ArticlePDF AvailableLiterature Review

Tissue and Organ Decellularization in Regenerative Medicine

Wiley
Biotechnology Progress
Authors:

Abstract and Figures

The advancement and improvement in decellularization methods can be attributed to the increasing demand for tissues and organs for transplantation. Decellularized tissues and organs, which are free of cells and genetic materials while retaining the complex ultrastructure of the extracellular matrix (ECM), can serve as scaffolds to subsequently embed cells for transplantation. They have the potential to mimic the native physiology of the targeted anatomic site. ECM from different tissues and organs harvested from various sources have been applied. Many techniques are currently involved in the decellularization process, which come along with their own advantages and disadvantages. This review focuses on recent developments in decellularization methods, the importance and nature of detergents used for decellularization, as well as on the role of the ECM either as merely a physical support or as a scaffold in retaining and providing cues for cell survival, differentiation and homeostasis. In addition, application, status and perspectives on commercialization of bioproducts derived from decellularized tissues and organs are addressed. This article is protected by copyright. All rights reserved.
Content may be subject to copyright.
Tissue and Organ Decellularization in Regenerative Medicine
Rajesh Guruswamy Damodaran
Laboratoire de bio-ingénierie et de biophysique de lUniversité de Sherbrooke, Department of Chemical and Biotechnological Engineering,
Université de Sherbrooke, 2500 Boul. de lUniversité, Sherbrooke, QC, J1K 2R1, Canada
Pharmacology Institute of Sherbrooke, Faculté de médecine et des sciences de la santé, 3001 12
ième
Avenue Nord, Sherbrooke, Québec,
J1H 5N4, Canada
Research Centre on Aging, Institut universitaire de gériatrie de Sherbrooke, 1036 rue Belvédère Sud, Sherbrooke, Québec, J1H 4C4,
Canada
Patrick Vermette
Laboratoire de bio-ingénierie et de biophysique de lUniversité de Sherbrooke, Department of Chemical and Biotechnological Engineering,
Université de Sherbrooke, 2500 Boul. de lUniversité, Sherbrooke, QC, J1K 2R1, Canada
Pharmacology Institute of Sherbrooke, Faculté de médecine et des sciences de la santé, 3001 12
ième
Avenue Nord, Sherbrooke, Québec,
J1H 5N4, Canada
Research Centre on Aging, Institut universitaire de gériatrie de Sherbrooke, 1036 rue Belvédère Sud, Sherbrooke, Québec, J1H 4C4,
Canada
DOI 10.1002/btpr.2699
Published online 0, 2018 in Wiley Online Library (wileyonlinelibrary.com)
The advancement and improvement in decellularization methods can be attributed to the
increasing demand for tissues and organs for transplantation. Decellularized tissues and organs,
which are free of cells and genetic materials while retaining the complex ultrastructure of the
extracellular matrix (ECM), can serve as scaffolds to subsequently embed cells for transplanta-
tion. They have the potential to mimic the native physiology of the targeted anatomic site. ECM
from different tissues and organs harvested from various sources have been applied. Many tech-
niques are currently involved in the decellularization process, which come along with their own
advantages and disadvantages. This review focuses on recent developments in decellularization
methods, the importance and nature of detergents used for decellularization, as well as on the role
of the ECM either as merely a physical support or as a scaffold in retaining and providing cues
for cell survival, differentiation and homeostasis. In addition, application, status, and perspectives
on commercialization of bioproducts derived from decellularized tissues and organs are
addressed. © 2018 American Institute of Chemical Engineers Biotechnol. Prog., 2018
Keywords: decellularization, recellularization, articial organs, extracellular matrix (ECM),
commercialization
Introduction
Increased demand for tissues and organs for transplantation
has expanded the elds of research on decellularization. Prep-
aration of decellularized tissues and organs involves removal
of cells and genetic materials from the tissue or organ and
retention of the complex ultrastructure of the extracellular
matrix (ECM) to serve as a natural scaffold. The ECM is pro-
duced by the native cells and serves as a mechanical support
and cues for cell migration, attachment, differentiation and
function.
1,2
Many tissues and organs have been successfully
decellularized and used for research and commercial purposes.
Different techniques including physical, chemical and biologi-
cal methods are currently applied in decellularization and
these have their own pros and cons.
The two important criteria to be fullled during a decellu-
larization procedure are: (i) retention of the native structures
and (ii) removal of a maximum of cell components. Several
tissues and organs such as the small intestine, bladder,
placenta, and liver, only to name a few, have been decellular-
ized and used as ECM scaffolds.
36
Recent advances in regen-
erative medicine and tissue engineering have paved the path
for investigating the use of whole decellularized organs for
transplantation. Many in vitro and in vivo studies support that
ECM-based scaffolds made of decellularized tissues and
organs have benecial effects on cells, including stem cells, in
terms of their attachment, proliferation, viability and function-
ality. In favor to that, these scaffolds have a low immunoge-
nicity, which facilitates their in vivo applications.
6,7
Over the past 20 years, decellularized tissues and organs
have been commercially available for wound healing. Compa-
nies are working to commercialize ECM-based products for
organ transplantation, which some are in clinical trials.
The objective of this review is to give an update on the
most important tissues and organs that are used for decellulari-
zation, the importance of the ECM as a complete set of bio-
molecules, the techniques and chemicals applied in
decellularization operations, rationale behind using ECM-
based bioproducts, as well as applications, perspectives and
path to commercialization.
Correspondence concerning this article should be addressed to
P. Vermette at: patrick.vermette@usherbrooke.ca
© 2018 American Institute of Chemical Engineers 1
Major tissues and organs used for decellularization
Liver
The liver is one of the important organs, which has been
decellularized for regenerative medicine. The rst successful
liver decellularization was done by surface treatment with
detergents and the resulting bioproduct was recellularized with
rat hepatocytes to assess functionality.
8
Surface treatment
refers to decellularizing a tissue or an organ by immersion in
decellularizing solutions as opposed to perfusion decellulariza-
tion, which involves perfusing solutions within the tissue or
organ. Studies on liver decellularization report the perfusion
of detergents through the network of veins and arteries, then
its recellularization and in vivo transplantation.
911
For exam-
ple, seeding rat hepatocytes into decellularized liver and sub-
sequent transplantation in rats showed that less damages
occurred to the cells following an 8 h transplantation, as
revealed by TUNEL staining.
9
In a separate study, when
human liver stem cells were seeded in decellularized rat liver,
differentiation into functional hepatocytes was observed as
well as the presence of epithelial and endothelial-like cells
reported.
12
Research on achieving human-sized whole liver
has also been attempted with pigs by decellularizing the organ
with SDS (sodium dodecyl sulfate).
13,14
Heart
Heart decellularization techniques started initially with heart
valves by surface decellularization using detergents and subse-
quent recellularization with different cell types.
1517
Intro-
duced in 2008, decellularization of whole hearts from rats was
done by perfusion methods and the resulting scaffolds were
seeded with cardiac and endothelial cells; contraction in cell
patches was observed after 4 days.
18
Recent advances in heart
decellularization aim to improve perfusion processes during
decellularization and cell seeding with progenitors as well as
to achieve decellularization of organs from larger animals.
1923
Allo-transplanted porcine heart seeded with mesenchymal
stem cells showed thrombosis in arteries as well as the pres-
ence of inammatory cells and the observation was similar to
decellularized heart transplanted without mesenchymal stem
cells.
24
In addition, a 14-day culture of human cardiomyocytes
in human decellularized heart showed visible contraction when
electrically stimulated.
25
Lungs
Decellularized lungs were considered as a source of bioarti-
cial organs for transplantation in end-stage lung diseases. The
rst successful perfused decellularization was reported in 2010
following methods initially applied on heart and liver.
26
Recent
works on decellularizing lung involve the improvement of
decellularization process and cell seeding methods, as well as
studies on the role of stem cells.
2628
Attempts to decellularize
human lungs are step closer to ll the gap in the demand for
organ transplantation.
29,30
Stabilization of endothelial cells by
seeding adipose-derived stem cells into decellularized lungs
could be a potential solution to regenerate the vasculature of
decellularized lungs.
31
Seeding mesenchymal stromal cells in
decellularized lung in suspension bioreactor resulted in differen-
tiation of the cells into collagen-1 alpha-1 producing cells.
32
In
another study, human-induced pluripotent stem cells (iPSCs)-
derived endothelium and ventralized iPSC were seeded in
decellularized lungs resulting in retention of epithelial progeni-
tors phenotype by expressing Nkx21 and endothelial phenotype
by expressing CD31.
33
Transplantation of decellularized rat
lung seeded with epithelial and endothelial cells maintained the
animals alive without ventilation for 6 h.
26
Pancreas
The rst perfused decellularization of pancreas was carried
out in 2009 with a porcine organ,
34
and then applied in
mouse.
35
The decellularized tissues were used to seed cells and
islets and their in vitro functions were studied as well as the
biocompatibility of these bioarticial pancreata was investigated
in vivo.
3437
Later, rat pancreata were decellularized and a new
method was introduced to infuse islets into the ducts, veins and
arteries.
38
Decellularized pancreata were seeded with human
stem cells, an AR427 acinal cell line, MIN-6 cells and islets,
and cell attachment as well as basic cell functions was
studied.
3538
Recently published work from our group demon-
strated that, islets infused into the ductal system of decellular-
ized pancreata were functional by releasing insulin in response
to glucose stimulation after 48 days in vitro.
39
Small intestinal submucosa
One of the rst usages of small intestinal submucosa (SIS)
was during 1989, as a vascular graft for dogs.
40
SIS biomate-
rials are widely used in regenerative medicine, for example to
reconstruct urethra, cornea, esophageal, and heart.
4144
Recently, SIS seeded with urothelial cells showed formation
of new vessels, proliferation of smooth muscle cells, epitheli-
zation and maintenance of the opening of urethra when com-
pared with unseeded scaffold.
45
Reconstruction of cornea was
accomplished successfully in 106 cases by preserving vision
at third month of postsurgery using porcine SIS.
46
Seeding
autologous oral mucosa epithelial cells along with SIS resulted
in muscular regeneration and re-epithelization after 8 weeks of
surgery and the results were superior to SIS with no cells.
47
Applying Food and Drug Administration (FDA)-approved SIS
ECM seeded with mesenchymal stem cells resulted in a lower
response of the adaptive immune system in a porcine model,
when compared with SISECM with no cells.
48
Bladder
Urinary bladder has been one of the rst organs selected for
decellularization. Recent studies on ECM derived from decel-
lularized bladder have indicated a benecial effect on neuron
survival, stem cells, spinal cord, and open wounds.
4952
Uri-
nary bladder-derived ECM has shown greater advantage than
cardiac-derived ECM in remodelling and for attracting site-
specic cells (cardiomyocytes) after implantation.
53
The
implantation of large segments (>24cm
2
) of urinary bladder
into porcine bladder showed inltration of smooth muscle
cells and promoted angiogenesis after 30 days.
54
Vesicles
derived from urinary bladder ECM had a positive effect on
neuron neurite growth and on spinal cord injury repair.
50,51
Urinary bladder-derived ECM also had a better effect on the
treatment of open wounds when compared with conventional
therapies.
49
Bioproducts made from urinary bladder-derived
ECM are available in different formulations such as sheets
(Cytal
®
Burn Matrix, also marketed as MatriStem
®
Burn
Biotechnol. Prog., 20182
Matrix, ACELL (Columbia, USA); the product is applied
directly on wounds), powder (MicroMatrix
®
, ACELL, Colum-
bia, USA) and hydrogels (not available commercially).
Kidneys
The initial method of decellularization of whole kidneys
was achieved by perfusion techniques, and the resulting prod-
ucts were used to seed and differentiate stem cells.
34,55
Advances in kidney decellularization include in vitro and
in vivo studies on rat and mouse kidneys to investigate the
functionality of recellularized kidneys with endothelial, epithe-
lial and renal progenitor cells.
56,57
Bones and cartilages
The successful removal of cells from bones and cartilages
was achieved in 2011.
58
Recent advancements in cartilage and
bone repair include utilizing adipose-derived stem cells, multi-
potent stromal cells seeded on decellularized cartilage ECM to
study the mechanical properties and bone regenerative
capacity.
59,60
Plants
Recent advancements in tissue and organ decellularization
have extended their boundaries to apply perfused decellulari-
zation protocols on plant leaves and eventually recellularize
them with cardiomyocytes resulting in contractile function
successfully for 21 days.
61
Recent decellularization methods
Over the years, several studies have been conducted to
understand the utility of decellularized tissues and organs in
regenerative medicine. Table 1 provides a summary of recent
studies highlighting the various techniques and their corre-
sponding results on decellularized tissues and organs.
Chemicals used for decellularization
Any decellularizing procedure involves disruption of the
ECM. The selection of decellularizing agents causing the least
possible damage to the ECM is vital. The choice of decellular-
izing agents depends on the nature of the tissue or organ. A
decellularization operation involves notable changes in the tis-
sue and organ color, starting from its original color to become
translucent. The process consists in removing cellular compo-
nents and nuclear materials. Destruction of the cell membrane,
dissolution of cellular components and removal of nucleic acid
materials are the key steps.
Alkalis and acids dissolve cellular components and wash
nucleic acids.
80
Detergents solubilize cell membranes and
remove cellular components and nucleic acids.
81
Detergents are soluble amphiphiles, which can solubilize
biological membranes. They have both hydrophilic and hydro-
phobic groups with a more hydrophilic nature than biological
membranes. They can be classied according to their charges:
nonionic, anionic, cationic, and zwitterionic detergents.
82,83
Triton X-100, a nonionic detergent, is commonly used for cell
lysis and permeabilization.
84,85
The hydrogen bonds in lipid
bilayers are disrupted by the polar head group of Triton X-
100, breaking the cell plasma membrane.
82
In some studies,
Triton X-100 has been reported to be the best decellularizing
agent compared with other detergents and that for many tis-
sues and organs including ligaments, small intestine, annulus
brosus, liver and aortic valves.
42,8689
On the other hand,
some studies demonstrated that Triton X-100 had poor cell
removal capacity for many tissues and organs such as veins,
cornea, urethra, heart and kidneys.
18,9093
A detergent commonly used in decellularization is the SDS.
SDS is an anionic detergent, used in commercial applications
such as in cleaning and in the making of cosmetics. SDS is
also well known in biochemistry for its use in the SDS-PAGE
technique. Its application in decellularization can be counterin-
tuitive when aiming to preserve the tissue or organ ECM
structure intact, as SDS is known to denature proteins.
94
SDS
solubilizes membrane proteins and thus helps effectively in
the decellularization process. SDS incorporates into the outer
membrane layer by increasing its curvature, creating a stress
which results in the membrane disruption.
95
SDS proved to be
superior when compared with other detergents in removing
cells and genetic materials in many tissues and organs includ-
ing aorta, veins, heart, kidneys and urethra.
18,90,92,93,96
Triton
X-100 and SDS have also shown negative inuence on tissues
and organs used for decellularization but, on the other hand,
when these two detergents were used in combination, the mix-
ture allowed efcient decellularization and preservation of
ECM structures and this has been tested on tendons, kidneys,
SIS, and liver.
11,43,97,98
Another ionic detergent/bile salt frequently used in decellu-
larization is sodium deoxycholate (SDC). After adhering to
the membrane, SDC incorporates its cholate moiety into the
lipid membrane, resulting in pore formation and membrane
disruption.
99
SDC, alone or in combination with other deter-
gents, has shown to be effective in removing cells while
retaining ECM structures in decellularized tissues.
16,100
CHAPS, 3-([3-cholamidopropyl]dimethylammonio)-
1-propanesulfonate hydrate, is another detergent used in decel-
lularization. CHAPS is a zwitterionic detergent with a hydro-
philic side and a hydrophobic back. CHAPS does not result in
pore formation, instead the molecules seem to directly break
the lipid membrane.
101
CHAPS has proven to be milder and
to have greater ability to retain mechanical strength in
lungs.
27,30,102
Other chemicals applied in decellularization protocols are
acids and bases. Both acids and bases had a negative impact
on ECM mechanical strength.
103
Acetic acid-treated bovine
pericardia had reduced tensile strength, but on the other hand,
it supported the growth of human mesenchymal stem cells and
hence proved to be biocompatible.
104
Peracetic acid along
with ethanol was used to sterilize decellularized scaffold mate-
rials, a well-known and FDA-approved method for medical
devices.
6
However, the use of peracetic acid for decellulariza-
tion resulted in incomplete cell removal from tumor ECM,
porcine liver and kidneys.
105,106
Decellularization of skin
using NaOH had a cytotoxic effect on broblasts, however,
prolonged washing of the scaffold helped in decreasing cyto-
toxicity.
107
Alcohols or acetone solutions can be used in pre-
treatment of tissues for lipid removal. It is easier to lyse and
remove cells after eliminating lipid contents.
108
Acetone/alco-
hol combinations had a negative effect on tissue dehydration
and greater inuence on modifying mechanical properties and
morphology of temporomandibular joint from pig jaws, as
compared with SDS-treated tissues.
109
Biotechnol. Prog., 2018 3
Table 1. Recent Studies Involving Decellularization Methods and Their Outcomes
Organ or
Tissue Species Applications Methods Chemicals Results References
Liver Mouse Culture of mouse hepatocytes Nonthermal irreversible
electroporation
- Cell integration into the host liver
parenchyma
62
Rat Hepatocyte seeding and
transplantation into rat
Perfusion through portal vein SDS (0.01, 0.1 and 1%) Preservation of functional and
structural characteristics of
microvascular network and display
of liver-specic functions
9
Decellularization using
increasing detergent
concentration
Perfusion through portal vein 1, 2 and 3% Triton X-100 +
0.1% SDS
Retention of laminin in the basement
membrane and collagen IV
63
Minimizing ECM damage Arterial perfusion under
oscillation pressure
conditions
1% Triton X-100 + 1% SDS Fast, homogenous, higher
concentration of hepatocyte growth
factor compared with the native
tissue, high GAG concentration and
gentle method of decellularization
64
Pig Comparison of three detergent
mixtures for decellularization
and recellularization with rat
hepatocytes
Perfusion decellularization and
freezethaw
1% SDS;
1% Triton X-100 + 1% SDC;
1% SDC +1% SDS
Cell removal, preservation of ECM
structure and biocompatibility,
achieved by 1% Triton X-100 + 1%
SDC
11
Kidneys Pig Study on decellularization
methods
Perfusion using a
high-throughput
decellularization apparatus
water and 0.5% SDS Preservation of important ECM
components, intact vasculature tree,
in vitro biocompatibility for cells
and preservation of gross anatomy
after 2 weeks of implantation
65
Heart Rat Recellularization with aortic
endothelial cells and rat
neonatal cardiomyocytes
Coronary perfusion 1% SDS Performance of basic functions 66
Zebra sh
+ Mouse
In vitro proliferation and
migration of human cardiac
precursor cells
Freezethaw cycles - Exhibition of pro-proliferative and
chemotactic effect with human
cardiac precursor cells by zebra sh
ECM including structural
preservation and cardiac contractile
function
67
Pig Comparison of two detergents Surface treatment 1% Triton X-100 + nucleases +
trypsin and SDC with and
without nucleases
Complete cell removal and
preservation of ECM structures by
Triton X-100 + nucleases + trypsin
68
Myocardium Pig Comparison of decellularizing
agents and recellularization
with rat myocardial
broblasts
Surface treatment 1% SDS;
1% Triton X-100; 0.5% trypsin
Effective decellularization by SDS and
retention of ECM microstructures.
Display of different beating
magnitudes of seeded cells: largest
in trypsin-, moderate in SDS- and
none in Triton X-100-treated
scaffolds
69
Teeth Human Comparison of
decellularization methods
Surface treatment 10% formaldehyde, PBS +
EDTA +2.5% sodium
hypochlorite; PBS + EDTA
+ 40 v hydrogen peroxide;
PBS + 2.5% sodium
hypochlorite + Ryozime
®
;
PBS + EDTA + 40 v
hydrogen peroxide +
Ryozime
®
Maximum removal of biological
particles and less damage to the
structures obtained with
PBS + EDTA +40 v hydrogen
peroxide + Ryozime
®
70
Biotechnol. Prog., 20184
Bones and
cartilages
Rat Calvaria regeneration in vivo
and recellularization with
mesenchymal stem cells
Surface treatment 0.5% SDS + 0.1% ammonium
hydroxide
Formation of new bone and merging
with the host bone after 3 months.
Proliferation and osteogenic
differentiation of mesenchymal stem
cells
71
Pig Tissue characterization and
recellularization with murine
broblasts and porcine
chondrocytes
Surface treatment 1% SDS Retention of compatible tension
properties of intact tissue but
reduction in compression. No
cytotoxicity on recellularized cells
72
Bovine Tissue characterization and
recellularization with human
adipose mesenchymal stem
cells
Surface treatment 10 mM
ethylenediaminetetraacetic
acid disodium salt dihydrate
(Na
2
EDTA, pH 7.27.4)
Chondrogenic differentiation of human
adipose mesenchymal stem cells
73
Skeletal
Muscle
Pig Comparison of
decellularization methods
and formulation of hydrogel
scaffolds
Surface treatment 1% SDS; 1% SDS + 0.2%
SDC; 1% SDS + 1% Triton
X-100; 1. 0.2%
Trypsin/0.1% EDTA,
2. 0.5% Triton X-100, 3. 1%
Triton X-100/0.2% SDC
Formation of jelly-like hydrogel by
using 0.2% trypsin/0.1% EDTA,
0.5% Triton X-100, and 1% Triton
X-100/0.2% SDC treatment
74
Nerve Rat Reconstruction of long gap
nerve injury
Surface treatment (Dulbeccos modied Eagle
medium +10% fetal bovine
serum and 4% penicillin/
streptomycin/amphotericin
B) + (DMEM with 10% fetal
bovine serum and 2%
penicillin/streptomycin/
amphotericin) + PBS
Exhibition of functional nerve
regeneration in vivo
75
Skin Mouse Comparison of detergent-free
decellularization with ionic
and anionic detergents for
decellularization
Surface treatment (50 nM latrunculin B +
Dulbeccos modied Eagle
medium) + 0.6 mol/L
potassium chloride +1.0 M
potassium iodide
Retention of matrix composition and
biomechanical properties with
detergent-free decellularization
method, but all methods resulted in
comparable bio-functionality
76
Trachea Rabbit In vivo transplantation Freeze-drying, detergent and
sonication
1% SDS Exhibition of necrosis and animal
death in 724 days and integration
of decellularized scaffold into the
host structure was observed
77
Spleen Rat Recellularization of bone
marrow mesenchymal stem
cells (BMSCs).
Freezethaw cycles and
perfusion decellularization
0.1% trypsin, 0.05% EDTA,
3% Triton X-100
Differentiation of BMSCs into
functional hepatocyte-like cells
78
Culture of rat hepatocytes Perfusion decellularization 0.1% SDS Survival and secretion of urea and
albumin for 10 days in culture
79
Pancreas Mouse Recellularization of MIN-6
cells and AR42J
Perfusion decellularization 0.5% SDS Upregulation of insulin gene in vitro,
less immune reaction and
angiogenesis after 14 days in vivo
35
Recellularization of MIN-6
cells and in vivo
implantation
Freezethaw at 80Cfor
1 day and followed by
perfusion decellularization
1% Triton X-100 + 0.1%
ammonium hydroxide
Biocompatibility and angiogenesis
induction
37
Rat Three perfusion routes for
decellularization and
recellularization of islets
Perfusion decellularization 1% Triton X-100 + 0.5% SDS Reach of islets into parenchymal space
of the pancreas after infusion
through ductal system
38
BMSCs, bone marrow mesenchymal stem cells; ECM, extracellular matrix; GAG, glycosaminoglycans; PBS, phosphate-buffered saline; SDS, sodium dodecyl sulfate.
Biotechnol. Prog., 2018 5
Along with chemicals, the use of enzymes in decellularizing
techniques can be advantageous. However, it is not possible to
decellularize tissues or organs only with enzymes. Enzymes
can be used in the removal of cell debris. The most commonly
used enzymes are trypsin, nucleases, collagenase, thermolysin,
α-galactosidase and dispase.
103,110
Rationale for using ECM-based products
ECM proteins have shown positive inuence on cell
growth, function, and differentiation and examples of biomi-
metic surfaces have been reviewed by us.
111
An important class of ECM-derived proteins is that of colla-
gens, as reviewed elsewhere.
112
Collagen is in fact an abun-
dant source of ECM proteins. Collagen microspheres can
differentiate oligodendrocyte progenitors into oligodendro-
cytes and collagen-immobilized nanowires had positive inu-
ence on human microvascular endothelial cells.
113,114
Fibronectin and laminin showed positive effects on cell sur-
vival and functions both in vitro and in vivo.
115,116
In the past
years, our group has veried the positive effects of various
ECM peptides and proteins. For example, immobilization of
bronectin, brinogen and brin on polymer surfaces
enhanced the proliferation of smooth muscle cells.
117
Also,
embedding pancreatic islets and endothelial cells in brin
showed increased insulin secretion and preservation of islet
integrity.
118
Culturing young porcine islets in brin exhibited
the protective nature of brin towards hydrogen peroxide.
119
In addition to whole ECM proteins, functional peptides
derived from the ECM can inuence cell behaviors.
120123
Peptide motifs from collagen I (GTPGPQGIAGQRGVV), col-
lagen IV (MNYYSNS), bronectin (PHSRN), laminin
(YIGSR) can enhance cell attachment.
124,125
Arginine-glycine-
aspartic acid (RGD), a peptide sequence found in bronectin,
vitronectin, laminin, collagen types I and IV, has a high afn-
ity for integrin-mediated cell attachment and spreading, func-
tionality and tissue development.
126130
The RGD sequence
co-immobilized with the SVVYGLR sequence was used to
enhance the adhesion of endothelial cells.
123
Three ECM-
derived peptides, laminin (IKLLI and PDSGR) and cadherin
(HAVDI) supported the adhesion and survival of insulin-
containing cultures for 5 days.
122
Rat insulinoma INS-1 cells
cultured on immobilized ECM proteins and peptides (RGD
and CDPGYIGSR) showed higher glucose-stimulated insulin
secretion compared with the surfaces bearing no peptides.
120
Hyaluronic acid, a polysaccharide, helped in neuron sur-
vival and controlled self-renewal and differentiation of human
embryonic stem cells.
131,132
Many studies demonstrated that
proteoglycans from the ECM are involved in molecular events
including cell adhesion, proliferation and migration.
133
These are just examples of the effects ECM components
can have on cells allowing appreciating the potential impact of
bioproducts made from decellularized tissues and organs. As
reviewed by us and others,
111,134,135
other ECM proteins have
been applied to produce biomimetic materials and surfaces
and these include: collagen types I, III, and IV, gelatin, bro-
nectin, vitronectin, polysaccharide nanobers, proteoglycans,
nidogen, and laminins, as well as ECM-mimicking peptides
including YIGSR,RGD, DGEA, KRSR, IKVAV, P15, and
GFOGER, to list a few.
Furthermore, following their implantation, decellularized
scaffolds are repopulated by host cells and even degraded,
resulting in a functional tissue containing site-specic cells.
136
Decellularized tissues or organs, with or with no cells, can
chemo-attract progenitor cells towards the implantation
site.
137,138
The degrading ECM molecules are also involved in
various biological activities and growth factor signaling.
139,140
Therefore, these matrices made from decellularized tissues and
organs act as bioactive scaffolds by modifying the host envi-
ronment to create a more suitable solution for healing, repair,
and even regeneration.
ECM scaffolds made from decellularized tissues and organs
also provide mechanical support in many conditions. They
possess different physical characteristics, which are often dif-
cult to reproduce with synthetic materials.
141
The ECM pro-
vides a mechanical support for the recellularizing cells to
attach to the matrix. It is very important for scaffolds made
from decellularized tissues and organs to have certain mechan-
ical properties matching the site of implantation of the tissue
or organ to be substituted. Creating less damage to the tissue
during decellularization could yield better mechanical proper-
ties and more intact vasculature structure, facilitating the
induction of angiogenesis post-implantation.
37
Also, the pres-
ence of ECM proteins and functional peptides in 3D scaffolds
allows recreating environments more physiological for cells,
as opposed to traditional 2D culture systems on at surfaces.
Immunogenicity of ECM-based scaffolds
Most decellularization protocols involve removing cell
materials, which could cause the major immunogenic reac-
tions: hyperacute, acute immune, and chronic immune rejec-
tion as well as inammatory reactions. Hyperacute rejection is
a severe reaction of vascularized xenografts, triggered a few
minutes to a few hours after implantation.
142,143
This hypera-
cute rejection is mainly caused by the α-Gal epitope
(galactose-α[1,3]-galactose) expressed on many cell surfaces,
and is less common in human and old-world monkeys. Pigs
have been modied genetically to remove the Gal epitope to
avoid xenograft rejections, but the application of this method
is resulting into an acute immune rejection by non-Gal anti-
bodies circulating at low levels, a few days to a few weeks
after implantation.
144
Chronic immune rejection is associated
with allografts transplantation by introducing donor-specic
antibodies.
145
Also, the presence of DNA in decellularized tis-
sues and organs can potentially cause inammatory
reactions.
146
Collagens with lower immunogenicity is achieved by
removing N- and C-terminal telopeptides by pepsin type-I
treatment, which are referred to as atelocollagens.
147
Atelocol-
lagens have been clinically applied in wound healing and as
bone substitutes.
147
Host response towards bioproducts made from ECM mate-
rials are different and are based on the product properties, spe-
cies and methods of preparation; ve different commercially
available products were tested on rats and resulted in distinct
morphological appearance of the implantation site.
148
Simi-
larly, the immune reaction from the decellularized heart tissue
(SynerGraft
®
) had a mixed reaction after implantation. In one
study,
149
SynerGraft
®
exhibited much less human leukocyte
antigen Classes I and II antibodies, as claimed by the com-
pany; however, in another study,
110
macrophage inltration
after implantation was observed into the tissue decellularized
using the SynerGraft
®
process.
142
In addition, porcine SIS had
both positive and negative inammatory responses when
implanted into rodents.
150152
Biotechnol. Prog., 20186
Another important factor to be investigated is the response
of the host innate and adaptive immune responses towards
implanted xenograft materials. The polarization of T helper
cells 1 (Th1) and macrophages (M1) results in a proinamma-
tory response and T helper cells 2 (Th2) and macrophages
(M2) are involved in an anti-inammatory response, which
leads to constructive tissue remodelling.
153
Many studies have different conclusions on host-
decellularized tissues/organs interactions and responses.
Therefore, a detailed scientic understanding of the outcomes
following decellularized products implantation including the
responses of host tissues towards the implanted decellularized
materials is vital.
154
Applications, status, and perspective of decellularized
tissues and organs
Engineered substitutes could be a possible cure for patients
in the last stage of tissue or organ failure. The increasing
demand for tissues and organs for transplantation and the
encountered immune rejection of xenograft transplantations
have pushed researchers to nd alternative tissue and organ
sources.
Decellularized tissues and organs have a great potential to
serve as carriers for transplanting autologous tissue made from
the host cells or site-specic functional cells. Regardless of
the availability of genetically engineered animals to harvest
tissues or organs for transplantation, the rejection of the trans-
planted graft by the host appears inevitable.
155
This situation
can potentially be solved with scaffolds and other bioproducts
made from decellularized tissues and organs since antigenic
elements of cell components are supposed to be completely
removed. Recent advances involving recellularization of
human-iPSCs in decellularized scaffolds could be a way to
avoid the intake of immunosuppressive drugs required after
organ transplantation.
33,156
Achieving a functional tissue or organ is the goal of any
decellularized scaffolds carrying either functional cells or stem
cells. The absence of re-endothelialization of the organ with a
functional endothelium is one of the main reasons for organ
loss in transplantation and a major hindrance in developing
functional decellularized scaffolds. However, many studies
have shown promising results on re-endothelialization and
could be considered as a potential solution to solve re-
endothelialization problems.
157,158
Materials and scaffolds made from decellularized tissues
and organs have been commercially available for many appli-
cations. ECM-derived products from animal sources have been
commercially available for more than 20 years and are applied
in the treatment of many tissue regeneration processes and sta-
tistically, many years ago, over 200,000 patients have been
implanted with xenogeneic decellularized scaffolds.
159
Many
animal and human tissue-derived decellularized commercial
products are applied in wound healing and these include:
Oasis
®
(porcine small submucosa, Cook Biotech, Inc.,
Indiana, USA), GraftJacket
®
(human dermis, Acelity L.P. Inc.,
Texas, USA), DermACELL
®
(human dermis, Novadaq Tech-
nologies Inc., Mississauga, Canada), Alloderm
®
(human der-
mis, Allergan plc [NYSE: AGN, Dublin, Ireland]),
NeoForm(human dermis, California, USA), Strattice
(porcine dermis, Allergan plc [NYSE: AGN, Dublin, Ireland]),
Restore(porcine small intestine [DePuy Orthopedics, Inc.,
Indiana, USA]), PrimaPlus (porcine heart valve, Edwards
Life Sciences LLC, California,USA), AlloSkinAC (human
dermis, AlloSource, Centennial, CO, USA), MatriStem
®
(mucosa of urinary bladder, ACell, Columbia, USA),
Biodesign
®
(small intestine, COOK MEDICAL INC., Bloom-
ington, IN, USA), Lyoplant
®
(pericardium, Aesculap, Inc.,
Center Valley, PA, USA).
160,161
Most of the biological scaffolds are marketed as surgical
mesh devices.
161
Scaffolds made from decellularized tissues or
organs have a potential to be marketed under the category of
510(K) of the FDA.
162
Under a 510(K) clearance, a company
must register to notify (PreMarket Notication) the FDA
90 days in advance to market a medical device. FDA would
assess the notied device to make sure it falls under already
existing three classications of devices in the market, for which
a premarketing approval is not required. The claim must be
made by the company to support their device is substantially
equivalent to the ones already available in the market. 510
(K) devices can fall under three classes: Classes IIII, depend-
ing on the assurance of safety and effectiveness (Table 2).
Oasis
®
, MiroMesh
®
, and PhotoFix
®
are few examples of
510(K)-cleared commercially available bioproducts derived
from decellularized tissues and organs.
160,164
Decellularized tissues which are intended to be implanted
or transplanted have a potential to be marketed under the cate-
gory of Human Cells, Tissues, and Cellular and Tissue-Based
Products (HCT/Ps) by FDA. HCT/Ps are the products regu-
lated by two different divisions namely the Center for Devices
and Radiological Health and the Center for Biologics Evalua-
tion and Research, or sometimes as a combination of both.
165
Table 2. Regulatory Requirements for 510(k) Devices
163
Regulatory Requirements Class I Class II Class III
General controls All general controls are
complied with and provide
reasonable assurance
General controls are
insufcient to provide
reasonable assurance
General controls are insufcient to
provide reasonable assurance
Secondary controls No further secondary controls
are required
Information required to
perform secondary controls
are sufcient and hence
secondary controls are
performed for gaining
reliable assurance of the
device
Information required to perform
secondary controls are
insufcient and hence
secondary controls cannot be
performed for gaining reliable
assurance of the device
Premarket notication/
approval
Eligible directly for premarket
notication
Eligible directly for premarket
notication
Requires premarket approval prior
to notication
Device/product risk Low Medium High
Note: General controls refer to a comprehensive set of regulatory authorities to be complied for by any medical device to be marketed. Special controls
are controls to be complied where general controls are insufcient.
Biotechnol. Prog., 2018 7
DermACELL
®
(a decellularized human dermis) and Allopatch
HD
®
(an acellular human dermis) are examples of HCT/Ps
products.
160,166
Miromatrix Medical Inc. (Eden Prairie, Min-
neapolis), a USA-based biotechnology company, is involved
in developing fully functional human organs by perfusion
technology, primarily applied to the liver and upon completion
extended to the kidneys. Miromeshand Miroderm
(derived from porcine liver) are products launched by the
company. AxoGen Inc. (Alachua, Florida) from USA,
develops a technology to regenerate peripheral nerve injuries.
AxoGuard
®
is derived from pigs and is used as a nerve con-
nector. Another USA-based company, Humacyte Inc.
(Morrisville, North Carolina), develops acellular grafts to
repair vascular damages; some of their products are under
phase III clinical trials.
Conclusions
The selection of a technique or chemicals to decellularize a
specic tissue or organ is an important decision to achieve
complete decellularization and to retain the ECM as intact as
possible. Perfusion decellularization has allowed fast and com-
plete decellularization of many whole tissues and organs using
either SDS or Triton X-100. Many whole organs have been
successfully decellularized and subsequently seeded with dif-
ferent cell types. Recent advances in recellularizing decellular-
ized tissues and organs involve the use of iPSCs to attain the
native function of the tissue. Decellularized tissues or organs
offer both mechanical support and cues from the ECM for cell
growth and function. Many bioproducts derived from decellu-
larized tissues and organs are commercially available, along
with that, regenerative medicine companies are working to
market functional whole organs to meet the demand. Extract-
ing puried proteins from ECM or synthesizing new motifs
are costly affairs compared with decellularizing a tissue, there-
fore, decellularized tissues and organs could be a potential
source of substitutes for transplantation, provided they are
devoid of cellular components, they maintain the necessary
functionality and they have acceptable immunogenicity.
Acknowledgments
This research project was supported by the Université de
Sherbrooke and NSERC through a Discovery Grant awarded
to Patrick Vermette (Grant no. 250296-2012).
Literature Cited
1. Vorotnikova E, McIntosh D, Dewilde A, Zhang J, Reing JE,
Zhang L, Cordero K, Bedelbaeva K, Gourevitch D,
Heber-Katz E, Badylak SF, Braunhut SJ. Extracellular
matrix-derived products modulate endothelial and progenitor
cell migration and proliferation in vitro and stimulate regenera-
tive healing in vivo. Matrix Biol. 2010;29:690700.
2. Cukierman E, Pankov R, Stevens DR, Yamada KM. Taking
cell-matrix adhesions to the third dimension. Science. 2001;294:
17081712.
3. Kimmel H, Rahn M, Gilbert TW. The clinical effectiveness in
wound healing with extracellular matrix derived from porcine
urinary bladder matrix: a case series on severe chronic wounds.
J Am Col Certif Wound Spec. 2010;2:5559.
4. Choi JS, Kim JD, Yoon HS, Cho YW. Full-thickness skin
wound healing using human placenta-derived extracellular
matrix containing bioactive molecules. Tissue Eng Part A. 2013;
19:329339.
5. Sandusky GE, Lantz GC, Badylak SF. Healing comparison of
small intestine submucosa and ePTFE grafts in the canine
carotid artery. J Surg Res. 1995;58:415420.
6. Brown B, Lindberg K, Reing J, Stolz DB, Badylak SF. The
basement membrane component of biologic scaffolds derived
from extracellular matrix. Tissue Eng. 2006;12:519526.
7. Ma R, Li M, Luo J, Yu H, Sun Y, Cheng S, Cui P. Structural
integrity, ECM components and immunogenicity of decellular-
ized laryngeal scaffold with preserved cartilage. Biomaterials.
2013;34:17901798.
8. Lin P, Chan WC, Badylak SF, Bhatia SN. Assessing porcine
liver-derived biomatrix for hepatic tissue engineering. Tissue
Eng. 2004;10:10461053.
9. Uygun BE, Soto-Gutierrez A, Yagi H, Izamis ML,
Guzzardi MA, Shulman C, Milwid J, Kobayashi N, Tilles A,
Berthiaume F, Hertl M, Nahmias Y, Yarmush ML, Uygun K.
Organ reengineering through development of a transplantable
recellularized liver graft using decellularized liver matrix. Nat
Med. 2010;16:814820.
10. Sabetkish S, Kajbafzadeh AM, Sabetkish N, Khorramirouz R,
Akbarzadeh A, Seyedian SL, Pasalar P, Orangian S, Beigi RS,
Aryan Z, Akbari H, Tavangar SM. Whole-organ tissue engineer-
ing: decellularization and recellularization of three-dimensional
matrix liver scaffolds. J Biomed Mater Res A. 2015;103:
14981508.
11. Wu Q, Bao J, Zhou YJ, Wang YJ, Du ZG, Shi YJ, Li L, Bu H.
Optimizing perfusion-decellularization methods of porcine livers
for clinical-scale whole-organ bioengineering. Biomed Res Int.
2015;2015:785474.
12. Navarro-Tableros V, Herrera Sanchez MB, Figliolini F,
Romagnoli R, Tetta C, Camussi G. Recellularization of rat liver
scaffolds by human liver stem cells. Tissue Eng Part A. 2015;
21:19291939.
13. Yagi H, Fukumitsu K, Fukuda K, Kitago M, Shinoda M,
Obara H, Itano O, Kawachi S, Tanabe M, Coudriet GM,
Piganelli JD, Gilbert TW, Soto-Gutierrez A, Kitagawa Y.
Human-scale whole-organ bioengineering for liver transplanta-
tion: a regenerative medicine approach. Cell Transplant. 2013;
22:231242.
14. Barakat O, Abbasi S, Rodriguez G, Rios J, Wood RP, Ozaki C,
Holley LS, Gauthier PK. Use of decellularized porcine liver for
engineering humanized liver organ. J Surg Res. 2012;173:
e11e25.
15. Kasimir MT, Rieder E, Seebacher G, Silberhumer G, Wolner E,
Weigel G, Simon P. Comparison of different decellularization
procedures of porcine heart valves. Int J Artif Organs. 2003;26:
421427.
16. Rieder E, Kasimir MT, Silberhumer G, Seebacher G, Wolner E,
Simon P, Weigel G. Decellularization protocols of porcine heart
valves differ importantly in efciency of cell removal and sus-
ceptibility of the matrix to recellularization with human vascular
cells. J Thorac Cardiovasc Surg. 2004;127:399405.
17. Kasimir MT, Weigel G, Sharma J, Rieder E, Seebacher G,
Wolner E, Simon P. The decellularized porcine heart valve
matrix in tissue engineering: platelet adhesion and activation.
Thromb Haemost. 2005;94:562567.
18. Ott HC, Matthiesen TS, Goh SK, Black LD, Kren SM,
Netoff TI, Taylor DA. Perfusion-decellularized matrix: using
natures platform to engineer a bioarticial heart. Nat Med.
2008;14:213221.
19. Ng SL, Narayanan K, Gao S, Wan AC. Lineage restricted pro-
genitors for the repopulation of decellularized heart. Biomate-
rials. 2011;32:75717580.
20. Akhyari P, Aubin H, Gwanmesia P, Barth M, Hoffmann S,
Huelsmann J, Preuss K, Lichtenberg A. The quest for an opti-
mized protocol for whole-heart decellularization: a comparison
of three popular and a novel decellularization technique and
their diverse effects on crucial extracellular matrix qualities. Tis-
sue Eng Part C Methods. 2011;17:915926.
21. Remlinger NT, Wearden PD, Gilbert TW. Procedure for decel-
lularization of porcine heart by retrograde coronary perfusion. J
Vis Exp. 2012;70:e50059.
Biotechnol. Prog., 20188
22. Wainwright JM, Czajka CA, Patel UB, Freytes DO, Tobita K,
Gilbert TW, Badylak SF. Preparation of cardiac extracellular
matrix from an intact porcine heart. Tissue Eng Part C Methods.
2010;16:525532.
23. Park KM, Woo HM. Porcine bioengineered scaffolds as new
frontiers in regenerative medicine. Transplant Proc. 2012;44:
11461150.
24. Kitahara H, Yagi H, Tajima K, Okamoto K, Yoshitake A,
Aeba R, Kudo M, Kashima I, Kawaguchi S, Hirano A, Kasai M,
Akamatsu Y, Oka H, Kitagawa Y, Shimizu H. Heterotopic trans-
plantation of a decellularized and recellularized whole porcine
heart. Interact Cardiovasc Thorac Surg. 2016;22:571579.
25. Guyette JP, Charest JM, Mills RW, Jank BJ, Moser PT,
Gilpin SE, Gershlak JR, Okamoto T, Gonzalez G, Milan DJ,
Gaudette GR, Ott HC. Bioengineering human myocardium on
native extracellular matrix. Circulation Research. 2016;118:
5672.
26. Ott HC, Clippinger B, Conrad C, Schuetz C, Pomerantseva I,
Ikonomou L, Kotton D, Vacanti JP. Regeneration and orthotopic
transplantation of a bioarticial lung. Nat Med. 2010;16:
927933.
27. Tsuchiya T, Mendez J, Calle EA, Hatachi G, Doi R, Zhao L,
Suematsu T, Nagayasu T, Niklason LE. Ventilation-based decel-
lularization system of the lung. Biores Open Access. 2016;5:
118126.
28. Cortiella J, Niles J, Cantu A, Brettler A, Pham A, Vargas G,
Winston S, Wang J, Walls S, Nichols JE. Inuence of acellular
natural lung matrix on murine embryonic stem cell differentia-
tion and tissue formation. Tissue Eng Part A. 2010;16:
25652580.
29. Wagner DE, Bonenfant NR, Parsons CS, Sokocevic D,
Brooks EM, Borg ZD, Lathrop MJ, Wallis JD, Daly AB,
Lam YW, Deng B, DeSarno MJ, Ashikaga T, Loi R, Weiss DJ.
Comparative decellularization and recellularization of normal
versus emphysematous human lungs. Biomaterials. 2014;35:
32813297.
30. ONeill JD, Anfang R, Anandappa A, Costa J, Javidfar J,
Wobma HM, Singh G, Freytes DO, Bacchetta MD, Sonett JR,
Vunjak-Novakovic G. Decellularization of human and porcine
lung tissues for pulmonary tissue engineering. Ann Thorac Surg.
2013;96:10461055. discussion 1055-1056.
31. Doi R, Tsuchiya T, Mitsutake N, Nishimura S,
Matsuu-Matsuyama M, Nakazawa Y, Ogi T, Akita S,
Yukawa H, Baba Y, Yamasaki N, Matsumoto K, Miyazaki T,
Kamohara R, Hatachi G, Sengyoku H, Watanabe H, Obata T,
Niklason LE, Nagayasu T. Transplantation of bioengineered rat
lungs recellularized with endothelial and adipose-derived stro-
mal cells. Sci Rep. 2017;7:8447.
32. Crabbe A, Liu Y, Sarker SF, Bonenfant NR, Barrila J, Borg ZD,
Lee JJ, Weiss DJ, Nickerson CA. Recellularization of decellular-
ized lung scaffolds is enhanced by dynamic suspension culture.
PLoS One. 2015;10:e0126846.
33. Gilpin SE, Ren X, Okamoto T, Guyette JP, Mou H,
Rajagopal J, Mathisen DJ, Vacanti JP, Ott HC. Enhanced lung
epithelial specication of human iPSCs on decellularized lung
matrix. Ann Thorac Surg. 2014;98:17211729.
34. Baptista PM, Orlando G, Mirmalek-Sani SH, Siddiqui M,
Atala A, Soker S. Whole organ decellularization - a tool for
bioscaffold fabrication and organ bioengineering. Conf Proc
IEEE Eng Med Biol Soc. 2009;2009:65266529.
35. Goh SK, Bertera S, Olsen P, Candiello JE, Halfter W, Uechi G,
Balasubramani M, Johnson SA, Sicari BM, Kollar E,
Badylak SF, Banerjee I. Perfusion-decellularized pancreas as a
natural 3D scaffold for pancreatic tissue and whole organ engi-
neering. Biomaterials. 2013;34:67606772.
36. Mirmalek-Sani SH, Orlando G, McQuilling JP, Pareta R,
Mack DL, Salvatori M, Farney AC, Stratta RJ, Atala A,
Opara EC, Soker S. Porcine pancreas extracellular matrix as a
platform for endocrine pancreas bioengineering. Biomaterials.
2013;34:54885495.
37. Wu D, Wan J, Huang Y, Guo Y, Xu T, Zhu M, Fan X, Zhu S,
Ling C, Li X, Lu J, Zhu H, Zhou P, Lu Y, Wang Z. 3D culture
of MIN-6 cells on decellularized pancreatic scaffold: in vitro
and in vivo study. Biomed Res Int. 2015;2015:432645.
38. Napierala H, Hillebrandt KH, Haep N, Tang P, Tintemann M,
Gassner J, Noesser M, Everwien H, Seiffert N, Kluge M,
Teegen E, Polenz D, Lippert S, Geisel D, Reutzel Selke A,
Raschzok N, Andreou A, Pratschke J, Sauer IM, Struecker B.
Engineering an endocrine neo-pancreas by repopulation of a
decellularized rat pancreas with islets of Langerhans. Sci Rep.
2017;7:41777.
39. Damodaran RG, Vermette P. Decellularized pancreas as a native
extracellular matrix scaffold for islets. J Tissue Eng Regen Med.
2018;12:12301237. https://doi.org/10.1002/term.2655.
40. Badylak SF, Lantz GC, Coffey A, Geddes LA. Small intestinal
submucosa as a large diameter vascular graft in the dog. J Surg
Res. 1989;47:7480.
41. Fiala R, Vidlar A, Vrtal R, Belej K, Student V. Porcine small
intestinal submucosa graft for repair of anterior urethral stric-
tures. Eur Urol. 2007;51:17021708. discussion 1708.
42. Oliveira AC, Garzon I, Ionescu AM, Carriel V, Cardona Jde L,
Gonzalez-Andrades M, Perez Mdel M, Alaminos M, Campos A.
Evaluation of small intestine grafts decellularization
methods for corneal tissue engineering. PLoS One. 2013;8:
e66538.
43. Syed O, Walters NJ, Day RM, Kim HW, Knowles JC. Evalua-
tion of decellularization protocols for production of tubular
small intestine submucosa scaffolds for use in oesophageal tis-
sue engineering. Acta Biomater. 2014;10:50435054.
44. Crapo PM, Wang Y. Small intestinal submucosa gel as a poten-
tial scaffolding material for cardiac tissue engineering. Acta Bio-
mater. 2010;6:20912096.
45. Zhang L, Du A, Li J, Pan M, Han W, Xiao Y. Development of
a cell-seeded modied small intestinal submucosa for urethro-
plasty. Heliyon. 2016;2:e00087.
46. Goulle F. Use of porcine small intestinal submucosa for corneal
reconstruction in dogs and cats: 106 cases. Journal of Small
Animal Practice. 2012;53:3443.
47. Wei RQ, Tan B, Tan MY, Luo JC, Deng L, Chen XH, Li XQ,
Zuo X, Zhi W, Yang P, Xie HQ, Yang ZM. Grafts of porcine
small intestinal submucosa with cultured autologous oral muco-
sal epithelial cells for esophageal repair in a canine model. Exp
Biol Med (Maywood). 2009;234:453461.
48. Chang CW, Petrie T, Clark A, Lin X, Sondergaard CS,
Grifths LG. Mesenchymal stem cell seeding of porcine small
intestinal submucosal extracellular matrix for cardiovascular
applications. PLoS One. 2016;11:e0153412.
49. Lanteri Parcells A, Abernathie B, Datiashvili R. The use of uri-
nary bladder matrix in the treatment of complicated open
wounds. Wounds. 2014;26:189196.
50. Tukmachev D, Forostyak S, Koci Z, Zaviskova K, Vackova I,
Vyborny K, Sandvig I, Sandvig A, Medberry CJ, Badylak SF,
Sykova E, Kubinova S. Injectable extracellular matrix hydrogels
as scaffolds for spinal cord injury repair. Tissue Eng Part A.
2016;22:306317.
51. Faust A, Kandakatla A, van der Merwe Y, Ren T, Huleihel L,
Hussey G, Naranjo JD, Johnson S, Badylak S, Steketee M. Uri-
nary bladder extracellular matrix hydrogels and matrix-bound
vesicles differentially regulate central nervous system neuron
viability and axon growth and branching. J Biomater Appl.
2017;31:12771295.
52. Li J, Wang W, An H, Wang F, Rexiati M, Wang Y. In vitro cul-
ture of rat hair follicle stem cells on rabbit bladder acellular
matrix. Springerplus. 2016;5:1461.
53. Remlinger NT, Gilbert TW, Yoshida M, Guest BN,
Hashizume R, Weaver ML, Wagner WR, Brown BN, Tobita K,
Wearden PD. Urinary bladder matrix promotes site appropriate
tissue formation following right ventricle outow tract repair.
Organogenesis. 2013;9:149160.
54. Merguerian PA, Reddy PP, Barrieras DJ, Wilson GJ,
Woodhouse K, Bagli DJ, McLorie GA, Khoury AE. Acellular
bladder matrix allografts in the regeneration of functional blad-
ders: evaluation of large-segment (> 24 cm2) substitution in a
porcine model. BJU International. 2000;85:894898.
55. Ross EA, Williams MJ, Hamazaki T, Terada N, Clapp WL,
Adin C, Ellison GW, Jorgensen M, Batich CD. Embryonic stem
cells proliferate and differentiate when seeded into kidney scaf-
folds. J Am Soc Nephrol. 2009;20:23382347.
Biotechnol. Prog., 2018 9
56. Song JJ, Guyette JP, Gilpin SE, Gonzalez G,
Vacanti JP, Ott HC. Regeneration and experimental orthotopic
transplantation of a bioengineered kidney. Nat Med. 2013;19:
646651.
57. Du C, Narayanan K, Leong MF, Ibrahim MS, Chua YP,
Khoo VM, Wan AC. Functional kidney bioengineering with
pluripotent stem-cell-derived renal progenitor cells and decellu-
larized kidney scaffolds. Adv Healthc Mater. 2016;5:
20802091.
58. Kheir E, Stapleton T, Shaw D, Jin Z, Fisher J, Ingham E. Devel-
opment and characterization of an acellular porcine cartilage
bone matrix for use in tissue engineering. J Biomed Mater
Res A. 2011;99A:283294.
59. Kang H, Peng J, Lu S, Liu S, Zhang L, Huang J, Sui X,
Zhao B, Wang A, Xu W, Luo Z, Guo Q. In vivo cartilage repair
using adipose-derived stem cell-loaded decellularized cartilage
ECM scaffolds. J Tissue Eng Regen Med. 2014;8:442453.
60. Gawlitta D, Benders KE, Visser J, van der Sar AS,
Kempen DH, Theyse LF, Malda J, Dhert WJ. Decellularized
cartilage-derived matrix as substrate for endochondral bone
regeneration. Tissue Eng Part A. 2015;21:694703.
61. Gershlak JR, Hernandez S, Fontana G, Perreault LR,
Hansen KJ, Larson SA, Binder BY, Dolivo DM, Yang T,
Dominko T, Rolle MW, Weathers PJ, Medina-Bolivar F,
Cramer CL, Murphy WL, Gaudette GR. Crossing kingdoms:
using decellularized plants as perfusable tissue engineering scaf-
folds. Biomaterials. 2017;125:1322.
62. Chang TT, Zhou VX, Rubinsky B. Using non-thermal irrevers-
ible electroporation to create an in vivo niche for exogenous cell
engraftment. Biotechniques. 2017;62:229231.
63. Shupe T, Williams M, Brown A, Willenberg B, Petersen BE.
Method for the decellularization of intact rat liver. Organogene-
sis. 2010;6:134136.
64. Struecker B, Butter A, Hillebrandt K, Polenz D,
Reutzel-Selke A, Tang P, Lippert S, Leder A, Rohn S, Geisel D,
Denecke T, Aliyev K, Johrens K, Raschzok N, Neuhaus P,
Pratschke J, Sauer IM. Improved rat liver decellularization by
arterial perfusion under oscillating pressure conditions. J Tissue
Eng Regen Med. 2017;11:531541.
65. Orlando G, Farney AC, Iskandar SS, Mirmalek-Sani SH,
Sullivan DC, Moran E, AbouShwareb T, De Coppi P,
Wood KJ, Stratta RJ, Atala A, Yoo JJ, Soker S. Production and
implantation of renal extracellular matrix scaffolds from porcine
kidneys as a platform for renal bioengineering investigations.
Ann Surg. 2012;256:363370.
66. Guyette JP, Gilpin SE, Charest JM, Tapias LF, Ren X, Ott HC.
Perfusion decellularization of whole organs. Nat Protoc. 2014;9:
14511468.
67. Chen WC, Wang Z, Missinato MA, Park DW, Long DW,
Liu HJ, Zeng X, Yates NA, Kim K, Wang Y. Decellularized
zebrash cardiac extracellular matrix induces mammalian heart
regeneration. Sci Adv. 2016;2:e1600844.
68. Roosens A, Somers P, De Somer F, Carriel V, Van Nooten G,
Cornelissen R. Impact of detergent-based decellularization
methods on porcine tissues for heart valve engineering. Ann
Biomed Eng. 2016;44:28272839.
69. Ye X, Wang H, Gong W, Li S, Li H, Wang Z, Zhao Q. Impact
of decellularization on porcine myocardium as scaffold for tis-
sue engineered heart tissue. J Mater Sci Mater Med. 2016;
27:70.
70. de Sousa Iwamoto LA, Duailibi MT, Iwamoto GY, Juliano Y,
Duailibi MS, Ossamu Tanaka FA, Duailibi SE. Tooth tissue
engineering: tooth decellularization for natural scaffold. Future
Sci OA. 2016;2:FSO121.
71. Lee DJ, Diachina S, Lee YT, Zhao L, Zou R, Tang N, Han H,
Chen X, Ko CC. Decellularized bone matrix grafts
for calvaria regeneration. J Tissue Eng. 2016;7:
2041731416680306.
72. Gao S, Yuan Z, Xi T, Wei X, Guo Q. Characterization of decel-
lularized scaffold derived from porcine meniscus for tissue engi-
neering applications. Frontiers of Materials Science. 2016;10:
101112.
73. Erten E, Sezgin Arslan T, Derkus B, Arslan YE. Detergent-free
decellularization of bovine costal cartilage for chondrogenic
differentiation of human adipose mesenchymal stem cells
in vitro. RSC Advances. 2016;6:9423694246.
74. Fu Y, Fan X, Tian C, Luo J, Zhang Y, Deng L, Qin T, Lv Q.
Decellularization of porcine skeletal muscle extracellular matrix
for the formulation of a matrix hydrogel: a preliminary study. J
Cell Mol Med. 2016;20:740749.
75. Vasudevan S, Huang J, Botterman B, Matloub HS, Keefer E,
Cheng J. Detergent-free decellularized nerve grafts for long-gap
peripheral nerve reconstruction. Plastic and Reconstructive Sur-
gery Global Open. 2014;2:e201.
76. Farrokhi A, Pakyari M, Nabai L, Pourghadiri A, Hartwell R,
Jalili R, Ghahary A. Evaluation of detergent-free and
detergent-based methods for decellularization of murine skin.
Tissue Eng Part A. 2018;24:955967. https://doi.org/10.1089/
ten.TEA.2017.0273.
77. Hung SH, Su CH, Lin SE, Tseng H. Preliminary experiences in
trachea scaffold tissue engineering with segmental organ decel-
lularization. Laryngoscope. 2016;126:25202527.
78. Xiang J, Zheng X, Liu P, Yang L, Dong D, Wu W, Liu X, Li J,
Lv Y. Decellularized spleen matrix for reengineering functional
hepatic-like tissue based on bone marrow mesenchymal stem
cells. Organogenesis. 2016;12:128142.
79. Gao R, Wu W, Xiang J, Lv Y, Zheng X, Chen Q, Wang H,
Wang B, Liu Z, Ma F. Hepatocyte culture in autologous decel-
lularized spleen matrix. Organogenesis. 2015;11:1629.
80. Hasan A. Tissue Engineering for Articial Organs: Regenera-
tive Medicine, Smart Diagnostics and Personalized Medicine.
Doha: Wiley; 2017.
81. Gilpin A, Yang Y. Decellularization strategies for regenerative
medicine: from processing techniques to applications. BioMed
Research International. 2017;2017:9831534.
82. Koley D, Bard AJ. Triton X-100 concentration effects on mem-
brane permeability of a single HeLa cell by scanning electro-
chemical microscopy (SECM).Proc Natl Acad Sci U S A. 2010;
107:1678316787.
83. Kalipatnapu S, Chattopadhyay A. Membrane protein solubiliza-
tion: recent advances and challenges in solubilization of seroto-
nin1A receptors. IUBMB Life. 2005;57:505512.
84. Jamur MC, Oliver C. Permeabilization of cell membranes.
Methods Mol Biol. 2010;588:6366.
85. Ji H. Lysis of cultured cells for immunoprecipitation. Cold
Spring Harb Protoc. 2010;8:pdb prot5466.
86. Vavken P, Joshi S, Murray MM. TRITON-X is most effective
among three decellularization agents for ACL tissue engineer-
ing. J Orthop Res. 2009;27:16121618.
87. Xu H, Xu B, Yang Q, Li X, Ma X, Xia Q, Zhang Y, Zhang C,
Wu Y, Zhang Y. Comparison of decellularization protocols for
preparing a decellularized porcine annulus brosus scaffold.
PLoS One. 2014;9:e86723.
88. Meyer SR, Chiu B, Churchill TA, Zhu L, Lakey JR, Ross DB.
Comparison of aortic valve allograft decellularization techniques
in the rat. J Biomed Mater Res A. 2006;79:254262.
89. Ren H, Shi X, Tao L, Xiao J, Han B, Zhang Y, Yuan X,
Ding Y. Evaluation of two decellularization methods in the
development of a whole-organ decellularized rat liver scaffold.
Liver Int. 2013;33:448458.
90. Bertanha M, Moroz A, Jaldin RG, Silva RAM, Rinaldi JC,
Golim MA, Felisbino SL, Domingues MAC, Sobreira ML,
Reis PP, Deffune E. Morphofunctional characterization of decel-
lularized vena cava as tissue engineering scaffolds. Exp Cell
Res. 2014;326:103111.
91. González-Andrades M, Carriel V, Rivera-Izquierdo M,
Garzón I, González-Andrades E, Medialdea S, Alaminos M,
Campos A. Effects of detergent-based protocols on decellulari-
zation of corneas with sclerocorneal limbus. Evaluation of
regional differences. Transl Vis Sci Technol. 2015;4:13.
92. Simoes IN, Vale P, Soker S, Atala A, Keller D, Noiva R,
Carvalho S, Peleteiro C, Cabral JM, Eberli D, da Silva CL,
Baptista PM. Acellular urethra bioscaffold: decellularization of
whole urethras for tissue engineering applications. Sci Rep.
2017;7:41934.
93. Nakayama KH, Batchelder CA, Lee CI, Tarantal AF. Decellu-
larized rhesus monkey kidney as a three-dimensional scaffold
Biotechnol. Prog., 201810
for renal tissue engineering. Tissue Eng Part A. 2010;16:
22072216.
94. Bhuyan AK. On the mechanism of SDS-induced protein dena-
turation. Biopolymers. 2010;93:186199.
95. Lichtenberg D, Ahyayauch H, Goni FM. The mechanism of
detergent solubilization of lipid bilayers. Biophys J. 2013;105:
289299.
96. Helder MRK, Stoyles NJ, Tefft BJ, Hennessy RS,
Hennessy RRC, Dyer R, Witt T, Simari RD, Lerman A.
Xenoantigenicity of porcine decellularized valves. J Cardi-
othorac Surg. 2017;12:56.
97. Xu K, Kuntz LA, Foehr P, Kuempel K, Wagner A, Tuebel J,
Deimling CV, Burgkart RH. Efcient decellularization for tissue
engineering of the tendon-bone interface with preservation of
biomechanics. PLoS One. 2017;12:e0171577.
98. Caralt M, Uzarski JS, Iacob S, Obergfell KP, Berg N,
Bijonowski BM, Kiefer KM, Ward HH, Wandinger-Ness A,
Miller WM, Zhang ZJ, Abecassis MM, Wertheim JA. Optimiza-
tion and critical evaluation of decellularization strategies to
develop renal extracellular matrix scaffolds as biological tem-
plates for organ engineering and transplantation.
Am J Transplant. 2015;15:6475.
99. Iglic A. Advances in Planar Lipd Bilayers and Liposomes, Vol
14. Oxford: Academic Press; 2011.
100. Hellstrom M, El-Akouri RR, Sihlbom C, Olsson BM,
Lengqvist J, Backdahl H, Johansson BR, Olausson M,
Sumitran-Holgersson S, Brannstrom M. Towards the develop-
ment of a bioengineered uterus: comparison of different proto-
cols for rat uterus decellularization. Acta Biomater. 2014;10:
50345042.
101. Rodi PM, Bocco Gianello MD, Corregido MC, Gennaro AM.
Comparative study of the interaction of CHAPS and Triton
X-100 with the erythrocyte membrane. Biochim Biophys Acta.
1838;2014:859866.
102. Petersen TH, Calle EA, Colehour MB, Niklason LE. Matrix
composition and mechanics of decellularized lung scaffolds.
Cells Tissues Organs. 2012;195:222231.
103. Crapo PM, Gilbert TW, Badylak SF. An overview of tissue and
whole organ decellularization processes. Biomaterials. 2011;32:
32333243.
104. Dong X, Wei X, Yi W, Gu C, Kang X, Liu Y, Li Q, Yi D.
RGD-modied acellular bovine pericardium as a bioprosthetic
scaffold for tissue engineering. J Mater Sci Mater Med. 2009;
20:23272336.
105. Wang Y, Bao J, Wu Q, Zhou Y, Li Y, Wu X, Shi Y, Li L,
Bu H. Method for perfusion decellularization of porcine whole
liver and kidney for use as a scaffold for clinical-scale bioengi-
neering engrafts. Xenotransplantation. 2015;22:4861.
106. Lu WD, Zhang L, Wu CL, Liu ZG, Lei GY, Liu J, Gao W,
Hu YR. Development of an acellular tumor extracellular matrix
as a three-dimensional scaffold for tumor engineering. PLoS
One. 2014;9:e103672.
107. Morris AH, Chang J, Kyriakides TR. Inadequate processing of
decellularized dermal matrix reduces cell viability in vitro and
increases apoptosis and acute inammation in vivo. BioResearch
Open Access. 2016;5:177187.
108. Albanna MZ, Holmes JH. Skin Tissue Engineering and Regen-
erative Medicine. Oxford: Academic Press; 2016.
109. Lumpkins SB, Pierre N, McFetridge PS. A mechanical evalua-
tion of three decellularization methods in the design of a xeno-
geneic scaffold for tissue engineering the temporomandibular
joint disc. Acta Biomater. 2008;4:808816.
110. Sayk F, Bos I, Schubert U, Wedel T, Sievers HH. Histopatho-
logic ndings in a novel decellularized pulmonary homograft:
an autopsy study. Ann Thorac Surg. 2005;79:17551758.
111. Dubiel EA, Martin Y, Vermette P. Bridging the gap between
physicochemistry and interpretation prevalent in cell-surface
interactions. Chem Rev. 2011;111:29002936.
112. Bonnans C, Chou J, Werb Z. Remodelling the extracellular
matrix in development and disease. Nat Rev Mol Cell Biol.
2014;15:786801.
113. Yao L, Phan F, Li Y. Collagen microsphere serving as a cell
carrier supports oligodendrocyte progenitor cell growth and
differentiation for neurite myelination in vitro. Stem Cell Res
Ther. 2013;4:109.
114. Leszczak V, Baskett DA, Popat KC. Endothelial cell growth
and differentiation on collagen-immobilized polycaprolactone
nanowire surfaces. J Biomed Nanotechnol. 2015;11:10801092.
115. Tate CC, Shear DA, Tate MC, Archer DR, Stein DG,
LaPlaca MC. Laminin and bronectin scaffolds enhance neural
stem cell transplantation into the injured brain. J Tissue Eng
Regen Med. 2009;3:208217.
116. Weber LM, Hayda KN, Anseth KS. Cell-matrix interactions
improve β-cell survival and insulin secretion in
three-dimensional culture. Tissue Eng Part A. 2008;14:
19591968.
117. Bramfeldt H, Vermette P. Enhanced smooth muscle cell adhesion
and proliferation on protein-modied polycaprolactone-based
copolymers. J Biomed Mater Res A. 2009;88:520530.
118. Dubiel EA, Lakey JR, T, Lamb MW, Vermette P. Culturing
free-oating and brin-embedded islets with endothelial cells:
effects on insulin secretion and apoptosis. Cel. Mol. Bioeng.
2014;7:243.
119. Kuehn C, Lakey JRT, Lamb MW, Vermette P. Young porcine
endocrine pancreatic islets cultured in brin show improved
resistance toward hydrogen peroxide. Islets. 2013;5:207215.
120. Kuehn C, Dubiel EA, Sabra G, Vermette P. Culturing INS-1
cells on CDPGYIGSR-, RGD- and bronectin surfaces
improves insulin secretion and cell proliferation. Acta Biomater.
2012;8:619626.
121. Dubiel EA, Kuehn C, Wang R, Vermette P. In vitro morphogen-
esis of PANC-1 cells into islet-like aggregates using
RGD-covered dextran derivative surfaces. Colloids Surf B
Biointerfaces. 2012;89:117125.
122. Andersen PL, Vermette P. Biomimetic surfaces supporting dis-
sociated pancreatic islet cultures. Colloids Surf B Biointerfaces.
2017;159:166173.
123. Monchaux E, Vermette P. Bioactive microarrays immobilized
on low-fouling surfaces to study specic endothelial cell adhe-
sion. Biomacromolecules. 2007;8:36683673.
124. Cooke MJ, Phillips SR, Shah DS, Athey D, Lakey JH,
Przyborski SA. Enhanced cell attachment using a novel cell cul-
ture surface presenting functional domains from extracellular
matrix proteins. Cytotechnology. 2008;56:7179.
125. Nguyen H, Qian JJ, Bhatnagar RS, Li S. Enhanced cell attach-
ment and osteoblastic activity by P-15 peptide-coated matrix in
hydrogels. Biochem Biophys Res Commun. 2003;311:179186.
126. Antonova LV, Seifalian AM, Kutikhin AG, Sevostyanova VV,
Matveeva VG, Velikanova EA, Mironov AV, Shabaev AR,
Glushkova TV, Senokosova EA, Vasyukov GY, Krivkina EO,
Burago AY, Kudryavtseva YA, Barbarash OL, Barbarash LS.
Conjugation with RGD peptides and incorporation of vascular
endothelial growth factor are equally efcient for biofunctionali-
zation of tissue-engineered vascular grafts. Int J Mol Sci. 2016;
17:1920.
127. Dumbleton J, Agarwal P, Huang H, Hogrebe N, Han R,
Gooch KJ, He X. The effect of RGD peptide on 2D and minia-
turized 3D culture of HEPM cells, MSCs, and ADSCs with algi-
nate hydrogel. Cell Mol Bioeng. 2016;9:277288.
128. Hunt NC, Hallam D, Karimi A, Mellough CB, Chen J,
Steel DH, Lako M. 3D culture of human pluripotent stem cells
in RGD-alginate hydrogel improves retinal tissue development.
Acta Biomater. 2017;49:329343.
129. Park SH, Zheng JH, Nguyen VH, Jiang SN, Kim DY,
Szardenings M, Min JH, Hong Y, Choy HE, Min JJ. RGD pep-
tide cell-surface display enhances the targeting and therapeutic
efcacy of attenuated salmonella-mediated cancer therapy. Ther-
anostics. 2016;6:16721682.
130. Sobers CJ, Wood SE, Mrksich M. A gene expression-based
comparison of cell adhesion to extracellular matrix and
RGD-terminated monolayers. Biomaterials. 2015;52:385394.
131. Gerecht S, Burdick JA, Ferreira LS, Townsend SA, Langer R,
Vunjak-Novakovic G. Hyaluronic acid hydrogel for controlled
self-renewal and differentiation of human embryonic stem cells.
Proc Natl Acad Sci U S A. 2007;104:1129811303.
132. Schizas N, Rojas R, Kootala S, Andersson B, Pettersson J,
Hilborn J, Hailer NP. Hyaluronic acid-based hydrogel enhances
Biotechnol. Prog., 2018 11
neuronal survival in spinal cord slice cultures from postnatal
mice. J Biomater Appl. 2014;28:825836.
133. Wight TN, Kinsella MG, Qwarnstrom EE. The role of proteo-
glycans in cell adhesion, migration and proliferation. Curr Opin
Cell Biol. 1992;4:793801.
134. von der Mark K, Park J, Bauer S, Schmuki P. Nanoscale engi-
neering of biomimetic surfaces: cues from the extracellular
matrix. Cell Tissue Res. 2010;339:131153.
135. Higuchi A, Ling QD, Hsu ST, Umezawa A. Biomimetic cell
culture proteins as extracellular matrices for stem cell differenti-
ation. Chem Rev. 2012;112:45074540.
136. Gilbert TW, Stewart-Akers AM, Simmons-Byrd A, Badylak SF.
Degradation and remodeling of small intestinal submucosa in
canine Achilles tendon repair. J Bone Joint Surg Am. 2007;89:
621630.
137. Beattie AJ, Gilbert TW, Guyot JP, Yates AJ, Badylak SF. Che-
moattraction of progenitor cells by remodeling extracellular
matrix scaffolds. Tissue Eng Part A. 2009;15:11191125.
138. Brennan EP, Tang XH, Stewart-Akers AM, Gudas LJ,
Badylak SF. Chemoattractant activity of degradation products of
fetal and adult skin extracellular matrix for keratinocyte progeni-
tor cells. J Tissue Eng Regen Med. 2008;2:491498.
139. Reing JE, Zhang L, Myers-Irvin J, Cordero KE, Freytes DO,
Heber-Katz E, Bedelbaeva K, McIntosh D, Dewilde A,
Braunhut SJ, Badylak SF. Degradation products of extracellular
matrix affect cell migration and proliferation. Tissue Eng
Part A. 2009;15:605614.
140. Tran KT, Lamb P, Deng JS. Matrikines and matricryptins:
implications for cutaneous cancers and skin repair. J Dermatol
Sci. 2005;40:1120.
141. Mahfouz W, Elsalmy S, Corcos J, Fayed AS. Fundamentals of
bladder tissue engineering. African Journal of Urology. 2013;
19:5157.
142. Wong ML, Grifths LG. Immunogenicity in xenogeneic scaf-
fold generation: antigen removal versus decellularization. Acta
Biomater. 2014;10:18061816.
143. Vadori M, Cozzi E. The immunological barriers to xenotrans-
plantation. Tissue Antigens. 2015;86:239253.
144. Chen G, Qian H, Starzl T, Sun H, Garcia B, Wang X, Wise Y,
Liu Y, Xiang Y, Copeman L, Liu W, Jevnikar A, Wall W,
Cooper DK, Murase N, Dai Y, Wang W, Xiong Y, White DJ,
Zhong R. Acute rejection is associated with antibodies to
non-Gal antigens in baboons using Gal-knockout pig kidneys.
Nat Med. 2005;11:12951298.
145. Campbell PM, Salam A, Ryan EA, Senior P, Paty BW,
Bigam D, McCready T, Halpin A, Imes S, Al Saif F, Lakey JR,
Shapiro AM. Pretransplant HLA antibodies are associated with
reduced graft survival after clinical islet transplantation.
Am J Transplant. 2007;7:12421248.
146. Zheng MH, Chen J, Kirilak Y, Willers C, Xu J, Wood D. Por-
cine small intestine submucosa (SIS) is not an acellular collage-
nous matrix and contains porcine DNA: possible implications in
human implantation. J Biomed Mater Res B Appl Biomater.
2005;73:6167.
147. Shim G, Kim MG, Park JY, Oh YK. 11 - Small interfering
RNAs (siRNAs) as cancer therapeutics. Biomaterials for Cancer
Therapeutics. Cambridge: Woodhead Publishing Limited; 2013:
237269.
148. Valentin JE, Badylak JS, McCabe GP, Badylak SF. Extracellu-
lar matrix bioscaffolds for orthopaedic applications. A compara-
tive histologic study. J Bone Joint Surg Am. 2006;88:
26732686.
149. Hawkins JA, Hillman ND, Lambert LM, Jones J, Di Russo GB,
Profaizer T, Fuller TC, Minich LL, Williams RV,
Shaddy RE. Immunogenicity of decellularized cryopreserved
allografts in pediatric cardiac surgery: comparison with standard
cryopreserved allografts. J Thorac Cardiovasc Surg. 2003;126:
247252. discussion 252-253.
150. Konstantinovic ML, Lagae P, Zheng F, Verbeken EK, De
Ridder D, Deprest JA. Comparison of host response to polypropyl-
ene and non-cross-linked porcine small intestine serosal-derived
collagen implants in a rat model. Bjog. 2005;112:15541560.
151. Prevel CD, Eppley BL, Summerlin DJ, Jackson JR, McCarty M,
Badylak SF. Small intestinal submucosa: utilization for repair of
rodent abdominal wall defects. Ann Plast Surg. 1995;35:374380.
152. Wang D, Ding X, Xue W, Zheng J, Tian X, Li Y, Wang X,
Song H, Liu H, Luo X. A new scaffold containing small intesti-
nal submucosa and mesenchymal stem cells improves pancreatic
islet function and survival in vitro and in vivo. Int J Mol Med.
2017;39:167173.
153. Keane TJ, Badylak SF. The host response to allogeneic and
xenogeneic biological scaffold materials. J Tissue Eng Regen
Med. 2015;9:504511.
154. Aamodt JM, Grainger DW. Extracellular matrix-based biomate-
rial scaffolds and the host response. Biomaterials. 2016;86:
6882.
155. Sandrin MS, McKenzie IF. Gal alpha (1,3)Gal, the major
xenoantigen(s) recognised in pigs by human natural antibodies.
Immunol Rev. 1994;141:169190.
156. Lu TY, Lin B, Kim J, Sullivan M, Tobita K, Salama G,
Yang L. Repopulation of decellularized mouse heart with human
induced pluripotent stem cell-derived cardiovascular progenitor
cells. Nat Commun. 2013;4:2307.
157. Stabler CT, Caires LC, Mondrinos MJ, Marcinkiewicz C,
Lazarovici P, Wolfson MR, Lelkes PI. Enhanced
re-endothelialization of decellularized rat lungs. Tissue Eng Part
C Methods. 2016;22:439450.
158. Lichtenberg A, Tudorache I, Cebotari S, Ringes-Lichtenberg S,
Sturz G, Hoefer K, Hurscheler C, Brandes G, Hilker A,
Haverich A. In vitro re-endothelialization of detergent decellu-
larized heart valves under simulated physiological dynamic con-
ditions. Biomaterials. 2006;27:42214229.
159. Badylak SF. Xenogeneic extracellular matrix as a scaffold for
tissue reconstruction. Transplant Immunology. 2004;12:
367377.
160. Dickinson LE, Gerecht S. Engineered biopolymeric scaffolds
for chronic wound healing. Frontiers in Physiology. 2016;7:341.
161. Ren-Ke Li RDW. Cardiac Regeneration and Repair: Biomate-
rials and Tissue Engineering. Cambridge: Woodhead Publishing
Limited; 2014.
162. U.S. Food and Drug Administration, (2017). 510(k) Clearances.
https://www.fda.gov/MedicalDevices/
ProductsandMedicalProcedures/
DeviceApprovalsandClearances/510kClearances/default.htm
(August 8),
163. U.S. Food and Drug Administration, (2017). Classication of
510 (k) medical devices. https://www.fda.gov/MedicalDevices/
DeviceRegulationandGuidance/Overview/ClassifyYourDevice
(15th September August).
164. U.S. Food and Drug Administration, (2017). FDA Regulation of
Human Cells, Tissues, and Cellular and Tissue-Based Products
(HCT/Ps) Product List 2017. https://www.fda.gov/
biologicsbloodvaccines/tissuetissueproducts/regulationoftissues/
ucm150485.htm.
165. U.S. Food and Drug Administration, (2017). Evaluating Sub-
stantial Equivalence in Premarket Notications [510(k)] https://
www.fda.gov/downloads/MedicalDevices/.../UCM284443.pdf.
166. Snyder DL, Sullivan N, Schoelles KM. Skin Substitutes for
Treating Chronic Wounds. Agency for Health Care Research
and Quality, Rockville (MD); 2012.
Manuscript received Jan. 25, 2018, revision received Apr. 30, 2018.
Biotechnol. Prog., 201812
... Decellularization is the meticulous process of eliminating cellular and genetic elements from tissues or organs, yielding a matrix enriched with proteins, microfibrils, growth factors, and glycosaminoglycans (GAGs). The primary objective of this method is to maintain the original 3D structure and mechanical properties, achieved through the use of diverse agents [10,11]. These biomaterials are essential in tissue engineering due to their biocompatibility and preservation of vascular structures, providing vital cues for cell recruitment and tissue repair [12,13]. ...
... Detergents are soluble amphipathic molecules usually with a phospholipidic head with hydrophilic properties and a non-polar hydrophobic chain capable of interacting with the lipid bilayer membrane [42]. Detergents induce cell membrane solubilization, contributing to the process of cell removal from the ECM while maintaining structural, biochemical, and biomechanical properties [2,11,41,43]. Moreover, the use of detergents has shown to contribute to DNA and protein dissociation, improving their effectiveness as decellularization agents [43]. ...
... Acidic and alkaline solutions play a crucial role in tissue decellularization due to the pH sensitivity of intracellular biochemical processes. These solutions facilitate decellularization by inducing the hydrolysis of cytoplasmic components and degrading or denaturing nucleic acids [11,47]. It is vital to carefully control the concentration and exposure time of these solutions to prevent toxic effects on the tissue and preserve essential elements like GAGs and mechanical cues [40]. ...
Article
Full-text available
There is a persistent an urgent need to solve tissue and organ donor shortage issues. Decellularized tissues represent a promising alternative to other biologic and synthetic treatments that have been developed, since they aim to retain native tissue characteristics that would help in the regenerative processes such as prolifera-tion, cellular adhesion, and the presence of growth factors, while minimizing the chances of an unwanted host immune response. In the present review, we describe the most common methodologies for decellularization pro-cesses, as well as the clinical applications of these biomaterials.
... Being free of cells and, consequently, also free of genetic materials, decellularized xenografts have low immunogenicity and, therefore, are expected to be tolerated by the recipient's immune system [12]. Despite decellularization, when the ECM ultrastructure remains intact, it resembles the anatomical and physiological characteristics of native tissue [13]. Therefore, decellularized xenografts are reassuring substitutions for valve replacement with storage potential and, consequently, commercial availability [14]. ...
Article
Full-text available
One of the most important medical interventions for individuals with heart valvular disease is heart valve replacement, which is not without substantial challenges, particularly for pediatric patients. Due to their biological properties and biocompatibility, natural tissue-originated scaffolds derived from human or animal sources are one type of scaffold that is widely used in tissue engineering. However, they are known for their high potential for immunogenicity. Being free of cells and genetic material, decellularized xenografts, consequently, have low immunogenicity and, thus, are expected to be tolerated by the recipient’s immune system. The scaffold ultrastructure and ECM composition can be affected by cell removal agents. Therefore, applying an appropriate method that preserves intact the structure of the ECM plays a critical role in the final result. So far, there has not been an effective decellularization technique that preserves the integrity of the heart valve’s ultrastructure while securing the least amount of genetic material left. This study demonstrates a new protocol with untraceable cells and residual DNA, thereby maximally reducing any chance of immunogenicity. The mechanical and biochemical properties of the ECM resemble those of native heart valves. Results from this study strongly indicate that different critical factors, such as ionic detergent omission, the substitution of Triton X-100 with Tergitol, and using a lower concentration of trypsin and a higher concentration of DNase and RNase, play a significant role in maintaining intact the ultrastructure and function of the ECM.
... These scaffolds may be natural or synthetic; however, the natural ones are reported to be more biocompatible [8]. One common strategy to generate natural scaffolds is through the decellularization of xenogeneic or allogeneic organ sources [9,10]. The ultimate goal of whole liver decellularization is to thoroughly remove the cellular materials with minimized damage to extracellular matrix (ECM) ultrastructure and constituents [11,12]. ...
Article
As the only reliable treatment option for end-stage liver diseases, conventional liver transplantation confronts major supply limitations. Accordingly, the decellularization of discarded livers to produce bioscaffolds that support recellularization with progenitor/stem cells has emerged as a promising translational medicine approach. The success of this approach will substantially be determined by the extent of extracellular matrix (ECM) preservation during the decellularization process. Here, we assumed that the matrix metalloproteinase (MMP) inhibition could reduce the ECM damage during the whole liver decellularization of an animal model using a perfusion-based system. We demonstrated that the application of doxycycline as an MMP inhibitor led to significantly higher preservation of collagen, glycosaminoglycans, and hepatic growth factor (HGF) contents, as well as mechanical and structural features, including tensile strength, fiber integrity, and porosity. Notably, produced bioscaffolds were biocompatible and efficiently supported cell viability and proliferation in vitro. We also indicated that produced bioscaffolds efficiently supported HepG2 cell function upon seeding onto liver ECM discs using albumin and urea assay. Additionally, MMP inhibitor pretreated decellularized livers were more durable in contact with collagenase digestion compared to control bioscaffolds in vitro. Using zymography, we confirmed the underlying mechanism that results in these promising effects is through the inhibition of MMP2 and MMP9. Overall, we demonstrated a novel method based on MMP inhibition to ameliorate the ECM structure and composition preservation during liver decellularization as a critical step in fabricating transplantable bioengineered livers.
... To investigate age-associated changes to the ovarian proteome, we isolated ovaries from reproductively young (6-12 weeks) and old mice (10-12 months) and analyzed them both in their native state and enriched for the ECM to specifically determine how aging alters the ovarian matrix ( Figure 1). We, therefore, first optimized an ECM enrichment strategy using a decellularization approach, which removes cellular and nuclear material while preserving the composition and structure of the ECM [30,31]. Decellularized scaffolds are often used for bioengineering applications, as they can be reseeded with allogenic cells, which has the benefit of preventing an immune response while maintaining tissue-specific cell phenotypes and functions [32,33]. ...
Article
The ovarian microenvironment becomes fibrotic and stiff with age, in part due to increased collagen and decreased hyaluronan. However, the extracellular matrix (ECM) is a complex network of hundreds of proteins, glycoproteins, and glycans which are highly tissue specific and undergo pronounced changes with age. To obtain an unbiased and comprehensive profile of age-associated alterations to the murine ovarian proteome and ECM, we used a label-free quantitative proteomic methodology. We validated conditions to enrich for the ECM prior to proteomic analysis. Following analysis by data-independent acquisition (DIA) and quantitative data processing, we observed that both native and ECM-enriched ovaries clustered separately based on age, indicating distinct age-dependent proteomic signatures. We identified a total of 4,721 proteins from both native and ECM-enriched ovaries, of which 383 proteins were significantly altered with advanced age, including 58 ECM proteins. Several ECM proteins upregulated with age have been associated with fibrosis in other organs, but to date their roles in ovarian fibrosis are unknown. Pathways regulating DNA metabolism and translation were downregulated with age, whereas pathways involved in ECM remodeling and immune response were upregulated. Interestingly, immune-related pathways were upregulated with age even in ECM-enriched ovaries, suggesting a novel interplay between the ECM and the immune system. Moreover, we identified putative markers of unique immune cell populations present in the ovary with age. These findings provide evidence from a proteomic perspective that the aging ovary provides a fibroinflammatory milieu, and our study suggests target proteins which may drive these age-associated phenotypes for future investigation.
Article
Full-text available
Tissue-engineered organs, based on native extracellular matrix (ECM) scaffolds, could be a game changer in regenerative medicine applications. Decellularization technology provides such scaffolds with organ-typic ECM composition and architecture. Despite limitations such as the requirement of huge cell numbers and finding the optimal route of entry, recellularized scaffolds provide alternative grafts for transplantation. In this study we assessed whether decellularized scaffolds, when implanted, are repopulated from the adjacent tissue. Since the vasculature plays an important role in tissue functionality, our main focus was to evaluate in situ repopulation of decellularized veins in a pig model. For this, porcine inferior vena cava grafts were decellularized and orthotopically implanted in recipient pigs (n = 12). To evaluate possible immune responses to the scaffolds and to assess potential thrombus formation, cellular allogeneic vena cava grafts were transplanted in control pigs (n = 8). Within 28 days after implantation, the decellularized veins were fully recellularized with endothelial cells and smooth muscle cells. Quantitative histological analysis showed a comparable amount of smooth muscle actin in the repopulated decellularized grafts similar to the native IVC. Lymphocyte infiltrates representing signs of graft rejection were not detected in the pigs, as opposed to the control group that received the allogeneic grafts. The decellularized grafts provoked a higher incidence of thrombosis in comparison with allogeneic grafts (33.3 vs. 12.5%). With this study, we show efficient in situ repopulation of decellularized vein grafts. These findings are insightful and promising to further explore the use of decellularized tissue without the need for full pre-transplant recellularization. Graphical abstract
Article
The development of methods for culturing cells in three-dimensional systems is an urgent focus of modern cell biology. When cultured in the 3D system, a tissue-specific architecture is reproduced and the real microenvironment and cell behavior in vivo are more precisely recreated. Human mesenchymal stem/stromal cells (MSCs) are typically isolated and cultured as a monolayer 2D culture. In this work, we developed a method for three-dimensional cultivation and tissue-specific decidual differentiation of MSCs isolated from human endometrial tissue using a matrix derived from decellularized apple. Decellularized apple matrices have sufficient mechanical strength, are biocompatible, accessible, easy to use, and have ample scope for surface modification. This cell culture system is suitable for both confocal microscopy and flow cytometry studies. The model we developed can become the basis for the creation of new cell products and tissue-engineering structures in the field of regenerative biomedicine.
Preprint
Full-text available
Tissue-engineered organs, based on native extracellular matrix (ECM) scaffolds, could be a game changer in regenerative medicine applications. Decellularization technology provides such scaffolds with organ-typic ECM composition and architecture. Despite limitations such as the requirement of huge cell numbers and finding the optimal route of entry, recellularized scaffolds provide alternative grafts for transplantation. In this study we assessed whether decellularized scaffolds, when implanted, are repopulated from the adjacent tissue. Since the vasculature plays an important role in tissue functionality, our main focus was to evaluate in situ repopulation of decellularized veins in a pig model. For this, porcine inferior vena cava grafts were decellularized and orthotopically implanted in recipient pigs (n=12). To evaluate possible immune responses to the scaffolds and to assess potential thrombus formation, cellular allogeneic vena cava grafts were transplanted in control pigs (n=8). Within 28 days after implantation, the decellularized veins were fully recellularized with endothelial cells and smooth muscle cells. Quantitative histological analysis showed a comparable amount of smooth muscle actin in the repopulated decellularized grafts similar to the native IVC. Lymphocyte infiltrates representing signs of graft rejection were not detected in the pigs, as opposed to the control group that received the allogeneic grafts. The decellularized grafts provoked a higher incidence of thrombosis in comparison with allogeneic grafts (33.3 vs. 12.5%). With this study, we show efficient in situ repopulation of decellularized vein grafts. These findings are insightful and promising to further explore the use of decellularized tissue without the need for full pre-transplant recellularization.
Article
Hydrogels are insoluble three-dimensional polymer networks that are water insoluble but can absorb large amounts of water or biological fluid. They can be prepared using a variety of polymers, including both synthetic and natural ones. Either physical crosslinking, chemical crosslinking, or both are used for the preparation of polymeric hydrogels. Hydrogels made from natural polymers like chitosan, gelatin, alginate, cellulose, and hyaluronic acid are promising biomaterials for applications in tissue engineering, cosmetics, agriculture, wound healing, etc. because of their high biocompatibility and biodegradability. The present work reviews the classification of natural polymer-based hydrogels, properties, cross-linking methods, characterization, and their biomedical applications.
Article
Full-text available
Diabetes Mellitus involves the loss of function and/or absolute numbers of insulin-producing β-cells in pancreatic islets. Islet transplantation is currently being investigated as a potential cure, and advances in tissue engineering methods can be used to improve pancreatic islets survival and functionality. Transplanted islets experience anoikis, hypoxia and inflammation-mediated immune response, leading to early damage and subsequent failure of the graft. Recent development in tissue engineering enables the use of decellularized organs as scaffolds for cell therapies. Decellularized pancreas could be a suitable scaffold as it can retain the native extracellular matrix (ECM) and vasculature. In this study, mouse pancreata were decellularized by perfusion using 0.5% sodium dodecyl sulphate (SDS). Different characterizations revealed that the resulting matrix was free of cells and retained part of the pancreas ECM including the vasculature and its internal elastic basal lamina, the ducts with their basal membrane as well as the glycosaminoglycan and collagen structures. Islets were infused into the ductal system of decellularized pancreata and glucose-stimulated insulin secretion (GSIS) results confirmed their functionality after 48h. Also, re-cellularizing the decellularized pancreas with GFP-tagged INS-1 cells and culturing the system over 120 days confirmed the biocompatibility and non-toxic nature of the scaffold. GFP-tagged INS-1 cells formed pseudo-islets which were, over time, budding out of the decellularized pancreata. Decellularized pancreatic scaffolds seeded with endocrine pancreatic tissue could be a potential bioengineered organ for transplantation.
Article
Full-text available
Bioengineered lungs consisting of a decellularized lung scaffold that is repopulated with a patient's own cells could provide desperately needed donor organs in the future. This approach has been tested in rats, and has been partially explored in porcine and human lungs. However, existing bioengineered lungs are fragile, in part because of their immature vascular structure. Herein, we report the application of adipose-derived stem/stromal cells (ASCs) for engineering the pulmonary vasculature in a decellularized rat lung scaffold. We found that pre-seeded ASCs differentiated into pericytes and stabilized the endothelial cell (EC) monolayer in nascent pulmonary vessels, thereby contributing to EC survival in the regenerated lungs. The ASC-mediated stabilization of the ECs clearly reduced vascular permeability and suppressed alveolar hemorrhage in an orthotopic transplant model for up to 3 h after extubation. Fibroblast growth factor 9, a mesenchyme-targeting growth factor, enhanced ASC differentiation into pericytes but overstimulated their proliferation, causing a partial obstruction of the vasculature in the regenerated lung. ASCs may therefore provide a promising cell source for vascular regeneration in bioengineered lungs, though additional work is needed to optimize the growth factor or hormone milieu for organ culture.
Article
Full-text available
Background The xenoantigenicity of porcine bioprosthetic valves is implicated as an etiology leading to calcification and subsequent valve failure. Decellularization of porcine valves theoretically could erase the antigenicity of the tissue leading to more durable prosthetic valves, but the effectiveness of decellularization protocols in regard to completely removing antigens has yet to be verified. Our hypothesis was that decellularization would remove the more abundant α-gal antigens but not remove all the non α-gal antigens, which could mount a response. Methods Porcine aortic valves were decellularized with 1% sodium dodecyl sulfate for 4 days. Decellularized cusps were evaluated for α-gal epitopes by ELISA. To test for non α-gal antigens, valves were implanted into sheep. Serum was obtained from the sheep preoperatively and 1 week, 1 month, and 2 months postoperatively. This serum was utilized for anti-porcine antibody staining and for quantification of anti-pig IgM and IgG antibodies and complement. Results Decellularized porcine cusps had 2.8 ± 2.0% relative α-gal epitope as compared to fresh porcine aortic valve cusps and was not statistically significantly different (p = 0.4) from the human aortic valve cusp which had a 2.0 ± 0.4% relative concentration. Anti-pig IgM and IgG increased postoperatively from baseline levels. Preoperatively anti-pig IgM was 27.7 ± 1.7 μg/mL and it increased to 71.9 ± 12.1 μg/mL average of all time points postoperatively (p = 0.04). Preoperatively anti-pig IgG in sheep serum was 44.9 ± 1.5 μg/mL and it increased to 72.6 ± 6.0 μg/mL average of all time points postoperatively (p = 0.01). There was a statistically significant difference (p = 0.00007) in the serum C1q concentration before valve implantation (2.5 ± 0.2 IU/mL) and at averaged time points after valve implantation (5.3 ± 0.3 IU/mL). Conclusions Decellularization with 1% sodium dodecyl sulfate does not fully eliminate non α-gal antigens; however, significant reduction in α-gal presence on decellularized cusps was observed. Clinical implications of the non α-gal antigenic response are yet to be determined. As such, evaluation of any novel decellularized xenografts must include rigorous antigen testing prior to human trials. Electronic supplementary material The online version of this article (doi:10.1186/s13019-017-0621-5) contains supplementary material, which is available to authorized users.
Article
Full-text available
The critical shortage of donor organs has spurred investigation of alternative approaches to either generate replacement organs or implant exogenous cells for treatment of end-stage organ failure. Non-thermal irreversible electroporation (NTIRE), which uses brief high electric field pulses to induce irreversible permeabilization of cell membranes, has emerged as a technique for tumor ablation. Here, we introduce a new application for NTIRE that employs in situ cell ablation to create a niche within a solid organ for engraft­ment of exogenous cells in vivo. We treated the livers of mice with NTIRE and subsequently implanted exogenous congenic hepato­cytes within the zone of cell ablation. Donor hepatocytes engraft­ed and integrated with host liver parenchyma pre-treated with NTIRE. This new approach should have value for studying the ef­fects of a native matrix scaffold on in vivo cell growth and may pioneer a new type of minimally-invasive regenerative surgery.
Article
Full-text available
As the gap between donors and patients in need of an organ transplant continues to widen, research in regenerative medicine seeks to provide alternative strategies for treatment. One of the most promising techniques for tissue and organ regeneration is decellularization, in which the extracellular matrix (ECM) is isolated from its native cells and genetic material in order to produce a natural scaffold. The ECM, which ideally retains its inherent structural, biochemical, and biomechanical cues, can then be recellularized to produce a functional tissue or organ. While decellularization can be accomplished using chemical and enzymatic, physical, or combinative methods, each strategy has both benefits and drawbacks. The focus of this review is to compare the advantages and disadvantages of these methods in terms of their ability to retain desired ECM characteristics for particular tissues and organs. Additionally, a few applications of constructs engineered using decellularized cell sheets, tissues, and whole organs are discussed.
Book
A comprehensive overview of the latest achievements, trends, and the current state of the art of this important and rapidly expanding field. Clearly and logically structured, the first part of the book explores the fundamentals of tissue engineering, providing a separate chapter on each of the basic topics, including biomaterials stem cells, biosensors and bioreactors. The second part then follows a more applied approach, discussing various applications of tissue engineering, such as the replacement or repairing of skins, cartilages, livers and blood vessels, to trachea, lungs and cardiac tissues, to musculoskeletal tissue engineering used for bones and ligaments as well as pancreas, kidney and neural tissue engineering for the brain. The book concludes with a look at future technological advances. An invaluable reading for entrants to the field in biomedical engineering as well as expert researchers and developers in industry.
Article
Acute and chronic wounds contribute to increased morbidity and mortality in affected people and impose significant financial burdens on healthcare systems. For these challenging wounds, Acellular Dermal Matrices (ADMs) have been used as a biological wound coverage. Unlike engineered dermal matrices, ADMs are prepared through the removal of cells from skin, while preserving the extracellular matrix structure and function. In this study, our primary objective was to develop a detergent-free method of decellularization of the skin to mitigate chemical stress on matrix molecules. Then we performed a set of in vitro and in vivo experiments to compare this method with non-ionic and anionic detergent methods. All decellularization methods satisfactorily removed cells and supported fibroblast growth and migration in vitro. Sulfated glycosaminoglycan (sGAGs) content was reduced significantly (P < 0.05) only in ionic detergent treatment group. In contrast to detergent-free method, all detergent-based methods significantly reduced scaffold mechanical strength and elastin content (P < 0.05). Three weeks after transplantation, the results showed reepithelialization, angiogenesis, and migration of host cell into scaffolds with no induction of immunogenic reaction in all ADMs groups tested. In our study, detergent-free method showed better preservation of matrix composition and biomechanical properties, but after transplantation all methods of ADM preparation resulted in equally biofunctional matrices as wound coverage.
Article
This study describes a method to screen biomimetic surfaces based on intracellular insulin content of either fully or partly dissociated primary endocrine islet tissue. It is challenging to maintain endocrine pancreatic islets and more so, using dissociated ones. Physiological activity of isolated islet cells in vitro declines due to loss of cell-to-cell and cell-to-extracellular matrix interactions. An in vitro model was developed to evaluate specific extracellular binding components potentially affecting islet biology, with the intention to identify in vivo-like peptides promoting survival and function. Synthetic peptides were bound to low-fouling carboxy-methyl-dextran surfaces, effectively presenting defined surfaces while minimizing non-specific interactions. These biomimetic surfaces were screened based on intracellular insulin content of applied mouse primary islet tissue by analysis with an anti-insulin cell-ELISA. Three active biomimetic surfaces were identified, two laminin- (IKLLI and PDSGR) and one cadherin (HAVDI)-derived, which supported adhesion and survival of insulin-containing cultures for 5 days, respectively suggesting a benefit from both cell-extracellular matrix and cell-cell interactions. Cells from dissociated islets show progression over 10 days on the HAVDI-biomimetic for the insulin immunoreactivity and cell density. The three surfaces did not act additively or synergistically. A favorable reaction to glucose-stimulated insulin secretion on the cadherin-biomimetic indicated the cultures were physiologically functional. This supportive role of biomimetic peptides represents initial progress in defining minimal extracellular binding requirements influencing islet cell physiology. This will influence further optimization of growth surfaces and promote the basic understanding of islet biology. Low-fouling biomimetics are predicted to be applicable to additional diverse culture systems.