Vous êtes sur la page 1sur 8

Technical Reports

Organ reengineering through development of


a transplantable recellularized liver graft using
decellularized liver matrix
Basak E Uygun1, Alejandro Soto-Gutierrez1,6, Hiroshi Yagi1,6, Maria-Louisa Izamis1, Maria A Guzzardi1,2,
Carley Shulman1, Jack Milwid1, Naoya Kobayashi3, Arno Tilles1, Francois Berthiaume1,4, Martin Hertl5,
Yaakov Nahmias1,6, Martin L Yarmush1,4 & Korkut Uygun1
© 2010 Nature America, Inc. All rights reserved.

Orthotopic liver transplantation is the only available treatment Tissue engineering approaches have thus far been able to partially
for severe liver failure, but it is currently limited by organ improve cell engraftment in animal models by enhancing cell-to-cell
shortage. One technical challenge that has thus far limited the contact and providing nonimmunogenic matrices before transplan­
development of a tissue-engineered liver graft is oxygen and tation15. However, these approaches have been limited by oxygen
nutrient transport. Here we demonstrate a novel approach to and nutrient diffusion to thin, two-dimensional structures that are
generate transplantable liver grafts using decellularized liver transplanted into highly vascularized regions16–18. Practical consid­
matrix. The decellularization process preserves the structural eration of primary hepatocyte oxygen consumption requires that
and functional characteristics of the native microvascular any hepatic mass, sufficiently large to provide metabolic function,
network, allowing efficient recellularization of the liver matrix contains an extensive microvascular network and be connected
with adult hepatocytes and subsequent perfusion for in vitro to the blood supply, or the internal mass of the cells will suffer
culture. The recellularized graft supports liver-specific function ischemic damage19,20.
including albumin secretion, urea synthesis and cytochrome Decellularization is an attractive technique for scaffold preparation
P450 expression at comparable levels to normal liver in vitro. in tissue engineering, as the resulting material can potentially retain
The recellularized liver grafts can be transplanted into rats, the architecture of the original tissue, including the functional aspects
supporting hepatocyte survival and function with minimal of the native microvasculature21. The potential applications of decellu­
ischemic damage. These results provide a proof of principle larized matrix in tissue engineering have been demonstrated for a
for the generation of a transplantable liver graft as a potential number of tissues, including bladder22, artery23, esophagus24, skin25
treatment for liver disease. and trachea26. More recently, another group reported the decellulari­
zation of an entire heart through perfusion, preserving the original
About thirty million people in the United States have liver dis­ architecture and original microvascular network and allowing for
order, with roughly 27,000 deaths registered annually due to liver extensive recellularization27.
disease1. The only definitive treatment for severe hepatic failure In this work, we modified and applied their perfusion decellulariza­
is transplantation. However, critical shortage of organs causes a tion technique27 to prepare whole liver grafts, and we further intro­
growing deficit of approximately 4,000 livers per year 2. Hepatocyte duced perfusion-seeding and culture techniques for the preparation
transplantation offers an alternative way to treat patients with liver of recellularized liver matrix for transplantation. A key advantage of
diseases3,4, and years of laboratory and clinical studies have demon­ this methodology is the preservation of liver-specific extracellular
strated its efficacy 5–7. However, a limited cell supply and minimal matrix and three-dimensional architecture, providing crucial cues
engraftment efficiency have limited this approach3,8,9. Whereas the for hepatocyte engraftment, survival and long-term function. Most
cell supply limitation can potentially be resolved by a number of importantly, the decellularized liver matrix (DLM) has the underlying
alternative cell sources 10–13, low engraftment efficiency 14 remains matrix of the vascular network, which can be readily connected to
a major concern. the circulation, facilitating rapid oxygen and nutrient delivery after

1Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals for Children, Boston, Massachusetts, USA.
2Sector of Medicine, Scuola Superiore Sant’Anna, Pisa, Italy. 3Department of Surgery, Okayama University Graduate School of Medicine and Dentistry, Shikata-cho,
Okayama, Japan. 4Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, USA. 5Transplantation Unit, Massachusetts General Hospital,
Boston, Massachusetts, USA. 6Current addresses: Center for Innovative Regenerative Therapies, Department of Surgery, Transplantation Section, Children’s Hospital
of Pittsburgh, McGowan Institute for Regenerative Medicine and University of Pittsburgh, Pittsburgh, Pennsylvania, USA (A.S.-G.), Department of Surgery, Keio
University School of Medicine, Shinjuku-ku, Tokyo, Japan (H.Y.) and the Selim and Rachel Benin School of Computer Science and Engineering, The Hebrew University
of Jerusalem, Jerusalem, Israel (Y.N.). Correspondence should be addressed to K.U. (kuygun@partners.org).

Received 1 July 2009; accepted 3 February 2010; published online 13 June 2010; doi:10.1038/nm.2170

814 VOLUME 16 | NUMBER 7 | JULY 2010  nature medicine


Technical Reports

a b c d e

f H&E Collagen I Collagen IV Fibronectin Laminin

Normal liver Normal liver Normal liver Normal liver Normal liver

H&E Collagen I Collagen IV Fibronectin Laminin


© 2010 Nature America, Inc. All rights reserved.

Decellularized liver Decellularized liver Decellularized liver Decellularized liver Decellularized liver

Figure 1  Decellularization of ischemic rat livers. (a–e) Representative images of ischemic rat livers during decellularization process at 0 h (a), 18 h
(b), 48 h (c), 52 h (d) and 72 h (e). The livers were perfused through the portal vein using SDS as described in the Online Methods. (f) Comparison of
normal liver (top) and DLM (bottom). Left to right: H&E, collagen I (red), collagen IV (red), fibronectin (red) and laminin (red) staining. Sections were
counterstained with DAPI (blue). Scale bars: 10 mm (a–e) and 100 μm (f).

transplantation. The large pool of donor organs unsuitable for trans­ observed both collagen type IV and fibronectin in ­ decellularized
plantation, such as donors after cardiac death, provides a source for sinusoidal spaces, whereas we saw laminin-β1 in the basement
decellularized liver matrices28, rendering this approach feasible. membrane of the larger vessels (Fig. 1f). We found that 100% of the
Our work demonstrates the decellularization of ischemic rat ­livers fibrillar collagen and approximately 50% of the glycosaminoglycans
by portal vein perfusion and its characterization. We show the preser­ of native liver were retained after decellularization (Supplementary
vation of both structural and basement membrane–based compo­ Table 1). Residual DNA content in the DLM was less than
nents of the native liver extracellular matrix, as well as the retention 3% (Supplementary Table 1).
of a functional microvascular network. DLM supported efficient To show that the decellularization protocol was successful in retain­
engraftment and in vitro metabolic function of adult primary hepato­ ing a functional vascular bed, we characterized its perfusion with
cytes. Retention of vascular structures allowed for transplantation Allura Red dye. Structural components of the vascular tree were
of ­recellularized liver graft in rats, and post-transplantation analysis clearly apparent in the translucent matrix (Fig. 2a,b). The dye injected
demonstrated preservation of hepatocyte structure and function with through the portal vein flowed as would be expected inside the vas­
minimal indications of ischemic damage. Preliminary recellulariza­ cular network, gradually moving from larger vessels to smaller capil­
tion with endothelial cells also indicated attachment and viability laries, suggesting the microvasculature remained intact. We further
within the recellularized matrix in vitro. Overall, the methods and characterized the intricate vascular tree by generating a corrosion cast
techniques developed in this study present what is to our knowledge of DLM (Fig. 2c,d). Portal (red) and venous (blue) vasculature casting
the first step toward manufacture of auxiliary liver grafts as an alter­ of both normal (Fig. 2c) and decellularized (Fig. 2d) livers showed
native source of organs for liver transplantation. that larger portal and venous circulation system of vessels and the vast
majority of the smaller microcirculatory branches were preserved,
RESULTS indicating that physiologic flow could be achieved by traversing the
Characterization of decellularized liver matrix portal venous system and emptying into the systemic venous circu­
We achieved whole-organ decellularization by portal perfusion with lation via the hepatic vein and inferior vena cava (IVC). Moreover,
SDS, an anionic detergent that lyses cells and solubilizes cytoplasmic scanning electron microscopy (SEM) images confirmed the presence
components27. After 72 h of decellularization, a translucent acel­ of large vessels within DLM (Fig. 2e), and we also observed remnants
lular scaffold, which retained the gross shape of liver, was generated of small vessels closely resembling portal triad (Fig. 2f). The honey­
(Fig. 1a–e). Histological evaluation revealed no nuclei or cyto­ comb of about 30-μm diameter gaps in the ECM is thought to be the
plasmic staining in DLM compared to normal rat liver (Fig. 1f). footprint of hepatocytes removed during decellularization (Fig. 2g).
Immunostaining for four extracellular matrix (ECM) proteins,
collagen type I, collagen type IV, fibronectin and laminin-β1, indi­ Recellularization of decellularized liver matrix
cated that both structural and basement membrane components of The presence of a functional vascular bed in the DLM offers the abil­
the ECM were retained similarly to native liver (Fig. 1f). A lack of ity to control hepatocyte engraftment and characterize liver-specific
DAPI staining in DLM confirmed the absence of cells (Fig. 1f). We metabolic function in vitro before transplantation of the ­recellularized

nature medicine  VOLUME 16 | NUMBER 7 | JULY 2010 815


Technical Reports

markers in engrafted hepatocytes was similar


a b c d to that in normal livers (Fig. 3h).
To assess the metabolic activity of engrafted
hepatocytes, we quantified hepatocyte albu­
min production and urea synthesis. The
cumulative urea amounts in the recellular­
ized liver system were statistically higher than
in hepatocyte sandwich culture during the
e f g 5-d culture period (P = 0.0017) (Fig. 4a,b).
The difference in cumulative albumin levels
was statistically not significant (P = 0.0176)
(Fig. 4a,b). The albumin production rate by
the recellularized liver graft was 27.6 ± 7.0 μg
million cells per day as compared to 140 μg
million cells per day by the normal adult
rat liver29.
Figure 2  DLM retains intact lobular structure and vascular bed. (a) Representative photograph Analysis of the expression of drug metabo­
of decellularized left lateral and median lobes of rat liver, with the vascular tree visible. (b) The lism enzymes (Supplementary Table 2) via
vascular tree, after perfusion with Allura Red AC dye. (c,d) Corrosion cast model of left lobe of a quantitative RT-PCR at 2 d revealed that
© 2010 Nature America, Inc. All rights reserved.

normal liver (c) and the DLM (d), with portal (red) and venous (blue) vasculature. (e–g) SEM images
expression levels of drug metabolism enzymes
of a vessel (e), a section featuring bile duct–like small vessels (arrows) (f), extracellular matrix
within the parenchyma (g), with hepatocyte-size free spaces. Scale bars: 10 mm (a,b), 5 mm (c,d) in the recellularized liver were similar to those
and 20 μm (e–g). measured in sandwich hepatocyte cultures
(P = 0.05) (Fig. 4c,d). As expected at this
stage in culture, gene transcription levels were
graft. We introduced primary rat hepatocytes via portal vein per­ overall much lower than those of freshly isolated hepatocytes (Fig. 4c).
fusion recirculation. Roughly 12.5 million cells were introduced at However, Cyp2c11 (encoding cytochrome P450, subfamily 2, polypep­
each step, for a total of four steps, with 10-min intervals between each tide 11) Gstm2 (glutathione S-transferase mu 2), Ugt1a1 (encoding
step (Fig. 3a). Cell viability and distribution in the parenchyma after UDP glucuronosyltransferase-1 family, polypeptide A1) and Cyp1a1
this four-step seeding protocol was superior to single-step infusion (encoding cytochrome P450, family-1, subfamily a, polypeptide 1)
of 50 million cells for 40 min (Supplementary Fig. 1a). Engraftment were expressed in the recellularized liver at similar levels to those in
efficiency with the four-step protocol was 95.6% ± 3.4%. After seed­ normal liver (Fig. 4e–h). Adh1 (encoding alcohol dehydrogenase-1)
ing, we transferred the recellularized liver grafts (Fig. 3b,c) into a and Cyp3a18 (encoding cytochrome P450, family 3, subfamily a,
specially designed perfusion chamber (Supplementary Fig. 1b) for polypeptide 18) expression levels were higher in recellularized liver
in vitro culture. The perfusion chamber features two hermetically than in sandwich plate culture control, although they were much lower
sealed silicon sheets, forming a pouch filled with culture medium; than in normal liver (Fig. 4i,j). Primary hepatocytes are known to
this design avoids rigid surfaces, preventing development of pressure require between 7 and 10 d in culture before their metabolic activity
spots, while enabling sterile culture of the recellularized grafts up to and gene transcription levels stabilize30.
2 weeks in vitro (data not shown). Histological staining of recellular­
ized sections at 4 h and at 1 d, 2 d and 5 d of in vitro perfusion culture Further scalability of recellularization methodology
revealed that at 4 h, the majority of the cells remain in and around Ultimately, reconstruction of liver grafts in vitro requires the addi­
the vessels, whereas at 1 d and 2 d, they leave the vessels and get tion of nonparenchymal cells. As a preliminary test, we attempted to
­distributed throughout the matrix (Supplementary Fig. 1c). We did seed a nonparenchymal component to the recellularized liver graft
not observe any change in the distribution of the cells within the by incorporating microvascular endothelial cells to the hepatocyte-
matrix at 5 d compared to previous days (Supplementary Fig. 1c). repopulated graft and tested it by perfusion culture for up to 5 d to
We continuously perfused the recellularized liver graft for 5 d. allow for endothelial cell engraftment. Histological analysis showed
Hepatocyte viability was maintained during culture, and quantifica­ that endothelial cells were capable of lining the vasculature encir­
tion of TUNEL-positive cells revealed that less than 20% of the cells cled by hepatocytes at 3 d of culture (Supplementary Fig. 2a–d).
were apoptotic on the first 2 d of culture (Fig. 3d,e). Lactate dehydro­ Hepatocytes remained viable, as indicated by TUNEL staining and
genase (LDH) release during perfusion was comparable and statis­ immunohistochemistry (Supplementary Fig. 2e–i).
tically not different between groups, indicating minimal cell death To test the scalability of the seeding approach, as well as the limits
(P = 0.0455) (Fig. 3f). SEM and histological analysis (Fig. 3g) showed of the perfusion-culture system, we repopulated the median lobe
that hepatocytes engrafted around the larger vessels, populating the of DLM with 200 million hepatocytes and observed the perfusion
surrounding parenchyma, suggesting the cells migrated beyond the culture for 10 d. This cellular amount corresponds to approximately
matrix barrier to reach decellularized sinusoidal spaces. We assessed 20% of a rat’s liver mass, more than double the minimum hepatic mass
the functional characteristics of engrafted hepatocytes in the decellu­ necessary for therapeutic intervention6. In these studies, we injected
larized matrix via immunostaining at 2 d of culture of UDP glucuron­ 50 million hepatocytes at each of the four steps mentioned above. We
osyltransferase 1 family, polypeptide A1 (Ugt1a), a sensitive enzyme assessed the metabolic activity of the recellularized liver graft through
with a short half-life (about 50 min) whose presence indicated hepato­ albumin production, urea secretion and total bile acid synthesis, and
cyte viability and function, glucose-6-phosphatase, catalytic subunit the cumulative levels of production of these meta­bolites were found
(G6pc) and albumin (Fig. 3h). The level of immunostaining for these to be at similar levels to those of static sandwich-culture controls

816 VOLUME 16 | NUMBER 7 | JULY 2010  nature medicine


Technical Reports

a –30 min 0 min 10 min 20 min 30 min 40 min d Recellularized liver Negative control Positive control

Perfusion with Inject Inject Inject Inject Incubation


culture medium 12.5 million 12.5 million 12.5 million 12.5 million starts
starts cells cells cells cells

b c e

TUNEL-positive cells (%)

(ng per 106 cells per 24 h)


100
f 20

Lactate dehydrogenase
Recellularized liver
80 Static sandwich culture
15
60
10
40
5
20
0 0
0 1 2 3 4 5 6

N ive

h
iv

24

48
t
at
si
Time (d)

eg
Po
g h H&E Albumin G6pc Ugt1a
© 2010 Nature America, Inc. All rights reserved.

Normal liver Normal liver Normal liver Normal liver

H&E Albumin G6pc Ugt1a

Recellularized liver Recellularized liver Recellularized liver Recellularized liver

Figure 3  Repopulation of rat DLM with adult rat hepatocytes. (a) Recellularization scheme of the DLM. (b,c) Decellularized whole liver matrix (b) and
same liver after recellularization with about 50 million hepatocytes (c). (d) TUNEL staining of recellularized liver grafts. Left to right: recellularized
graft at 48 h in culture (arrows indicate positive cells), fresh isolated liver (negative control), DNase-treated normal liver (positive control). Insets show
Hoechst 33258 counterstain of the same sections. (e) TUNEL-positive cell percentage in recellularized liver grafts as a function of perfusion-culture
time. (f) LDH release from recellularized livers during perfusion culture (P = 0.0455 by Friedman’s test). (g) SEM micrographs of recellularized liver
graft after 2 d in culture. (h) Immunohistochemical staining of the recellularized liver graft (bottom) in comparison to normal liver (top); left to right:
H&E, albumin (red), glucose 6-phosphatase (red) and Ugt1a (green). Sections were counterstained with Hoechst 33258 (blue). Scale bars: 20 mm
(b,c), 200 μm (d) and 100 μm (h). All error bars represent s.e.m. (n = 6).

(P = 0.5249, P = 0.5271 and P = 0.0114, respectively), indicating c­ onfirmed that hepatic function was also retained in the transplanted
the scalability of the techniques we introduce here, as well as the grafts, with minimal indications of ischemic damage (Fig. 5b).
usability of the perfusion-culture system as an in vitro model To investigate early graft function beyond 8 h, we adopted an
(Supplementary Fig. 3). ex vivo whole-blood perfusion technique as a surrogate model of trans­
plantation, which has been shown to be representative of early graft
Transplantation of recellularized liver grafts performance and to predict primary graft failure in an orthotopic rat
An available functional vascular structure offers the opportunity to liver transplant model31,32. The ex vivo perfusion system was similar
transplant the recellularized liver graft by connecting the graft to the to the in vitro perfusion system, except that the perfusate consisted of
blood supply, allowing for the transplantation of a critical hepatocyte whole rat blood diluted with perfusion medium (hematocrit 20%). The
mass while avoiding ischemic damage due to poor graft perfusion. recellularized liver graft was perfused for 24 h, during which the blood
Briefly, recipient rats underwent unilateral nephrectomy to prepare a was replenished three times, once every 8 h to keep the hematocrit
viable site for auxiliary liver graft transplantation. We used the renal level at 20% (Supplementary Fig. 5b). TUNEL staining and histologi­
vein and artery as ports to create blood flow within the graft. Upon cal analysis showed that hepatocytes remained viable (22.6% ± 13.8%
unclamping of the artery, the graft was perfused, quickly filling with TUNEL-positive cells, Supplementary Fig. 5c) and preserved their
blood, and appropriate efflux was established within 5 min (Fig. 5a). morphology and parenchymal position at the end of ex vivo blood per­
The recellularized graft was kept in vivo for 8 h before harvesting fusion (Supplementary Fig. 5d). In addition, hepatocytes remained
for further analysis. TUNEL staining revealed that there was mini­ metabolically active during the 24 h ex vivo blood perfusion, as evi­
mal damage to the hepatocytes due to the arterial blood flow and the denced by urea and albumin secretion (Supplementary Fig. 5e,f).
resulting shear stress during 8 h of transplantation (19.7% ± 13.7% of
TUNEL-positive cells after transplantation versus 21.4% ± 5.8% of DISCUSSION
TUNEL-positive cells before transplantation, P = 0.55, Supplementary These results present the first step toward development of recellular­
Fig. 4). Histological staining indicated that the hepatocytes retained ized liver matrix as an auxiliary graft for transplantation. Previous
both normal morphology and their parenchymal positions (Fig. 5b). attempts, including those of our group, have failed in creating ­complex
Immunohistochemical staining for albumin, G6pc and Ugt1a tissue-engineered constructs for liver transplantation16,18. Here we

nature medicine  VOLUME 16 | NUMBER 7 | JULY 2010 817


Technical Reports

Figure 4  Hepatic function of the recellularized


liver graft in vitro. (a,b) Albumin synthesis (a)
a c e f
1.2 1.2

Normalized mRNA

Normalized mRNA
and urea secretion (b) in comparison to static 250 Recellularized liver 1.0 1.0

Albumin (µg per 106 cells)


Static sandwich culture

ce ult . liv r
0.8 0.8

lls ur er
sandwich culture (P values are 0.0176 for

C ll e
R al ls
200

e v
m l

level

level
ed
ec li
or e
0.6 0.6

N hc
albumin and 0.0017 for urea, by Friedman’s

es
150 0.4 0.4

Fr
test). (c) Gene expression analysis of hepatocytes 0.2 0.2
in the recellularized liver graft (2 d) compared 100 0 0

el ltu ich

el ltu ch

er
or liv zed
er
or ve d
to normal liver, fresh hepatocytes and sandwich

lu re

lu re
N li rize
50

al r
ec u i

liv
m r
liv
ec u w

R c ndw

m e
ri
R c d

al

la
la
n
culture hepatocytes (2 d) for phase 1 and phase 2

sa

sa
0

ic

ic

N
drug metabolism enzymes. (d) Scatter plot com­

at

at
St

St
0 1 2 3 4 5 6
paring gene expression of phase 1 and phase 2
g h

Phase 1
Time (d)
drug metabolism enzymes at 2-d recellularized
b 1.6 50

Normalized mRNA
Normalized mRNA
liver graft and cultured hepatocytes (P = 0.0499 500 1.4
Recellularized liver
by Friedman’s test). (e–j) Normalized gene 1.2

Urea (µg per 106 cells)


Static sandwich culture 1.0
400

level
level
expression of Cyp2c11 (e), Gstm2 (f), Ugt1a1 (g), 0.8 40
0.6 1
Cyp1a1 (h), Adh1 (i) and Cyp3a18 (j). All error 300 0.4
bars represent s.e.m. (n = 3). 0.2
200 0 0

el ltu ch

el ltu ch
er

or ve d
or iv d

er
N li ize
N l rize
lu re

lu re
liv

m r
ec u i

m er

ec u i

liv
100

R c ndw

R c ndw

r
al

al
la
la
successfully demonstrate the preservation

sa

sa
Phase 2
0

ic

ic
at

at
of the liver’s three-dimensional architecture,

St

St
0 1 2 3 4 5 6
functional vasculature and native matrix d 100 Time (d)
i j
© 2010 Nature America, Inc. All rights reserved.

composition upon decellularization. Further,


Recellularized liver

1.2 1.20

Normalized mRNA

Normalized mRNA
10
1.0
we present a technique for efficient in vitro 1 0.8 1.00
recellularization of the graft, which main­ 0.6

level

level
0.1 0.80
0.02
tains cell viability and function. Finally, we 0.01
0.2

demonstrate the feasibility of transplanting 0.001 0 0


these recellularized liver grafts in vivo with 0.001 0.01 0.1 1 10 100

el ltu ich

el ltu ich
er
or iv d

or iv d

er
N l rize

N l rize
lu re

lu re
m er
liv

m er
liv
ec cu dw

ec u w
R c nd
al
Static sandwich culture

la
minimal ischemic damage.

la

al
n
sa

sa
ic

ic
Oxygen and nutrient transport limitations

at

at
R
St

St
have thus far restricted the development of
tissue-engineered liver constructs to the diffusion limit, about 200 μm. distance from the nearest blood vessel17. It is a crucial configuration
The high rates of oxygen consumption 33–36 by primary hepato­ that should ideally be replicated in vitro before transplantation.
cytes result in ischemic damage in cells found further away from This report builds on the previously reported technique developed
the blood within minutes of transplantation. Owing to high oxygen for heart perfusion decellularization27, demonstrating its applicabil­
demands, the liver is the only organ that receives both arterial and ity for rat liver decellularization and further extending its innova­
venous blood supply, whereas hepatocytes are located less than 2 μm tive contributions by introducing a perfusion-seeding method and
an extracorporeal perfusion-culture system.
Additionally, we further optimize the decel­
a lularization for liver.
Our work shows that portal vein perfusion
decellularization creates a translucent liver
matrix while preserving the three-dimensional
architecture, the native structural and basement

Figure 5  Transplantation of the recellularized


liver graft. Decellularized and recellularized rat
liver was transplanted as auxiliary heterotopic
graft with portal vein arterialization, and
graft viability was determined 8 h after
transplantation. (a) Representative images of
graft transplantation; top, left to right: transplant
b Normal liver Normal liver Normal liver Normal liver site, recellularized graft at the transplant site,
transplanted graft before blood reperfusion,
the graft right after declamping the renal
artery; bottom, left to right: transplanted graft
1 min, 2 min and 4 min after declamping of
H&E Albumin G6pc Ugt1a the renal artery, and auxiliary graft in contrast
Recellularized liver Recellularized liver Recellularized liver Recellularized liver with the native liver. (b) Immunohistochemical
staining of recellularized auxiliary graft 8 h after
transplantation (bottom) compared to native liver
(top); left to right: H&E, albumin (red), glucose
6-phosphatase (red), Ugt1a (green). Sections
were counterstained with Hoechst 33258 (blue).
H&E Albumin G6pc Ugt1a
Scale bars: 10 mm (a) and 100 μm (b).

818 VOLUME 16 | NUMBER 7 | JULY 2010  nature medicine


Technical Reports

membrane matrix, and the functional vascular network of the original Methods
organ. Portal vein dye perfusion, corrosion casting, SEM and immuno­ Methods and any associated references are available in the online ­version
staining show the presence and function of the microvascular network of the paper at http://www.nature.com/naturemedicine/.
of the liver post decellularization. Quantification of extracellular matrix
Note: Supplementary information is available on the Nature Medicine website.
proteins, SEM analysis and immunostaining suggests that the matrix
of both the large vessels (collagen type I, laminin-β1) and the sinusoids Acknowledgments
(collagen type IV, fibronectin) remained relatively intact. We note that This work was supported by grants from the US National Institutes of Health,
the main structural protein of the liver, collagen type I, was retained in R01DK59766 and R01DK084053 to M.L.Y., K99/R00 DK080942 to K.U. and
K99DK083556 to A.S.-G., and US National Science Foundation CBET-0853569 to
its entirety during the decellularization process. However, we also note K.U. We thank the support of the Shriners Hospitals for Children to B.E.U. (grant
the marked loss of half of the glycosaminoglycan content of the liver no. 8503), the American Liver Foundation to A.S.-G. and Massachusetts General
during decellularization. This loss is not surprising, as many glycosamino­ Hospital Junior Faculty Grant to K.U. and the Shriners Hospitals for Children. We
glycans are associated with cellular membranes that are solubilized in the would like to thank A. Vitalo, C. Calhoun and B. Crowther for technical support.
decellularization process. AUTHOR CONTRIBUTIONS
We have found that the DLM can be seeded with primary rat K.U. had full access to all of the data in the study and takes responsibility for the
hepatocytes via a multistep perfusion technique, with efficiency integrity of the data and the accuracy of the data analyses. B.E.U., A.S.-G., H.Y.
greater than 90%. As expected, this grafting efficiency is much and K.U., study concept and design; B.E.U., A.S.-G., H.Y., M.A.G., acquisition of
data; B.E.U., A.S.-G., Y.N. and K.U., analysis and interpretation of data; M.-L.I.,
higher than in vivo levels of engraftment after hepatocyte trans­
C.S. and B.E.U., rat liver harvest, decellularization and hepatocyte isolation; J.M.
plantation, possibly as a result of the lack of endothelial barrier and and B.E.U., design and construction of the recellularized liver chamber; B.E.U.,
the abundant empty space. We seeded 50 million primary rat hepato­ A.S.-G., H.Y. and M.A.G., recellularization; A.S.-G. and H.Y., histology and
© 2010 Nature America, Inc. All rights reserved.

cytes, roughly 5% of the cells in native rat liver, as this amount was transplantation studies; B.E.U., A.S.-G., H.Y. and K.U., drafting of the manuscript;
B.E.U., A.S.-G., H.Y., M.L.Y., Y.N., A.T., F.B., M.H., N.K. and K.U., critical revision
previously shown to be sufficient to restore liver function in animal
of the manuscript for intellectual content; B.E.U. and K.U., statistical analysis;
models6. We have also shown that this number can be increased M.L.Y., K.U. and A.S.-G., obtained funding; M.-L.I., C.S., J.M., Y.N., A.T. and
up to 200 million to account for 20% of the native rat liver hepatic F.B., administrative, technical or material support. All authors contributed to the
mass, corresponding to repopulation of approximately 50% of the preparation of the report.
original hepatic mass within the lobe used for transplantation.
COMPETING FINANCIAL INTERESTS
After engraftment, hepatocyte viability and metabolic function was The authors declare no competing financial interests.
maintained in perfusion culture, as assessed by TUNEL staining,
LDH release, synthetic activity and gene expression. Engrafted cells Published online at http://www.nature.com/naturemedicine/.
Reprints and permissions information is available online at http://npg.nature.com/
showed albumin production and gene expression equivalent to cells
reprintsandpermissions/.
from the same isolation cultured in a collagen-sandwich configu­
ration, a culture configuration previously shown to maintain high
liver-specific function for up to a month in vitro37. Overall, albu­ 1. Heron, M. et al. Deaths: Final Data for 2006. in National Vital Statistics Report,
Vol. 57 (Centers for Disease Control and Prevention, Hyattsville, Maryland, 2009).
min production and gene expression were roughly 20% and 30% of
2. Punch, J.D., Hayes, D.H., LaPorte, F.B., McBride, V. & Seely, M.S. Organ donation
in vivo levels, respectively. and utilization in the United States, 1996–2005. Am. J. Transplant. 7, 1327–1338
We have shown that rat livers can be depleted of their cellular com­ (2007).
3. Fox, I.J. & Roy-Chowdhury, J. Hepatocyte transplantation. J. Hepatol. 40, 878–886
ponents and used to engineer sophisticated hepatic structures that (2004).
may recover many functions of normal liver. Despite its advantages, 4. Strom, S.C. et al. Hepatocyte transplantation as a bridge to orthotopic
further improvements of the decellularization and recellularization of liver transplantation in terminal liver failure. Transplantation 63, 559–569
(1997).
liver matrix are necessary. Engineering of an entire liver will require 5. Horslen, S.P. et al. Isolated hepatocyte transplantation in an infant with a severe
addition of nonparenchymal cells such as liver sinusoidal endothelial urea cycle disorder. Pediatrics 111, 1262–1267 (2003).
6. Fox, I.J. et al. Treatment of the Crigler-Najjar syndrome type I with hepatocyte
cells, stellate cells, biliary epithelial cells and Kupffer cells. Particularly, transplantation. N. Engl. J. Med. 338, 1422–1426 (1998).
we observed the presence of hepatocytes outside the microvasculature 7. Fisher, R.A. & Strom, S.C. Human hepatocyte transplantation: worldwide results.
system, mainly distributed in the parenchymal spaces, possibly owing Transplantation 82, 441–449 (2006).
8. Habibullah, C.M., Syed, I.H., Qamar, A. & Taher-Uz, Z. Human fetal hepatocyte
to loss of sinusoidal integrity in the absence of the liver sinusoidal transplantation in patients with fulminant hepatic failure. Transplantation 58,
endothelium and other nonparenchymal cells. We speculate that the 951–952 (1994).
9. Nagata, H. et al. Prolonged survival of porcine hepatocytes in cynomolgus monkeys.
nonparenchymal cells of the liver will be essential for reacquisition of Gastroenterology 132, 321–329 (2007).
vascular integrity. Our ongoing studies are directed toward optimizing 10. Basma, H. et al. Differentiation and transplantation of human embryonic stem
seeding strategies, re-endothelialization and biliary tract reconstruc­ cell–derived hepatocytes. Gastroenterology 136, 990–999 (2009).
11. Cho, C.H. et al. Homogeneous differentiation of hepatocyte-like cells from embryonic
tion. However, our preliminary results on re-endothelialization of the stem cells: applications for the treatment of liver failure. FASEB J. 22, 898–909
graft indicate the feasibility of this approach. Addition of nonparen­ (2008).
12. Totsugawa, T. et al. Survival of liver failure pigs by transplantation of reversibly
chymal cells and modification of the perfusate may further enhance immortalized human hepatocytes with tamoxifen-mediated self-recombination.
the reconstitution of a functional liver graft and even lead to hepatic J. Hepatol. 47, 74–82 (2007).
repopulation by imitating the regenerative behavior of the liver. Thus, 13. Shafritz, D.A., Oertel, M., Menthena, A., Nierhoff, D. & Dabeva, M.D. Liver stem
cells and prospects for liver reconstitution by transplanted cells. Hepatology 43,
we envision the possible use of DLM ultimately as a model for the S89–S98 (2006).
study of both liver development and liver regeneration. 14. Smets, F., Najimi, M. & Sokal, E.M. Cell transplantation in the treatment of liver
diseases. Pediatr. Transplant. 12, 6–13 (2008).
In summary, this report provides a foundation for efficient develop­
15. Langer, R. & Vacanti, J.P. Tissue engineering. Science 260, 920–926 (1993).
ment of auxiliary liver grafts for transplantation and practical, unique 16. Wang, S., Nagrath, D., Chen, P.C., Berthiaume, F. & Yarmush, M.L. Three-
techniques to prepare recellularized liver graft. Further studies are dimensional primary hepatocyte culture in synthetic self-assembling peptide
hydrogel. Tissue. Eng. Part A 14, 227–236 (2008).
required to determine whether the techniques described here can be 17. Nahmias, Y., Berthiaume, F. & Yarmush, M.L. Integration of technologies for hepatic
scaled up for use in humans. tissue engineering. Adv. Biochem. Eng. Biotechnol. 103, 309–329 (2007).

nature medicine  VOLUME 16 | NUMBER 7 | JULY 2010 819


Technical Reports

18. Ohashi, K. et al. Engineering functional two- and three-dimensional liver systems 28. Safar, P. Clinical death symposium. Crit. Care Med. 16, 919–920 (1988).
in vivo using hepatic tissue sheets. Nat. Med. 13, 880–885 (2007). 29. Hoffenberg, R. Measurement of the synthesis of liver-produced plasma proteins
19. Kulig, K.M. & Vacanti, J.P. Hepatic tissue engineering. Transpl. Immunol. 12, with particular reference to dietary protein and amino acid supply. Biochem. J.
303–310 (2004). 129, 3P (1972).
20. Nahmias, Y., Schwartz, R.E., Hu, W.S., Verfaillie, C.M. & Odde, D.J. Endothelium- 30. Khetani, S.R. & Bhatia, S.N. Microscale culture of human liver cells for drug
mediated hepatocyte recruitment in the establishment of liver-like tissue in vitro. development. Nat. Biotechnol. 26, 120–126 (2008).
Tissue Eng. 12, 1627–1638 (2006). 31. Tolboom, H. et al. Recovery of warm ischemic rat liver grafts by normothermic
21. Badylak, S.F. The extracellular matrix as a biologic scaffold material. Biomaterials extracorporeal perfusion. Transplantation 87, 170–177 (2009).
28, 3587–3593 (2007). 32. Uygun, K. et al. Diluted blood reperfusion as a model for transplantation
22. Yoo, J.J., Meng, J., Oberpenning, F. & Atala, A. Bladder augmentation using of ischemic rat livers: alt is a direct indicator of viability. Transplant. Proc. (in the
allogenic bladder submucosa seeded with cells. Urology 51, 221–225 (1998). press).
23. Dahl, S.L., Koh, J., Prabhakar, V. & Niklason, L.E. Decellularized native and 33. Cho, C.H. et al. Oxygen uptake rates and liver-specific functions of hepatocyte and
engineered arterial scaffolds for transplantation. Cell Transplant. 12, 659–666 3T3 fibroblast co-cultures. Biotechnol. Bioeng. 97, 188–199 (2007).
(2003). 34. Rotem, A. et al. Oxygen is a factor determining in vitro tissue assembly: effects on
24. Nieponice, A., Gilbert, T.W. & Badylak, S.F. Reinforcement of esophageal attachment and spreading of hepatocytes. Biotechnol. Bioeng. 43, 654–660
anastomoses with an extracellular matrix scaffold in a canine model. Ann. Thorac. (1994).
Surg. 82, 2050–2058 (2006). 35. Foy, B.D. et al. Optimization of hepatocyte attachment to microcarriers: importance
25. Schechner, J.S. et al. Engraftment of a vascularized human skin equivalent. FASEB J. of oxygen. Biotechnol. Bioeng. 42, 579–588 (1993).
17, 2250–2256 (2003). 36. Balis, U.J. et al. Oxygen consumption characteristics of porcine hepatocytes. Metab.
26. Macchiarini, P. et al. Clinical transplantation of a tissue-engineered airway. Lancet Eng. 1, 49–62 (1999).
372, 2023–2030 (2008). 37. Dunn, J.C., Tompkins, R.G. & Yarmush, M.L. Long-term in vitro function of adult
27. Ott, H.C. et al. Perfusion-decellularized matrix: using nature’s platform to engineer hepatocytes in a collagen sandwich configuration. Biotechnol. Prog. 7, 237–245
a bioartificial heart. Nat. Med. 14, 213–221 (2008). (1991).
© 2010 Nature America, Inc. All rights reserved.

820 VOLUME 16 | NUMBER 7 | JULY 2010  nature medicine


ONLINE METHODS Recellularized liver graft perfusion system. For in vitro perfusion-culture
Rats. Female Lewis rats (150–200 g) (Charles River Laboratories) were used experiments, the recellularized liver was transferred to a clean chamber, part
for hepatocyte isolation and liver harvest. Male Lewis rats (250–300 g) (Charles of a perfusion system previously developed for donors-after-cardiac-death
River Laboratories) were used as recipients. The rats were cared for in accord­ organ resuscitation31 that consisted of a peristaltic pump, bubble trap and
ance with the guidelines set by the Committee on Laboratory Resources, US oxygenator. The system was placed in an incubator for temperature control,
National Institutes of Health, and Subcommittee on Research Animal Care and and the oxygenator was connected to atmospheric gas mixture. The graft was
Laboratory Animal Resources of Massachusetts General Hospital. continuously perfused through the portal vein at 15 ml min−1 with continuous
oxygenation that delivered an inflow partial oxygen tension of ~300 mm Hg.
Liver decellularization. Frozen livers were thawed at 4 °C and washed with The medium was changed daily.
PBS overnight through perfusion via portal vein at 1 ml min−1. Liver was per­
fused with SDS (Sigma) in distilled H2O for 72 h starting with 0.01% SDS for Heterotopic recellularized liver graft transplantation. The left renal artery
24 h and then with 0.1% SDS for 24 h and with 1% SDS for 24 h. Subsequently, and vein of the recipient rat were isolated, and the left ureter was ligated
liver was washed with distilled H2O for 15 min and with 1% Triton X-100 and transected. After clamping the left renal artery and vein at their origin,
(Sigma) for 30 min to remove residual SDS. Decellularized livers were washed nephrectomy was performed. The recellularized graft was placed in the left
with PBS for 1 h. Unless otherwise mentioned, caudal, left lateral and right renal space of the recipient. One milliliter of saline containing 100 U of heparin
lateral lobes were ligated and resected, and the median lobe was kept for recel­ and 0.25 mg per kg body weight of the Fab fragment of a chimeric monoclonal
lularization. DLM was sterilized in 0.1% peracetic acid (Sigma) in PBS for 3 h. antibody, a glycoprotein IIb/IIIa inhibitor (ReoPro, Eli Lilly), was injected
DLM was washed extensively with sterile PBS and preserved in PBS containing into the penile vein to prevent coagulation. An 8-mm stent (0.3-mm inner
antibiotics at 4 °C for up to 7 d. diameter) was introduced into the donor’s portal vein, and another 8-mm
stent (0.6-mm inner diameter) was introduced and secured in the inferior vena
Hepatocyte seeding. DLM was connected to the perfusion system through cava. The stent in the portal vein was inserted into the left renal artery, allow­
© 2010 Nature America, Inc. All rights reserved.

portal vein cannulation. The inferior vena cava and superior vena cava were ing for arterial-venous anastomosis with standardized flow-regulated inflow
left open for outlet. DLM was kept immersed in medium in a perfusion rates38. The portal vein and renal artery were connected with two sutures. The
chamber while it was perfused through the portal vein. We recirculated 50 ml donor’s inferior vena cava and the stent were introduced and sutured in the
medium in the perfusion system. Portal pressure was maintained between left renal vein. The arterial clamp and then the venous clamp was removed
10 and 15 cm H2O. The perfusate consisted of Williams’ E (Sigma), 5% FBS to start reperfusion.
(Hyclone), 0.5 U ml−1 insulin (Eli Lilly), 20 ng ml−1 epidermal growth factor
(Invitrogen), 14 ng ml−1 glucagon (Bedford Laboratories), 7.5 μg ml−1 hydro­ Statistical analyses. Statistical differences of collagen, glycosaminoglycans
cortisone (Pharmacia), 100 U ml−1 penicillin and 100 μg ml−1 streptomycin and DNA in DLM and comparison of TUNEL-positive cells were evaluated by
(Invitrogen). For recellularization with 200 million hepatocytes (10-d culture two-tailed unpaired Student’s t test at α = 0.01. Statistical analyses of albumin,
period), the perfusate was high-glucose DMEM and 10% FBS with other sup­ urea, total bile acid and LDH in the recellularized grafts were performed with
plements remaining the same to support survival and function of hepatocytes Friedman’s test in MATLAB (v7.7.0), a nonparametric alternative to two-way
for more than 7 d in vitro37. After a 30-min perfusion with medium, a total of analysis of variance, which does not assume normal distribution, with multiple
50 or 200 million primary adult rat hepatocytes with over 90% viability were replicates at α = 0.01.
infused into the circuit in four steps at 10-min intervals and were recirculated
in the system at 25 °C. At the end of 40 min, the perfusate was collected and the Additional methods. Detailed methodology is described in the Supplementary
number and viability of cells not retained in the liver was determined with a Methods.
hemacytometer and trypan blue exclusion. The total number of cells retained
38. Schleimer, K. et al. Improved technique of heterotopic auxiliary rat liver
in DLM was the difference between the initial number of cells seeded and the transplantation with portal vein arterialization. Langenbecks Arch. Surg. 391,
number of cells present in the perfusate after seeding. 102–107 (2006).

doi:10.1038/nm.2170 nature medicine

Vous aimerez peut-être aussi