64.
64. Dorin RP, Pohl HG, De Filippo RE, Yoo JJ, Atala 78. Mikami H, Kuwahara G, Nakamura N, Yamato M,
A. Tubularized urethral replacement with unseeded Tanaka M, Kodama S. Two-layer tissue engineered
matrices: what is the maximum distance for normal urethra using oral epithelial and muscle derived cells.
tissue regeneration? World J Urol. 2008;26:323–6. J Urol. 2012;187:1882–9.
9 Urethra 235

79. Li C, Xu YM, Liu ZS, Li HB. Urethral reconstruction 86. Kim BS, Mooney DJ. Development of biocompatible
with tissue engineering and RNA interference tech- synthetic extracellular matrices for tissue engineering.
niques in rabbits. Urology. 2013;81:1075–80. Trends Biotechnol. 1998;16:224–30.
80. Xie M, Xu Y, Song L, Wang J, Lv X, Zhang Y. Tissue-­ 87. Silver FH, Pins G. Cell growth on collagen: a review
engineered buccal mucosa using silk fibroin matrices of tissue engineering using scaffolds containing
for urethral reconstruction in a canine model. J Surg extracellular matrix. J Long-Term Eff Med Implants.
Res. 2014;188:1–7. 1992;2:67–80.
81. Osman NI, Patterson JM, MacNeil S, Chapple
88. Andrich DE, Mundy AR. Non-transecting anasto-

CR. Long-term follow-up after tissue-engineered buc- motic bulbar urethroplasty: a preliminary report. BJU
cal mucosa urethroplasty. Eur Urol. 2014;66:790–1. Int. 2012;109:1090–4.
82. Kim BD, Ver Halen JP, Grant DW, Kim JY. Anesthesia 89. Kannan RY, Salacinski HJ, Sales K, Butler P, Seifalian
duration as an independent risk factor for postop- AM. The roles of tissue engineering and vascularisa-
erative complications in free flap surgery: a review tion in the development of micro-vascular networks: a
of 1,305 surgical cases. J Reconstr Microsurg. review. Biomaterials. 2005;26:1857–75.
2014;30:217–26. 90.
Rouwkema J, Rivron NC, van Blitterswijk
83. Zhang Y, McNeill E, Tian H, Soker S, Andersson CA. Vascularization in tissue engineering. Trends
KE, Yoo JJ, et al. Urine derived cells are a potential Biotechnol. 2008;26:434–41.
source for urological tissue reconstruction. J Urol. 91. Wu S, Wang Z, Bharadwaj S, Hodges SJ, Atala A,
2008;180:2226–33. Zhang Y. Implantation of autologous urine derived
84. Bharadwaj S, Liu G, Shi Y, Markert C, Andersson stem cells expressing vascular endothelial growth fac-
KE, Atala A, et al. Characterization of urine-derived tor for potential use in genitourinary reconstruction. J
stem cells obtained from upper urinary tract for use in Urol. 2011;186:640–7.
cell-based urological tissue engineering. Tissue Eng 92. Kulkarni S, Barbagli G, Sansalone S, Lazzeri M. One-­
Part A. 2011;17:2123–32. sided anterior urethroplasty: a new dorsal onlay graft
85. Sievert KD, Selent-Stier C, Wiedemann J, Greiner technique. BJU Int. 2009;104:1150–5.
TO, Amend B, Stenzl A, et al. Introducing a large 93. Barbagli G, Sansalone S, Djinovic R, Romano G,
animal model to create urethral stricture similar to Lazzeri M. Current controversies in reconstructive
human stricture disease: a comparative experimental surgery of the anterior urethra: a clinical overview. Int
microscopic study. J Urol. 2012;187:1101–9. Braz J Urol. 2012;38:307–16. discussion 16
Urethral Sphincter: Stress Urinary
Incontinence 10
Eun Sang Yoo and Jun Nyung Lee

10.1 Introduction 10.1.1.2 D  efinition of Stress Urinary


Incontinence
10.1.1 Stress Urinary Incontinence The International Continence Society defined
stress urinary incontinence as the involuntary
10.1.1.1 Introduction of Stress leakage of urine on effort, exertion, sneezing, or
Urinary Incontinence coughing. It is estimated that one third of the
Including Symptoms procedures that are performed to treat stress uri-
Urinary incontinence is described as the invol- nary incontinence are performed on patients
untary loss of urine and is a common condition with recurrent disease. Currently accepted pro-
in middle-aged and elderly women and men [1]. cedures for stress urinary incontinence are usu-
Urinary incontinence can be generally classified ally based on compensatory and nonphysiological
into the following three: (1) stress urinary mechanisms. Although the complete success of
incontinence, (2) urge urinary incontinence, and long-­term management of any condition is based
(3) a mixed form of (1) and (2) [1]. Stress uri- on an understanding of its pathophysiology, the
nary incontinence occurs when increased intra-­ pathophysiology of stress urinary incontinence
abdominal pressure causes bladder pressure to is not well defined. Therefore, further investiga-
exceed urethral pressure, resulting in involun- tions including the development of animal mod-
tary leakage of urine. In stress urinary inconti- els to properly understand the condition and
nence, urine leakage can be observed during alternative managements based on pathophysio-
coughing, sneezing, laughing, lifting, and exer- logical changes are necessary.
cising. Classically stress urinary incontinence
often relies on distinguishing between intrinsic 10.1.1.3 Prevalence of Stress Urinary
sphincter deficiency and urethral malposition or Incontinence
hypermobility [2]. However, this is controver- Inappropriate micturition accounts for a large
sial as each component may contribute in vary- portion of the affected patients. Especially, uri-
ing proportion to the occurrence of stress urinary nary incontinence is a major health problem
incontinence. causing a significant social and economic impact
affecting more than 200 million people world-
wide [3]. The prevalence rises with age, and
therefore the magnitude of this problem is
E.S. Yoo (*) • J.N. Lee expected to substantially increase as the life
Department of Urology, School of Medicine,
Kyungpook National University, Daegu, South Korea expectancy and aging population continue to rise
e-mail: uroyoo@knu.ac.kr; ljnlover@gmail.com in the world [3]. The incidence of incontinence is

© Springer Nature Singapore Pte Ltd. 2018 237


B.W. Kim (ed.), Clinical Regenerative Medicine in Urology, DOI 10.1007/978-981-10-2723-9_10
238 E.S. Yoo and J.N. Lee

increasing by more than one third of middle-aged striated and smooth muscle fibers. As a result,
females report leakage at least weekly with 10% patients often do not become symptomatic until
reporting daily or severe leakage [4]. years after the initial trauma of childbirth due to
a cascade of events which continues to occur with
10.1.1.4 E  tiology of Stress Urinary age. Therefore, relevant animal models are also
Incontinence essential in understanding these events and in the
Voiding dysfunction represents a diverse spec- development of preventative interventions.
trum of urologic conditions including stress uri-
nary incontinence and overactive bladder: two of 10.1.1.5 M echanism of Stress Urinary
the most common and challenging conditions Incontinence
faced by urologists today. Several of these disor- Maintaining urinary continence involves several
ders are precipitated by an acute injury such as aspects:
vaginal delivery in female stress urinary inconti-
nence or radical prostatectomy in male stress 1. A stable bladder with adequate capacity and
urinary incontinence. The underlying pathophys- accommodation,
iology comprises a complex interplay of injuries 2. An anatomically normal and functionally

or altered function at the tissue, nerve, and vascu- competent continence mechanism (consisting
lar level that is then exacerbated by underlying of a bladder neck, urethra, urethral sphincter
comorbidities, including obesity, increasing age, [itself consisting of striated and smooth mus-
and diabetes mellitus. cle layers, neuronal innervations, a vascular
plexus, submucosa, and epithelium], endopel-
Male vic fascia, arcus tendineus, and pelvic muscle
The most common cause in the development of support)
male stress urinary incontinence is iatrogenic 3. The correct integrity of somatic and auto-

damage due to radical prostatectomy, transure- nomic innervation of the structures involved
thral resection of the prostate, and radiation
therapy. Postprostatectomy stress urinary incon- The muscular structures are controlled by
tinence is of particular concern because data three sets of major nerves: (1) parasympathetic
shows that anywhere from 2% to 66% of males sacral nerves (pelvic nerves), (2) sympathetic
are affected depending on the chosen group of thoracolumbar nerves innervating the periure-
patients and methods of measurement [5]. thral smooth muscle, and (3) somatic sacral
Radical prostatectomy may cause direct injury to nerves (pudendal nerves) innervating the urethral
the urethral sphincter via transection, penetra- striated muscles including the external periure-
tion, a combination of both, or indirect neuro- thral sphincter and the pelvic floor muscles.
muscular damage. Assuming proper bladder function, damage to the
urethral sphincter muscle or its innervation can
Female be produced by various ways in both men and
In women, the most influential factors for stress women, resulting in urethral sphincter damage or
urinary incontinence are vaginal delivery and deficit and ultimately stress urinary incontinence.
increased age [6]. The exact mechanism of injury Thus, the mechanism of human stress urinary
is not well understood but is likely multifactorial. incontinence is a complex and usually multifac-
Vaginal childbirth produces mechanical and neu- torial process sometimes combining denervation,
rovascular injury to the pelvic floor, and aging muscle degeneration and apoptosis, chronic mus-
plays a negative role in the structure and function cle atrophy, fibrosis, and connective tissue disor-
of the pelvic floor. Combined with other potential ders, among others, which makes the generation
risk factors such as parity, obesity, and meno- of an in vivo model that integrates all pathophysi-
pause, vaginal childbirth contributes to a decrease ological aspects of stress urinary incontinence
in the number and diameter of the periurethral difficult, if not impossible.
10  Urethral Sphincter: Stress Urinary Incontinence 239

The pathophysiology of stress urinary inconti- performed annually. Postprostatectomy inconti-


nence has not been fully understood. Several nence results from a failure to store urine second-
hypotheses have been proposed to explain the ary to inadequate resistance of the outlet
mechanism of urethral closure [7]. One school of sphincter. Surgical damage to the urethral sphinc-
thought considers the rhabdosphincter (a ter occurs due to direct injury to the sphincter
horseshoe-­shaped muscle which sits on the dor- itself, as well as to surrounding nerves and sup-
sal part of the midurethra), along with the ure- portive tissue [10].
thral smooth muscle and the vascular elements
within the submucosa, as the principal structures 10.1.1.6 Current Clinical
for continence. The second school of thought Management of Stress
considers that stress urinary incontinence results Urinary Incontinence
from the weakening or loss of the structures
which support the bladder neck and proximal Male
urethra (ligaments, fascias, and muscles) [8] and Over time diverse treatments for stress urinary
that the rhabdosphincter plays a minor role in incontinence have been used. As in females, sur-
closure [8]. Probably the majority of women with gical therapies are the mainstay of therapy and
stress urinary incontinence have varying degrees include transurethral bulking agents, bulbar ure-
of both intrinsic sphincter weakness and loss of thral slings, and artificial urinary sphincters.
the normal anatomic support to the bladder neck These treatments aim to augment a deficient uri-
and proximal urethra resulting in hypermobility nary sphincter by allowing urethral coaptation
[9]. The primary cause of male stress urinary during periods of increased abdominal pressure
incontinence is damage to the urethral sphincter without causing outlet obstruction during void-
during surgical procedures, mostly radical ing. Currently, the gold standard in treatment for
prostatectomy. moderate male stress urinary incontinence is the
In females, urinary continence relies on an use of the male sling or, in the case of severe
intact urethral sphincteric mechanism. Multiple incontinence, implantation of an artificial sphinc-
factors contribute to urethral pressure including ter. However, 5 years post-artificial urinary sphinc-
bladder neck position, urethral sphincter muscu- ter implantation, up to 24% of men still report
lature, sphincter innervation, and surrounding severe incontinence, and 28% will have undergone
vascular supply and tissue support. Pregnancy an artificial urinary sphincter revision [11]. In
and childbirth are well-recognized risk factors addition, there are many complications associated
for stress urinary incontinence, and four related with these procedures. Although treatment for
major mechanisms of injury have been identified: male stress urinary incontinence due to sphincter
(1) injury to connective tissue support during damage has evolved in the past 40 years, this dis-
vaginal delivery, (2) vascular damage due to fetal ease will continue to be an unresolved social health
compression of surrounding pelvic structures, (3) problem. The development of new treatments
traumatic injury to pelvic nerves and muscula- focusing on cellular sphincter regeneration could
ture, and (4) direct injury to the lower urinary play a very important role in the years to come.
tract during childbirth.
In postprostatectomy stress urinary inconti- Female
nence, contemporary series report improved con- Clinical treatment of stress urinary incontinence is
tinence rates following prostatectomy although complex and includes conservative techniques,
wide variation continues to exist due to discrepan- pharmacologic therapy, and surgical procedures.
cies in data collection methodology and patient- Patients with stress urinary incontinence can ben-
versus surgeon-reported outcomes. Nonetheless, efit from initial conservative measures, including
while treatment options have improved in recent pelvic floor physiotherapy, biofeedback, and elec-
years, the burden of disease remains high due to trical stimulation. The goal of surgical ­procedures
the increasing numbers of radical prostatectomies is anatomical and functional restoration of the
240 E.S. Yoo and J.N. Lee

p­ elvic structures. Urethral slings and suspensions 15% of cases. Because of not sustainable efficacy
aim to correct stress urinary incontinence by cor- and complications of standard therapies of stress
recting hypermobility and augmenting intrinsic urinary incontinence, there is a need to find a
sphincter deficiency by allowing urethral com- new, effective, and minimally invasive method of
pression during periods of increased intra-abdom- treatment. Although injection of a bulking agent
inal pressure without causing obstruction during is attempted as a minimal alternative of stress uri-
voiding. As of now, more than 100 surgical meth- nary incontinence treatment, the long-term cure
ods have been performed for treatment of stress rates are poor [20].
urinary incontinence, with various success rates.
These techniques may be broadly subcategorized
into four major types: periurethral bulking agent 10.1.2 Regenerative Medicine
injection, retropubic suspension, sling and tape for Stress Urinary
procedures, and artificial urinary sphincter devices Incontinence
[12]. In contemporary practice, the use of midure-
thral slings has shown cost-effective objective 10.1.2.1 P  urpose of Regenerative
results and subjective success [13]. Currently, the Medicine
gold standard for surgical treatment of all types of Despite significant developments in the underly-
stress urinary incontinence in women is midure- ing pathophysiology of stress urinary inconti-
thral slings. The surgical options remain the main- nence, current gold standard therapies remain
stay for cases nonresponsive to conservative lacking in their ability to target these fundamental
measures [14, 15], with a success rate for different mechanisms of injury or etiology of disease. To
sling operations of up to 80–90% and with obser- date, none of the standard therapies focus on halt-
vation times up to 17 years [16]. In cases of severe ing disease progression or reversing underlying
incontinence such as intrinsic sphincter deficiency, injury. Regenerative medicine concepts have
artificial urinary sphincter implantation is an emerged as an exciting means of fulfilling this
appropriate treatment option [17] since other treat- therapeutic void by restoring and maintaining
ments such as placement of suburethral bands or normal function through direct effects on injured
injection of bulking agents have not proved effec- or dysfunctional tissues. Over the past decade, the
tive enough for this condition [13]. Although the use of stem cells has shown promise for a host of
use of artificial sphincters is highly effective in urologic disorders including applications in lower
severe incontinence cases, the overall complica- urinary tract dysfunction, ureteral and bladder
tion rates are not negligible, and recurrence of trauma, erectile dysfunction, and renal disease.
stress urinary incontinence can occur. Stem cells are classically thought to improve
tissue repair through multilineage differentiation
10.1.1.7 L  imitation of Current and self-renewal. Stem cells may also exert a
Therapy therapeutic effect through the secretion of bioac-
Although the tension-free vaginal tape is accepted tive factors that have antiapoptotic, antiscarring,
as gold standard procedure for surgical treatment neovascularization, and immunomodulatory
of stress urinary incontinence in women, midure- effects on innate tissues and can direct innate
thral slings are associated with complications stem and progenitor cells to the area of injury.
including overactive bladder/urgency; urinary Multiple treatment avenues using stem cells for
retention; bladder, urethral, or vaginal perfora- voiding dysfunction, especially stress urinary
tion; erosion; infections; persistent groin/supra- incontinence, have been evaluated with preclini-
pubic pain; and dyspareunia [18, 19]. Incidences cal animal models and clinical trials ­demonstrating
of complications after midurethral slings are ero- their potential to restore function via direct
sion rates in up to 23% of cases, permanent reten- effects on the underlying mechanisms that lead to
tion in up to 5% of cases, as well as wound incontinence or voiding dysfunction. Currently,
complications, bladder perforation, persistent many challenges remain to translate these prom-
groin/ suprapubic pain, and dyspareunia in up to ising results to clinical practice.
10  Urethral Sphincter: Stress Urinary Incontinence 241

10.1.2.2 A  pplication of Regenerative defining characteristics: (1) the ability to self-­


Medicine for Stress Urinary renewal, (2) multipotent differentiation potential
Incontinence or the ability to differentiate into a number of dif-
Currently, the most promising method of stress ferent cell types, and (3) clonogenicity or the
urinary incontinence treatment is cell implanta- ability to form clonal cell populations derived
tion which is based on regeneration of the dam- from a single stem cell. It is these unique abilities
aged rhabdosphincter. The main concept of for differentiation and self-renewal that give
cell-based therapies for stress urinary inconti- these cells the potential for restoration of func-
nence involves the implantation of cells into the tion in multiple tissue types.
damaged parts of the urethral sphincter complex, Regenerative medicine may offer such an
which can restore its proper function through the alternative by using a cell-based therapeutic
induction of muscle and nerve regeneration. This approach to regenerate the sphincter muscle. This
is an active, complex process that involves matrix approach has the advantage of treating the cause
remodeling necessary for restoration of func- and not only the symptoms [23–25] and may
tional cells responsible for the proper function of improve the external (striated muscle) and inter-
the sphincter complex and ultimately continence. nal (smooth muscle) urethral sphincter muscle
This effect is achieved by stem cell differentia- function, the neuromuscular transmission, and
tion into new striated muscle cells or neurons that blood supply [24, 26]. Since the majority of
replace injured parts or by induced regeneration women with stress urinary incontinence may
of host tissue, triggered by trophic factors have hypermobility rather than intrinsic sphincter
released by implanted cells. Therefore, a cell-­ deficiency, this would limit the cell-based
based regenerative therapy could be established approach to patients with intrinsic sphincter defi-
as a novel treatment option for future urethral ciency. Clearly, some pathological processes
sphincter muscle regeneration. place a woman at risk of both sphincter defi-
ciency and urethral hypermobility/malposition
10.1.2.3 Purpose of This Chapter (e.g., childbirth injuries) and an age-dependent
In this chapter, we provide the current state of loss of both striated muscle thickness and con-
knowledge regarding cell therapy for stress uri- tractility [27].
nary incontinence including stem cell sources, the Many recent reviews have discussed different
potential mechanisms of stem cell therapy, experi- aspect of stem cell treatment of stress urinary
mental trials, and clinical trials, as well as new incontinence [14, 23–25], and most of these
approaches of using regenerative pharmacology reviews emphasize the potential of this approach,
as an adjunct or replacement for cell therapy. but also that additional research is required.

10.2.1.2 S  ource of Stem Cells


10.2 Body for Stress Urinary
Incontinence Therapy
10.2.1 Regenerative Therapy Currently, four broad categories describe the
of Stress Urinary Incontinence diversity of stem cells being investigated in
As Alternative Approach regenerative medicine: embryonic stem cells,
stem cells derived from placenta or amniotic
10.2.1.1 O  verview of Cell Therapy fluid, induced pluripotent stem cells, and adult
for Stress Urinary stem cells [21, 22, 28–30]. Embryonic stem cells
Incontinence are stem cells isolated from an early-stage
The field of stem cell research began with the dis- embryo [22]. They represent a pluripotent cell
covery of mouse embryonic stem cells in the source and can differentiate into all adult cell
early 1970s and with the description of human types. Although embryonic stem cells have great
embryonic stem cells in 1998 [21, 22]. Stem cells therapeutic potential, their use is limited by ethi-
comprise a unique population of cells with three cal factors. In addition, their clinical application
242 E.S. Yoo and J.N. Lee

is limited because they represent an allogeneic issues with embryonic stem cells. A disadvantage
cell source with the potential to provoke an of induced pluripotent stem cells is the time
immune response and because of concerns involved in resetting the cells to a pluripotent
regarding potential tumorigenicity. state followed by the additional time required to
induce the cells to differentiate into the desired
Embryonic Stem Cells lineage [36]. Furthermore, there remain concerns
Embryonic stem cells are currently being investi- that full transition to this new desired lineage
gated for application to type 1 diabetes mellitus may not occur [36]. To date, IPSCs have not
and cardiomyopathy [31, 32]. However, embry- nearly been used for urologic applications.
onic stem cells are not being investigated as a
treatment for voiding dysfunction or urinary Adult Stem Cells
incontinence because of several limitations stated Adult stem cells are the most well-understood
above. cell type in the field of stem cell therapy and are
the most studied stem cell type in urologic fields
Amniotic Fluid Stem Cells [28–30, 37]. Over the past decade, adult stem
Amniotic fluid stem cells are a new class of stem cells have been identified throughout the body
cells with properties intermediate to those of and are thought to act as tissue-specific progeni-
embryonic stem cells and adult stem cells [33]. tors that repair damage and restore function
They represent a heterogeneous stem cell popu- locally. Mesenchymal stem cells, also known as
lation derived from the amniotic fluid and pla- multipotent adult progenitor cells, are a unique
cental membrane of the developing fetus. Cells subset of adult stem cells and are described for
found in these tissues include mesenchymal the first time by Friedenstein et al. in the 1970s
stem cells as well as multipotent amniotic fluid [38]. Classically, mesenchymal stem cells were
stem cells that possess extensive self-renewal isolated from bone marrow stroma although more
capacity. In addition, amniotic fluid stem cells contemporary studies have demonstrated that
can be induced to differentiate into cells of all they may also be found in other well-­vascularized
three germ cell layers including cells of adipo- tissues including adipose, muscle, endometrium,
genic, osteogenic, myogenic, endothelial, neu- and kidney [39]. Unlike tissue-specific progeni-
ral, and hepatic lineages. Amniotic fluid stem tor cells, they can be induced to differentiate into
cells are currently being investigated for a vari- multiple cell lineages including bone, neuronal,
ety of applications including treatment of uri- adipose, muscle, liver, lungs, spleen, and gastro-
nary incontinence, acute tubular necrosis, intestinal tissues [39].
cardiac valve regeneration for the early correc-
tion of congenital cardiac malformations, and as Mesenchymal Stem Cells
a source of immunomodulatory cells for a vari- Currently, mesenchymal stem cells are the pri-
ety of immunotherapies [34]. mary source of stem cells tested for therapeutic
benefit in urologic applications. However, this
Induced Pluripotent Stem Cells cell population is relatively rare in the bone mar-
Induced pluripotent stem cells are a unique class row (approximately 1 per 10,000 cells), and tra-
of stem cells recently discovered by Takahashi ditional bone marrow procurement is a painful
and Yamanaka who demonstrated that specific and underproductive procedure and requires gen-
transcription factors could be used to reprogram eral or spinal anesthesia [40]. Alternative cell
differentiated cells to a pluripotent state [35]. sources that have been investigated for urologic
Induced pluripotent stem cells have been shown application include both muscle-derived stem
to possess capacity for multipotent differentia- cells and adipose-derived stem cells which can be
tion and self-renewal. In addition, they can be obtained via less invasive biopsies and in larger
used autologously, and their derivation from quantities under local anesthesia [28].
adult cells obviates concerns regarding ethical Investigators continue to identify novel, less
10  Urethral Sphincter: Stress Urinary Incontinence 243

invasive cell sources for urologic application harvest the cells. Following procurement, the tis-
including cells derived from hair follicles, men- sue specimen is then transported to a regulated
strual fluid, and urine [41–43]. facility for isolation from other cell types. Here,
the cells are grown via a process of ex vivo stem
Urine-Derived Stem Cells cell expansion involving multiple cycles of dif-
Urine-derived stem cells are of particular interest ferentiation and senescence until adequate num-
for potential urologic applications [43–48]. In bers are reached. Finally, cell sorting may or may
recent studies, Zhang et al. have reported the suc- not be used to isolate stem cells prior to therapeu-
cessful isolation and expansion of stem cells tic application, as this process is often labor-­
from voided human urine [43]. These cells, intensive and expensive. Thus, the cells that are
thought to be mesenchymal stem cells or peri- ultimately administered to the patient may com-
cytes from the kidney [45], can be obtained non- prise a heterogeneous combination of stem cells
invasively from human urine specimens and and differentiated cells. Although still investiga-
demonstrate stem cell-like features including clo- tional, it is feasible during this process to manip-
nogenicity, self-renewal, and multipotent differ- ulate the cells for a variety of therapeutic
entiation capacity [47]. Single clones of applications. For example, cells can be prediffer-
urine-derived stem cells have been shown to have entiated toward a specific cell lineage, preex-
the capacity to expand to yield a large population. posed to an environment similar to the in vivo
Comprehensive characterization studies of these posttransplant environment, or transfected to pro-
urine-derived stem cells demonstrate their ability duce specific cytokines or growth factors.
to differentiate into multiple cell lineages at the
gene/protein expression, cellular, and tissue lev- 10.2.1.4 Stem Cell Homing
els [43, 47]. In vitro studies show that urine-­ The process of innate systemic stem cell delivery
derived stem cells derived from a single clone to the site of injury is termed “homing” and can be
were able to differentiate into smooth muscle and taken advantage in delivering cells systemically
urothelial lineages as evidenced by histologic rather than locally. In contrast to tissue-­specific
examination and gene/protein marker assays progenitor cells, mesenchymal stem cells derived
[46]. Urine-derived stem cells can also be induced from the bone marrow traverse the circulatory sys-
to differentiate into osteogenic, adipogenic, and tem with access to all tissues in the body but then
chondrogenic lineages using different differenti- migrate to specific locations such as areas of acute
ation protocols [46]. In vivo studies using BMP-­ injury following chemokine gradients where they
2- and BMP-9-transduced urine-derived stem can engraft and facilitate healing and regeneration
cells also demonstrated their ability to form bone, [50]. Much of the insight regarding stem cell hom-
fat, and cartilage tissue in immunodeficient ing derives from literature regarding leukocyte
female mice [49]. Urine-derived stem cells hold migration into injured tissue, metastatic cancer
several advantages as a cell source for inconti- cells, and hematopoietic stem cells [50]. Similar to
nence and voiding dysfunction [43]. They are leukocytes, mesenchymal stem cells express cell
easily harvested and do not require invasive sur- surface receptors and adhesion molecules respon-
gical techniques to obtain. Furthermore, they are sible for directing cellular migration and homing
easily isolated with a significant cost advantage. to particular tissues, including the chemokine
Finally, they can be used autologously, obviating receptor CXCR4 and its binding partner CXCL12
potential ethical issues or the potential of adverse [51] as well as the chemokine ligands: CCR1,
immune reactions. CCR4, CCR7, CCR10, CCR9, CXCR5, and
CXCR6 [52]. Mesenchymal stem cells are hypoth-
10.2.1.3 Stem Cell Procurement esized to migrate to target tissues via a process
The procurement and isolation of stem cells for similar to that of leukocyte migration: initial local-
therapeutic use typically occurs in three stages. ization by means of chemoattraction, adhesion to
The patient initially undergoes a procedure to vascular endothelial cells at the target site, and,
244 E.S. Yoo and J.N. Lee

finally, transmigration across the endothelium to cells following bladder injury were associated
the site of injury. Integrins and selectins are other with increased chemokine ligand 2 (CCL2)
classes of cell surface molecules that direct migra- expression in the affected tissue [60].
tion and adhesion of a variety of cells, including In therapeutic applications, stem cell homing is
mesenchymal stem cells [53]. While the role of affected by a variety of factors including age and
these molecules in leukocyte-endothelial adhesion passage number of the cells, culture conditions,
is well established, their exact role in facilitating and the delivery method [50]. Rombouts and
mesenchymal stem cells interaction with endothe- Ploemacher demonstrated that with increasing age
lium is less well characterized. Ruster and col- and passage number, the efficiency of mesenchy-
leagues found that binding and rolling of mal stem cells engraftment decreases, possibly
mesenchymal stem cells were mediated by due to rapid aging of the cells with increased
P-selectin, while migration involved binding of the in vitro multiplication [61]. Reproduction of the
integrin VLA-4 on mesenchymal stem cells with innate mesenchymal stem cells microenvironment
VCAM-1 on endothelial cells [54]. in vitro is challenging and plays a significant role
A large body of literature utilizing animal in stem cell homing potential. For example,
models has demonstrated the ability of mesen- in vitro expression of matrix metalloproteinases
chymal stem cells to home to injured tissues in (MMPs), key factors to stem cell migration, is
several disease models including cardiac injury, affected by cell culture factors such as hypoxia and
renal failure, and skin wounds. In addition, recent culture confluence and may be upregulated with
work investigating mesenchymal stem cells for the addition of certain inflammatory cytokines,
stress urinary incontinence has shown that che- such as transforming growth factor beta 1 (TGFβ1),
mokine ligand 7 (CCL7), a homing factor for interleukin (IL)-1β, and tumor necrosis factor
mesenchymal stem cells, is upregulated in both (TNF)-α [62]. Despite the appeal of delivering
the urethra and vagina after vaginal distension, cells systemically rather than locally, systemically
suggesting that intravenously administered mes- infused mesenchymal stem cells often suffer from
enchymal stem cells could have the potential to a first-­
pass effect whereby these larger cells
home to sites of injury in stress urinary inconti- become trapped in capillary beds of various tis-
nence [55–57]. Subsequent literature examining sues, especially the lungs, liver, and spleen,
intravenous injection of mesenchymal stem cells decreasing their therapeutic bioavailability and
in a vaginal distension rat model of simulated functionality [63]. To overcome this, recent inves-
childbirth injury showed that mesenchymal stem tigations have utilized an intraperitoneal delivery
cells home to the urethra and vagina and facilitate route for mesenchymal stem cells and have
recovery of continence as measured by leak point obtained higher yield in target tissues [64].
pressure [58, 59]. Recent work by Lenis and col-
leagues used a rat model of childbirth injury with 10.2.1.5 Stem Cell Differentiation
both virgin rats that had undergone vaginal dis- The mechanism of action of stem cells was ini-
tension and postpartum rats to further investigate tially thought to primarily derive from their ability
the expression of chemokines and receptors to differentiate into multiple cell types and regen-
involved in stem cell homing and tissue repair erate damaged tissues. To date, treatment avenues
[57]. They showed that vaginal distension in vir- for voiding dysfunction and urinary incontinence
gin and postpartum rats resulted in upregulation have focused on this potential to restore function
of urethral CCL7 expression. In addition, preg- via differentiation to replace injured or diseased
nancy and delivery was found to upregulate the tissues such as smooth or striated muscle for ure-
chemokine receptor CD191 but to decrease the thral sphincter regeneration and urothelial tissue
expression of hypoxia inducible factor 1α and for bladder, urethral, and upper urinary tract
vascular endothelial growth factor. Similarly, in a reconstruction. It is thought that mesenchymal
mouse model of overactive bladder via bladder stem cells restore function in stress urinary incon-
outlet obstruction, Woo and colleagues showed tinence primarily by their ability to differentiate
that intravenously injected mesenchymal stem into multiple cell lineages, with animal studies
10  Urethral Sphincter: Stress Urinary Incontinence 245

demonstrating increases in urethral muscle, transplant host [73]. Mesenchymal stem cells have
nerves, and connective tissue following mesen- also been shown to play a role in suppressing
chymal stem cells injection [65–67]. Mesenchymal immune responses by modulation of T cell activa-
stem cells have also been induced to differentiate tion and proliferation through both direct cell to
into a smooth muscle phenotype when exposed to cell interaction and via the action of soluble factors
conditioned media from smooth muscle cell cul- [73]. These immunomodulating properties are cur-
tures or when induced with specific myogenic rently being investigated in myriad applications
growth factors for application to bladder recon- including prevention of graft versus host disease
struction [68]. However, more recent researches following allogeneic transplantation and Crohn’s
suggest a more complex role of stem cells in func- disease [74]. Clinical trials in cardiology have
tional recovery, with some studies demonstrating taken advantage of these properties of mesenchy-
that paracrine secretions of mesenchymal stem mal stem cells to investigate the efficacy of non-
cells play an important role in the regeneration type-matched allogeneic MSC transplantation [75,
process [59, 69]. 76]. In a study by Hare et al. 1 year after intrave-
nous administration of allogeneic human mesen-
10.2.1.6 B  ioactive Effects of Stem chymal stem cells in reperfused myocardial
Cells infarction patients, recovery, as measured by global
Recent research has demonstrated that stem cells symptom score, ejection fraction, ambulatory elec-
likely exert a therapeutic effect via the secretion of trocardiogram monitoring, and pulmonary func-
bioactive factors since few mesenchymal stem tion testing, was significantly improved in treated
cells engraft and remain long term in target tissues, patients compared with those that received a pla-
in contrast to their large therapeutic effect [59, 69]. cebo [75]. In addition, no signs of rejection were
Mesenchymal stem cells also activate and direct observed and adverse event rates were comparable
endogenous stem and progenitor cells to areas of between treated and placebo arms.
injury via the secretion of cytokines and chemo- A recent study by Dissaranan et al. demon-
kines [70]. In addition, their secretions have anti- strates that the secretome of mesenchymal stem
apoptotic, antiscarring, and neovascularization cells can facilitate recovery of continence as
effects, as well as immunomodulatory properties measured by leak point pressure following vagi-
[71]. The investigation of this protein milieu or nal distension in a rat model. Moreover, rats
“secretome” is a subject of growing interest with treated locally with mesenchymal stem cells sec-
the increasing recognition of the paracrine/auto- retome, as contained in conditioned media,
crine role of cell secretions in the regulation of exhibited an increase in elastin fibers and ure-
many physiological processes and their potential thral smooth muscle, which may have contrib-
for therapeutic application. The investigation of uted to the restoration of continence [59]. A
these cell-specific proteins often begins in cell cul- study of hamsters with heart failure suggests that
ture. While in vitro studies cannot fully capture mesenchymal stem cells act systemically as well
and test the totality of mesenchymal stem cells as locally [77]. In this study mesenchymal stem
secretions in the in vivo microenvironment, cells injected into the hamstrings of affected ani-
researchers seek to replicate the effects of the mes- mals were unable to migrate from the injection
enchymal stem cells secretome via the use of site; however, the authors found that treated ani-
media conditioned by the mesenchymal stem cells mals still benefited from stem cell injection
and containing their secretions [72]. based on histologic and functional analysis of
Mesenchymal stem cells also possess immuno- the myocardium. More recent work by Timmers
modulatory and immunological tolerance inducing et al. utilizes human mesenchymal stem cells
characteristics. These cells typically express secretions collected as conditioned medium in a
MHC-I but lack expression of MHC-II, CD40, pig model of myocardial infarction [78]. Pigs
CD80, and CD86. Owing to the lack of co-stimula- underwent left circumflex coronary artery and
tory cell surface molecules, mesenchymal stem were then administered intravenous conditioned
cells fail to induce an immune response by the media for 7 days. At 3 weeks following initial
246 E.S. Yoo and J.N. Lee

cardiac injury and treatment, pigs treated with recovery in a rat model of overactive bladder from
mesenchymal stem cells-conditioned media partial bladder outlet obstruction despite limited
were found not only to have increased myocar- engraftment 4 weeks posttreatment [79]. Further
dial vascular density but also reduced infarction investigations into the use of stem cell bioactive
size and more preserved cardiac function. factors could someday obviate the need for cellu-
These findings have been borne out in other uro- lar injections in future stem cell therapies.
logic investigations as well. Lin and colleagues
investigated rats subjected to vaginal distension
and ovariectomy that were subsequently treated 10.2.2 Experimental Trials
with intraurethral injection of adipose-­derived stem
cells; despite limited cell engraftment on histologic 10.2.2.1 Animal Model
analysis, these rats demonstrated significant func- In the last 45 years, several animal models for
tional recovery [67]. Similarly, a study by Song urethral dysfunction have emerged (Table 10.1).
and colleagues utilizing bladder wall injections of These models include vaginal distension, pudendal
mesenchymal stem cells demonstrated functional nerve crush, urethrolysis, periurethral cauteriza-

Table 10.1  Regenerative therapy targeting the urethral sphincter animal models
Animal
Cell type Biomaterial model SUI model Continence assessment
BMSC [66, 108] – Rodent PNT LPP
BMSC [92] – Rodent Urethrolysis/cardiotoxin ALPP
BMSC [109] Alginate/Calcium Rodent PNT LPP
ADSC [67, 113] – Rodent VD Bladder capacity, LPP
ADSC [106] PLGA ± FNG Rodent PNT Bladder capacity, LPP,
RUPP
ADSC [65] – Rodent VD Bladder capacity, LPP
DFAT [114] – Rodent VD LPP
MDC [116] – Rodent Sciatic nerve resection and LPP, EFS
T9 spinal cord transection
MDSC [105] Fibrin gel Rodent PNT LPP
MPC [98] – Rodent Sphincterotomy LPP, CP
MPC [117] – Swine None UPP
MPC [96] – Rodent Electrocoagulation Cystometry
MPC [152] – Canine Sphincterotomy DLP, UPP, USP,
cystourethrogram,
electrical stimulation
Human MDC [119] – Rodent Sciatic nerve resection and LPP
T9 spinal cord transection
Human UCB [94] – Rodent Electrocauterization and LPP
T9 spinal cord transection
Human AFSC [120] – Rodent Bilateral PNT and T9 LPP, CP
spinal cord transection
Human myoblasts [121] – Rodent Botulinum-A toxin Urine volume
Human AFSC [110, 120] – Rodent Bilateral PNT and T9 LPP, CP
spinal cord transection
ADSC adipose-derived stem cells, AFSC amniotic fluid stem cells, ALPP abdominal leak point pressure, ASMA alpha
smooth muscle actin, BMSC bone marrow mesenchymal stems cells, CP closure urethral pressure, DFAT mature
adipocyte-­derived dedifferentiated fat, DLP lowest bladder pressure, EFS electrical field stimulation, FNG nerve growth
factor, LPP leak point pressure, MDC muscle-derived cells, MPC muscle precursor cells, MSCs mesenchymal stem
cells, PLGA polylactic-co-glycolic acid, RUPP retrograde urethral perfusion pressure, UCB umbilical cord blood, UPP
urethral pressure curve, USP urethral pressure curve with abdominal compression
10  Urethral Sphincter: Stress Urinary Incontinence 247

tion, urethral sphincterotomy, pudendal nerve method that introduced an intravaginal balloon
transection, and toxins injected into the sphincter. filled with 2 mLl of saline using a transurethral
Some of these models were based on pathophysi- 12 F catheter left in situ for 4 h. This murine
ological theories of urethral sphincter dysfunc- model showed a significant decrease in the peri-
tion. For instance, the vaginal distention model urethral striated and smooth muscle and c-Fos
was developed to simulate maternal childbirth immunostaining neurons in the L6 to S1 spinal
trauma and its related direct sheering, ischemic, cord segments, indicating that irritation causes
or neurogenic effects on the function of the ure- nerve and muscle injury. This finding paved the
thral sphincter. The pudendal nerve crush model way for the next studies using vaginal distension
is a more specific model of neurogenic urethral methods for animal models of stress urinary
dysfunction and isolates the injuries to the puden- incontinence and recovery after childbirth [81].
dal nerve. Both models are unique for the charac- Since then, many authors have modified the
teristic of reversibility where the dysfunction in vaginal distension method in rats or mice using
the urethral function spontaneously resolves. different sized catheters or balloons or by
This characteristic is ideal for researchers who changing the distension time. However, the
are primarily interested in understanding mecha- results have always been similar with short-term
nisms of injury and repair in the pathophysiology effects including urethral dysfunction, a
of urethral dysfunction. Researchers interested in decrease in urethral resistance, neuromuscular
generating a durable model of dysfunction have damage of structures responsible for continence
developed non-pathophysiologic-based models with edema in the pelvic musculature, and over-
such as urethrolysis, cauterization, and pudendal expression of hypoxia-inducible factors and cel-
nerve transection. A durable model is generally lular markers of innate repair. The main
defined as stress urinary incontinence lasting limitation of this model is the short durability of
3 months or longer. Therefore what is advanta- the functional, structural, and biomechanical
geous to one group may be disadvantageous to defects due to vaginal distension, with a recov-
the other group. The necessity of animal models ery within 10 days [82] and up to 6 weeks [81],
is essential to determine the best cell therapeutic depending on the vaginal distension time. This
approach to treatment of stress urinary inconti- limitation makes the vaginal distension method
nence. However, animal models are limited in useful for mechanistic studies of childbirth-­
that we do not have their cooperation, and thus induced tissue injury and recovery, but it is chal-
we assess continence through various surrogate lenging for testing the efficacy of stress urinary
outcome measurements that also constitute a incontinence treatments beyond 6 weeks. The
source of interlaboratory variability. In this sec- strength of this model is that it recapitulates
tion, the different models of stress urinary incon- childbirth injury which is one of the strongest
tinence urethral dysfunction are introduced. known risk factors for the development of stress
urinary incontinence. However, to the best of
10.2.2.2 Pathophysiological Animal our knowledge, the long-term effects of simu-
Models of Reversible lated childbirth injury have not been investi-
Incontinence gated in animals. Of the women who have full
remission of stress urinary incontinence after
Vaginal Distension Model delivery, almost half redevelop stress urinary
incontinence 5 years later, and many more
Murine Vaginal Distension Model develop stress urinary incontinence decades
One of the first animal models to study the later in menopause. For this reason it is impor-
pathophysiological mechanisms of stress uri- tant to test the effect of age in vaginally dis-
nary incontinence associated with childbirth tended rodents, since they should be expected to
was developed 15 years ago by Lin et al. [80] redevelop stress urinary incontinence as a con-
and performed in rats using a vaginal distension sequence of their altered function or support.
248 E.S. Yoo and J.N. Lee

Cell Therapy in Vaginal Distension Murine developed a murine model to study external ure-
Model thral sphincter dysfunction associated with stress
Vaginal distension rat models have been used to urinary incontinence due to pudendal nerve dam-
study stem cell regeneration therapies. For exam- age during childbirth [84]. The pudendal nerve
ple, autologous mesenchymal stem cells were was accessed dorsally and then crushed bilater-
shown to home to the urethra and vagina after tail ally in the ischiorectal fossa. The electromyo-
vein injection [59]. Although this study showed grams showed motor units undergoing typical
improvement in continence as measured by leak denervation changes followed by regeneration
point pressure 1 week posttreatment, the external and recovery. Pan et al. used a similar model [85]
urethral sphincter function as measured by elec- and demonstrated greater alterations in histologi-
tromyography was not improved at that time. cal muscle atrophy in diabetic rats, as well as a
Therefore, regenerative treatment investigations reversible reduction in leak point pressure versus
may be limited by the short durability exhibited the control group. Recently, Castiglione et al.
in the vaginal distension models. Other authors [86] used a rat model for pudendal neuropraxia to
have combined vaginal distension with bilateral test betamethasone as a treatment for bladder
ovariectomy in rats [67] to study tail vein or ure- dysfunction and showed that pudendal nerve
thral injection of adipose-derived stem cells and crush leads to stress urinary incontinence with
showed that 66.7% of the animals had normal irregular micturition by involuntary muscle
voiding function after 1 month of treatment as contractions.
measured by four-channel conscious cystometry.
Fecal Incontinence
Cell Therapy in Vaginal Distension Larger This model has been used to test stem cells as a
Animal Model therapy in a rat model of fecal incontinence [87].
The vaginal distension technique has been estab- However, no significant differences in either rest-
lished in larger animals as well. Burdzin’ska et al. ing or peak pressure of the anal sphincter follow-
developed the vaginal distension method in pigs ing treatment at 10 days postinjection with
and showed a decrease in maximal urethral closure mesenchymal stem cells were observed. Although
pressure (50%) and functional urethral length this model is useful for investigating mechanisms
(52%) 28 days post-vaginal distension [83]. The of neuromuscular recovery and for testing neuro-
advantage of this animal model is that the external regenerative agents and can be used in conjunc-
urethral sphincter of pigs and humans consists of a tion with other pathological animal models to
high percentage of slow-twitch myofibers and uro- assess mechanisms of injury and recovery in pop-
dynamic evaluations can be carried out using the ulations at increased risk for stress urinary incon-
same procedure used for human patients, whereas tinence, its short-term durability could represent a
this is not the case in rodents or dogs [83]. The dis- handicap in cell-based therapy investigations.
advantage of larger animal models is the rate of
animal growth during the study and the mainte- Vaginal Distension Combined with Pudendal
nance costs of larger animals compared to murine Nerve Crush Model
models. However, it is important to have both small Other animal models combine the two method-
and large animal models available since regulatory ologies vaginal distension and pudendal nerve
requirements that apply to novel therapies usually crush to mimic stress urinary incontinence after
involve completion of a full nonclinical develop- childbirth. Song et al. developed a murine model
ment program. combining vaginal distension and pudendal nerve
crush to study the long-term effects of damage
Pudendal Nerve Crush associated with childbirth [88]. Six weeks after
Pudendal Nerve Crush Model Introduction surgery, there was no difference in the leak point
The pudendal nerve that controls external ure- pressure data between the vaginal distension/
thral sphincter activity can be injured during vag- pudendal nerve crush and the sham group.
inal delivery. Among other authors, Kerns et al. However, after 9 weeks distal nerve regions were
10  Urethral Sphincter: Stress Urinary Incontinence 249

diffuse and were innervated by tortuous and mul- urethrolysis that resulted in neuromuscular and
tiple axons, demonstrating that reinnervation of functional differences between incontinent and
the area was still in progress and could explain sham-operated rats [90]. Urethrolysis was
female recurrent stress urinary incontinence after ­performed by making a lower abdominal midline
the recovery following the first childbirth. incision. After the bladder and the urethra were
identified, the proximal urethra was detached cir-
Conclusion of Reversible Incontinence Model cumferentially by incising the endopelvic fascia,
In conclusion, reversible stress urinary inconti- and the remaining urethra was detached from the
nence models such as vaginal distension, pudendal anterior vagina and the pubic bone. They
nerve crush, or a combination of both may be use- observed a significant decrease in retrograde ure-
ful to evaluate pathophysiological mechanisms thral perfusion pressure. The mean leak point
associated with stress urinary incontinence after pressure and retrograde urethral perfusion pres-
childbirth, urethral damage and recovery, and neu- sure decreased to 9.8 cm H2O and 11.2 cm H2O,
romuscular recovery to test potentially neurore- respectively, at 1 week post-urethrolysis, and
generative treatments and to identify favoring these changes were maintained for up to
situations for recurrence of stress urinary inconti- 24 weeks. After urethrolysis, histological analy-
nence. Cell therapy investigations using the vagi- sis showed a 65% reduction in urethral smooth
nal distension model would be interesting, but due muscle with a high correlation between retro-
to their reversibility in the absence of treatment, grade urethral perfusion pressure and muscular
they must be combined with other stress urinary atrophy. The number of apoptotic cells in the ure-
incontinence models to ensure structural and func- thra and bladder was also significantly higher,
tional viability of the injected cells. with the most obvious apoptosis rate in the sub-
mucosa and muscle layers. Since then, others
10.2.2.3 Pathophysiological Animal have improved this technique showing the repro-
Models of Durable ducibility of the urethrolysis mechanism as an
Incontinence animal model for lasting stress urinary inconti-
nence with decreased urethral resistance and con-
Durable Animal Model nective tissue damage for at least 8 weeks and up
Another strategy to generate stress urinary inconti- to 12 weeks [91]. Recently, Skaff et al. developed
nence in animal models consists of simulation of a durable animal model of stress urinary inconti-
periurethral sphincter damage through direct or nence in rabbits by using urethrolysis which
indirect mechanisms that produce a longer-­lasting resulted in a significant decrease in the leak point
form of stress urinary incontinence. Among other pressure (from 33 to 12 cm H2O) and a decrease
direct mechanisms, it is important to mention in 22% of the smooth muscle density for up to
sphincter damage by cauterization and periurethral 12 weeks according to histological analysis [91].
damage by sphincterotomy. Indirect mechanisms
include bilateral pudendal nerve transection. Limitation of Urethrolysis
Although the urethrolysis model induces a severe
Urethrolysis type of incontinence that is not entirely represen-
tative of the complex etiologic factors associated
First Study of Urethrolysis and Method with stress urinary incontinence, and it is not
Including Durability known if full physiological recovery to the nor-
In 1999, Kato et al. developed a canine model to mal preoperative status could occur in such a
evaluate the effects of urethrolysis and periure- destructive model, these studies show how the
thral nerve resection on the capacity and compli- alteration of anatomical structural components of
ance of the bladder [89]. Their experiments laid the continence mechanism plays a key role in the
the groundwork for others, such as Rodriguez development of stress urinary incontinence. The
et al., to establish a murine model of stable and urethrolysis model could represent severe forms
durable urethral dysfunction by transabdominal of incontinence associated with non-sparing
250 E.S. Yoo and J.N. Lee

prostatectomy during cystectomy or urethral zation was developed by Chermansky et al. in


intrinsic sphincter deficiency or apoptosis of the which cauterization of the periurethral region
muscle that correlates with stress urinary inconti- 1 cm from the bladder neck to the upper face of
nence and age. However this model may or may the symphysis of the pubis for 30 s at an elevated
not be reflective of stress urinary incontinence in temperature (up to 1204°C) was performed in
women over a lifetime. rats [93]. Sphincter function was assessed by
measuring the leak point pressure through a
Enhanced Model of Urethrolysis (Urethrolysis suprapubic bladder catheter. Before the measure-
with Cardiotoxin Injection) ment, the spinal cord was transected at the T9–
Kinebuchi et al. used this methodology in a rat T10 level to eliminate the micturition reflex with
model of stress urinary incontinence which com- increased intravesical pressure. Six weeks after
bined urethrolysis with cardiotoxin injection and surgery, cauterized rats with sphincter damage
tested the use of bone-marrow-derived mesenchy- demonstrated a significant decrease in leak point
mal stem cells as a cell-based therapy [92]. In this pressure versus the sham-operated group, and
model cultured autologous bone-marrow-­derived differences remained for 16 weeks. Histological
mesenchymal stem cells were injected into the evaluation of the midurethra at 6 and 16 weeks
periurethral tissues, and leak point pressure was post-electrocauterization showed an alteration in
measured before injury and during the follow-up neuromuscular periurethral striated fibers, which
for up to 13 weeks postinjection. Despite that the was even more evident in the later evaluation.
authors view that the bone-­marrow-­derived mes- Although to date self-regeneration of striated mus-
enchymal stem cells group transplanted cells sur- cles after electrocauterization has not been docu-
vived and differentiated into striated muscle cells mented and therefore one may think that this is a
and peripheral nerve cells and that there was a good model to test cell-based therapies, we should
clear trend toward recovery of leak point pressure note that this model mainly reproduces damage of
in bone-marrow-derived mesenchymal stem cells- the urethral sphincter that can occur accidentally
transplanted urethras, no significant effect was during a surgical procedure, such as during prosta-
detected. Although this study did not result in tectomy in men or any procedure approaching the
improved continence, the significant reduction in urethra and anterior vagina in women. Therefore,
smooth muscle and correlation with urethral pres- this type of stress urinary incontinence could rep-
sure observed in this model [90, 91] suggests that resent a conventional model.
it may be useful for investigators interested in
muscular regeneration of the internal urethral Limitation of Electrocauterization
sphincter composed of smooth muscle. This may However, it is not fully representative of the com-
be especially important since it has been sug- plex etiologic factors associated with stress uri-
gested that the density of the circular smooth nary incontinence, and like urethrolysis, it is
muscle decreases up to 50% with age which may unclear if full physiological recovery to the pre-
account for age-related declines in urethral clo- operative state occurs. Having said that, regener-
sure pressure, a predominant factor associated ative therapies have been tested in rat models of
with stress urinary incontinence and aging in stress urinary incontinence induced by
women more often than loss of urethral support. electrocauterization.

Electrocauterization Electrocauterization Studies


For example, Lim et al. used mononuclear cells
Method of Electrocauterization and Durability isolated from human umbilical cord blood as a
Like urethrolysis, electrocauterization achieves a source of stem cells [94]. Four weeks after injec-
similar outcome of stress urinary incontinence tion, leak point pressure values were significantly
but via a different method. The most representa- higher in the experimental group, and the external
tive animal model of periurethral electrocauteri- urethral sphincter of the experimental group
10  Urethral Sphincter: Stress Urinary Incontinence 251

remained well organized and intact, while the surgery and an absence of muscle cells [99].
saline-treated control group showed severe degen- Furthermore, in vivo pudendal nerve stimulation
eration and disruption of the sphincter muscle lay- confirmed the loss of sphincter tissue function.
ers. However, the mechanism as to how the Therefore, if this model [99] is reproducible in
injected human umbilical cord blood mononu- other animals and results in long-lasting stress
clear cells acted on the urethral sphincter is not urinary incontinence, it may represent a tool for
explained in this work. Although they found the evaluation of methods reproducing sphincter
human cells in the lamina propria and in the stri- function and intending to regenerate smooth and
ated sphincter layers at 2 weeks, they could not striated muscle of the internal and external ure-
find human cells at 4 weeks. In a previous study, thral sphincter, respectively.
intraurethral injection of muscle-­ derived cells
improved urethral sphincter function in an elec- Limitation of Sphincterotomy
trocauterized rat model of intrinsic sphincter Although this model does not represent the multi-
deficiency [95]. Despite that they showed faceted etiology of stress urinary incontinence,
improvement in sphincter function after 4 weeks one needs to consider that a major postoperative
and that the muscle-derived cells had integrated complication of radical prostatectomy is damage
within the striated muscle layer of the cauterized to the muscle-nerve-blood vessel units around the
tissue, they could not answer the question whether urethra. Vaginal delivery also causes varying
muscle-derived cells were bulking the urethra or degrees of neuromuscular damage. Such a micro-
promoting the functional recovery of the injured surgical approach to remove the sphincter muscle
sphincter. In another study the use of muscle pre- offers the advantage that it produces durable dam-
cursor cells isolated from limb myofibers of rats age and that the surgical techniques are relatively
were used to regenerate the sphincter in an elec- simple and reproducible. In addition, it can gener-
trocauterized rat model of stress urinary inconti- ate a direct muscle injury without pronounced
nence and showed partial muscle regeneration in nerve damage therefore potentially allowing a
the context of denervation and fibrosis as demon- better direct assessment of a muscle regenerative
strated by histologic staining and immunofluores- therapy with easier electromyographic measure-
cence detection of beta-­galactosidase expressed ments. However, like the other destructive mod-
in the injected muscle precursor cells [96]. els, it is not known if full physiological recovery
to the normal preoperative status can occur.
Urethral Sphincterotomy
Pubourethral Ligament and Pudendal
Method of Sphincterotomy and Durability Nerve Transection
Similar models of direct sphincter damage by
muscle resection have been developed as a model Durability of Pubourethral Ligament
not only for stress urinary incontinence but also and Pudendal Nerve Transection
for fecal incontinence [97]. In 2007 Praud et al. Animal models of incontinence by transection of
developed three mouse models of stress urinary the pubourethral ligament, which removes the
incontinence (freezing, longitudinal sphincterot- structural support of the urethra, and/or transec-
omy, and periurethral injection) and observed tion of the pudendal nerve generated stress uri-
that only sphincterotomy caused stress urinary nary incontinence for up to 1 month. Kefer et al.
incontinence as measured by a decrease in ure- compared the two methods and transected the
thral closure pressure 21 days after surgery [98]. pubourethral ligament between the middle ure-
Eberli et al. described a durable canine model of thra and posterior symphysis of the rat and the
sphincter deficiency by microsurgical abdominal pudendal nerve via a dorsal incision and the
sectioning of 25% of the periurethral sphincter bilateral ischiorectal opening [100, 101]. After
(smooth and skeletal) muscle that resulted in a 28 days, a decrease in leak point pressure was
decrease in urethral profiles up to 7 months post-­ observed in the rats with pubourethral ligament
252 E.S. Yoo and J.N. Lee

or pudendal nerve transection compared to the based on voiding efficiency. Other non-rodent
sham-operated group. Following pubourethral animal models of stress urinary incontinence
ligament transection, histologic studies demon- have been developed as well, including cats,
strated an absent pubourethral ligament for up to dogs, and more recently nonhuman primates.
4 weeks post-injury. Therefore, the long-term Badra et al. developed a model for stress urinary
durability of this model is unclear at this point. incontinence in premenopausal female primates
Applications directed at treating hypermobility by abdominal bilateral transection of the puden-
of the urethra could benefit from the use of this dal nerve and cauterization of the pudendal nerve
model. Pubourethral ligament could have similar [103]. Urodynamic investigations were per-
effects to the urethrolysis model, since transec- formed before injury and 3, 6, and 12 months
tioning the ligaments may damage vascular, after injury. Electromyography prior to necropsy
nerve, and possibly muscle structures, and could showed decreased levels in the transection of the
also be combined with other models to investi- pudendal nerve group versus the sham group.
gate cell therapies, such as transection of the Histological and inmunohistochemical analysis
pudendal nerve, favoring the neurogenic atrophy showed a decrease in smooth and striated muscle
and decreased periurethral neurofilaments. fibers, increased collagen, and decreased vascu-
larization in injured animals. Cystogram results
Bilateral Pudendal Nerve Transection were consistent with those results showing
(Transection of the Pudendal Nerve) greater amplitude and incompetence of the blad-
der outlet and the external urethral sphincter.
Durability of Bilateral Transection
of the Pudendal Nerve Advantage of Transection of the Pudendal
Bilateral pudendal nerve transection has been Nerve
used in several animal models as a mechanism of This model offers the advantage that it may be
a stress urinary incontinence model due to indi- representative of neurogenic damage that occurs
rect periurethral sphincter damage. In 2003 in radical prostate surgery or urethral denervation
Kamo et al. demonstrated the importance of the and/or pudendal nerve and muscular damage that
pudendal nerve in the continence mechanism in a probably occurs during vaginal delivery [102].
murine model by demonstrating an 80% decrease These studies demonstrate the feasibility of using
in the muscle response at the midurethral level this model in cell therapy approaches investigat-
when transectioning the bilateral pudendal nerves ing regeneration of neuromuscular tissue. It also
and the nerves to the iliococcygeus and pubococ- demonstrates that transection of the pudendal
cygeus muscles, but not by transection of the vis- nerve affects the structural and functional ana-
ceral branches of the pelvic nerves and tomical capacity of the periurethral urethral
hypogastric nerves [101]. Their data also showed sphincter in a human-like animal model.
that in stress conditions such as sneezing, the
contribution of pudendal nerve-mediated striated Limitation of Transection of the Pudendal
muscle activity in the external urethral sphincter Nerve
to the continence mechanism may be greater than Despite the success of such stress urinary inconti-
that of iliococcygeus and pubococcygeus muscle nence animal models, most if not all cell therapy
activity. Peng et al. investigated the effect of the regenerative assessments have been performed to
unilateral versus the bilateral transection of the date in murine models (rats or mice) using different
pudendal nerve in rats [102]. After 6 weeks they cell types such as human or allogeneic skeletal
observed a significant decrease in leak point pres- muscle stem cells [104, 105], autologous or alloge-
sure and striated muscle atrophy in rats that neic adipose-derived stem cells [106, 107], or allo-
underwent unilateral transection of the pudendal geneic bone-marrow-­ derived mesenchymal stem
nerve, which was even more prominent in the cells [66, 108]. These aspects of regenerative cell
bilateral transection of the pudendal nerve group therapies will be discussed in the next section.
10  Urethral Sphincter: Stress Urinary Incontinence 253

10.2.2.4 R  egeneration of the Urethral continence (Valsalva leak point pressure) as mea-


Sphincter Using Cell Therapy sured by cystometry at 4 weeks [66, 108].
in Animal Models Mesenchymal stem cells survived and were via-
The feasibility and efficacy of different strategies ble in the injected area based on immunohisto-
of cell therapy, at times combined with bioengi- chemistry analysis. Histopathologic regeneration
neering techniques, have been tested in various of striated muscle was observed by desmin and
animal models of sphincter regeneration. Given myosin staining, respectively. However this
the legal and ethical issues on the use of embry- approach is far from optimal since it is well
onic stem cells and their higher tumor potential, known that fluorescent dyes coming out of trans-
the strategy in the last decade has been based planted cells can be transferred to surrounding
largely on the use of adult stem cells derived cells and can mistakenly be taken as evidence for
from muscle, adipose tissue, and bone marrow. differentiation. Other authors have evaluated the
use of biomaterials combined with mesenchymal
Mesenchymal Stem Cell-Based Study stem cells for sphincter regeneration. Du et al.
evaluated submucosal injection at the level of the
Mesenchymal Stem Cell Homing Study urethra and bladder neck of an alginate/calcium
Therapies using mesenchymal stem cells have gel alone or in combination with mesenchymal
been used in stress urinary incontinence rodent stem cells or muscle-like predifferentiated cells
models of vaginal distension and various strate- in vitro with 5-azacytidine in a rat stress urinary
gies of bilateral transection of the pudendal incontinence model that used a bilateral transec-
nerve. Dissaranan used bone-marrow-derived tion of the pudendal nerve model combined with
mesenchymal stem cells in a vaginal distension transectioning of the nerves innervating the ilio-
rat model to investigate homing of mesenchymal coccygeus/pubococcygeus muscle [109]. The
stem cells to pelvic organs after i.v. injection and three experimental groups showed an improve-
recovery from simulated childbirth injury [59]. ment in leak point pressure at 1, 4, and 8 weeks
However, a limitation of this study was that their posttreatment compared to the control group, and
assessments were short term (after 1 week postin- immunohistochemical analysis resulted in des-
jection) and mesenchymal stem cells from pas- min- and alpha-SMA-positive cells in all three
sage 16 were used. Interestingly though, they did groups, without observing clear and obvious spe-
show that the vaginal distension model disrupted cific muscle regeneration. In an analysis per-
the smooth and striated muscle of the urethral formed at 8 weeks after injection, the gel still
sphincter and that mesenchymal stem cells pref- maintained a certain volume, and therefore it
erentially engrafted in the smooth muscle of the cannot be ascertained whether the improvement
urethra and vagina. Although there was an in leak point pressure was due to a bulking effect
improvement in leak point pressure, the external since there was no group injected with cells
urethral sphincter function was not improved. alone. A remarkable fact is that addition of the
While this study suggests that mesenchymal stem calcium alginate composite gel seemed to stimu-
cells can home to the damaged tissue and may aid late angiogenesis with a higher density of newly
in some level of recovery in continence, other formed microvessels in the cell plus gel group,
methods of injection such as direct injection into which was not observed in the gel group alone.
the sphincter may produce different results.
Amniotic Fluid Stem Cells
Mesenchymal Stem Cells + Periurethral In a combined urethrolysis and cardiotoxin
Injection injection stress urinary incontinence female rat
Corcos et al. and Kim et al. tested periurethral model, bone-marrow-derived mesenchymal
injection of mesenchymal stem cells in rats that stem cells differentiated into striated muscle
underwent bilateral transection of the pudendal cells and peripheral nerve cells in vivo, but only
nerve and reported an improvement in urinary resulted in a small improvement in urodynamics
254 E.S. Yoo and J.N. Lee

[92]. A recent study tested different combinations delivery products may enhance such an effect,
of what was defined by the authors as “early” but are they enough to reinnvervate the tissue?
muscle, neuron, and endothelial cells that were
differentiated from human amniotic fluid stem Adipose-Derived Stem Cell-Based Study
cells for 7–21 days in vitro prior to injection in a
transection of the pudendal nerve female mouse Adipose-Derived Stem Cell Introduction
model [110]. Histological and immunohisto- (Advantage and Disadvantage)
chemical analysis revealed that regeneration of The use of adipose-derived stem cells or deriva-
the sphincter muscle was accelerated in the mus- tives has been evaluated in different murine mod-
cle group through fusion with damaged cells, as els of stress urinary incontinence using vaginal
well as activation of the local muscle progenitor distension or transection of the pudendal nerve
cells, but the enhancement was less pronounced models [67, 106, 113, 114]. In most of the cases,
and the regenerated muscle quality was lower the functional assessment after adipose-derived
compared to the muscle/neuron/endothelial cell stem cells injection was evaluated by cystometric
group. The muscle/neuron and muscle/neuron/ parameters (bladder capacity, leak point pressure,
endothelial cell groups showed increased muscle and retrograde urethral perfusion pressure) and
bundles in the urethral sphincter, while the triple reported a significant increase in these values in
combination group showed areas of connective the cell-injected groups compared to control
tissue masses at the urethral sphincter and higher groups. However, the muscle regenerative capac-
nerve and endothelial cell marker expression. ity or the in vivo mechanisms of these cell sources
Real-time PCR results also demonstrated higher to achieve such results are not well defined. The
levels of myogenic, neuronal, and endothelial incontinence models most commonly used with
marker expression in the muscle/neuron/endothe- this cellular source were investigated using the
lial cell group. At 4 weeks, the leak point pres- vaginal distension model as shown by Obinata
sure and closure pressure was significantly higher et al. [114]. Although their group demonstrated a
for each of the three cell injection groups versus recovery in muscle atrophy starting at 2 weeks,
the control which the authors propose may be due which was fully evident at day 28, it is difficult to
to tissue edema and increased outflow resistance assess to what extent muscle tissue regeneration
due to over injection. This suggests that in addi- of the damaged tissue was achieved since muscle
tion to the effect on myogenesis, mesenchymal regeneration markers included collagen I/III, elas-
stem cells may have a synergistic effect on neuro- tic fibers, and alpha-SMA which do not guarantee
genesis, the formation of neuromuscular junc- the regeneration of a quality and morpho-­
tions and angiogenesis during regeneration. functionally competent smooth and/or striated
muscle. What is evident in these models is the cell
Limitation viability and the paracrine capacity of adipose-­
The limitation of these studies was a short-term derived stem cells at the injection site.
follow-up (4–8 weeks) for functional analyses.
Another consideration is the injury models used. Adipose-Derived Stem Cell Several Studies
For example, if a denervated urethral sphincter Zhao et al. [106] and Obinata et al. [114] reported
(e.g., after bilateral transection of the pudendal cell survival and viability of adipose-derived
nerve) is treated with a cell-based therapy, can stem cells in the injection area. In a vaginal dis-
the regenerated tissue become innervated in a tension stress urinary incontinence model, Zhao
physiological manner? Reinnervation of muscle et al. [106] demonstrated how the use of a
tissue after injury in such a model is necessary biomaterial-­derived hydrogel (polylactic-co-­
for functional recovery to occur. Mesenchymal glycolic acid, PLGA), in conjunction with cell
stem cells are known to generate neurotrophic therapy (mature adipocyte-derived dedifferenti-
factors and can promote endogenous neuronal ated fat cells), can be used to generate micropar-
growth [111, 112], and the use of biomaterials or ticles (growth factor delivery “vehicles”) that
10  Urethral Sphincter: Stress Urinary Incontinence 255

allow the introduction of certain factors such as pressure [116]. However, electromyographic
nerve growth factor to improve the microenviron- activity demonstrated that only the muscle-­
ment and generate neurofilaments. However, it is derived cells had the ability to differentiate into
not clearly defined if the improvement in bladder myotubes and myofibrils and integrated into the
capacity of this group (mature adipocyte-derived sphincter tissue. This model also suggests that a
dedifferentiated fat cells + PLGA/nerve growth transient bulking effect, but not morphological
factor) versus control is due to the self-­ and functional regeneration, could occur by
improvement of the cellular microenvironment injection of fibroblasts since the improvement
in vivo or an increased bulking effect. Li et al. reported was dose dependent and a linear increase
[67, 113] observed that some adipose-derived in leak point pressure at increasing fibroblast
stem cells may exhibit a vascular endothelial doses occurred, except at high doses of fibroblast
growth factor paracrine activity. Moreover this injection which caused urinary retention. In order
group showed increased vascular density and to improve the disadvantages of using muscle-­
increased expression of p-ERK1/2, which could derived stem cells including migration and
induce favorable changes in the extracellular absorption, Xu et al. studied the effect of adding
matrix and cause trophic effects of immunomod- a biodegradable fibrin glue to the periurethral
ulation and cell survival, thus improving the muscle-derived stem cells injection therapy
microenvironment for regeneration. Mature [105]. They observed that although both groups
adipocyte-­derived dedifferentiated fat cells alone injected with muscle-derived stem cells or
have also promoted functional recovery from spi- muscle-­derived stem cells + fibrin glue showed a
nal cord injury-induced motor dysfunction in rats significant improvement in the leak point pres-
[115], suggesting reinnervation may occur with sure, only the muscle-derived stem cells
adipose-derived stem cells or mature adipocyte-­ implanted in combination with the fibrin glue
derived dedifferentiated fat cell treatment. appeared to integrate into the host tissue and
result in an increase in survival and a tendency to
Innervation Comment form multinucleated myofibrils. These results
However, the same question arises as with mesen- demonstrated that the addition of biomaterials
chymal stem cells; can cell-based therapy lead to such as fibrin may not be necessary to generate
innervation in a physiological manner after bilat- an early functional improvement based on the
eral transection of the pudendal nerve or pudendal analysis of the leak point pressure, but may pro-
nerve crush (which may in fact be more represen- mote the implantation and differentiation of
tative of postpartum stress urinary incontinence)? muscle-­derived stem cells into striated muscle.
Some groups have suggested that factors such as
brain-derived neurotrophic factor may need to be Muscular Precursor Cell-Based Study
delivered with the stem cells to lead to such an
effect. Myoblast Researches
Other strategies have included the isolation and
Muscle-Derived Cells and Muscle-Derived implantation of muscular precursor cells from
Stem Cell-Based Study muscle biopsies. Some results suggest that the
use of myoblasts could be a potential treatment
MDS Researches for damaged urethral sphincter muscle regenera-
Cell therapy with muscle-derived cells and tion. Praud et al. performed periurethral injec-
muscle-­ derived stem cells has been tested in tions of skeletal myoblasts in a stress urinary
murine models of stress urinary incontinence by incontinence murine model of sphincterotomy
pudendal or sciatic denervation [105, 116]. Kwon and reported a significant improvement in ure-
et al. showed that injection of muscle-derived thral closure pressure [98]. However, there was
cells, fibroblasts, or both can produce a func- only fusing of myoblasts and regeneration of
tional improvement of continence in leak point new myofibers when the cells were grafted into
256 E.S. Yoo and J.N. Lee

normal striated skeletal muscle, which was not showed that nerve fibers of different sizes were
observed in the incontinent animals. Due to a large present in between the newly formed muscle tis-
similarity between human and porcine rhabdo- sue in the cell-treated group. Urodynamic evalu-
sphincter muscle cells, Mitterberger et al. investi- ations showed an improvement in static, stress,
gated myoblast injection in a non-­incontinent pig and detrusor leak point pressure, electromyo-
model [117]. They showed cell dose-dependent graphic parameters demonstrated an improve-
functional effects. Thus, injection of high cell ment in pressure during stimulation of the
numbers (7.8 × 107) led to a clear increase in ure- pudendal nerve, and anatomical parameters (cys-
thral pressure values at 3 weeks compared with tourethrogram) showed a urethrovesical mor-
low cell dose groups (4.4 × 107). However, the phology in the muscle precursor cell-injected
mechanisms underlying these effects were not group comparable to normal anatomy at 1, 3, and
explained. In the histological analysis, integration 6 months postinjection. Again, this highlights the
and differentiation of myoblasts was observed, as importance in survival and integration of the cells
well as an increase in the formation of new myofi- using a cell carrier in the context of cell therapy.
brils in the high cell dose injected groups.
Likewise, Yiou et al. demonstrated that periure- Human Cell-Based Study
thral injection of muscle precursor cells in a Finally, other groups have gone a step further and
murine stress urinary incontinence model caused instead of using allogeneic or autologous animal
by electrocauterization promoted the formation of cells in animal models, they used human cells
myotubes and neuromuscular junctions [118]. implanted in animal models of incontinence.
Muscle precursor cell injection resulted in forma-
tion of myotubes and 40% restoration of the Human Muscle Precursor Cell Studies
sphincter function 1 month after the injection. Kim et al. evaluated the feasibility of using
human muscle precursor cells that were injected
Muscle Precursor Cell Researches periurethrally in a sciatic sectioning murine
While most studies on cell therapy using myo- model of incontinence [119]. They showed hom-
blasts injected into the bladder and urethra were ing of the muscle precursor cells to the sphincter
performed in murine models, recently Eberli by immunofluorescence using human-specific
et al. investigated the use of muscle precursor antibodies. An improvement in leak point pres-
cells in a sphincterotomy canine model of stress sure at 4 weeks posttreatment was reported.
urinary incontinence [99]. Their group showed
that injection of autologous muscle precursor Human-Human Umbilical Cord Blood Studies
cells, with a low concentration of collagen used Lim et al. used human umbilical cord blood as a
as a cell carrier to provide a microenvironment treatment for stress urinary incontinence [94]. A
for the muscle precursor cells, significantly significant improvement in leak point pressure
decreased urethral pressure values and was struc- 4 weeks after cell injection was reported, as well
turally and functionally viable after 6 months. as homing of DiI-labeled injected cells to the lam-
Histological analysis revealed the capacity of not ina propria and the muscular urethral sphincter at
only skeletal muscle development but also nerve 2 weeks postinjection. However, 4 weeks after
development, demonstrating that the injected injection human cells could not be detected in the
muscle precursor cells are able to survive in vivo urethral tissue, perhaps because the iron oxide
for an extended period of time. Thus, they showed label of the injected cells was already metabolized
histologically how injected muscle precursor at that point or because the injected cells were
cells grow in clusters and form new myofibrils phagocytized and lost. Histologically, desmin
starting 1 month after the treatment and at 3 and staining in the control group showed a markedly
6 months new muscle fiber formation occurred, disrupted sphincter muscle with atrophic muscle
although thinner and less organized than native layers and collagen deposition, while the sphinc-
muscle tissue. Moreover, staining with S100b ter muscle was restored without damage in the
10  Urethral Sphincter: Stress Urinary Incontinence 257

cell-treated group. Despite a functional improve- treatment of muscle, neuron, and endothelial cells
ment in the human umbilical cord blood group, derived from human amniotic fluid stem cells as a
the mechanism as to how the injected human cell therapy in the same mouse model of stress uri-
umbilical cord blood mononuclear cells act on the nary incontinence described above [110]. This
urethral sphincter is not explained in this work. time they established four experimental groups:
(1) amniotic fluid stem cells, (2) a single-cell
Human Amniotic Fluid Stem Cell Studies group containing early muscle progenitor differen-
Recently, human amniotic fluid stem cells have tiated cells, (3) a double-cell group containing
been proposed as a stem cell source for various muscle/endothelial or muscle/neuron progenitor
cell therapies and tissue engineering. Thus, Kim cells, and (4) a triple-cell-combination group with
et al. used amniotic fluid stem cells in a bilateral muscle/neuron/endothelial progenitor cells.
transection of the pudendal nerve murine model of Functional evaluation of stress urinary inconti-
stress urinary incontinence and demonstrated an nence as evaluated by leak point pressure and clo-
improvement in the leak point pressure and clo- sure pressure reported a significant improvement
sure pressure (using the vertical tilt/intravesical in all experimental groups compared to the control
pressure clamp model) in the cell-transplanted group, with the triple-cell-injected group showing
group after 4 weeks of injury [120]. In this work, superlative improvement compared to other exper-
FACS analysis showed a lower expression of imental groups. Again CD8 T cell evaluations
HLADR in the human amniotic fluid stem cells, demonstrated a low immunoreaction in the experi-
and their injection did not stimulate CD8 T cell mental groups versus the control group. The cell
infiltration into the injected area or tumor forma- tracking method used in the previous work showed
tion after 8 weeks postinjection, suggesting that the best result in terms of homing for the triple-
these cells are immunologically tolerated and/or cell-combination injection. Using the same tech-
result in an immunosuppression effect and have niques as in the previous work, muscle regeneration
low tumorigenic potential. By immunohistochem- and increased formation of blood vessels were
istry using a human-specific antihuman nuclei reported in all groups but to a higher extent in the
antibody, they confirmed that the injected cells triple-cell-injected group. This work reinforces
were able to survive; however, at days 3–7 cell their previous work and adds the option of com-
migration from the injection site to the periurethral bining human progenitor cells obtained from
area was reported, and at day 14 a loss of trans- human amniotic fluid stem cells, suggesting that
planted cells was observed. This is especially mesenchymal stem cells may have a synergic
interesting since in vivo cell tracking was per- effect on myogenesis, neuromuscular junction for-
formed and showed homing of the injected cells mation, and angiogenesis during the regeneration
and cell clustering at the injection site during the of the damaged urethral sphincter.
first 10 days postinjection. Moreover, they reported
muscle regeneration in the periurethral region in Human Myoblast Studies
the experimental group by H&E staining and con- Finally, Bandyopadhyay et al. reported the effec-
firmed this result by MyoD antibody staining and tiveness of human myoblasts injection in a botu-
real-time PCR of early and late striated muscle cell linum-­A toxin injection murine model of stress
marker expression. Alpha-­ bungarotoxin for the urinary incontinence [121]. Significant improve-
acetylcholine receptor expression was reported to ment in the recovery of urination volume and
be similar between the experimental and control muscle atrophy was observed. Following myo-
group. The experimental group also showed higher blast injection, GFP-positive myoblast cells and
neurogenic gene expression (nestin, vimentin, urethral staining for human desmin demonstrated
neurofilament, microtubule-associated protein 2, β homing of the cells to the urethra. They also dem-
III tubulin, glial fibrillary acidic protein) than the onstrated formation of myotubes and an increase
control group. Subsequently in 2014, the same in the thickness of the periurethral muscle in the
authors published the use of a triple combination treatment group compared to the control group.
258 E.S. Yoo and J.N. Lee

10.2.3 Clinical Trials Cell Status (Number, Injection Routes,


Duration) on Clinical Trials
10.2.3.1 Overview of Clinical Trials The number of cells used for the treatment of
Several clinical trials in human subjects with stress urinary incontinence differed considerably
stress urinary incontinence were conducted between the studies and ranged from 0.6 mln to
using cell-based therapy in the last decade. 1020 mln cells. However, it is still unclear by
Generally, the therapy was concluded to be safe which route the implantation of cells should take
and effective in the short term. Disregarding place: peri- or transurethral, injected into the
several studies, where doubts have been raised sphincter or submucosa, or both? Should injec-
about the reliability of the results [14], reports tions be performed in single or repeated doses?
on the efficacy of cellular therapy in humans How long after the first series of injections should
have shown cure rates between 40% and 75%. the following one be performed? Which are bet-
However, these figures are based on small, non- ter to implant: freshly isolated or expanded
controlled studies using different methodolo- in vitro cells? These questions remain
gies, different cell types, and varying unanswered.
terminology, and a reliable overall assessment
of the efficacy of treatment is not possible [122]. Overall Outcomes Including Efficacy
In addition, the cell sources, methods of cell and Safety on Clinical Trials
processing, cell number, and implantation tech- Recent review article [138] showed that the clini-
niques differed considerably between studies, cal outcomes of cell therapy in stress urinary
so a comparison was very difficult. Multiple cell incontinence based on 616 patients indicated that
types were evaluated for treatment of stress uri- mean cure and improvement rates over a
nary incontinence. In several studies, no distinc- 12 month follow-up were 37.2 ± 29.7% and
tion has been made to establish the mechanism 33.1 ± 14.3% (patients), respectively (Table 10.2).
of stress urinary incontinence at baseline, i.e., A comparative analysis of cross-sex outcomes
the patients have not been categorized as having indicated that the cure rate was significantly
ISD of hypermobility. higher in female (41 ± 30.7%) compared to male
patients (28.7 ± 31.5%) (p < 0.01).
Cell Type on Clinical Trials No serious adverse events or major complica-
Different types of stem cells have been used for tions related to the treatment were reported. The
stress urinary incontinence treatment in humans most common adverse events were worsening of
as well as in animal models including skeletal incontinence, urinary retention (<24 h), pain at the
muscle derived (myoblasts, satellite cells, muscle injection site, cystitis, and urinary tract infection.
progenitor cells, and muscle-derived stem cells),
bone marrow stem cells (bone-marrow-derived Regarding Usage of Bulking Agents
mesenchymal stem cells), human umbilical When the outcomes were analyzed separately for
mononuclear cells, adipose-derived stem cells, studies where only cells were implanted from
and modified/sorted stem cells which show vari- those together with a bulking agent (collagen,
ous efficacy [23]. Skeletal muscle-derived cells, adipose tissue), the cure rate over the 12 month
including myoblasts, muscle-derived stem cells, follow-up was significantly lower (p < 0.001) for
and fibroblasts, were the most commonly used in the cell-only group (21.7 ± 8.9%) compared to
recent clinical trials [123–133]. Adipose-derived the cell-bulking agent combination (60.8 ± 36%).
regenerative cells [134, 135], cord blood stem This trend was observed in both male and female
cells [136], and total nucleated cells and platelets patients. So in fact, the results of cell therapy in
[137] were tested less frequently. Almost authors stress urinary incontinence are comparable or
have not reported any significant advantage from even worse to those observed in conventional
the use of any particular type of cells thus far. urethral bulking [20].
10  Urethral Sphincter: Stress Urinary Incontinence 259

Table 10.2  Clinical trials of regenerative therapy on stress urinary incontinence


Follow
Bulking up
Study Cell type agent Female, n Male, n SUI type months Cu% I% Assessment
Mitterberger M, F C 123 0 II or III 12 79 13 Objective
et al. [117, 123]
Mittenberger M, F C 0 63 Iatrogenic 12 65 27 Objective
et al. [124]
Mittenberger M, F C 20 0 III 24 89 11 Objective
et al. [125]
Carr et al. [126] MDSCs None 8 0 Not 12 20 40 Objective
classified (PT)
Lee et al. [136] CBSCs None 39 0 II, III, or 12 36 36 Subjective
II and III
Sebe et al./Cornu M None 12 0 III 72 17 0 Objective
et al. [127, 128] (PT)
Blaganje et al. M None 38 0 Not 6 24 53 Subjective
[129, 140] classified
Gerullis et al. M None 0 222 Iatrogenic 12 12 42 Subjective
[130]
Yamamoto et al. ADSCs AT 0 3 Iatrogenic 6 33 67 Objective
[134] (PT)
Shirvan et al. TNCs-Ps None 9 0 II or III 6 89 11 Objective
[137] (PT)
Carr et al. [132] MDSCs None 38 0 Not 18 20 40 Objective
classified (PT)
Stangel-­ MDSCs None 16 0 Not 24 50 25 Subjective
Wojcikiewicz classified
et al. [131]
Gotoh et al. ADSCs AT 0 11 Iatrogenic 12 9 55 Objective
[135] (PT)
Peters et al. MDSCs None 80 0 Not 12 24 24 Objective
[133] classified (PT)
Female SUI was classified into three basic types: type I defined as urine loss occurring in the absence of urethral hyper-
mobility, type II defined as urine loss occurring due to the urethral hypermobility, and type III defined as urine leakage
occurring from an intrinsic sphincter deficiency (ISD)
ADSCs adipose-derived stem cells, AT adipose tissue, CBSCs cord blood stem cells, C collagen, Cu cure, F fibroblasts,
I improvement, M myoblasts, MDCs muscle-derived cells, MDSCs muscle-derived stem cells, PT pad test, TNCs-Ps
total nucleated cells platelets

10.2.3.2 F  irst Clinical Trial Using by myoblast and fibroblast isolation, expansion,
Myoblast and Fibroblast and implantation. The myoblasts were implanted
The first series of clinical trials utilizing cell ther- into the urethral rhabdosphincter, while the fibro-
apy for stress urinary incontinence were conducted blasts were mixed with collagen and implanted
by a group of investigators from Austria [123– into the urethral submucosa. This innovative strat-
125]. However, their resultant two publications egy resulted in a 65–90% cure rate over a 12-month
were retracted due to ethical and legal irregulari- follow-up, but their results raised several questions
ties. The first reported an approach to stress uri- [123–125]. A 1 year follow-­up seems to be too
nary incontinence treatment involving performing short to indisputably prove the superior efficacy of
the muscle biopsy from the patients’ arm, followed injecting autologous fibroblasts and myoblasts
260 E.S. Yoo and J.N. Lee

into the urethral submucosa and the rhabdosphinc- The same group of investigators later confirmed
ter versus delivering a bulking agent as therapy for the safety and efficacy of this method of treatment
stress urinary incontinence. The efficiency of in a larger dose-ranging study [132]. A total of 38
bulking agents is estimated at 8–12 months, and women with stress urinary incontinence under-
long-term continence is dependent on multiple went intrasphincter injection with low (1, 2, 4, 8,
booster injections. It is highly probable that the or 16 mln) or high (32, 64, or 128 mln) doses of
bulking effect is partially responsible for the muscle-derived stem cells. Of these, 32 patients
reported improvement of continence. Periurethral selected a second treatment of the same dose after
delivery of a solution of mixed stem cells and a 3 month follow-up. Patients who received two
collagen might lead to gradual coaptation of the muscle-derived stem cell implantations exhibited
urethra, caused by spontaneous proliferation of a significant reduction in pad weight (61.5% and
the implanted cells and inflammatory reaction. 88.9% in the low- and high-dose group, respec-
Although it has been shown that collagen is tively) and stress leaks (53.3% and 77.8% in the
absorbed 3–6 months after injection, and since low- and high-dose group, respectively) at
the 1 year cure rates are poor, the addition of a 18 months. Only four patients (two low dose and
high amount of fibroblast could affect collagen two high dose) reported urinary continence at
degradation and activate an intrinsic fibrosis 18 month follow-­up. All four patients who received
reaction similar to scar formation [139]. only a single muscle-derived stem cell implanta-
tion reported a 50% or greater reduction in the
10.2.3.3 M
 uscle-Derived Stem Cells incidence of stress leakage, including one patient
and Muscle-Derived Cell who was dry at 12 months [132].
Clinical Trial
Limitation of Muscle-Derived Stem Cell
Muscle-Derived Stem Cell Trials Trial
In 2008 Carr et al. published the first American The major weakness of the study design concerns
clinical trial of muscle-derived stem cell therapy the number of patients in the subgroups (three to
in eight female patients [126]. Muscle-derived four patients), which was insufficient to achieve
stem cells were isolated from the lateral thigh, statistical significance after data acquisition. The
expanded and implanted by periurethral or trans- authors did not present their results for the vari-
urethral routes. The three patients were with- ous subgroups, namely, low (1, 2, 4, 8, or 16 mln)
drawn from the study after 1 month follow-up. and high (32, 64, or 128 mln) muscle-derived
Partial improvement in symptoms was observed stem cell dosage groups, respectively. This led to
in the remaining five patients, but only one an oversimplification of their results and the inac-
patient achieved total continence. The authors curate conclusion that increased effectiveness of
concluded from their results that both periure- stress urinary incontinence treatment results from
thral and transurethral routes of cell implantation usage of high doses of muscle-derived stem cells
could be successfully used for stress urinary compared with low doses of muscle-derived stem
incontinence treatment [126]. Numerous vari- cells. Determining the minimal number of cells
ables, including cell implantation routes (peri- providing therapeutic effect is fundamental to
urethral or transurethral), injection positions (3, establishing any state-of-the-art method in the
9 or 3, 6, 9, 12 o’clock positions), and number of field of cell-based therapies for stress urinary
cells (with or without reinjection 3 months fol- incontinence. Despite the analyses of study
lowing the first injection), make the results of results, it remains unclear what constitutes the
this small study very difficult to compare and minimal number of implanted cells allowing
discuss. Furthermore, as the muscle-derived improvement of outcomes in stress urinary incon-
stem cells were shipped frozen to the clinic, the tinence treatment—32, 64, 128, or 256 mln?
eventual number of viable cells implanted is How incredibly large would the muscle biopsy
unknown. have to be to obtain 256 mln muscle-derived stem
10  Urethral Sphincter: Stress Urinary Incontinence 261

cells? As the authors did not present data con- However, the power of this study is reduced by
cerning the phenotype analysis of the isolated the small number of patients in the experimental
cells, it is unknown whether muscle-derived stem groups, non-confirmed phenotype of isolated
cells or a co-culture of other cell types were used cells, cell freezing, and only two injection posi-
in this study. Furthermore, as muscle-derived tions (3 and 9 o’clock). Surprisingly, a long-term
stem cells were shipped frozen to their clinic, 6 year follow-up study indicated that two patients
again the number of viable cells implanted is categorized as cured at 1 year evaluation were
unknown. Besides, distribution of muscle-derived still dry at last follow-up. However, of the five
stem cells into the muscle tissue following their patients considered improved at 1 year, all wors-
injection into only two areas of sphincter was ened during follow-up [128].
certainly very difficult, especially when such a Similar studies utilizing myoblasts or muscle-­
high dose of cells was implanted. The most derived stem cells in stress urinary incontinence
important observation from this study was that no treatment were conducted by Blaganje et al. [129,
serious adverse events or major complications 140], Gerullius et al. [130], and Stangel-­
were related to the treatment [132]. Wojcikiewicz [131]. All of them confirmed that
cell therapy provides encouraging results in stress
Muscle-Derived Cell Trial and Limitation urinary incontinence treatment. The delayed
More recently, the same group assessed the safety improvement suggests rather detrusor regenera-
and efficacy of muscle-derived cells for urinary tion than bulking effect. Unfortunately, the num-
sphincter repair in 80 women with stress urinary ber of cells utilized for stress urinary incontinence
incontinence [133]. Patients received intrasphinc- treatment differs considerably in each of the stud-
ter injections of 10, 50, 100, or 200 mln autolo- ies, e.g., 1–50 mln [129, 140], 1.2–19.2 mln
gous muscle-derived cells. However, as the [130], and 0.6–25 mln [131]. This makes the
muscle-derived cells were shipped frozen to the results very difficult to compare and interpret.
clinic, the exact number of viable cells implanted The time of cell culture was very long, either
into the patients is unknown. Although the 5–14 weeks [129, 140] or 2–17 weeks [130],
­differences in efficacy between the groups were which suggests that the methods of cell isolation
not statistically significant, the higher doses (100 and culture were not standardized. The regenera-
and 200 mln muscle-derived cells) tended to be tion potential of cells between second and 17th
associated with a greater percentage of patients weeks can differ considerably.
with at least a 50% reduction in stress leaks and Blaganje et al. combined ultrasound-guided
pad weight at 12-month follow-up. No serious injection of autologous myoblasts into the exter-
adverse events were reported in this study [133]. nal urethral sphincter followed by electrical stim-
ulation of the sphincter in 38 women with stress
Myoblast Trial urinary incontinence (those with severe hypermo-
Another dose-ranging study utilizing myoblasts bility were excluded) [140]. The rationale for per-
for treatment of stress urinary incontinence was forming the electrical stimulation was a previous
performed by Sebe et al. [127]. Twelve female report that electrical stimulation accelerated the
patients randomly divided into three dose-related maturation and organization of myoblasts into
groups (n = 4 each) underwent myoblast implan- myotubes. After 6 weeks five patients were con-
tation into the urethral sphincter. Five of 12 sidered cured and 29 (78.4%) reported improve-
patients improved (>50% of improvement) after ment. The short observation time and the lack of
12 month follow-up, but only three patients were control group reduce the impact of these results.
totally continent. Interestingly, there was no cor-
relation either with cell dosage (10, 25, 50 mln) Minced Skeletal Muscle Tissue (MSMT)
or urinary incontinence severity (serve, moder- Freshly harvested, minced autologous skeletal
ate, and mild) [127]. These results appear to con- muscle tissue with its inherent content of regen-
tradict the studies by Carr et al. [132, 133]. erative cells were injected intraurethrally in 20
262 E.S. Yoo and J.N. Lee

and 15 women with uncomplicated and compli- larger group of patients are necessary to evaluate
cated stress urinary incontinence, respectively the efficacy of this method of treatment [134].
[141]. They were followed for 1 year. Significant In addition, Kuismanen et al. [142] injected
reductions were observed in each group in the five women with autologous adipose-derived
mean number of leakages (p < 0.01). In the stem cells combined with bovine collagen gel
uncomplicated group, cure or improvement was and saline. Patients were evaluated, 3, 6, and
observed in 25% and 63% of patients and in the 12 months after the injections. The primary end-
complicated group 7% and 57%. No voiding dys- point was a cough test. At 6 months one patient
function developed, and only minor adverse was continent and two more at 12 months. No
events were noted. adverse effects of treatment were reported. This
pilot study is limited by the low number of
10.2.3.4 A  dipose-Derived Stem Cell patients, but may support that the treatment was
Trial safe and possibly that there is a therapeutic poten-
Another source of cells used for stress urinary tial for cells other than muscle precursor cells.
incontinence treatment is adipose tissue.
Yamamoto et al. reported in 2010 two initial 10.2.3.5 U  mbilical Cord Blood
cases of adipose-derived stem cell implantation Mononuclear Cell Trial
for treatment of stress urinary incontinence fol- In another trial, allogenic umbilical cord blood
lowing radical prostatectomy [134]. This paper mononuclear cells were injected into the urethral
was initially retracted from the published journal submucosa of 39 women with stress urinary
due to ethical irregularities, but eventually pub- incontinence. Lee et al. [136] used umbilical cord
lished 2 years later in the same journal with minor stem cells in 39 women with stress urinary incon-
changes [134]. Freshly isolated adipose-derived tinence. The injections were performed by one
stem cells from the abdominal adipose tissue operator at a single hospital. Twenty-two of the
were implanted immediately without in vitro patients had urethral hypermobility, eight had
expansion into the urethral rhabdosphincter. intrinsic sphincter deficiency, and nine had mixed
Additionally, probably 2.2–3.8 mln of adipose-­ stress urinary incontinence. After 12 months,
derived stem cells in 4 mL of vehicle were com- there was a more than 50% subjective improve-
bined with intact adipose tissue and subsequently ment in 72% of the patients evaluated by a patient
injected into the urethral submucosa. Interestingly, satisfaction test. The investigators noted a pro-
urinary incontinence improved significantly a gressive improvement over time with more than
few days after injection, deteriorated subse- 90% improvement at 12 months. No adverse
quently, and progressively improved up to effects were reported. The study supports that the
6 months. This could indicate a loss of adipose approach used may be safe. However, the evalua-
tissue mass in the initial period and regeneration tion of efficacy data is not convincing, and the
of sphincter muscle by implanted adipose-derived results did not allow an assessment of efficacy
stem cells at a later stage. However, MRI showed differences between the different types of stress
no change in the volume of injected adipose tis- urinary incontinence.
sue between 4 days and 12 weeks following
implantation, proving, in fact, that the bulking 10.2.3.6 T  otal Nucleated Cells
effect was the dominant factor responsible for with Platelet-Rich Plasma
success. Also, the rapid improvement is a charac- Trial
teristic for bulking therapy. The regeneration of In a recently published trial, autologous total
the sphincter complex induced by implanted cells nucleated cells with platelet-rich plasma were
needs time to occur. The acute phase of inflam- used in the treatment of nine women with stress
mation and tissue remodeling lasts at least 4 urinary incontinence [137]. Total nucleated cells
weeks, and during this time the regeneration pro- isolated from peripheral blood were implanted
cess continues. Both long-term follow-up and a into the rhabdosphincter, while platelets were
10  Urethral Sphincter: Stress Urinary Incontinence 263

implanted into the submucosa. There was a sig- deemed to have only a minor role in continence
nificant improvement at 6 month follow-up, with control [8]. In some clinical trials evaluating
eight patients attaining full continence. However, cell-based therapy in stress urinary inconti-
it is not known which of the agents, total nucle- nence, the cells were implanted into the ure-
ated cells or platelet-rich plasma, were responsi- thral rhabdosphincter to treat incontinence
ble for the improvement. Future research is caused by urethral hypermobility [123, 136,
needed to distinguish their effects. Moreover, 137]. In order to evaluate the effectiveness of
larger trial and long-term follow-up are needed to cell implantation, those patients with combined
confirm the safety and efficacy of this method of stress urinary incontinence (urethral hypermo-
treatment. bility and ISD) should not be included in the
study groups. Urethral hypermobility as a cause
10.2.3.7 A
 nalysis of Clinical of stress urinary incontinence is beyond the
Outcomes and Adverse scope of treatment with cell injection within the
Events of Cell-Based Therapy sphincter complex. At least some cell stress uri-
in Urinary Incontinence nary incontinence operations should be focused
on the reconstruction of the structures, which
Summary and Limitation of Clinical Trials support the bladder neck and proximal urethra.
In review of clinical trials, cell-based therapy, as The treatment of stress urinary incontinence
practiced today, is a safe but ineffective method should be based on targeted therapy. The
for stress urinary incontinence treatment. The patients with stress urinary incontinence type
present article provides an indication of new II, caused by urethral hypermobility, should be
management for stress urinary incontinence treated by implantation of tissue-engineered
patients, accurate diagnosis of specific anatomi- pubourethral ligament. The treatment of stress
cal defects, and application of targeted therapy: urinary incontinence type III caused by urethral
cell implantation to restore the smooth muscle sphincter insufficiency or iatrogenic sphincter
and nerve cells and tissue engineering to restore injury should focus on cell implantation into
the collagen, the key structural component of the the urethral sphincter. The mixed etiology stress
pubourethral ligament. There are other issues, urinary incontinence (types II and III), how-
such as identifying the most effective sources of ever, should be treated using both methods of
cells; optimizing the methods of their isolation, ligament and sphincter regeneration.
culture, and implantation; and, finally, random-
ized blinded controlled trials with long-term fol- Limitations of (Pre-) Clinical Trials
low-­up comparing any new cell therapy with a What has delayed further development of cell
bulking agent and/or a midurethral sling. This treatment of stress urinary incontinence?
technology has some way to go before it becomes Undoubtedly, the withdrawal of several initial
a viable clinical option. studies for various reasons has created a doubtful
attitude to the approach among clinicians,
Discussion/Approach of Clinical Trials researchers, and the authorities. Importantly, the
Cell-based therapy as practiced today is safe published reports, although preliminary, have
but only moderately effective, comparable to shown varying effects, and there is no apparent
conventional urethral bulking. A fundamental consensus on how studies should be designed,
question needs to be addressed before any cell what cells should be injected, and how effects
therapy is performed: what is the etiology of should be evaluated. Furthermore, very few pre-
stress urinary incontinence? The huge success clinical studies simulate the patient condition
of the midurethral sling operation in all types of which is chronic and not an acute injury.
stress urinary incontinence indicates that it is Nonetheless, there seems to be a general agree-
the pubourethral ligament, which should be the ment that to accelerate clinical progress more pre-
focus of attention, not the rhabdosphincter, dictive preclinical research should be performed.
264 E.S. Yoo and J.N. Lee

This, in turn, focuses interest on what predictive human condition. However, preclinical studies
animal models and cell types/fractions should be have identified an exciting new approach to the
used. regeneration of the urinary sphincter by using the
components of cells (secretomes) or chemokines
Limitations of Mesenchymal Stem Cells that home reparative cells to the sites of injury.
As mentioned, a number of different cell types
have been used in the treatment of stress urinary
incontinence both clinically and experimentally 10.3.2 Future Perspectives
[24–26, 143]. Mesenchymal stem cells which can
be isolated from many different tissues are Preclinical research in animal models of intrinsic
defined by their ability to indefinitely self-renew, sphincter deficiency/stress urinary incontinence
differentiate into multiple cell lineages, and form and studies in humans have shown the feasibility
clonal cell populations [72, 144–146]. The appli- of cellular therapy as a treatment alternative.
cation of mesenchymal stem cells as part of a However, in view of the promising, but rather,
therapeutic strategy for functional regeneration preliminary clinical results, it seems reasonable
of the urinary sphincter complex opens two ways that before starting new randomized controlled
of action, by application of mesenchymal stem trials based on existing information, novel
cells capable to differentiate and functionally approaches should be evaluated in optimized pre-
replace defect striated and smooth muscular cells clinical models, using well-defined cells or secre-
of the urethral sphincter complex and by taking tomes. Such endeavors may aid in the
advantage of the growth factors released by the development of clinically applicable cellular
mesenchymal stem cells injected in or deposited therapy with satisfactory long-term outcome.
around the urethral wall. However, based on the
current knowledge, a differentiation of adult 10.3.2.1 O  ptimization of In Vivo
mesenchymal stem cells to yield functional Animal Models
­striated or smooth muscle cells is very unlikely to Most of the animal experiments for the evalua-
occur at reasonable efficacy. tion of cell therapy have been performed in
murine models with a more destructive or perma-
nent form of stress urinary incontinence.
10.3 Conclusion However, other groups have used reversible
incontinence models. One of the best predictors
10.3.1 Summary of later development of stress urinary inconti-
nence is postpartum incontinence, whether it
Although cell therapy will continue to be resolves or not. Therefore, investigating therapies
explored, a regenerative pharmacologic approach for postpartum incontinence should have rele-
to treatment of stress urinary incontinence holds vance to the development of later incontinence as
the promise of bypassing the lengthy and expen- well. The mechanism of human stress urinary
sive process of cell isolation and increase avail- incontinence is a complex and usually multifac-
ability of treatment in many clinical settings. torial process. Moreover, the pathological
However, this approach will require careful pre- changes associated with these mechanisms are
clinical/clinical modeling and attention to its variable and complex, combining denervation,
health benefit/risk ratio. The clinical trials have muscle degeneration and apoptosis, chronic mus-
included small numbers of patients and results cle atrophy, fibrosis, and connective tissue disor-
vary depending on the patient cohorts and the ders, among others. Hence, integrating all
cells used. Results of preclinical studies also conditions of stress urinary incontinence into a
vary, but report a more favorable outcome. This single model is very difficult, if not impossible.
difference is most likely explained by animal Therefore, the specific animal models should
modeling not being directly translatable to the take into account the potential patient to be
10  Urethral Sphincter: Stress Urinary Incontinence 265

treated and the target tissue to be regenerated. Sloff et al. [151] performed a systematic review
Besides that, more efforts must be done in order of preclinical results on the use of stem cells in
to be able to perform long-term functional, clini- the tissue engineering of the urinary bladder and
cal, urodynamic, and electromyogram assess- concluded that the translational predictability of
ments in such animal models to demonstrate cell models, particularly those based on small ani-
survival, accurate homing of the cells, viability, mals, might be questioned. This may be the case
and cell regeneration capacities by use of more also for cellular therapy for stress urinary incon-
specific cell lineage markers. A special effort tinence. It seems reasonable to suggest that new
must be done to incorporate human cell therapy approaches should be evaluated in large animal
studies in animal models as previous steps to models of stress urinary incontinence before
clinical trials. Key phenomena such as neurogen- being applied clinically. As expected, rodent
esis and angiogenesis should be evaluated in the models do not always recapitulate the character-
animal models, as well as proper muscular istics of stress urinary incontinence seen in
regeneration. patients.
A useful large animal model that replicates the
Outline of Animal Models for Cell Therapy irreversible loss of external sphincter function
Many animal models of stress urinary inconti- found in humans should fulfill several criteria.
nence have been described and discussed in detail However, the disadvantages of large animal mod-
[23, 147–150]. Most of the studies, investigating els are the rate of animal growth during the study
the effects of cell therapy in these models, have and the specialized facility and husbandry
been performed in small animals such as rodents required. An important issue and a limiting factor
and rabbits. However, large animals such as pigs, are the maintenance costs of larger animals com-
dogs, and nonhuman primates have also been pared to rodent models. However, it is important
used. The applicability of the different models is to have both small and large animal models avail-
dependent on what is to be studied: effects of the able since regulatory requirements that apply to
intervention on acute injury, effects on recovery/ novel therapies usually involve completion of a
regeneration, or pathophysiological mechanisms. full nonclinical development program.
The durability of the model is of major impor-
tance from a translational point of view. In an Canine Model
acute situation such as traumatic childbirth, cel- Eberli et al. [99] described a canine model of irre-
lular therapy may be expected to have a prophy- versible (12 month duration) urethral sphincter
lactic effect, but can hardly be considered an insufficiency. Subgroups of the dogs were sub-
option clinically. To mimic a clinical situation, jected standard urodynamic and radiographic
the model should be in a stable chronic state studies before and at 1, 2, 3, 4, and 7 months after
before intervention, i.e., a condition where spon- surgery. The urodynamic studies, which included
taneous recovery is not expected. If the interven- a static urethral pressure profile, stress urethral
tion shows improvement under these conditions, profile, and detrusor leak point pressure, showed
the results may have a clinical impact. a significant and sustained decrease in sphincter
function, indicating that the model was a reliable
Various Animal Models and Limitation and reproducible model. Using this model, Eberli
of Small or Large Animal Model et al. [152] injected autologous skeletal muscle
A number of animal models of urethral dysfunc- precursor cells into the damaged sphincter mus-
tion have emerged, including vaginal distension, cles of 12 animals. The animals were followed up
pudendal nerve crush, urethrolysis, periurethral for up to 6 months after injection, and urody-
cauterization, urethral sphincterotomy, pudendal namic studies, functional organ bath studies, and
nerve transection, and toxins. In particular, the ultrastructural and histological examinations
shortcomings and reliability issues with measur- were performed. Animals receiving cell injec-
ing techniques should be emphasized [147, 150]. tions had sphincter pressures of approximately
266 E.S. Yoo and J.N. Lee

80% of normal values, while the pressures in the heart disease, osteoporosis, breast/uterine cancer,
control animals without cells dropped and and cognitive decline [156], and also share a
remained at 20% of normal values. Histological well-defined menarche, premenopause (charac-
analysis indicated that the implanted cells sur- terized by a 28 day menstrual cycle), perimeno-
vived and formed tissue, including new inner- pause, and postmenopause [155]. The upright
vated muscle fibers, within the injected region of posture and pelvic location of the bladder and
the sphincter. These results would indicate that urethra, ultrastructure of the sphincter complex,
muscle precursor cells may be able to restore oth- and pelvic floor support are similar to those of
erwise irreversibly damaged urinary sphincter women [156].
function clinically. Further study revealed that Badra et al. [103] developed and characterized
injection of cells resulted in no significant locala nonhuman primate model of intrinsic sphincter
or systemic pathologic features within the dose deficiency. Injury to the sphincter complex was
range that improved sphincter pressures [153]. created by cauterizing and then transecting its
pudendal innervation. Urodynamic studies were
Pig Model performed before and during pudendal and hypo-
Burdzinska et al. [83] developed a model of gastric nerve stimulation at baseline and 3, 6, and
intrinsic sphincter deficiency in female pigs by 12 months after injury. Sphincter function was
balloon distension of the urethra. The urethral studied in vivo by cystourethrography and ex vivo
pressure profile was evaluated before injury, by quantitative histology and immunohistochem-
immediately post-injury, and at day 28 post-­ istry at these time points. The inflicted injury pro-
injury in the experimental group. The maximal duced a 47–50% decrease in maximal urethral
urethral closure pressure, the functional urethral pressure versus baseline [103]. It also abolished
length, and the area under curve of the urethral the increase in maximal urethral pressure in
pressure profile were measured. At 28 days post-­ response to pudendal and hypogastric nerve stim-
urethral distension, there was a decrease in maxi- ulation, and this effect persisted for more than
mal urethral closure pressure (50%) and 12 months after injury. Urodynamic changes
functional urethral length (52%). As pointed out were consistent with decreased skeletal and
by the authors, an advantage of this animal model smooth muscle content [103], decreased nerve
is that the urinary sphincter of pigs and humans responses, and an associated decrease in somatic
consists of a high percentage of slow-twitch and adrenergic innervation in the sphincter
myofibers and urodynamic evaluations can be complex.
carried out using the same procedure as used for Using this model, Badra et al. [26] assessed
human patients, whereas this is not the case in the long-term efficacy of autologous muscle pre-
rodents or dogs. Unfortunately, after injection of cursor cell therapy. Maximal urethral pressure
autologous muscle-derived cells into the urethral measurement and corresponding histological
sphincter using a urethrocystoscope, urethral analysis of the structural and cellular compo-
pressure profilometry demonstrated no signifi- nents of the sphincter complex were performed
cant differences between urethral closure pres- up to 12 months after injection. It was found that
sure in the transplanted group and the control cell treatment produced sustained (12 months)
group at day 28 [154]. increases in resting somatic nerve-stimulated
and adrenergic nerve-stimulated maximal ure-
Monkey Model thral pressure. Treated group showed a greater
Despite some differences in the encircling stri- percentage of sphincter area occupied by the
ated muscle in the urethra [155], the outflow muscle and a decreased collagen content com-
region of nonhuman primates is the closest to the pared to the untreated group. A translational
human anatomy. Female cynomolgus monkeys weakness of this study was that the cells were
share with women comorbidities common to age- injected only 6 weeks after muscle biopsy, which
and hormone-related health problems, including makes the approach more prophylactic than
10  Urethral Sphincter: Stress Urinary Incontinence 267

therapeutic, since a chronic, irreversible sphinc- continue to be developed, attention will need to
ter injury may not have been achieved. As be given to these different patient cohorts and
pointed out previously, the animal models com- therapies possibly tailored to their needs.
monly used to simulate stress urinary inconti-
nence in women are acute in nature and not 10.3.2.4 A  re There Alternatives
durable and tend to recover spontaneously in a to Cells?
short period, while stress urinary incontinence in The broad plasticity of mesenchymal stem cells
women most often is a chronic symptom, and was originally believed to define their therapeutic
spontaneous recovery is not expected. Ongoing potential. However, the remarkable array of bio-
studies in monkeys are designed to reveal active molecules produced by stem cells, the
whether or not this is the case. “secretome,” comprises a host of diverse cyto-
kines, chemokines, angiogenic factors, and
10.3.2.2 T  he Adequate Cell Type growth factors that may mediate the diverse
Needed for Regeneration effects of the mesenchymal stem cells [26, 57,
of Tissue 157, 158]. The autocrine/paracrine role of these
What is the best cell type to use for sphincter molecules is being increasingly recognized as
regeneration? Throughout the years different cell key to the regulation of many physiological pro-
types have been studied and tested in animal cesses including directing endogenous and pro-
models, and in most cases, with successful mea- genitor cells to sites of injury as well as mediating
surable results. If so, why have we failed to stan- antiapoptosis, angiogenesis and revasculariza-
dardize their potential use? Maybe not all cell tion, immune and inflammatory modulation, and
types have the same ability to successfully regen- wound healing and tissue repair [26]. In a rat
erate a particular target or tissue. Perhaps we model of simulated childbirth injury (pudendal
need to customize the cell therapy approach to crush + vaginal distension ), Deng et al. [159]
the target. found that mesenchymal stem cells and concen-
trated conditioned media from cultured rat bone-­
10.3.2.3 S  ubtyping the Stress Urinary marrow-­ derived mesenchymal stem cells
Incontinence Population provided similar protective effects on nerves and
The stress urinary continence mechanism is mul- other structures. This suggested that mesenchy-
tifactorial and complex, and stress urinary incon- mal stem cells act via their secretions to produce
tinence can result from multiple etiologies and their effects. Thus, the cell and secretome
pathophysiological processes. Therefore we need approaches to therapy can be said to represent
actions aimed and focused at understanding the “regenerative pharmacology” defined as “the
etiological and pathophysiological mechanisms application of pharmacological sciences to accel-
of specific types of stress urinary incontinence to erate, optimize, and characterize (either in vitro
improve future results aimed at therapy because or in vivo) the development, maturation, and
not all patients can be expected to respond function of bioengineered and regenerating tis-
equally to cellular therapy. This could be an sues” [160].
underlying cause of the variation in clinical Chemokines and receptors expressed during
results versus results seen in preclinical studies. tissue injury have been suggested to be upregu-
Personalized treatment is advocated in other uro- lated by vaginal distension, and simulated birth
logic conditions, and characterization of the sta- trauma induced urinary incontinence [57, 157].
tus of the sphincter and clinical investigations One of these chemokines is CXCL12, which
focused on understanding the pathophysiologi- plays a major role in cell trafficking and homing
cal mechanisms of stress urinary incontinence of progenitor cells to sites of injury through a
on an individualized basis could be expected to receptor mechanism and enhancing cell survival
improve future results. As regenerative medicine once at the injury site [158, 159, 161]. Cells
approaches to treatment of pelvic floor disorders from the injured organ highly express CXCL12,
268 E.S. Yoo and J.N. Lee

which causes an increase of local CXCL12 lev- 15. Koski ME, Enemchukwu EA, Padmanabhan
els, and peripheral and bone marrow progenitor P, Kaufman MR, Scarpero HM, Dmochowski
RR. Safety and efficacy of sling for persistent
cells follow the chemical gradient to the organ stress urinary incontinence after bulking injection.
[159, 161]. Urology. 2011;77(5):1076–80.
16. Nilsson CG, Palva K, Aarnio R, Morcos E, Falconer
C. Seventeen years’ follow-up of the tension-free
vaginal tape procedure for female stress urinary
References incontinence. Int Urogynecol J. 2013;24(8):1265–9.
17. Phe V, Benadiba S, Roupret M, Granger B, Richard
1. Abrams P, Cardozo L, Fall M, Griffiths D, Rosier F, Chartier-Kastler E. Long-term functional out-
P, Ulmsten U, et al. The standardisation of ter- comes after artificial urinary sphincter implantation
minology of lower urinary tract function: report in women with stress urinary incontinence. BJU Int.
from the Standardisation Sub-committee of the 2014;113(6):961–7.
International Continence Society. Neurourol 18. Stanford EJ, Paraiso MF. A comprehensive review of
Urodyn. 2002;21(2):167–78. suburethral sling procedure complications. J Minim
2. Kalejaiye O, Vij M, Drake MJ. Classification Invasive Gynecol. 2008;15(2):132–45.
of stress urinary incontinence. World J Urol. 19. Osborn DJ, Dmochowski RR, Harris CJ, Danford
2015;33(9):1215–20. JJ, Kaufman MR, Mock S, et al. Analysis of patient
3. Norton P, Brubaker L. Urinary incontinence in and technical factors associated with midurethral
women. Lancet. 2006;367(9504):57–67. sling mesh exposure and perforation. Int J Urol.
4. Hunskaar S, Lose G, Sykes D, Voss S. The preva- 2014;21(11):1167–70.
lence of urinary incontinence in women in four 20. Mohr S, Siegenthaler M, Mueller MD, Kuhn
European countries. BJU Int. 2004;93(3):324–30. A. Bulking agents: an analysis of 500 cases and review
5. Carlson KV, Nitti VW. Prevention and management of the literature. Int Urogynecol J. 2013;24(2):241–7.
of incontinence following radical prostatectomy. 21. Martin GR. Isolation of a pluripotent cell line from
Urol Clin North Am. 2001;28(3):595–612. early mouse embryos cultured in medium condi-
6. Strohbehn K, Lauria MR. Risk of urinary inconti- tioned by teratocarcinoma stem cells. Proc Natl
nence after childbirth: a 10-year prospective cohort Acad Sci U S A. 1981;78(12):7634–8.
study. Obstet Gynecol. 2007;109(1):202; author 22. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz
reply-3. MA, Swiergiel JJ, Marshall VS, et al. Embryonic
7. Cundiff GW. The pathophysiology of stress urinary stem cell lines derived from human blastocysts.
incontinence: a historical perspective. Rev Urol. Science. 1998;282(5391):1145–7.
2004;6(Suppl 3):S10–8. 23. Lin CS, Lue TF. Stem cell therapy for stress uri-
8. Petros PEP, Ulmsten U. Urethral pressure increase nary incontinence: a critical review. Stem Cells Dev.
on effort originates from within the urethra, and 2012;21(6):834–43.
continence from musculovaginal closure. Neurourol 24. Hart ML, Izeta A, Herrera-Imbroda B, Amend B,
Urodyn. 1995;14(4):337–50. Brinchmann JE. Cell therapy for stress urinary
9. Kayigil O, Ahmed SI, Metin A. The coexistence incontinence. Tissue Eng B Rev. 2015;21(4):365–76.
of intrinsic sphincter deficiency with type II stress 25. Tran C, Damaser MS. The potential role of stem
incontinence. J Urol. 1999;162(4):1365–6. cells in the treatment of urinary incontinence. Ther
10. Radomski SB. Practical evaluation of post-­ Adv Urol. 2015;7(1):22–40.
prostatectomy incontinence. Cuaj-Can Urol Assoc. 26. Badra S, Andersson KE, Dean A, Mourad S,
2013;7(9-10):S186–S8. Williams JK. Long-term structural and functional
11. Dalkin BL, Wessells H, Cui HY. A national survey effects of autologous muscle precursor cell therapy
of urinary and health related quality of life out- in a nonhuman primate model of urinary sphincter
comes in men with an artificial urinary sphincter deficiency. J Urol. 2013;190(5):1938–45.
for post-radical prostatectomy incontinence. J Urol. 27. Klauser A, Frauscher F, Strasser H, Helweg G, Kolle
2003;169(1):237–9. D, Strohmeyer D, et al. Age-related rhabdosphinc-
12. Harding CK, Thorpe AC. The surgical treatment of ter function in female urinary stress incontinence:
female stress urinary incontinence. Indian J Urol. assessment of intraurethral sonography. J Ultrasound
2010;26(2):257–62. Med. 2004;23(5):631–7; quiz 8-9.
13. Mischinger J, Amend B, Reisenauer C, Bedke J, 28. Goldman HB, Sievert KD, Damaser MS. Will we
Naumann G, Germann M, et al. Different surgi- ever use stem cells for the treatment of SUI?: ICI-RS
cal approaches for stress urinary incontinence in 2011. Neurourol Urodyn. 2012;31(3):386–9.
women. Minerva Ginecol. 2013;65(1):21–8. 29. Vaegler M, Lenis AT, Daum L, Amend B, Stenzl
14. Hakim L, De Ridder D, Van der Aa F. Slings for uri- A, Toomey P, et al. Stem cell therapy for void-
nary incontinence and the application of cell-based ing and erectile dysfunction. Nat Rev Urol.
therapy. Adv Drug Deliv Rev. 2015;82–83:22–30. 2012;9(8):435–47.
10  Urethral Sphincter: Stress Urinary Incontinence 269

30. Kim JH, Lee SR, Song YS, Lee HJ. Stem cell therapy 45. Bharadwaj S, Liu GH, Shi YG, Markert C,
in bladder dysfunction: where are we? And where do Andersson KE, Atala A, et al. Characterization of
we have to go? BioMed Res Int. 2013;2013:1–10. urine-derived stem cells obtained from upper urinary
31. Godfrey KJ, Mathew B, Bulman JC, Shah O, tract for use in cell-based urological tissue engineer-
Clement S, Gallicano GI. Stem cell-based treatments ing. Tissue Eng A. 2011;17(15-16):2123–32.
for type 1 diabetes mellitus: bone marrow, embry- 46. Lang R, Liu GH, Shi YG, Bharadwaj S, Leng XY,
onic, hepatic, pancreatic and induced pluripotent Zhou XB, et al. Self-renewal and differentiation
stem cells. Diabet Med. 2012;29(1):14–23. capacity of urine-derived stem cells after urine pres-
32. Singla DK, Abdelli LS. Embryonic stem cells ervation for 24 hours. Plos One. 2013;8(1):e53980.
and released factors stimulate c-kit(+)/FLK-1(+) 47. Liu GH, Pareta RA, Wu RP, Shi YG, Zhou XB, Liu
progenitor cells and promote neovasculariza- H, et al. Skeletal myogenic differentiation of urine-­
tion in doxorubicin-­induced cardiomyopathy. Cell derived stem cells and angiogenesis using micro-
Transplant. 2015;24(6):1043–52. beads loaded with growth factors. Biomaterials.
33. Anker PS, Scherjon SA, Kleijburg-van der Keur C, 2013;34(4):1311–26.
Noort WA, Claas FH, Willemze R, et al. Amniotic fluid 48. Zhang Y, Atala A. Urothelial cell culture. Methods
as a novel source of mesenchymal stem cells for ther- Mol Biol. 2013;1037:27–43.
apeutic transplantation. Blood. 2003;102(4):1548–9. 49. Bharadwaj S, Liu G, Shi Y, Wu R, Yang B, He T,
34. Moorefield EC, McKee EE, Solchaga L, Orlando et al. Multipotential differentiation of human urine-­
G, Yoo JJ, Walker S, et al. Cloned, CD117 selected derived stem cells: potential for therapeutic applica-
human amniotic fluid stem cells are capable of tions in urology. Stem Cells. 2013;31(9):1840–56.
modulating the immune response. PLoS One. 50. Sohni A, Verfaillie CM. Mesenchymal stem cells
2011;6(10):e26535. migration homing and tracking. Stem Cells Int.
35. Takahashi K, Yamanaka S. Induction of plu- 2013;2013:130763.
ripotent stem cells from mouse embryonic and 51. Wynn RF, Hart CA, Corradi-Perini C, O’Neill L,
adult fibroblast cultures by defined factors. Cell. Evans CA, Wraith JE, et al. A small proportion of mes-
2006;126(4):663–76. enchymal stem cells strongly expresses functionally
36. Morris SA, Daley GQ. A blueprint for engineer- active CXCR4 receptor capable of promoting migra-
ing cell fate: current technologies to reprogram cell tion to bone marrow. Blood. 2004;104(9):2643–5.
identity. Cell Res. 2013;23(1):33–48. 52. Honczarenko M, Le Y, Swierkowski M, Ghiran
37. Wang HJ, Chuang YC, Chancellor MB. Development I, Glodek AM, Silberstein LE. Human bone mar-
of cellular therapy for the treatment of stress urinary row stromal cells express a distinct set of biologi-
incontinence. Int Urogynecol J. 2011;22(9):1075–83. cally functional chemokine receptors. Stem Cells.
38. Friedenstein AJ, Deriglasova UF, Kulagina NN, 2006;24(4):1030–41.
Panasuk AF, Rudakowa SF, Luria EA, et al. 53. Docheva D, Popov C, Mutschler W, Schieker
Precursors for fibroblasts in different populations of M. Human mesenchymal stem cells in contact with
hematopoietic cells as detected by the in vitro colony their environment: surface characteristics and the
assay method. Exp Hematol. 1974;2(2):83–92. integrin system. J Cell Mol Med. 2007;11(1):21–38.
39. Ding DC, Shyu WC, Lin SZ. Mesenchymal stem 54. Ruster B, Gottig S, Ludwig RJ, Bistrian R, Muller
cells. Cell Transplant. 2011;20(1):5–14. S, Seifried E, et al. Mesenchymal stem cells display
40. Krause DS, Theise ND, Collector MI, Henegariu coordinated rolling and adhesion behavior on endo-
O, Hwang S, Gardner R, et al. Multi-organ, multi-­ thelial cells. Blood. 2006;108(12):3938–44.
lineage engraftment by a single bone marrow-­ 55. Woo LL, Hijaz A, Kuang M, Penn MS, Damaser
derived stem cell. Cell. 2001;105(3):369–77. MS, Rackley RR. Over expression of stem cell hom-
41. Lavker RM, Sun TT, Oshima H, Barrandon Y, ing cytokines in urogenital organs following vaginal
Akiyama M, Ferraris C, et al. Hair follicle stem cells. distention. J Urol. 2007;177(4):1568–72.
J Investig Dermatol Symp Proc. 2003;8(1):28–38. 56. Wood HM, Kuang M, Woo L, Hijaz A, Butler RS,
42. Patel AN, Park E, Kuzman M, Benetti F, Silva FJ, Penn M, et al. Cytokine expression after vaginal
Allickson JG. Multipotent menstrual blood stromal distention of different durations in virgin Sprague-­
stem cells: isolation, characterization, and differen- Dawley rats. J Urol. 2008;180(2):753–9.
tiation. Cell Transplant. 2008;17(3):303–11. 57. Lenis AT, Kuang M, Woo LL, Hijaz A, Penn MS,
43. Zhang Y, McNeill E, Tian H, Soker S, Andersson Butler RS, et al. Impact of parturition on chemo-
KE, Yoo JJ, et al. Urine derived cells are a potential kine homing factor expression in the vaginal disten-
source for urological tissue reconstruction. J Urol. tion model of stress urinary incontinence. J Urol.
2008;180(5):2226–33. 2013;189(4):1588–94.
44. Wu S, Wang Z, Bharadwaj S, Hodges SJ, Atala A, 58. Cruz M, Dissaranan C, Cotleur A, Kiedrowski M,
Zhang Y. Implantation of autologous urine derived Penn M, Damaser M. Pelvic organ distribution of
stem cells expressing vascular endothelial growth mesenchymal stem cells injected intravenously after
factor for potential use in genitourinary reconstruc- simulated childbirth injury in female rats. Obstet
tion. J Urol. 2011;186(2):640–7. Gynecol Int. 2012;2012:612946.
270 E.S. Yoo and J.N. Lee

59. Dissaranan C, Cruz MA, Kiedrowski MJ, Balog 72. Stastna M, Van Eyk JE. Secreted proteins as a funda-
BM, Gill BC, Penn MS, et al. Rat mesenchymal mental source for biomarker discovery. Proteomics.
stem cell secretome promotes elastogenesis and 2012;12(4–5):722–35.
facilitates recovery from simulated childbirth injury. 73. Ryan JM, Barry FP, Murphy JM, Mahon
Cell Transplant. 2014;23(11):1395–406. BP. Mesenchymal stem cells avoid allogeneic rejec-
60. Woo LL, Tanaka ST, Anumanthan G, Pope JCT, tion. J Inflamm (Lond). 2005;2:8.
Thomas JC, Adams MC, et al. Mesenchymal stem 74. Duijvestein M, van den Brink GR, Hommes
cell recruitment and improved bladder function after DW. Stem cells as potential novel therapeutic
bladder outlet obstruction: preliminary data. J Urol. strategy for inflammatory bowel disease. J Crohns
2011;185(3):1132–8. Colitis. 2008;2(2):99–106.
61. Rombouts WJC, Ploemacher RE. Primary murine 75. Hare JM, Traverse JH, Henry TD, Dib N, Strumpf
MSC show highly efficient homing to the bone RK, Schulman SP, et al. A randomized, double-­
marrow but lose homing ability following culture. blind, placebo-controlled, dose-escalation study of
Leukemia. 2003;17(1):160–70. intravenous adult human mesenchymal stem cells
62. De Becker A, Van Hummelen P, Bakkus M, Broek (prochymal) after acute myocardial infarction. J Am
IV, De Wever J, De Waele M, et al. Migration of Coll Cardiol. 2009;54(24):2277–86.
culture-expanded human mesenchymal stem cells 76. Telukuntla KS, Suncion VY, Schulman IH, Hare
through bone marrow endothelium is regulated JM. The advancing field of cell-based therapy:
by matrix metalloproteinase-2 and tissue inhibi- insights and lessons from clinical trials. J Am Heart
tor of metalloproteinase-3. Haematol-Hematol J. Assoc. 2013;2(5):e000338.
2007;92(4):440–9. 77. Shabbir A, Zisa D, Suzuki G, Lee T. Heart failure
63. Fischer UM, Harting MT, Jimenez F, Monzon-­ therapy mediated by the trophic activities of bone
Posadas WO, Xue HS, Savitz SI, et al. Pulmonary marrow mesenchymal stem cells: a noninvasive ther-
passage is a major obstacle for intravenous stem cell apeutic regimen. Am J Physiol Heart Circ Physiol.
delivery: the pulmonary first-pass effect. Stem Cells 2009;296(6):H1888–97.
Dev. 2009;18(5):683–91. 78. Timmers L, Lim SK, Hoefer IE, Arslan F, Lai RC,
64. Ghionzoli M, Cananzi M, Zani A, Rossi CA, Leon van Oorschot AA, et al. Human mesenchymal stem
FF, Pierro A, et al. Amniotic fluid stem cell migration cell-conditioned medium improves cardiac function
after intraperitoneal injection in pup rats: implica- following myocardial infarction. Stem Cell Res.
tion for therapy. Pediatr Surg Int. 2010;26(1):79–84. 2011;6(3):206–14.
65. Fu Q, Song XF, Liao GL, Deng CL, Cui L. Myoblasts 79. Song YS, Lee HJ, Doo SH, Lee SJ, Lim I, Chang
differentiated from adipose-derived stem cells KT, et al. Mesenchymal stem cells overexpressing
to treat stress urinary incontinence. Urology. hepatocyte growth factor (HGF) inhibit collagen
2010;75(3):718–23. deposit and improve bladder function in rat model
66. Kim SO, Na HS, Kwon D, Joo SY, Kim HS, Ahn of bladder outlet obstruction. Cell Transplant.
Y. Bone-marrow-derived mesenchymal stem cell 2012;21(8):1641–50.
transplantation enhances closing pressure and leak 80. Lin AS, Carrier S, Morgan DM, Lue TF. Effect of
point pressure in a female urinary incontinence rat simulated birth trauma on the urinary continence
model. Urol Int. 2011;86(1):110–6. mechanism in the rat. Urology. 1998;52(1):143–51.
67. Lin GT, Wang GF, Banie L, Ning HX, Shindel 81. Pan HQ, Kerns JM, Lin DL, Liu S, Esparza N,
AW, Fandel TM, et al. Treatment of stress urinary Damaser MS. Increased duration of simulated
incontinence with adipose tissue-derived stem cells. childbirth injuries results in increased time to recov-
Cytotherapy. 2010;12(1):88–95. ery. Am J Physiol Regul Integr Comp Physiol.
68. Wu R, Liu G, Bharadwaj S, Zhang Y. Isolation and 2007;292(4):R1738–44.
myogenic differentiation of mesenchymal stem cells 82. Woo LL, Hijaz A, Pan HQ, Kuang M, Rackley
for urologic tissue engineering. Methods Mol Biol. RR, Damaser MS. Simulated childbirth inju-
2013;1001:65–80. ries in an inbred rat strain. Neurourol Urodyn.
69. Carvalho MM, Teixeira FG, Reis RL, Sousa 2009;28(4):356–61.
N, Salgado AJ. Mesenchymal stem cells in the 83. Burdzinska A, Crayton R, Dybowski B, Koperski
umbilical cord: phenotypic characterization, sec- L, Idziak M, Fabisiak M, et al. Urethral disten-
retome and applications in central nervous system sion as a novel method to simulate sphincter insuf-
regenerative medicine. Curr Stem Cell Res Ther. ficiency in the porcine animal model. Int J Urol.
2011;6(3):221–8. 2012;19(7):676–82.
70. Caplan AI, Dennis JE. Mesenchymal stem 84. Kerns JM, Damaser MS, Kane JM, Sakamoto
cells as trophic mediators. J Cell Biochem. K, Benson JT, Shott S, et al. Effects of pudendal
2006;98(5):1076–84. nerve injury in the female rat. Neurourol Urodyn.
71. Deans RJ, Moseley AB. Mesenchymal stem cells: 2000;19(1):53–69.
biology and potential clinical uses. Exp Hematol. 85. Pan HQ, Lin DL, Strauch C, Butler RS, Monnier
2000;28(8):875–84. VM, Daneshgari F, et al. Pudendal nerve injury
10  Urethral Sphincter: Stress Urinary Incontinence 271

reduces urethral outlet resistance in diabetic rats. 99. Eberli D, Andersson KE, Yoo JJ, Atala A. A canine
Am J Physiol Ren Physiol. 2010;299(6):F1443–50. model of irreversible urethral sphincter insuffi-
86. Castiglione F, Bergamini A, Bettiga A, Bivalacqua ciency. BJU Int. 2009;103(2):248–53.
TJ, Benigni F, Strittmatter F, et al. Perioperative 100. Kefer JC, Liu G, Daneshgari F. Pubo-urethral liga-
betamethasone treatment reduces signs of bladder ment injury causes long-term stress urinary inconti-
dysfunction in a rat model for neurapraxia in female nence in female rats: an animal model of the integral
urogenital surgery. Eur Urol. 2012;62(6):1076–85. theory. J Urol. 2009;181(1):397–400.
87. Salcedo L, Mayorga M, Damaser M, Balog B, 101. Kamo I, Torimoto K, Chancellor MB, de Groat WC,
Butler R, Penn M, et al. Mesenchymal stem cells can Yoshimura N. Urethral closure mechanisms under
improve anal pressures after anal sphincter injury. sneeze-induced stress condition in rats: a new ani-
Stem Cell Res. 2013;10(1):95–102. mal model for evaluation of stress urinary inconti-
88. Song QX, Balog BM, Kerns J, Li Lin D, Sun YH, nence. Am J Physiol Regul Integr Comp Physiol.
Damaser MS, et al. Long-term effects of simulated 2003;285(2):R356–65.
childbirth injury on function and innervation of the 102. Peng CW, Chen JJ, Chang HY, de Groat WC, Cheng
urethra. Neurourol Urodyn. 2015;34(4):381–6. CL. External urethral sphincter activity in a rat
89. Kato H, Igawa Y, Khaleque MA, Nishizawa model of pudendal nerve injury. Neurourol Urodyn.
O. Bladder dysfunction after proximal urethrolysis 2006;25(4):388–96.
in female dogs. Int J Urol. 1999;6(1):33–7. 103. Badra S, Andersson KE, Dean A, Mourad S, Williams
90. Rodriguez LV, Chen S, Jack GS, de Almeida F, JK. A nonhuman primate model of stable urinary
Lee KW, Zhang R. New objective measures to sphincter deficiency. J Urol. 2013;189(5):1967–74.
quantify stress urinary incontinence in a novel 104. Furuta A, Jankowski RJ, Pruchnic R, Egawa S,
durable animal model of intrinsic sphincter defi- Yoshimura N, Chancellor MB. Physiological effects
ciency. Am J Physiol Regul Integr Comp Physiol. of human muscle-derived stem cell implantation on
2005;288(5):R1332–R8. urethral smooth muscle function. Int Urogynecol J.
91. Skaff M, Pinto E, Leite KR, Almeida 2008;19(9):1229–34.
FG. Development of a rabbit’s urethral sphincter 105. Xu Y, Song YF, Lin ZX. Transplantation of muscle-­
deficiency animal model for anatomical-functional derived stem cells plus biodegradable fibrin glue
evaluation. Int Braz J Urol. 2012;38(1):17–24. restores the urethral sphincter in a pudendal
92. Kinebuchi Y, Aizawa N, Imamura T, Ishizuka O, nerve-transected rat model. Braz J Med Biol Res.
Igawa Y, Nishizawa O. Autologous bone-­marrow-­ 2010;43(11):1076–83.
derived mesenchymal stem cell transplantation 106. Zhao WM, Zhang C, Jin CJ, Zhang ZJ, Kong DL, Xu
into injured rat urethral sphincter. Int J Urol. WH, et al. Periurethral injection of autologous adi-
2010;17(4):359–68. pose-derived stem cells with controlled-release nerve
93. Chermansky CJ, Tarin T, Kwon DD, Jankowski growth factor for the treatment of stress urinary incon-
RJ, Cannon TW, de Groat WC, et al. Intraurethral tinence in a rat model. Eur Urol. 2011;59(1):155–63.
muscle-­derived cell injections increase leak point 107. Wu GZ, Song YF, Zheng X, Jiang ZQ. Adipose-­
pressure in a rat model of intrinsic sphincter defi- derived stromal cell transplantation for treat-
ciency. Urology. 2004;63(4):780–5. ment of stress urinary incontinence. Tissue Cell.
94. Lim JJ, Jang JB, Kim JY, Moon SH, Lee CN, Lee 2011;43(4):246–53.
KJ. Human umbilical cord blood mononuclear cell 108. Corcos J, Loutochin O, Campeau L, Eliopoulos
transplantation in rats with intrinsic sphincter defi- N, Bouchentouf M, Blok B, et al. Bone mar-
ciency. J Korean Med Sci. 2010;25(5):663–70. row mesenchymal stromal cell therapy for exter-
95. Chermansky CJ, Cannon TW, Torimoto K, Fraser nal urethral sphincter restoration in a rat model of
MO, Yoshimura N, de Groat WC, et al. A model of stress urinary incontinence. Neurourol Urodyn.
intrinsic sphincteric deficiency in the rat: electrocau- 2011;30(3):447–55.
terization. Neurourol Urodyn. 2004;23(2):166–71. 109. Du XW, Wu HL, Zhu YF, Hu JB, Jin F, Lv RP,
96. Yiou R, Yoo JJ, Atala A. Restoration of functional et al. Experimental study of therapy of bone mar-
motor units in a rat model of sphincter injury by row mesenchymal stem cells or muscle-like cells/
muscle precursor cell autografts. Transplantation. calcium alginate composite gel for the treatment
2003;76(7):1053–60. of stress urinary incontinence. Neurourol Urodyn.
97. Zutshi M, Salcedo LB, Zaszczurynski PJ, Hull 2013;32(3):281–6.
TL, Butler RS, Damaser MS. Effects of sphinc- 110. Chun SY, Kwon JB, Chae SY, Lee JK, Bae JS, Kim
terotomy and pudendal nerve transection on the BS, et al. Combined injection of three different lin-
anal sphincter in a rat model. Dis Colon Rectum. eages of early-differentiating human amniotic fluid-­
2009;52(7):1321–9. derived cells restores urethral sphincter function in
98. Praud C, Sebe P, Bierinx AS, Sebille A. Improvement urinary incontinence. BJU Int. 2014;114(5):770–83.
of urethral sphincter deficiency in female rats fol- 111. Joyce N, Annett G, Wirthlin L, Olson S, Bauer G, Nolta
lowing autologous skeletal muscle myoblasts graft- JA. Mesenchymal stem cells for the treatment of neuro-
ing. Cell Transplant. 2007;16(7):741–9. degenerative disease. Regen Med. 2010;5(6):933–46.
272 E.S. Yoo and J.N. Lee

112. Hardy SA, Maltman DJ, Przyborski incontinence: 1-year followup of 63 patients. J Urol.
SA. Mesenchymal stem cells as mediators of 2008;179(1):226–31.
neural differentiation. Curr Stem Cell Res Ther. 125. Mitterberger M, Pinggera GM, Marksteiner R,
2008;3(1):43–52. Margreiter E, Fussenegger M, Frauscher F, et al.
113. Li GY, Zhou F, Gong YQ, Cui WS, Yuan YM, Adult stem cell therapy of female stress urinary
Song WD, et al. Activation of VEGF and ERK1/2 incontinence. Eur Urol. 2008;53(1):169–75.
and improvement of urethral function by adipose-­ 126. Carr LK, Steele D, Steele S, Wagner D, Pruchnic
derived stem cells in a rat stress urinary incontinence R, Jankowski R, et al. 1-year follow-up of autolo-
model. Urology. 2012;80(4):953–e1. gous muscle-derived stem cell injection pilot study
114. Obinata D, Matsumoto T, Ikado Y, Sakuma T, to treat stress urinary incontinence. Int Urogynecol J
Kano K, Fukuda N, et al. Transplantation of mature Pelvic Floor Dysfunct. 2008;19(6):881–3.
adipocyte-­derived dedifferentiated fat (DFAT) cells 127. Sebe P, Doucet C, Cornu JN, Ciofu C, Costa P, de
improves urethral sphincter contractility in a rat Medina SG, et al. Intrasphincteric injections of
model. Int J Urol. 2011;18(12):827–34. autologous muscular cells in women with refractory
115. Ohta Y, Takenaga M, Tokura Y, Hamaguchi A, stress urinary incontinence: a prospective study. Int
Matsumoto T, Kano K, et al. Mature adipocyte-­ Urogynecol J. 2011;22(2):183–9.
derived cells, dedifferentiated fat cells (DFAT), 128. Cornu JN, Lizee D, Pinset C, Haab F. Long-term
promoted functional recovery from spinal cord follow-up after regenerative therapy of the urethral
injury-induced motor dysfunction in rats. Cell sphincter for female stress urinary incontinence. Eur
Transplant. 2008;17(8):877–86. Urol. 2014;65(1):256–8.
116. Kwon D, Kim Y, Pruchnic R, Jankowski R, Usiene 129. Blaganje M, Lukanovic A. Ultrasound-guided
I, De Miguel F, et al. Periurethral cellular injection: autologous myoblast injections into the extrinsic
comparison of muscle-derived progenitor cells and urethral sphincter: tissue engineering for the treat-
fibroblasts with regard to efficacy and tissue contrac- ment of stress urinary incontinence. Int Urogynecol
tility in an animal model of stress urinary inconti- J. 2013;24(4):533–5.
nence. Urology. 2006;68(2):449–54. 130. Gerullis H, Eimer C, Georgas E, Homburger M,
117. Mitterberger M, Pinggera GM, Marksteiner R, El-Baz AG, Wishahi M, et al. Muscle-derived
Margreiter E, Plattner R, Klima G, et al. Functional cells for treatment of iatrogenic sphincter dam-
and histological changes after myoblast injec- age and urinary incontinence in men. Sci World J.
tions in the porcine rhabdosphincter. Eur Urol. 2012;2012:1–6.
2007;52(6):1736–43. 131. Stangel-Wojcikiewicz K, Jarocha D, Piwowar M,
118. Yiou R, Dreyfus P, Chopin DK, Abbou CC, Jach R, Uhl T, Basta A, et al. Autologous muscle-­
Lefaucheur JP. Muscle precursor cell autografting in derived cells for the treatment of female stress urinary
a murine model of urethral sphincter injury. BJU Int. incontinence: a 2-year follow-up of a polish investi-
2002;89(3):298–302. gation. Neurourol Urodyn. 2014;33(3):324–30.
119. Kim YT, Kim DK, Jankowski RJ, Pruchnic R, 132. Carr LK, Robert M, Kultgen PL, Herschorn S, Birch
Usiene I, de Miguel F, et al. Human muscle-derived C, Murphy M, et al. Autologous muscle derived cell
cell injection in a rat model of stress urinary inconti- therapy for stress urinary incontinence: a prospective,
nence. Muscle Nerve. 2007;36(3):391–3. dose ranging study. J Urol. 2013;189(2):595–601.
120. Kim BS, Chun SY, Lee JK, Lim HJ, Bae JS, Chung 133. Peters KM, Dmochowski RR, Carr LK, Robert M,
HY, et al. Human amniotic fluid stem cell injection Kaufman MR, Sirls LT, et al. Autologous muscle
therapy for urethral sphincter regeneration in an ani- derived cells for treatment of stress urinary inconti-
mal model. BMC Med. 2012;10:94. nence in women. J Urol. 2014;192(2):469–76.
121. Bandyopadhyay B, Thakur A, Dave V, Viswanathan 134. Yamamoto T, Gotoh M, Kato M, Majima T,
C, Ghosh D. A non-invasive method to evaluate the Toriyama K, Kamei Y, et al. Periurethral injection of
efficacy of human myoblast in botulinum-A toxin autologous adipose-derived regenerative cells for the
induced stress urinary incontinence model in rats. treatment of male stress urinary incontinence: report
Urol J. 2013;10(4):1126–34. of three initial cases. Int J Urol. 2012;19(7):652–9.
122. Aref-Adib M, Lamb BW, Lee HB, Akinnawo E, Raza 135. Gotoh M, Yamamoto T, Kato M, Majima T,
MM, Hughes A, et al. Stem cell therapy for stress uri- Toriyama K, Kamei Y, et al. Regenerative treatment
nary incontinence: a systematic review in human sub- of male stress urinary incontinence by periurethral
jects. Arch Gynecol Obstet. 2013;288(6):1213–21. injection of autologous adipose-derived regenera-
123. Mitterberger M, Marksteiner R, Margreiter E, tive cells: 1-year outcomes in 11 patients. Int J Urol.
Pinggera GM, Colleselli D, Frauscher F, et al. 2014;21(3):294–300.
Autologous myoblasts and fibroblasts for female 136. Lee CN, Jang JB, Kim JY, Koh C, Baek JY, Lee
stress incontinence: a 1-year follow-up in 123 KJ. Human cord blood stem cell therapy for treat-
patients. BJU Int. 2007;100(5):1081–5. ment of stress urinary incontinence. J Korean Med
124. Mitterberger M, Marksteiner R, Margreiter E, Sci. 2010;25(6):813–6.
Pinggera GM, Frauscher F, Ulmer H, et al. Myoblast 137. Shirvan MK, Alamdari DH, Mahboub MD, Ghanadi
and fibroblast therapy for post-prostatectomy urinary A, Rahimi HR, Seifalian AM. A novel cell therapy
10  Urethral Sphincter: Stress Urinary Incontinence 273

for stress urinary incontinence, short-term outcome. 150. Herrera-Imbroda B, Lara MF, Izeta A, Sievert KD,
Neurourol Urodyn. 2013;32(4):377–82. Hart ML. Stress urinary incontinence animal models
138. Pokrywczynska M, Adamowicz J, Czapiewska as a tool to study cell-based regenerative therapies
M, Balcerczyk D, Jundzill A, Nowacki M, et al. targeting the urethral sphincter. Adv Drug Deliv Rev.
Targeted therapy for stress urinary incontinence: a 2015;82–83:106–16.
systematic review based on clinical trials. Expert 151. Sloff M, Simaioforidis V, de Vries R, Oosterwijk
Opin Biol Ther. 2016;16(2):233–42. E, Feitz W. Tissue engineering of the bladder –
139. Herschorn S, Radomski SB. Collagen injections for reality or myth? A systematic review. J Urol.
genuine stress urinary incontinence: patient selec- 2014;192(4):1035–42.
tion and durability. Int Urogynecol J Pelvic Floor 152. Eberli D, Aboushwareb T, Soker S, Yoo JJ, Atala
Dysfunct. 1997;8(1):18–24. A. Muscle precursor cells for the restoration of
140. Blaganje M, Lukanovic A. Intrasphincteric autolo- irreversibly damaged sphincter function. Cell
gous myoblast injections with electrical stimula- Transplant. 2012;21(9):2089–98.
tion for stress urinary incontinence. Int J Gynaecol 153. Williams JK, Eckman D, Dean A, Moradi M,
Obstet. 2012;117(2):164–7. Allickson J, Cline JM, et al. The dose-effect safety
141. Gras S, Klarskov N, Lose G. Intraurethral injec- profile of skeletal muscle precursor cell therapy in a
tion of autologous minced skeletal muscle: a simple dog model of intrinsic urinary sphincter deficiency.
surgical treatment for stress urinary incontinence. J Stem Cells Transl Med. 2015;4(3):286–94.
Urol. 2014;192(3):850–5. 154. Burdzinska A, Crayton R, Dybowski B, Idziak M,
142. Kuismanen K, Sartoneva R, Haimi S, Mannerstrom Gala K, Radziszewski P, et al. The effect of endo-
B, Tomas E, Miettinen S, et al. Autologous adipose scopic administration of autologous porcine muscle-­
stem cells in treatment of female stress urinary derived cells into the urethral sphincter. Urology.
incontinence: results of a pilot study. Stem Cells 2013;82(3):743.e1–8.
Transl Med. 2014;3(8):936–41. 155. Ganzer R, Kohler D, Neuhaus J, Dorschner W,
143. Klein G, Hart ML, Brinchmann JE, Rolauffs B, Stolzenburg JU. Is the rhesus monkey (Macaca
Stenzl A, Sievert KD, et al. Mesenchymal stromal mulatta) comparable to humans? Histomorphology
cells for sphincter regeneration. Adv Drug Deliv of the sphincteric musculature of the lower uri-
Rev. 2015;82–83:123–36. nary tract including 3D-reconstruction. Anat Histol
144. Ranganath SH, Levy O, Inamdar MS, Karp Embryol. 2004;33(6):355–61.
JM. Harnessing the mesenchymal stem cell secre- 156. Kaplan JR, Manuck SB. Ovarian dysfunction,
tome for the treatment of cardiovascular disease. stress, and disease: a primate continuum. ILAR J.
Cell Stem Cell. 2012;10(3):244–58. 2004;45(2):89–115.
145. Lavoie JR, Rosu-Myles M. Uncovering the 157. Hijaz AK, Grimberg KO, Tao M, Schmotzer B,
secretes of mesenchymal stem cells. Biochimie. Sadeghi Z, Lin YH, et al. Stem cell homing factor,
2013;95(12):2212–21. CCL7, expression in mouse models of stress urinary
146. Deng KL, Lin DL, Hanzlicek B, Balog B, Penn MS, incontinence. Female Pelvic Med Reconstr Surg.
Kiedrowski MJ, et al. Mesenchymal stem cells and 2013;19(6):356–61.
their secretome partially restore nerve and urethral 158. Lau TT, Wang DA. Stromal cell-derived fac-
function in a dual muscle and nerve injury stress tor-­
1 (SDF-1): homing factor for engineered
urinary incontinence model. Am J Physiol-Renal. regenerative medicine. Expert Opin Biol Ther.
2015;308(2):F92–F100. 2011;11(2):189–97.
147. Jiang HH, Damaser MS. Animal models of stress 159. Herberg S, Shi XM, Johnson MH, Hamrick MW,
urinary incontinence. Handb Exp Pharmacol. Isales CM, Hill WD. Stromal cell-derived factor-
2011;202:45–67. 1 beta mediates cell survival through enhancing
­
148. Hong SH, Piao S, Kim IG, Lee JY, Cho HJ, Kim autophagy in bone marrow-derived mesenchymal
SW, et al. Comparison of three types of stress uri- stem cells. Plos One. 2013;8(3):e58207.
nary incontinence rat models: electrocauteriza- 160. Christ GJ, Saul JM, Furth ME, Andersson KE. The
tion, pudendal denervation, and vaginal distension. pharmacology of regenerative medicine. Pharmacol
Urology. 2013;81(2):465.e1–6. Rev. 2013;65(3):1091–133.
149. Koike Y, Furuta A, Suzuki Y, Honda M, Naruoka T, 161. Nagasawa T. CXC chemokine ligand 12 (CXCL12)
Asano K, et al. Pathophysiology of urinary inconti- and its receptor CXCR4. J Mol Med. 2014;
nence in murine models. Int J Urol. 2013;20(1):64–71. 92(5):433–9.
Penis and Testis
11
Seok Cho and Jae Hyun Bae

11.1 Penis congenital anomaly, the patients need to estab-


lish morphological reconstruction. The second
11.1.1 Introduction one is the functional reconstruction. The
patients with erectile dysfunction which is not
Reconstruction or substitution of the corpora cav- response to medical treatment want to recover
ernosa to restore a cosmetically acceptable phal- their erectile function first. Of course, it will be
lus that would allow normal reproductive, sexual, ideal to regenerate both morphological and
and urinary function represents a very challeng- functional recovery.
ing task. Despite the development of many surgical
Conditions requiring penile reconstruction materials have been developed, current recon-
include penile malignancy, congenital anomaly, structive techniques are limited by issues of tis-
trauma, erectile dysfunction, Peyronie’s disease, sue availability and compatibility. These
gender reassignment surgery, and post penec- techniques use nonurologic host tissues (skin,
tomy status. Penile necrosis due to infection after gastrointestinal segments, or mucosa from mul-
penile surgery such as penile prosthesis insertion tiple body sites), donor tissues (cadaver fascia
is not common but not rare in diabetic patients. In or cadaver or living donor kidney), heterologous
those cases, partial or total penectomy has to be tissues (bovine collagen), or artificial materials
performed. (silicone, polyurethane, or polytetrafluoroethyl-
Penile reconstruction requires two tasks. ene) to reconstruct damaged organs. However,
The first one is the morphological reconstruc- all of these materials can lead to significant
tion. In cases of post-penectomy status or complications resulting from immunologic
rejection or functional mismatches between the
native and implanted tissues such as infection,
graft failure, and donor site morbidity have been
reported and in these cases, the functional
S. Cho (*) regeneration is impossible, therefore additional
The Department of Urology, Ilsanpaik Hospital, penile prosthesis insertion is needed and also,
Inje University College of Medicine, Goyang, additional complications can be developed. In
South Korea order to reduce or eliminate these complica-
e-mail: envystone@hanmail.net
tions, recent researchers have tried to regenerate
J.H. Bae penis to overcome the limitations of the free flap
The Department of Urology, Korea University Ansan
Hospital, Korea University College of Medicine, operation technique.
Ansan, South Korea The functional regeneration of erectile func-
e-mail: urobae@genetherapy.or.kr tion is now focusing on cell therapy such as stem

© Springer Nature Singapore Pte Ltd. 2018 275


B.W. Kim (ed.), Clinical Regenerative Medicine in Urology, DOI 10.1007/978-981-10-2723-9_11
276 S. Cho and J.H. Bae

cell injection with or without various growth role in the erectile process. In the flaccid state,
factors. these smooth muscles are tonically contracted,
In this chapter, various trials and results will allowing only a small amount of arterial flow for
be introduced. nutritional purposes. The blood partial pressure
of oxygen (pO2) is about 35 mmHg. The flaccid
penis is in a moderate state of contraction, as evi-
11.1.2 Anatomy
denced by further shrinkage in cold weather and
after phenylephrine injection.
The penis is complex and composed of three sep-
Sexual stimulation triggers release of neu-
arate cylinders: two paired cylinders called the
rotransmitters from the cavernous nerve termi-
corpora cavernosa on the dorsal side make up the
nals. This results in relaxation of these smooth
majority of the penis and are the primary structure
muscles and the following events: (1) dilation
involved in erectile function. These cylinders con-
of the arterioles and arteries by increased
sist of a network of large venous sinuses separated
blood flow in both the diastolic and systolic
by trabeculae, dense connective tissue. The cor-
phases; (2) trapping of the incoming blood by
pus spongiosum, the third cylinder is located on
the expanding sinusoids; (3) compression of
the ventral side and is the structure which wraps
the subtunical venous plexuses between the
the urethra for the protection. It has the same arte-
tunica albuginea and the peripheral sinusoids,
rial and venous relationship as the corpora caver-
reducing venous outflow; (4) stretching of the
nosa. Each of these cylinders is covered by a very
tunica to its capacity, which occludes the
tough thick sheath called the tunica albuginea. All
emissary veins between the inner circular and
three cylinders are encased in a thick membrane
outer longitudinal layers and further decreases
called as Buck’s fascia. Finally, Collie’s fascia,
venous outflow to a minimum; (5) an increase
which is continuous with the abdominal wall,
in PO2 (to about 90 mmHg) and intracavern-
covers the entire structure and makes this whole
ous pressure (around 100 mmHg), which
supporting structure of the penis very tough,
raises the penis from the dependent position to
allowing it to take quite a bit of force and trauma.
the erect state (the full-erection phase); and
The erectile tissue within the corpora contains
(6) a further pressure increase (to several hun-
arteries, nerves, muscle fibers, and venous sinuses
dred millimeters of mercury) with contraction
lined with flat endothelial cells, and it fills the
of the ischiocavernosus muscles (rigid-­
space of the corpora cavernosa. The cut surface
erection phase).
of the corpora cavernosa looks like a sponge.
The angle of the erect penis is determined
There is a thin layer of areolar tissue that sepa-
by its size and attachment to the puboischial
rates this tissue from the tunica albuginea.
rami (the crura) and the anterior surface of the
The shaft is covered by nearly hairless skin.
pubic bone (the suspensory and funiform liga-
Under the skin lies the dense connective tissue of
ments). In men with a long heavy penis or a
penile fascia. The tunica albuginea encircles all
loose suspensory ligament, the angle usually
three corpora, divides the corpora proximally, but
will not be greater than 90 degrees, even with
is incomplete distally.
full rigidity.
Three phases of detumescence have been
11.1.3 Hemodynamics reported in an animal study [1]. The first entails a
and Mechanism of Erection transient intracorporeal pressure increase, indi-
and Detumescence cating the beginning of smooth muscle contrac-
tion against a closed venous system. The second
The penile erectile tissue, specifically the cavern- phase shows a slow pressure decrease, suggest-
ous smooth musculature and the smooth muscles ing a slow reopening of the venous channels with
of the arteriolar and arterial walls, plays a key resumption of the basal level of arterial flow. The
11  Penis and Testis 277

third phase shows a fast pressure decrease with the feasibility of engineering human cartilage
fully restored venous outflow capacity. rods for potential use as penile prostheses [4].
Erection thus involves sinusoidal relaxation, Chondrocytes from the human ear were
arterial dilation, and venous compression. The seeded on rod-shaped biodegradable-polymer
importance of smooth muscle relaxation has scaffolds and implanted into athymic rats. After
been demonstrated in animal and human 2 months, chondrocytes formed flexible cartilagi-
studies. nous rods similar in size to the initial implants,
and with mature chondrocytes. The mechanical
properties of these cartilaginous rods were com-
11.1.4 Penile Reconstruction parable with those of commercially available sili-
cone prostheses. Autologous-engineered cartilage
Hu and colleagues suggested that patients with a constructed using cells derived from rabbit ear
penile defect are frequently extremely con- cartilage was used for penile reconstruction in
cerned about the extent to which their problem rabbits [5]. The autologous-cartilage cells were
might affect their social status and the fidelity of seeded on PGA and implanted into the corporal
their partners [2]. But, current therapies using space of 10 rabbits; cartilage structures were
microsurgical technology and free-flaps from formed within the corpora, with polymer degra-
autologous donor sites, combined with a penile dation within 2 months.
prosthetic implant have not adequately Beside these efforts to develop engineering
addressed various structural disorders of the penile prosthesis, there were experiments about
phallus. Penile implants are associated with culturing human corporal tissues. Corporal
complications including infection, erosion, and smooth muscle is one of the major components of
mechanical malfunction. Tissue-engineering the phallus. In the event of pathology, and during
approaches have been employed to address a surgical penile reconstruction for conditions such
number of penile disorders. Although the corpo- as ambiguous genitalia and exstrophy-epispadias
ral bodies only contain two primary cell types, (where there is a limited amount of native caver-
smooth muscle, and endothelium, engineering nosal tissue available), the creation of autologous
this tissue remains a challenge. and functional de novo corporal tissue would be
Only a few experimental studies have been beneficial.
published to date. In 1999, Park et al. investigated the possibil-
At first, investigators focused on morpho- ity of developing corporal tissue by combining
logic reconstruction of penis using rib carti- smooth muscle and endothelial cells. Normal
lage as a stiffener. However, this method was human cavernosal smooth muscle cells and
discouraged due to the unsatisfactory func- human endothelial cells were seeded on biode-
tional and cosmetic results, and then cartilage gradable polymer for implantation [6]. This is
would serve as an ideal prosthesis for penile the first demonstration in tissue engineering in
reconstruction, owing to its biomechanical which capillary formation was facilitated by the
properties. addition of endothelial cells in composite tissue
Yoo et al. demonstrated that the feasibility of in vivo. This study showed that human corporal
creating natural penile prostheses composed of muscle and endothelial cells seeded on biode-
cartilage [3]. Chondrocytes from articular surface gradable polymer scaffolds are able to form vas-
of calf shoulders were seeded onto preformed cularized cavernosal tissue when implanted
cylindrical polyglycolic acid (PGA) polymer rods in vivo.
and then implanted in athymic mice. Rod-­shaped Then initial investigation into the possible cul-
solid cartilaginous structures with mature chon- ture of human corporal SMCs combined with a
drocytes were observed in cell-seeded scaffolds. polymer to generate a bioengineered corpus cav-
A subsequent study was performed to determine ernosum has been reported [7].
278 S. Cho and J.H. Bae

Following these experiments, the same group, muscle-derived stem cells, then described an
Kwon et al. reported that autologous corpus cav- experiment in which they transduced, via a lenti-
ernosal smooth muscle and endothelial cells virus, the muscle-derived stem cells with vascu-
seeded on collagen matrices can form corpora lar endothelial growth factor (VEGF) and
cavernosa tissue structures in a rabbit model. demonstrated that after the in vivo implant, the
Engineered corpora cavernosa achieved adequate obtained construct showed increased content of
structural and functional parameters [8]. endothelial cells, smooth muscle cells, and capil-
The viability of engineered scaffolds was laries [12, 13].
proved in vivo in rabbits in which the corpus cav- Another approach to recover erectile function
ernosum was replaced with an acellular collagen by cell based therapy could be the injection of
matrix. Chen et al. reported that normal erectile functional cells into the corpus cavernosum, e.g.
function and the ability to mate after 1 month; in cases of severe endothelial dysfunction or
compared to the control animals, which were decrease of smooth muscle cells. Wessells et al.
implanted with acellular matrix itself and were suggested and experimentally investigated first
unable to reach erection [9]. Cavernosography for endothelial cell transplantation [14].
examination showed that the experimental corpo- Fluorescently labeled autologous endothelial
ral bodies had structural integrity and similar cells were injected into the rat corpus caverno-
pressure characteristics, when compared with the sum. After 2 days clusters of cells were still seen
normal controls. within the central corporal sinusoids, whereas
This work was recently taken one step further after one and 2 weeks remaining cells were only
by the same group. Chen et al. demonstrated the detected peripherally in the sinusoids beneath the
ability to bioengineer entire pendular penile tunica albuginea. Although this technique might
corporal bodies in a rabbit model [10]. be considered for direct repair of damaged corpo-
Decellularized donor corporal bodies were ral endothelium, this kind of cell based therapy
seeded with autologous smooth muscle and for erectile dysfunction, either with endothelial
endothelial cells in a stepwise fashion. Rabbits or with smooth muscle cells, only seems to be
then underwent complete excision of the corpo- promising when combined with cell manipula-
ral bodies followed by implantation of seeded tion by gene therapy prior to cell transfer
scaffolds. The group implanted with seeded [15–17].
scaffolds demonstrated normal intra-cavernosal Cavernous nerves can be easily damaged
pressures within 6 months of surgery, and 10 out during penile surgery, and simply exposing the
of 12 rabbits could successfully mate with cavernous nerve can lead to ED. Therefore,
female rabbits. methods of restoring nerve function in these
A further group seeded acellular corporal col- patients are necessary. Different regenerative
lagen matrices with SMCs derived from human strategies used until now for repair or replace-
umbilical arteries and implanted the cellularized ment of cavernous nerves include: use of sili-
implants into athymic mice. Song et al. [11] cone guidance tubes filled with primary
reported that the scaffolds became cellularized to Schwann cells, and nerve gap interposition with
develop tissue with anatomical and functional acellular nerve matrices, which showed signifi-
properties similar to corpus cavernosum smooth cant nerve regeneration and ED recoverability
muscle. [18]; use of sural nerve as a scaffold for cavern-
Another interesting approach has been pre- ous nerve regeneration after radical prostatec-
sented by Zhang et al.: they first created a tissue-­ tomy, where partial improvement of ED and
engineered corpus cavernosum seeded with continence recovery were confirmed in many
11  Penis and Testis 279

clinical studies [19–22]; and use of a nerve graft complications such as recurrence of disease and
procedure after radical prostatectomy—a large graft contracture.
clinical study in Japan indicated that this proce- Schultheiss et al. isolated porcine fibroblasts
dure might contribute to the recovery of urinary from open fascia biopsies and seeded them on
and sexual function [23]. decellularized collagen matrices [26]. The seeded
matrices were then cultivated in a bioreactor
11.1.4.1 Penile Enhancement under continuous multiaxial stress for 21 days.
Penile enhancement has been used for both ther- Mechanically strained cultures of fibroblasts
apeutic and esthetic purposes. In a study, autolo- showed a homogeneous multilayer matrix infil-
gous fibroblasts from scrotal skin were harvested tration and a regular cell alignment in the direc-
and seeded on poly acid-co-glycolide biode- tion of strain axis after 7 days as well as increased
gradable (Maxpol-T) scaffolds [24]. This con- production of extracellular matrix proteins com-
struct was implanted under Buck’s fascia of the pared to the static control.
penile shaft of 69 patients. Mean penile girth Similarly, Joo et al. performed using mucosal
was measured preoperatively and 6 months graft from pigs’ bladder [27]. A segment of the
postoperatively in the flaccid and erect states; tunica albuginea of nine rabbits was excised, and
the mean penile girth had increased significantly the defect was covered with porcine bladder acel-
after 6 months, and 94.2% of patients were sat- lular matrix. Two months after implantation, the
isfied with the procedure. In another study, 12 graft sites exhibited excellent healing without
patients underwent repeated penile widening contracture, and the fusion between the graft and
using biodegradable scaffolds enriched with the neighboring normal TA appeared to be well
expanded autologous scrotal dartos cells [25]; established. There were no significant histologi-
penile girth increased in both flaccid and erect cal differences between the implanted tunica and
states. Furthermore, 10–14 months after first the normal control tunica at 6 months after
surgery, biopsies showed highly vascularized implantation.
loose tissue with collagen deposition and some Recently, Imbeault et al. used human fibro-
inflammation, and after 22–24 months, biopsies blasts to form a fibroblast sheet—human umbil-
revealed almost no inflammation and a tissue ical vein endothelial cells (HUVECs) were
resembling normal dartos fascia. All patients seeded on fibroblasts sheets and wrapped around
were satisfied with the surgery. a tubular support, and HUVECs were seeded
into the lumen of the fibroblast sheets [25]. This
11.1.4.2 Tunica Albuginea endothelialized tubular graft was structurally
Reconstruction similar to normal tunica albuginea with ade-
Peyronie’s disease is a benign, acquired wound-­ quate mechanical resistance. In an attempt to
healing-­connective-tissue disorder of the tunica engineer tunica albuginea, autologous fibro-
albuginea that affects 3–9% of men [25]. Patients blasts were seeded onto a PGA scaffold and
with Peyronie’s disease can experience signifi- sutured into the same-­sized defect in rat cor-
cant pain and functional problems due to plaque pora. The seeded graft had features similar to
formation and curvature of the penis. Therapeutic the sham group, in terms of erectile response to
options for Peyronie’s disease include plication, cavernous nerve stimulation and retraction of
incision and grafting of the tunica albuginea with graft 4 months after surgery, while the unseeded-
materials such as small intestinal submucosa, fas- graft control group had poor outcomes for these
cia, and pericardium, and penile prosthesis. factors. Studies on the reconstruction of the
Graft-based therapies are associated with some penile tissue are shown in Table 11.1.
280 S. Cho and J.H. Bae

Table 11.1  List of study about penile tissue for which regenerative medicine therapies are being reported
Tissue Material Target Authors Years
Penile prosthesis
Cartilage Chondrocytes Athymic mice Yoo JJ et al. 1998
Cartilage Autologous cartilage cells Rabbit Yoo JJ et al. 1999
Cartilage Chondrocytes from Athymic rat Kim BS et al. 2002
human ear
Corporal tissues
Smooth muscle and Human cavernosal Athymic mice Park HJ et al. 1999
endothelial cells smooth muscle cells and
endothelial cells
Smooth muscle Human corporal smooth Athymic mice Kershen RT et al. 2002
cells
Smooth muscle and Rabbit cavernosal smooth Rabbit Kwon TG et al. 2002
endothelial cells muscle and endothelial
cells
Corpus Cavernosum Autologous cavernosal Rabbit Yoo JJ et al. 2006
tissue on cellular collagen
matrix
Smooth muscle cells Human umbilical artery Athymic mice Song LJ et al. 2009
smooth muscle cells
Penile corporal body Autologous cavernosal Rabbit Chen KL et al. 2010
tissue
Corpus cavernosum Muscle derived stem cells Rabbit Ji C et al. 2011
Corpus cavernosum Muscle derived stem cells Rabbit An G et al. 2013
with human vascular
endothelial growth factor
Cavernous nerve
Cavernous nerve Acellular nerve graft Rat Connolly SS et al. 2008
Penile enhancement
Fibroblast Human Djinovic R et al. 2011
Scrotal dartos cell Human Imbeault A et al. 2011
Tunica albuginea
Tunica albuginea Porcine fibroblast In vitro Shultheiss D et al. 2004
Tunica albuginea Porcine bladder acellular Rabbit Joo KW et al. 2006
matrix
Tunica albuginea Human fibroblast In vitro Imbeault A et al. 2011

11.2 Testis testes can be congenitally absent in conditions


such as unresolved cryptorchidism due to bilat-
11.2.1 Introduction eral maldescent or atrophy. Acquired causes of
anorchia include trauma, torsion, toxic damage
In males, androgens, in particular testosterone, from alcohol or chemotherapy and infections,
are known to have many important physiological such as mumps, orchitis or bilateral orchiec-
actions, including effects on muscle, bone, cen- tomy, which may be performed as a treatment
tral nervous system, prostate, bone marrow, and for primary testicular cancer. There are many
sexual function. There are many causes of a sequelae for patients without testes. Patients are
bilateral loss or congenital absence of the testi- sterile, have increased risk of osteoporosis and
cles, which compromises endocrine, reproduc- pathologic fractures, and are subject to further
tive, and psychological function in men. The complications, such as sexual dysfunction, mood
11  Penis and Testis 281

changes, severe psychological trauma, altered occurs in the seminiferous tubules. The seminifer-
fat distribution, reduced stamina, and muscle ous tubules consist of germ cells and supporting
wasting [28]. cells and are a unique environment for gamete
Current strategies of treatment in anorchic production. Support cells include the Sertoli cells
patients focus on the medical management of and fibrocyte and myoid cells of the basement
androgenic hormone replacement therapy to membrane. The germ cells include a slowly divid-
avoid the complications associated with hypogo- ing stem cell population, more rapidly proliferat-
nadism. And such therapy may increase muscle ing spermatogonia and spermatocytes, and
strength, stabilize bone density, improve osteo- metamorphosing spermatids. Sertoli cells have
porosis, and restore secondary sexual character- irregularly shaped nuclei, prominent nucleoli, low
istics, including libido and erectile function. mitotic index, and exhibit unique tight junctional
Surgically, the main intervention is the implanta- complexes between adjacent Sertoli cells. These
tion of a prosthetic testis, a procedure which is tight junctions are the strongest intercellular barri-
well accepted by patients and has been shown to ers in the body. They divide the seminiferous
subjectively improve body image [29]. tubule space into basal (basement membrane) and
adluminal (lumen) compartments. This anatomic
arrangement forms the basis for the “blood-testis
11.2.2 Anatomy barrier” and allows spermatogenesis to occur in
an immunologically privileged site. Sertoli cells
The testes are two glandular organs, which serve as “nurse” cells for spermatogenesis, nour-
secrete the semen, suspended in the scrotum by ishing developing germ cells that are arranged
the spermatic cords. In mammals, the testes are between Sertoli cell cytoplasmic projections.
located outside the body because spermatogene- The second functions of testis are secretion of
sis in mammals is more efficient at a temperature steroid hormones (androgens) by Leydig cells in
somewhat less than the core body. When the tem- the interstitial tissue. Testosterone blood levels
perature needs to be lowered, the cremasteric change dramatically during human fetal, neonatal,
muscle relaxes and the testicles are lowered away and adult life. A testosterone peak occurs in the
from the warm body and are able to cool. human fetus between 12 and 18 weeks of gesta-
The tunica albuginea has smooth muscle cells tion. Another testosterone peak occurs at approxi-
that course through predominantly collagenous mately 2 months of age. A third testosterone peak
tissue. These smooth muscle cells may impart occurs during the 2nd or 3rd decade of life. After
contractile capability to the capsule, may affect this, there is a plateau, and then a slow decline with
blood flow into the testis, and promote the flow of age. The testosterone peaks correspond temporally
seminiferous tubule fluid out of the testis. to the following developmental events: (1) the dif-
Under the tunica albuginea, a tough fibrous ferentiation and development of the fetal repro-
shell, the testis contains very fine coiled tubes ductive tract, (2) the neonatal organization or
called seminiferous tubules. The tubes are lined imprinting of androgen-dependent target tissues,
with a layer of cells that, from puberty into old (3) the masculinization of the male at puberty, and
age, performed spermatogenesis. (4) the maintenance of growth and function of
From the cellular point of view the human tes- androgen-dependent organs in the adult.
tis is a complex organ comprising germ cells and
a variety of somatic cells such as Sertoli, Leydig,
endothelial, fibroblast, macrophage, and peritu- 11.2.3 Transplantation of Testicular
bular myoid cells. Tissue
Testicles are components of both the reproduc-
tive system (being gonads) and the endocrine sys- Testicular transplantation had its foundation not
tem (being endocrine glands). The testis has two as a treatment modality for the recognized com-
functions: The first is spermatogenesis, which plications of anorchism, but for arguably less
282 S. Cho and J.H. Bae

ethical reasons. The origins emerged from the According to this report, testosterone was
quest to find the fountain of eternal youth; in this being produced by the autograft, but without the
pursuit, the science of endocrinology and experi- normal architecture of the seminiferous epithe-
mentation with testicular transplants began in the lium, it is hard to understand how germ cell trans-
late nineteenth century. The French physiologist fer could have restored spermatogenesis.
Charles-Édouard Brown-Séquard [30] presented Therefore, efforts to develop tissue grafting for
the results of an experiment about injecting him- the purpose of improving testosterone levels in
self with crushed testicular extracts from young hypogonadal men are more likely to succeed than
dogs and guinea pigs. He reported this technique are attempts at restoring fertility. The former goal
successfully made him feel younger, along with appears to be simple, requiring the transfer of
increasing his sexual potency and heightening his interstitial cells (Leydig cells), which are readily
intellect. This early work reinforced the belief isolated from the donor testes by means of colla-
that youth rejuvenation could be achieved genase. Interstitial cells grafted in castrated
through the replacement of endogenous sub- rodents resulted in partial restoration of body
stances, which then led to an increased interest in weight, and testosterone levels above those of
sex gland transplantation. controls [34–36]. A number of vehicles and sev-
Voronoff in 1923 was the first to use chimpan- eral implantation sites for interstitial cells have
zee and baboon organs for treating patients. The been tried, but none fully replaced testicular
success described in this case quickly resulted in androgen production.
the procedure gathering interest. But that times However, in 2009, Sun reported the successful
none of surgeons used microsurgery to join blood restoration of androgen production in prepubertal
vessels of the graft to the host’s circulation, rats undergoing Leydig cell transplantation.
resulting in ischemic necrosis preceded by organ Serum testosterone levels were reported as nor-
rejection [31]. mal 12 weeks post-transplantation with a high
With the advent of synthetic testosterone and rate of survival and functionality of transplanted
other steroid hormones in 1935, the clinical indi- cells [37].
cations of testicular transplantation as a form of In the twenty-first century, attention turned to
androgen therapy gradually became obsolete. testicular transplantation mainly as a therapeutic
In 1978, Silber [32] published a case report intervention for infertility. This scenario arises in
claiming the first entire human testis transplant. oncology patients who face courses of chemo-
He described the case of two genetically identical therapy that jeopardize their fertility.
male twins, one born with two normally func- Cryopreservation of sperm leaves these patients
tioning testicles and the other born with none. with a finite resource of fertility and is associated
Using microvascular techniques, Silber success- with only modest rates of successful future con-
fully transplanted a testicle from the healthy twin ception. Cryopreservation is also not a suitable
into the anorchic brother, and, within 2 h of the option for prepubertal patients who have not yet
operation, normal levels of testosterone were started to produce sperm [38].
measured in both the donor and the recipient. Complex immunologic principles provide a
FSH and LH levels also gradually normalized, further unique challenge in testicular transplanta-
and, over several months, the sperm count and tion. The testes are known to be a site of immuno-
motility also reached normal levels in the logic privilege, which means they can tolerate the
recipient. introduction of antigens without the development
The problems arising from the size of the tes- of an immune response.
tis and its fibrous capsule led some transplanters Spermatogonial transplantation and reconsti-
to use sliced or minced organs. Kearns, who tution of donor cell spermatogenesis was shown
reimplanted testicular tissue subcutaneously in a in recipient mice by Nagano et al. [39]. The same
victim of accidental castration, reported the most group was able to show the restoration of fertility
plausible case [33]. in mice with the transplantation of male germ-­line
11  Penis and Testis 283

stem cells [40]. Subsequently Lo et al. reported nonpulsatile testosterone therapy is not optimal
that spermatogonial stem cells can be isolated and and can cause multiple problems, including fluid
enriched with specific markers [41]. In 2003, and nitrogen retention, excessive erythropoiesis,
Shinohara et al. presented that Sertoli cells, the hypertension, and bone density changes. In addi-
main component of the testicular germ-­cell niche, tion, fluctuating serum testosterone levels may
were recovered and dissociated from testes of occur, and frequent treatments may be required.
donor male mice and microinjected into recipient Due to these problems, alternate treatment modali-
testes, forming mature seminiferous tubule struc- ties, involving more physiological and longer-act-
tures, and supporting spermatogenesis [42]. ing systems for androgen delivery, have been
Kanatsu-Shinohara et al. showed the germ-line pursued. To address the problem, a system was
niche transplantation was also able to restore fer- designed in which Leydig cells, which produce
tility in mice [43]. most of the testosterone in the male, were micro-
According to Fijak and Meinhardt, further encapsulated in an alginate-­poly-­L-lysine solution
research into the immune system of the testis and injected into castrated animals. Serum testos-
may open new avenues for future successful tes- terone was measured serially; the animals were
ticular transplantation. It is apparent, however, able to maintain testosterone levels in the long
that despite more than 100 years of effort, evi- term [46].
dence of successful entire testis transplantation Other studies have shown that testicular pros-
between humans remains scarce [44]. theses created with chondrocytes in bioreactors
More recently, Raya et al. reported that engi- could be loaded with testosterone, and that these
neered testicular prosthesis using chondrocytes prostheses could provide controlled testosterone
seeded onto testicular-shaped polymer scaffolds release into the bloodstream when implanted. The
can be implanted in vivo, and can release testos- prostheses were implanted in athymic mice with
terone for a prolonged period. Furthermore, the bilateral anorchia, and testosterone was released
levels of testosterone release can be maintained long term, maintaining the androgen level at a
within the physiologic range. Periodic reinjection physiologic range [45]. One could envision com-
may potentially provide permanent physiologic bining the Leydig cell technology described
hormonal replacement. This novel technology above with engineered prostheses for the long-
may be beneficial for patients who require tes- term functional replacement of androgen levels.
ticular prostheses and chronic hormone supple-
mentation [45]. This technique can be used in
prepubertal boys who undergo chemotherapy to
save fertility after puberty. References
Testosterone and sperm production are the
1. Bosch RJ, Bénard F, Aboseif SR, Stief CG,
main functions of the testis, and can be disrupted Lue TF, Tanagho EA. Penile detumescence. Urol. 1991:
by congenital or acquired problems. However, 867–71.
sperm production is still beyond our ability for 2. Hu W, Lu J, Zhang L, Wu W, Nie H, Zhu Y, et al.
A preliminary report of penile transplantation: part 2.
now.
Eur Urol. 2006;50:1115–6.
3. Yoo JJ, Lee I, Atala A. Cartilage rods as a poten-
tial material for penile reconstruction. J Urol.
11.2.4 Testosterone Delivery System 1998;160:1164–8.
4. Kim B-S, Yoo JJ, Atala A. Engineering of human car-
tilage rods: potential application for penile prostheses.
Patients with testicular dysfunction require andro- J Urol. 2002;168:1794–7.
gen replacement for somatic development. Con- 5. Yoo JJ, Park HJ, Lee I, Atala A. Autologous engi-
ventional treatment for testicular dysfunction neered cartilage rods for penile reconstruction. J Urol.
1999;162:1119–21.
consists of periodic intramuscular injections of
6. Park HJ, Yoo JJ, Kershen RT, Moreland R, Atala
chemically modified testosterone or, more recently, A. Reconstitution of human corporal smooth muscle
skin patch applications. However, long-­ term and endothelial cells in vivo. J Urol. 1999;162:1106–9.
284 S. Cho and J.H. Bae

7. Kershen RT, Yoo JJ, Moreland RB, Krane RJ, Atala 22. Sim HG, Kliot M, Lange PH, Ellis WJ, Takayama
A. Reconstitution of human corpus cavernosum TK, Yang CC. Two-year outcome of unilateral sural
smooth muscle in vitro and in vivo. Tissue Eng. nerve interposition graft after radical prostatectomy.
2002;8:515–24. Urology. 2006;68:1290–4.
8. Kwon TG, Yoo JJ, Atala A. Autologous penile cor- 23. Namiki S, Saito S, Nakagawa H, Sanada T, Yamada
pora cavernosa replacement using tissue engineering A, Arai Y. Impact of unilateral sural nerve graft on
techniques. J Urol. 2002;168:1754–8. recovery of potency and continence following radi-
9. Yoo JJ, Chen K-L, Atala A. Total penile corpora cav- cal prostatectomy: 3-year longitudinal study. J Urol.
ernosa replacement using tissue engineering tech- 2007;178:212–6.
niques. The FASEB Journal. 2006;20(5):A885–A. 24. Djinovic R. Penile corporoplasty in Peyronie's dis-
10. Chen K-L, Eberli D, Yoo JJ, Atala A. Bioengineered ease: which technique, which graft? Curr Opin Urol.
corporal tissue for structural and functional res- 2011;21:470–7.
toration of the penis. Proc Natl Acad Sci U S A. 25. Imbeault A, Bernard G, Ouellet G, Bouhout S, Carrier
2010;107:3346–50. S, Bolduc S. Surgical option for the correction of
11. Song L-J, Xu Y-M, Li C, Fu Q, Cui L, Hu
Peyronie's disease: an autologous tissue-engineered
X-Y. Construction of cavernosum smooth muscle endothelialized graft. J Sex Med. 2011;8:3227–35.
using umbilical artery smooth muscle cells seeded 26. Schultheiss D, Lorenz RR, Meister R, Westphal M,
on acellular corporal collagen matrices. Int J Androl. Gabouev AI, Mertsching H, et al. Functional tissue
2009;32:514–23. engineering of autologous tunica albuginea: a pos-
12. Ji C, Min F, Liang W, Chen Y, Pan S, Bin L, et al. sible graft for Peyronie's disease surgery. Eur Urol.
Construction of tissue-engineered corpus cavernosum 2004;45:781–6.
with muscle-derived stem cells and transplantation 27. Joo K-J, Kim B-S, Han J-H, Kim C-J, Kwon C-H,
in vivo. BJU Int. 2011;107:1638–46. Park H-J. Porcine vesical acellular matrix graft of
13. An G, Ji C, Wei Z, Chen H, Zhang J. Engineering of tunica albuginea for penile reconstruction. Asian J
corpus Cavernosum using vascular endothelial growth Androl. 2006;8:543–8.
factor-expressing muscle-derived stem cells seeded 28. Kumar P, Kumar N, Thakur DS, Patidar A. Male
on acellular corporal collagen matrices. Urology. hypogonadism: symptoms and treatment. J Adv
2013;81:424–31. Pharm Technol Res. 2010;1:297–301.
14. Wessells H, Williams SK. Endothelial cell transplan- 29. Incrocci L, Bosch JL, Slob AK. Testicular prosthe-
tation into the corpus cavernosum: moving towards ses: body image and sexual functioning. BJU Int.
cell-based gene therapy. J Urol. 1999;162:2162–4. 1999;84:1043–5.
15. Chancellor MB, Tirney S, Mattes CE, Tzeng E, Birder 30. Schultheiss D, Denil J, Jonas U. Rejuvenation in the
LA, Kanai AJ, et al. Nitric oxide synthase gene trans- early 20th century. Andrologia. 1997;29:351–5.
fer for erectile dysfunction in a rat model. BJU Int. 31. Deschamps JY, Roux FA, Saï P, Gouin E. History of xeno-
2003;91:691–6. transplantation. Xenotransplantation. 2005;12:91–109.
16. Deng WW, Bivalacqua TJ, Chattergoon NN, Hyman 32. Silber SJ. Transplantation of a human testis for anor-
AL, Jeter JR, Kadowitz PJ. Adenoviral gene trans- chia. Fertil Steril. 1978;30:181–7.
fer of eNOS: high-level expression in ex vivo 33. Kearns WM. Testicular transplantation: successful

expanded marrow stromal cells. Am J Phys Cell Phys. autoplastic graft following accidental castration. Ann
2003;285:C1322–C9. Surg. 1941;114:886.
17. Tirney S, Mattes CE, Yoshimura N, Yokayama T, 34.
FOX M, BOYLE PF, HAMMONDS
Ozawa H, Tzeng E, et al. Nitric oxide synthase gene JC. Transplantation of interstitial cells of the testis. Br
therapy for erectile dysfunction: comparison of plas- J Urol. 1973;45:696–701.
mid, adenovirus, and adenovirus-transduced myoblast 35. BOYLE PF, Fox M, Slater D. Transplantation of inter-
vectors. Mol Urol. 2001;5:37–43. stitial cells of the testis: effect of implant site, graft
18. Connolly SS, Yoo JJ, Abouheba M, Soker S, McDougal mass and ischaemia. Br J Urol. 1975;47:891–8.
WS, Atala A. Cavernous nerve regeneration using 36. Tai I, Sun A. Microencapsulation of recombinant

acellular nerve grafts. World J Urol. 2008;26:333–9. cells: a new delivery system for gene therapy. FASEB
19. Kim ED, Scardino PT, Hampel O, MILLS NL,
J. 1993;7:1061–9.
Wheeler TM, Nath RK. Interposition of sural nerve 37. Sun J, Xi Y-B, Zhang Z-D, Shen P, Li H-Y, Yin M-Z,
restores function of cavernous nerves resected during et al. Leydig cell transplantation restores androgen
radical prostatectomy. J Urol. 1999;161:188–92. production in surgically castrated prepubertal rats.
20. Kim ED, Nath R, Slawin KM, Kadmon D, Miles BJ, Asian J Androl. 2009;11:405.
Scardino PT. Bilateral nerve grafting during radi- 38. Blackhall FH, Atkinson A, Maaya M, Ryder WDJ,
cal retropubic prostatectomy: extended follow-up. Horne G, Brison DR, et al. Semen cryopreservation,
Urology. 2001;58:983–7. utilisation and reproductive outcome in men treated
21. Singh H, Karakiewicz P, Shariat SF, Canto EI, Nath RK, for Hodgkin's disease. Br J Cancer. 2002;87:381–4.
Kattan MW, et al. Impact of unilateral interposition 39. Nagano M, Brinster RL. Spermatogonial transplanta-
sural nerve grafting on recovery of urinary function tion and reconstitution of donor cell spermatogenesis
after radical prostatectomy. Urology. 2004;63:1122–7. in recipient mice. APMIS. 1998;106:47–57.
11  Penis and Testis 285

40. Ogawa T, Dobrinski I, Avarbock MR, Brinster 43. Kanatsu-Shinohara M, Miki H, Inoue K, Ogonuki
RL. Transplantation of male germ line stem cells restores N, Toyokuni S, Ogura A, et al. Long-term culture
fertility in infertile mice. Nat Med. 2000;6:29–34. of mouse male germline stem cells under serum-or
41. Lo KC, Brugh VM, Parker M, Lamb DJ. Isolation feeder-free conditions. Biol Reprod. 2005;72:985–91.
and enrichment of murine spermatogonial stem cells 44. Fijak M, Meinhardt A. The testis in immune privilege.
using rhodamine 123 mitochondrial dye. Biol Reprod. Immunol Rev. 2006;213:66–81.
2005;72:767–71. 45. Raya-Rivera AM, Baez C, Atala A, Yoo JJ. Tissue
42. Shinohara T, Orwig KE, Avarbock MR, Brinster
engineered testicular prostheses with prolonged tes-
RL. Restoration of spermatogenesis in infertile tosterone release. World J Urol. 2008;26:351–8.
mice by Sertoli cell transplantation. Biol Reprod. 46. Machluf M, Atala A. Emerging concepts for tissue
2003;68:1064–71. and organ transplantation. Graft. 1998;1:31–7.

You might also like