Accepted Manuscript Splenectomy attenuates murine liver fibrosis with hypersplenism stimulating hepatic accumulation of Ly-6Clo macrophages Akito Yada, Yuji Iimuro, Naoki Uyama, Yugo Uda, Toshihiro Okada, Jiro Fujimoto PII: DOI: Reference:
S0168-8278(15)00337-2 http://dx.doi.org/10.1016/j.jhep.2015.05.010 JHEPAT 5682
To appear in:
Journal of Hepatology
Received Date: Revised Date: Accepted Date:
19 January 2015 4 May 2015 5 May 2015
Please cite this article as: Yada, A., Iimuro, Y., Uyama, N., Uda, Y., Okada, T., Fujimoto, J., Splenectomy attenuates murine liver fibrosis with hypersplenism stimulating hepatic accumulation of Ly-6Clo macrophages, Journal of Hepatology (2015), doi: http://dx.doi.org/10.1016/j.jhep.2015.05.010
This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Splenectomy attenuates murine liver fibrosis with hypersplenism stimulating hepatic accumulation of Ly-6Clo macrophages.
Akito Yada, Yuji Iimuro, Naoki Uyama, Yugo Uda, Toshihiro Okada, Jiro Fujimoto.
Department of Surgery, Hyogo College of Medicine, Nishinomiya, JAPAN
Corresponding author:Yuji Iimuro MD, PhD, Department of Surgery, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, 663-8501 Japan E-mail:
[email protected] Phone: +81-798-45-6582 Fax:
+81-798-45-6581
Electronic word count: 5985 (Abstract: 250) Number of figures and tables: 8 figures, 0 table, and 3 supplementary figures Abbreviations: TAA, thioacetamide; MMP-9, matrix metalloproteinase -9; α-SMA, alpha smooth muscle actin; EpCAM, epithelial cell adhesion molecule; DAPI, 4',6-Diamidino-2-Phenylindole; HPLC, High-Performance Liquid Chromatography; CXCL9, chemokine (C-X-C motif) ligand 9; CXCR3, chemokine (C-X-C motif) receptor 3; Sfrp1, Secreted frizzled-related protein 1; Dkk2 Dickkopf 2; CTNNB1, catenin (cadherin-associated protein) beta 1; Dll4, delta like ligand 4; Jag2, jagged-2; Hes5, transcription factor HES-5; Hey1, hairy/enhancer-of-split related with YRPW motif protein 1.
Key words: splenectomy, liver fibrosis, monocyte/macrophage, progenitor cell Conflict of interest: The authors who have taken part in this study declared that they do not have anything to disclose regarding or conflict of interest with respect this manuscript.
Financial support: This study was supported in part by a Grant-in-Aid for Scientific Research (C) from the Japan Society for the Promotion of Science (JSPS), 25461966 and 22591510.
Author’s contributions: Study concept and design: AY, YI, TO, JF. Acquisition of data: AY, NU, YU. Drafting of the manuscript: AY, YI. Statistical analysis: AY. Study supervision: YI, JF. 1
Abstract Background & Aims: Splenectomy in cirrhotic patients has been reported to improve liver
function; however the underlying mechanism remains obscure. In the present study, we investigated
the mechanism using a murine model, which well represents the compensated liver cirrhosis. Methods: C57BL/6 male mice were allowed to drink water including thioacetamide (TAA: 300
mg/l) ad libitum for 32 weeks. After splenectomy at 32 weeks, mice were sacrificed on days 1, 7,
and 28, respectively, while TAA-administration was continued. Perioperative changes in peripheral
blood and liver tissues were analyzed. Results: TAA treatment of mice for 32 weeks reproducibly achieved advanced liver fibrosis with
splenomegaly, thrombocytopenia, and leukocytopenia. After splenectomy, liver fibrosis was
attenuated, and macrophages/monocytes were significantly increased in peripheral blood, as well as
in the liver. Progenitor-like cells expressing CK-19, EpCAM, or CD-133 appeared in the liver after
TAA treatment, and gradually disappeared after splenectomy. Macrophages/monocytes accumulated
in the liver, most of which were negative for Ly-6C, existed adjacent to the hepatic progenitor-like
cells, and quantitative RT-PCR indicated increased canonical Wnt and decreased Notch signals. As a result, a significant amount of β-catenin accumulated in the progenitor-like cells. Moreover, Ki67-positive relatively small hepatic cells were significantly increased. Protein expression of
2
MMP-9, to which Ly-6G-positive neutrophils contributed, was also increased in the liver after
splenectomy. Conclusions: The hepatic accumulation of macrophages/monocytes, most of which are Ly-6Clo,
the reduction of fibrosis, and the gradual disappearance of hepatic progenitor-like cells possibly play
significant roles in the tissue remodeling process in cirrhotic livers after splenectomy.
3
Introduction
Liver cirrhosis is the result of excessive accumulation of extracellular matrix in the liver, and is
one of the major causes of morbidity and mortality in many countries. Liver cirrhosis often leads to
portal hypertension and splenomegaly, and finally results in liver failure. Liver transplantation is the
only curative treatment for advanced cirrhosis. Meanwhile, splenectomy has been performed as a
palliative surgical treatment to attenuate hypersplenism [1, 2]. Recently, it has been reported that
liver function parameters improved after splenectomy, in addition to the amelioration of
thrombocytopenia in several clinical settings [3-6]. However, the mechanism underlying such a
phenomenon remains unclear, and the significant role of platelets in liver fibrosis has been
emphasized in some reports [7, 8]. One obstacle for the elucidation of the mechanism is that there
has been no suitable animal model of liver fibrosis that resembles a human situation, in which
thrombocytopenia appears in accordance with the advancement of liver fibrosis. Thus, we tried to
produce such a suitable animal model and found that thioacetamide (TAA) treatment of mice via
drinking water for a long period well reproduces thrombocytopenia in addition to advanced liver
fibrosis. In the present study, we analyzed the effect of splenectomy on liver fibrosis and its function,
and tried to investigate the underlying mechanism using this model. In this report, we indicate that
splenectomy induced the accumulation of macrophages/monocytes as well as platelets in the liver in
a TAA-induced murine liver fibrosis model, accelerating the resolution of fibrosis. The accumulated
4
macrophages/monocytes (F4/80+) were negative for Ly-6C; the macrophage subset which has been
reported to play important roles in resolution of liver fibrosis [9]. Meanwhile, the increased number
of hepatic progenitor-like cells after TAA-treatment gradually decreased after splenectomy with
up-regulated Wnt and down-regulated Notch signals in the liver.
5
Methods
Animal models
C57BL/6 male mice (4 weeks old) were obtained from Japan SLC Inc. (Shizuoka, Japan). Mice
were fed with a control chow diet and were allowed to drink water including thioacetamide (TAA;
300 mg/l, Sigma-Aldrich, St Louis, MO) ad libitum for 32 weeks. After the TAA treatment,
splenectomy was performed. The mice were sacrificed 1, 7 and 28 days after the operation, and
blood samples and liver tissues were collected. Control mice underwent sham operations without
splenectomy. TAA-treatment was continued even after splenectomy or sham operation until sacrifice.
All animals received human care, and all animal experiments were performed in accordance with the
guidelines of the Institutional Animal Care Committee, Hyogo College of Medicine.
Splenectomy
For splenectomy, the abdominal wall of mice was opened through a left subcostal minimal
incision under ether anesthesia. The splenic arteries and veins were ligated at the splenic hilum with
3-0 silk and divided. The resected spleen was removed, and the abdominal incision was closed. All
of the surgical procedures were performed under sterilized conditions. Under the sham operation, the
abdominal wall was similarly opened, and the wall was closed immediately after identifying the
spleen.
Microarray analysis
6
RNA was extracted from liver tissue using the RNeasy Mini kit (Qiagen, Hilden, Germany). RNA
degradation was analyzed by electrophoresis using a 2100 Bioanalyzer (Agilent Technologies, Santa
Clara, CA). One microgram of each total RNA sample was amplified using Ambion Amino Allyl
aRNA kit (Ambion, Carlsbad, CA). The amplified anti-sense RNAs were labeled with Cyanin5 (GE,
Amersham, Little Chalfont, Buckinghamshire, UK) and were subjected to hybridization. The
hybridization was performed using Mouse Oligo chips 24k (Toray industries Inc., Tokyo, Japan) The
signal for each gene was normalized using a global normalization method (median Cy3/Cy5 ratio =
1).
Quantitative RT-PCR (Real-time PCR)
Total RNA was extracted from liver tissues using the ISOGEN kit (NIPPON GENE, Tokyo,
Japan). Reverse transcription was carried out using the High-capacity cDNA archive kit (Applied
Biosystems, Foster City, CA). Quantitative RT-PCR was performed using the Applied Biosystems
7300 Real-Time PCR System (Applied Biosystems). Taqman gene expression assays including the
primers for Cxcl9 and Cxcr3 were purchased from Applied Biosystems. A predeveloped TaqMan
assay reagent for 18S ribosomal RNA (18S rRNA) (Applied Biosystems) was used as an internal
control. For analysis of the mRNA expression in the Wnt signaling pathway, PrimerArray® Wnt
signaling pathway (Takara Bio, Otsu, Japan) was employed. For analysis of the mRNA expression in
7
the Notch signaling pathway, RT 2 Profiler TM PCR Array Mouse Notch Signaling Pathway (Qiagen,
Hilden, Germany) was used.
Flow cytometry (FACS)
Cell surface expression analysis of immune cells was performed using the following antibodies: T
lymphocytes, B lymphocytes, monocytes and macrophages/monocytes were reacted with
anti-murine CD3e-APC, CD45R/B220-PerCp-Cy5.5, CD14-PE(BD Bioscience, San Diego, CA),
and F4/80-FITC (Biomedicals, Augst, Switzerland), respectively. Cells were incubated for 15
minutes on ice with anti-CD16/CD32 monoclonal antibody (Biolegend, San Diego, CA) to reduce
nonspecific binding. Cells were washed in buffer and incubated with each antibody for 30 min and
subjected to flow cytometry using the FACS Calibur (BD Biosciences, San Diego, CA). Liver
lymphomyeloid cells were obtained from mice liver as previously reported [10].
Histological analysis
Histological analysis was performed as previously reported [11]. Tissues were fixed in 10%
buffered formalin and embedded in paraffin. Sections were stained with hematoxylin-eosin and
examined using a light microscopy. Sirius red-fast green staining was also performed to evaluate
liver fibrosis. For the detection of activated hepatic stellate cells, a monoclonal antibody against α-SMA (Diagnostic Biosystems, Pleasanton, CA) was employed. Macrophages/monocytes or platelets were detected using an anti-F4/80 monoclonal antibody (BMA Biomedicals, Augst,
8
Switzerland) or a polyclonal anti-thrombocyte antibody (Acris Antibodies Inc., San Diego, CA),
respectively. For the detection of progenitor-like cells in the liver, a monoclonal anti-CK-19 (Abcam,
Cambridge, MA), a monoclonal anti-EpCAM (BD Biosciences, San Jose, CA), and a polyclonal
anti-CD133 (eBioscience, San Diego, CA) antibodies were used. Similarly, anti-β-catenin (Santa Cruz Biotechnology Inc, Dallas, TX), anti-Wnt-2 (Atlas Antibodies, Stockholm, Sweden), anti-Ki67
(Cell Signaling Technology Inc, Beverly, MA), or anti-MMP-9 (R&D Systems, Minneapolis, MN)
was used for each immunostaining. Frozen sections were used for MMP-9 immunostaining. For the
assessment of the number of Ki-67-positive cells, cells with Ki-67-positive nuclei were counted in
10 fields (x 200) randomly selected in each sample, and the mean values were calculated.
Image analysis
Image analysis was employed for semi-quantitative assessment of each immunostaining. At a
magnification of x40, x100, or x200, the ratio of the area positive for each immunostaining including
Sirius-red, α-SMA, F4/80, thrombocyte, CK-19, EpCAM, CD133 or MMP-9 was defined in 10
fields randomly selected in each sample using free software available from the National Institutes of
Health (Image J; http://rsb.info.nih.gov/ij). The mean value of the 10 fields was used as a
representative data point of the sample.
Immunofluorescence staining
9
Fluorescence immunohistochemistry was performed as previously reported [11]. An anti-CK19
mouse monoclonal antibody (Abcam) and an anti-F4/80 rat monoclonal antibody (BMA
Biomedicals,
Augst,
Switzerland)
were
used
to
investigate
the
relationship
between
macrophages/monocytes and CK19-positive cells in the liver. Alexa Fluor 555 tyramide (Invitrogen,
Carlsbad, CA, USA) was used to visualize the CK-19-positive cells. Alexa Fluor 488-conjugated
streptavidin (Invitrogen) was employed to visualize the F4/80-positive cells. DAPI staining was used
for the counterstaining of nuclei. In the same manner, fluorescence immunostaining was also
performed using an anti-Wnt2 mouse monoclonal antibody (Atlas Antibodies, Stockholm, Sweden),
an anti-β-catenin (CTNNB-1) antibody (Santa Cruz Biotechnology Inc.), an anti-MMP9 (R&D Systems) antibody, an anti-Ly6C antibody (Abcam), anti-CXCL9 antibody (Bioss Inc., Woburn, MA,
USA), or an anti-Ly-6G antibody (BD Biosciences). Fluorescence immunostaining was observed
using a confocal laser scanning microscope, LSM510 (Carl Zeiss, Jena, Germany).
Measurement of hydroxyproline content
Liver tissues were hydrolyzed with 12 N HCL for 12 h. After centrifugation, the hydroxyproline
content of these tissues was quantified using a High-Performance Liquid Chromatography (HPLC)
system (Nihon Bunko, Tokyo, Japan).
Statistical analysis
10
Data were expressed as the mean + SD, and the statistical significance of differences among groups
were assessed by Student’s t-test or the Mann–Whitney U-test as appropriate. P-values < 0.05 were
regarded as statistically significant.
11
RESULTS
A murine liver fibrosis model suitable for analyzing the effect of splenectomy in hypersplenism
After the treatment of mice with TAA in drinking water for 32 weeks, remarkable liver fibrosis was detected by Sirius-red staining (Suppl. Fig. 1A). Immunostaining for α-SMA also revealed a significant increase in the number of activated myofibroblasts in the liver (Suppl. Fig. 1A). The
spleen/body weight ratio was significantly increased, reaching almost twice (0.58% + 0.08) that of
normal untreated controls (0.32 + 0.01) (Suppl. Fig. 1B). Most importantly, the counts of platelets
and leukocytes were significantly decreased after the treatment, concomitant with a significant
increase in the serum concentrations of total bilirubin, resembling the situation of hypersplenism
with liver fibrosis in humans (Fig. 1D and Suppl. Fig. 1C). Additionally, the serum concentration of
albumin did not change, and the increase in the concentrations of transaminases was moderate after
TAA treatment. These results indicate that this TAA-induced murine liver fibrosis model seems to
resemble well the compensated liver cirrhosis with hypersplenism in humans as reported previously
[12].
Splenectomy attenuates the TAA-induced liver fibrosis as well as thrombocytopenia
After splenectomy, liver fibrosis was markedly attenuated (Fig. 1A). This phenomenon was also confirmed by semi-quantitative analysis of the Sirius red- or α-SMA-positive area in the liver (Fig. 1B) and by the quantitative measurement of the hydroxyplorine content in the liver (Fig. 1C). The
12
decreased counts of platelets and leukocytes in peripheral blood after TAA treatment were
dramatically increased after splenectomy with a peak around day 7, similar to the clinical situation in
humans (Fig. 1D). Interestingly, the increase in leukocytes was much more drastic (almost three
times) than that in platelets. Moreover, the increased concentration of serum total bilirubin was
significantly decreased after splenectomy, while the concentration of serum albumin did not
significantly change (Fig. 1D). While thrombocytosis has been well documented after splenectomy
in previous reports, the drastic increase in the leukocyte counts in peripheral blood was more
intriguing.
The mRNA expression of CXCL-9 is increased in the liver after splenectomy
Comparison of the mRNA expression in the TAA-treated liver before and 1 week after
splenectomy using microarray analysis revealed that only the mRNA expression of CXCL-9, a
chemokine mainly produced by macrophages or monocytes [13], was increased over 1.5-fold after
splenectomy. Real-time PCR using the mRNA extracted from liver tissues revealed a mild increase
in the mRNA expression of CXCL-9 after splenectomy (Suppl. Fig. 3A), while the mRNA
expression of CXCR3, the receptor of CXCL-9, did not change (data not shown). These results
implicated the possible participation of macrophages or monocytes during the attenuation process of
liver fibrosis after splenectomy in this model. However, double immunofluorescence staining for
13
CXCL9 and F4/80 or CXCL9 and Ly-6G revealed that the source of increased CXCL9 protein was
not macrophages/monocytes but neutrophils (Suppl. Fig. 3B) in this model.
The percentage of macrophages/monocytes is increased in the liver and peripheral blood after
splenectomy
To analyze the compositional change in the increased counts of leukocytes, we employed FACS to
analyze the peripheral and liver leukocytes. The percentage of F4/80-positive cells was significantly
increased in the peripheral blood on day 7 (Fig. 2A). In addition, the percentage of F4/80-positive
liver macrophages/monocytes was also significantly increased 7 days after splenectomy (Fig. 2B).
We also analyzed the changes in the percentage of T or B cells in peripheral and liver leukocytes.
The percentages of T or B cell in peripheral blood or in the liver did not change significantly (Fig.
2C). The percentage of other leukocytes, which consist largely of neutrophils, did not change
significantly in the peripheral blood or liver. Therefore, we thereafter focused on the dynamic change
in macrophages/monocytes in the liver tissues, rather than on the mild increase in hepatic neutrophil
infiltration, which was the source of CXCL9.
Macrophages as well as platelets accumulate in the fibrotic liver after splenectomy
To investigate the possible participation of macrophages/monocytes during the attenuation process
of liver fibrosis after splenectomy, we focused on these cells. Immunohistochemical analysis of liver
tissues revealed that F4/80-positive macrophages/monocytes (Fig. 3A) as well as platelets (Fig. 3C)
14
remarkably accumulated in the liver after splenectomy. Semi-quantitative image analysis of the
stain-positive area confirmed that the macrophages/monocytes and platelets accumulated in the liver
with a peak approximately 7 days after splenectomy, while the macrophage accumulation
predominated that of platelets (Fig. 3B and D). Interestingly, TAA treatment itself decreased the
intrahepatic accumulation of macrophages/monocytes but not platelets in this model, suggesting a
possible relationship between fibrosis and macrophages/monocytes in the liver. Intriguingly, the
detailed observation of the pattern of macrophage/monocyte accumulation in the liver revealed that
these cells predominantly existed along with the injured hepatic area including a fibrotic scar on day
7 and that they gradually became less obvious coinciding with the attenuation of liver fibrosis on day
28 (Fig. 3A). This accumulation pattern was not obvious concerning platelets on day 7 (Fig. 3C).
Appearance of hepatic progenitor-like cells after the TAA treatment
The participation of hepatic progenitor cells in the recovery process of advanced liver fibrosis has
been well documented recently [14, 15]. Thus, we investigated the existence of such cells in our
model. Immunohistochemical analysis revealed that the number of cells expressing hepatic
progenitor markers, including CK-19, EpCAM, or CD-133, was dramatically increased along with
the chronically injured hepatic area after TAA treatment (Fig. 4A). Interestingly, the number of these
cells was gradually decreased after splenectomy accompanied by the accumulation of F4/80-positive
macrophages/monocytes in the area (Fig. 4A and B). Double fluorescence immunostaining for
15
CK-19 and F4/80 revealed a close interaction between macrophages/monocytes and hepatic
progenitor-like cells in the injured hepatic area after splenectomy (Fig. 5A right), while only a few
F4/80-positive cells infiltrated into the area without splenectomy (Fig. 5A left).
The macrophages/monocytes accumulated in the liver were negative for Ly-6C One subset of macrophages responsible for the hepatic tissue remodeling, which express F4/80int Ly-6Clo, have been recently reported [9]. Thus, we analyzed whether the macrophages/monocytes
accumulated in the liver after splenectomy expressed Ly-6C in our model. Double
immunofluorescence staining for Ly-6C and F4/80 revealed that only small part of the accumulated
macrophages/monocytes expressed Ly-6C at day 7 after splenectomy (Fig. 5B middle), suggesting that these cells (Ly-6Clo) were responsible for tissue remodeling in this model. Meanwhile, a large
part of the splenic F4/80-positive macrophages expressed Ly-6C (Fig. 5B right). Intriguingly,
macrophages/monocytes marginally detected in the sham-operated liver did not express Ly-6C,
possibly reflecting no or mild acute inflammation in our TAA-induced animal model.
Canonical Wnt signaling is up-regulated after splenectomy in the TAA-treated liver
The interaction between infiltrated liver macrophages and hepatic progenitor-like cells through Wnt
signaling in murine liver fibrosis has been previously reported [16]. Thus, we analyzed changes in
Wnt signaling after splenectomy in this model. Real-time RT-PCR using mRNA obtained from
sham- or splenectomy-treated TAA-induced cirrhotic mice revealed that the mRNA expression of the
16
canonical Wnt molecules, including Wnt9b, Wnt2,
and Wnt16, dramatically increased after
splenectomy (Fig. 6A left). By contrast, the mRNA expression of Wnt4, a non-canonical Wnt
molecule, and negative regulators of Wnt signaling, including Secreted frizzled-related protein (Sfrp)
1 and Dickkopf (Dkk) 2, decreased more than 3 times after splenectomy (Fig. 6A right). These
results indicate that splenectomy up-regulates canonical Wnt signaling in the TAA-induced fibrotic
liver. Down-regulation of Notch signaling pathway is also reported in such a situation [16], so we
also performed real-time RT-PCR for Notch signals. The results showed the down-regulated Notch
signals, including Dll4, Jag2, Notch3, Notch4, Hes5, and Hey1 (Fig. 6B). To investigate the cell
sources of the canonical Wnt molecules in the liver after splenectomy, double immunofluorescence
staining for F4/80 and Wnt2 was performed. The expression of Wnt2 was markedly increased 7 days
after splenectomy, while only slight expression of Wnt2 was detected in sham-operated animals (Fig.
6C). This double immunofluorescence staining revealed that most of F4/80-positive cells expressed
Wnt2 after splenectomy, while other cells, including sinusoidal endothelial cells, were also positive
for Wnt2. Additionally, double immunofluorescence staining for CK19 and CTNNB1 (β-catenin) clearly demonstrated increased accumulation of CTNNB1 in the cytoplasm of CK19-positive
progenitor-like cells, indicating the activated canonical Wnt signaling pathway in these cells (Fig.
6D).
Ki67-positive cells increases after splenectomy in the TAA-treated liver
17
Next, we investigated whether the up-regulated Wnt signaling resulted in hepatic cell proliferation
after splenectomy in this model. TAA treatment for 32 weeks induced the occasional appearance of
Ki67-positive cells along with the injured area in the liver (Fig. 7A). After splenectomy, the number
of Ki67-positive cells started to increase along with the injured area and peaked on day 7, declining
thereafter (Fig. 7A and B). Interestingly, the number of Ki67-positive cells with relatively small
nuclei and cytoplasm was increased on days 1 and 7 after splenectomy, implying the stimulated
proliferation of immature or small hepatocytes (Fig. 7A). The mean number of Ki67-positive cells at
each time point was summarized in Fig. 7B. Co-expression of Ki67 and CTNNB1 was further
immunohistochemically analyzed in the liver (Fig. 7C). Brown-colored, Ki67-positive, relatively
small, round nuclei were merely overlapped with orange-colored CTNNB1-positive cells even after
splenectomy (white triangles), suggesting that proliferating hepatic cells were already not
CTNNB1-positive progenitor-like cells. Meanwhile, double immunohistochemistry for Ki67 and
EpCAM revealed that some of the cells with relatively small and round Ki67-positive nuclei weakly
expressed EpCAM at day 7 after splenectomy (Fig. 7D, white triangle), and that these cells existed
close or adjacent to narrow-shaped cells strongly expressing EpCAM, namely progenitor-like cells.
Expression of MMP-9 in the liver increases after splenectomy
Intrahepatic expression of MMP-9 was immunohistochemically analyzed. Expression of MMP-9
was occasionally detected after TAA treatment even without splenectomy (Fig. 8A). Additionally,
18
the expression obviously increased after splenectomy along with the chronically injured hepatic area.
Semi-quantitative image analysis revealed that the MMP-9 expression significantly increased after
splenectomy, peaking around day 7 and lasting until day 28 (Fig. 8B). Double immunofluorescence
staining for MMP-9 and F4/80 or Ly-6G revealed that MMP-9 was expressed on Ly-6G-positive
infiltrated neutrophils but not on F4/80-positive macrophages/monocytes after splenectomy (Fig. 8C
and D), suggesting a significant participation of neutrophils in the resolution process of liver fibrosis
in this model.
19
Discussion
Liver function parameters have been often improved after splenectomy, in addition to the
amelioration of thrombocytopenia in several clinical reports [3-6]; however the underlying
mechanism remains unclear. To investigate the mechanism, an appropriate animal model resembling
the clinical situation is required. In a popular liver fibrosis model induced by chronic intra-peritoneal
injection of CCl4, acute inflammation accompanied by CCl4 administration cannot be avoided [12]. Indeed, in our preliminary experiments, repeated i.p. injection with CCl4 for 8 weeks often caused peritonitis and acute liver damage, while the counts of platelets and leukocytes in peripheral blood
was rather increased compared with normal controls (data not shown), consistent with a previous
report [7]. Therefore, we used a murine liver fibrosis model induced by chronic treatment with TAA
in drinking water (300mg/l) in the present study [12]. In this model, a mild increase in ALT and AST
has been reported irrespective of the duration of TAA treatment without obvious parenchymal
damage. Moreover, thrombocytopenia in addition to liver fibrosis was well reproducible after a
32-week treatment with TAA in the present study. The spleen/body weight ratio was also
significantly increased, and even leukocytopenia was observed. Thus, we assumed that this murine
model is suitable for analyzing liver fibrosis with hypersplenism.
A significant role of thrombocytosis during the improving process of liver function after
splenectomy was suspected [7], and several reports have emphasized the role of platelets
20
accumulated in the liver or platelet-derived growth factors including HGF in this process [8, 17].
Indeed, we also observed the accumulation of platelets in the liver after splenectomy in the present
study. More intriguingly, we detected the drastic accumulation of macrophages/monocytes in fibrotic
livers after splenectomy for the first time, and the accumulation of these cells was prominent along
with the injured hepatic area. Although the sensitivity of microarray analysis employed in the present
study seemed insufficient, only CXCL9 was detected as an up-regulated gene in the liver after
splenectomy, leading us to focus on the significant role of macrophages/monocytes. Even though the
source of CXCL9 was neutrophils in our model, drastic accumulation of macrophages/monocytes in
the liver was confirmed. The accumulation of macrophages/monocytes was followed by the
resolution of liver fibrosis and cell proliferation. A similar observation was reported in a
macrophage-infusion experiment in mice, in which bone marrow-derived macrophages were
therapeutically delivered into injured fibrotic livers, improving fibrosis and stimulating regeneration
[18].
Ly-6C, a cell surface glycoprotein used to identify murine circulating monocytes, is differentially
expressed in diseased tissues, implying existence of functionally distinct macrophage population [19, 20]. As for the liver, an Ly-6Chi intrahepatic macrophage population, derived from recruitment of circulating Ly-6Chi monocytes, is critical for fibrogenesis [21]. Meanwhile, it was recently reported that an F4/80int Ly-6Clo macrophage population plays significant roles in liver fibrosis resolution [9].
21
Therefore, we analyzed whether the macrophages/monocytes accumulated in the liver after splenectomy in our model were Ly-6Chi or Ly-6Clo. The results showed that only small part of the
accumulated macrophages/monocytes expressed Ly-6C, suggesting that most of the accumulated macrophages/monocytes (Ly-6Clo) were responsible for tissue remodeling in this model. On the
other hand, most of the F4/80-positive splenic macrophages/monocytes expressed Ly-6C. From these results, it is possible to speculated that Ly-6Chi macrophages, which were increased in the circulating blood pool after splenectomy, were recruited to the liver, and differentiated to a Ly-6Clo
population, similar to the phenomenon observed in previous report [9].
In the present study, accumulated macrophages/monocytes were detected close to the hepatic
progenitor-like cells expressing CK-19, EpCAM, or CD-133. The pivotal roles of macrophages in
specifying hepatic progenitor cell fate have been reported in chronic liver injury [16]. In that report,
canonical Wnt signaling was determined to be a macrophage-derived factor that promotes the
progenitor’s specification to hepatocytes, opposing Notch signaling. In our model, the mRNA
expression of some canonical Wnt signaling molecules, such as Wnt2, Wnt9b, and Wnt16, was
dramatically increased in the liver after splenectomy. Moreover, an obvious increase in the protein
expression
of
Wnt2
was
also
histologically
confirmed,
and
many
F4/80-positive
macrophages/monocytes participated in its expression. Additionally, Notch signaling, including Dll4,
Jag2, Notch3, Notch4, Hes5, and Hey1, was markedly suppressed after splenectomy. As a result, the
22
increased protein accumulation of CTNNB1 (β-catenin) was detected in the cytoplasm of CK19-positive progenitor-like cells after splenectomy, indicating up-regulated Wnt signals in these
cells. It should be noted that cells undergoing Wnt signaling may display an overall rise in CTNNB1
protein without a clear nuclear preference in many cases [22]. The number of Ki67-positive
proliferating cells was significantly increased after splenectomy; particularly, the appearance of
relatively small-sized nuclei positive for Ki67 was prominent 7 days after splenectomy. However,
those Ki67-positive small cells were already negative for CTNNB1, implying those cells represent
transit-amplifying cells rather than hepatic progenitor cells [23]. Double immunohistochemistry for
Ki67 and EpCAM revealed that some of the cells with relatively small and round Ki67-positive
nuclei weakly expressed EpCAM, and that these cells existed close or adjacent to the narrow-shaped
EpCAM-positive progenitor-like cells. These phenomena may suggest the proliferation of immature
hepatocytes that were possibly derived from progenitor-like cells as a result of canonical Wnt-signal
activation. Although we could not provide direct evidence for such differentiation in our experiment,
the gradual decrease in the number of CK19-positive cells and increase in the number of
Ki67-positive cells after splenectomy suggest that the progenitor-like cells may possibly play
significant role in the regeneration process in cirrhotic livers after splenectomy.
How the macrophages/monocytes were recruited into the liver after splenectomy was unclear in
this study. One possible explanation is that the hepatic accumulation of these cells may be a passive
23
phenomenon in response to the prominent increase in F4/80-positive cells in the peripheral blood
after splenectomy. A significant increase in the number and percentage of monocytes and B cells in
peripheral blood has been reported in humans in earlier investigations [24-26]. The reason for this
increase has been explained such that cells homing to and circulating through the spleen accumulate
in the relatively small peripheral blood pool after splenectomy. An increased number of monocytes
in peripheral blood possibly resulted in the accumulation of these cells in the liver. The participation
of chemokines such as CCL2, a macrophage chemoattractant, in this process is strongly speculated
[18]. An alternative possible explanation for the macrophage/monocyte recruitment is that platelets
accumulated in the liver after splenectomy stimulated leukocyte recruitment into the liver. However,
immunohistochemical analysis of the liver in the present study revealed that the response after
splenectomy was much quicker in macrophages/monocytes than in platelets, and that
macrophages/monocytes accumulated mostly in the injured area but not platelets. Thus, the latter
explanation seems less persuasive. A more detailed investigation will be required to solve this issue
in the future.
In addition to cell proliferation, another important observation in the present study was the
resolution of liver fibrosis after splenectomy. We analyzed the expression of MMP-9, one of the
MMPs responsible for the degradation of the hepatic extracellular matrix [27]. The protein
expression of MMP-9 quickly increased after splenectomy and lasted until day 28. Additionally,
24
MMP-9 protein was produced by Ly-6G-positive neutrophils infiltrated into the liver, but not by
accumulated macrophages/monocytes. This phenomenon was similar to that observed in a previous
report concerning the macrophage-infusion experiment mentioned above [18, 27], implying the
important role of macrophage-mediated neutrophil recruitment in fibrosis resolution. Scar-associated
macrophages producing CXCL9 has been recently reported to contribute to the fibrosis resolution
process, expressing MMP-13 [28]. Meanwhile, neutrophils, but not macrophages, were positive for
CXCL9 in our model, and produced significant amount of MMP-9 after splenectomy. This
discrepancy is possibly derived from the difference in the animal models.
In summary, we newly detected macrophage/monocyte accumulation in the murine fibrotic liver
with hypersplenism after splenectomy, and these cells participated in the activation of the canonical
Wnt signaling pathway, which is a key component of tissue homeostasis. As a result, CTNNB1
accumulated in the neighboring hepatic progenitor-like cells. Additionally, the number of
Ki67-positive proliferating cells increased, and some of the cells were positive for EpCAM,
suggesting the important roles of progenitor-like cells in the regeneration process after splenectomy.
This mechanism, in addition to the resolution of fibrosis, possibly contributes to the ameliorating
liver function after splenectomy in cirrhotic patients with hypersplenism. It remains unclear whether
advanced liver fibrosis and portal hypertension completely disappears after radical eradication of
hepatitis viruses B or C. In such a situation, splenectomy might become a clinical choice if the
25
surgical procedure is minimally invasive and safe.
26
Acknowledgement
The authors thank Miss Keiko Mitani for her technical assistance in immunohistochemistry.
27
References
[1.]
Sugawara Y, Yamamoto J, Shimada K, Yamasaki S, Kosuge T, Takayama T, et al.
Splenectomy in patients with hepatocellular carcinoma and hypersplenism. J Am Coll Surg
2000; 190:446-450.
[2.]
Tomikawa M, Hashizume M, Akahoshi T, Shimabukuro R, Gotoh N, Ohta M, et al. Effects
of splenectomy on liver volume and prognosis of cirrhosis in patients with esophageal
varices. J Gastroenterol Hepatol 2002; 17:77-80.
[3.]
Elsebae MM, Abu-Zekri NB. A study of the effect of splenectomy on hepatic functional
reserve and structural damage in patients with chronic hepatitis C virus infection by
non-invasive serum markers. A prospective study. Int J Surg 2008; 6:362-366.
[4.]
Imura S, Shimada M, Utsunomiya T, Morine Y, Ikemoto T, Mori H, et al. Impact of
splenectomy in patients with liver cirrhosis: Results from 18 patients in a single center
experience. Hepatol Res 2010; 40:894-900.
[5.]
Ushitora Y, Tashiro H, Takahashi S, Amano H, Oshita A, Kobayashi T, et al. Splenectomy in
chronic hepatic disorders: portal vein thrombosis and improvement of liver function. Dig
Surg 2011; 28:9-14.
[6.]
Nomura Y, Kage M, Ogata T, Kondou R, Kinoshita H, Ohshima K, et al. Influence of
splenectomy in patients with liver cirrhosis and hypersplenism. Hepatol Res 2013.
28
[7.]
Watanabe M, Murata S, Hashimoto I, Nakano Y, Ikeda O, Aoyagi Y, et al. Platelets
contribute to the reduction of liver fibrosis in mice. J Gastroenterol Hepatol 2009; 24:78-89.
[8.]
Kodama T, Takehara T, Hikita H, Shimizu S, Li W, Miyagi T, et al. Thrombocytopenia
exacerbates
cholestasis-induced
liver
fibrosis
in
mice.
Gastroenterology
2010;
138:2487-2498, 2498 e2481-2487.
[9.]
Ramachandran P, Pellicoro A, Vernon MA, Boulter L, Aucott RL, Ali A, et al. Differential
Ly-6C expression identifies the recruited macrophage phenotype, which orchestrates the
regression of murine liver fibrosis. Proc Natl Acad Sci U S A 2012; 109:E3186-3195.
[10.]
Goossens PL, Jouin H, Marchal G, Milon G. Isolation and flow cytometric analysis of the
free lymphomyeloid cells present in murine liver. J Immunol Methods 1990; 132:137-144.
[11.]
Asano Y, Iimuro Y, Son G, Hirano T, Fujimoto J. Hepatocyte growth factor promotes
remodeling of murine liver fibrosis, accelerating recruitment of bone marrow-derived cells
into the liver. Hepatol Res 2007; 37:1080-1094.
[12.]
Salguero Palacios R, Roderfeld M, Hemmann S, Rath T, Atanasova S, Tschuschner A, et al.
Activation of hepatic stellate cells is associated with cytokine expression in
thioacetamide-induced hepatic fibrosis in mice. Lab Invest 2008; 88:1192-1203.
[13.]
Lalor PF, Shields P, Grant A, Adams DH. Recruitment of lymphocytes to the human liver.
Immunol Cell Biol 2002; 80:52-64.
29
[14.]
Greenbaum LE, Wells RG. The role of stem cells in liver repair and fibrosis. Int J Biochem
Cell Biol 2011; 43:222-229.
[15.]
Williams MJ, Clouston AD, Forbes SJ. Links between hepatic fibrosis, ductular reaction,
and progenitor cell expansion. Gastroenterology 2014; 146:349-356.
[16.]
Boulter L, Govaere O, Bird TG, Radulescu S, Ramachandran P, Pellicoro A, et al.
Macrophage-derived Wnt opposes Notch signaling to specify hepatic progenitor cell fate in
chronic liver disease. Nat Med 2012; 18:572-579.
[17.]
Takahashi K, Murata S, Fukunaga K, Ohkohchi N. Human platelets inhibit liver fibrosis in
severe combined immunodeficiency mice. World J Gastroenterol 2013; 19:5250-5260.
[18.]
Thomas JA, Pope C, Wojtacha D, Robson AJ, Gordon-Walker TT, Hartland S, et al.
Macrophage therapy for murine liver fibrosis recruits host effector cells improving fibrosis,
regeneration, and function. Hepatology 2011; 53:2003-2015.
[19.]
Lin SL, Castano AP, Nowlin BT, Lupher ML, Jr., Duffield JS. Bone marrow Ly6Chigh
monocytes are selectively recruited to injured kidney and differentiate into functionally
distinct populations. J Immunol 2009; 183:6733-6743.
[20.]
Nahrendorf M, Swirski FK, Aikawa E, Stangenberg L, Wurdinger T, Figueiredo JL, et al.
The healing myocardium sequentially mobilizes two monocyte subsets with divergent and
complementary functions. J Exp Med 2007; 204:3037-3047.
30
[21.]
Karlmark KR, Weiskirchen R, Zimmermann HW, Gassler N, Ginhoux F, Weber C, et al.
Hepatic recruitment of the inflammatory Gr1+ monocyte subset upon liver injury promotes
hepatic fibrosis. Hepatology 2009; 50:261-274.
[22.]
Clevers H. Wnt/beta-catenin signaling in development and disease. Cell 2006; 127:469-480.
[23.]
Okabe M, Tsukahara Y, Tanaka M, Suzuki K, Saito S, Kamiya Y, et al. Potential hepatic
stem cells reside in EpCAM+ cells of normal and injured mouse liver. Development 2009;
136:1951-1960.
[24.]
McBride JA, Dacie JV, Shapley R. The effect of splenectomy on the leucocyte count. Br J
Haematol 1968; 14:225-231.
[25.]
Durig M, Landmann RM, Harder F. Lymphocyte subsets in human peripheral blood after
splenectomy and autotransplantation of splenic tissue. J Lab Clin Med 1984; 104:110-115.
[26.]
Ferrante A, Drew PA, Kiroff GK, Zola H. Peripheral blood leucocyte subpopulations in
patients splenectomized for trauma. Clin Exp Immunol 1987; 70:158-163.
[27.]
Roderfeld M, Weiskirchen R, Wagner S, Berres ML, Henkel C, Grotzinger J, et al.
Inhibition of hepatic fibrogenesis by matrix metalloproteinase-9 mutants in mice. FASEB J
2006; 20:444-454.
[28.]
Yang L, Kwon J, Popov Y, Gajdos GB, Ordog T, Brekken RA, et al. Vascular endothelial
growth factor promotes fibrosis resolution and repair in mice. Gastroenterology 2014;
31
146:1339-1350 e1331.
32
Figure legends Figure 1. Splenectomy attenuated liver fibrosis, thrombocytopenia, and leukocytopenia. A:
Sirius-red staining of the liver tissues on days 1, 7 and 28 are shown. The original magnification was
x40.
B: Semi-quantitative analysis of the Sirius-red- or α-SMA-positive area in the liver is
summarized. C: The hydoxyproline contents in the liver (µg/g) on days 7 and 28 are shown. Perioperative changes in platelets (Plt), white blood cells (WBCs), serum total bilirubin (T.Bil), or serum albumin (s-Alb) are summarized in D. White or black bars represent the sham-operated or
splenectomy-performed mice, respectively. * P < 0.05, ** P< 0.01 compared with mice treated with
thiocetamide (TAA) for 32 weeks (TAA32W). Each experimental group contains 6-10 animals. Figure 2.
Flow cytometry analysis of peripheral or hepatic leukocytes revealed the increased
number of F4/80-positeive cells. A: Flow cytometry analysis of peripheral white blood cells. Red
circles indicate CD3-netative, CD220-negative, and F4/80-positive cells, and the percentages of these cells are summarized in the bar graphs in the right side. B: Flow cytometry analysis of hepatic
white blood cells. The percentages of CD3-negative, CD220-negative, and F4/80-positive cells are
similarly summarized. The percentages of T, B cells, or other leukocytes in peripheral and hepatic white blood cells are summarized in C. * P < 0.05 compared with sham-operated mice. Each
experimental group contains 4 animals. Figure 3. Immunohistochemical analysis of the liver indicated significant accumulation of
33
F4/80-positive macrophages/monocytes and platelets in the liver after splenectomy. A:
Immunostaining for F4/80 of the liver before and after splenectomy is shown. The original
magnifications are x 200 or x 400, as indicated. Results of the semi-quantitative image analysis of the F4/80-positive area (%) are summarized in B. Each experimental group contains 6-10 animals. White or black bars represent the sham-operated or splenectomy-performed mice, respectively. C:
Immunostaining for thrombocytes of the liver before and after splenectomy is shown. The original
magnifications are x 200 or x 400, as indicated. Results of the semi-quantitative image analysis of the thrombocyte area (%) are summarized in D. Each experimental group contains 6-10 animals.
White or black bars represent the sham-operated or splenectomy-performed mice, respectively. * P <
0.05, ** P< 0.01 compared with mice treated with thiocetamide (TAA) for 32 weeks (TAA32W). Figure 4. Immunohistochemical analysis of progenitor-like cells in the liver revealed the increased
number of progenitor-like cells after TAA treatment and their gradual disappearance after splenectomy. A: Immunostaining of the livers for CK-19, EpCAM, or CD-133 before and after splenectomy is shown. The original magnification was x 100. B: Semi-quantitative analysis of each
immunostainig (%) is summarized in B. Each experimental group contains 6-10 animals. White or
black bars represent the sham-operated or splenectomy-performed mice, respectively. * P < 0.05, **
P< 0.01 compared with mice treated with thiocetamide (TAA) for 32 weeks (TAA32W). ## P < 0.01
compared with sham-operated mice.
34
Figure 5. Double fluorescence immunostaining revealed the close interaction between
F4/80-positive macrophages/monocytes and CK-19-positive progenitor-like cells in the liver.
Representative figures in sham-operated control (Fig. 5A left) and splenectomy-performed mice (Fig.
5A right) on day 7 are shown. The original magnification was x 400. Double fluorescence
immunostaining for Ly-6C and F4/80 is shown in Fig. 5B. Only small part of the accumulated
macrophages/monocytes expressed Ly-6C at day 7 after splenectomy (Fig. 5B middle), while a large
part
of
the
splenic
F4/80-positive
macrophages
expressed
Ly-6C
(Fig.
5B
right).
Macrophages/monocytes detected in the sham-operated liver at day 7 did not express Ly-6C (Fig. 5B
left). The original magnification was x 400. Figure 6. Canonical Wnt signaling was up-regulated in the liver after splenectomy. A: Quantitative
RT-PCR for genes included in the Wnt signaling pathway revealed the up-regulated mRNA
expression of Wnt2, 9b, and 16 after splenectomy. Additionally, the mRNA expression of a
non-canonical Wnt molecule, Wnt4, or negative regulators of Wnt signaling, including Secreted
frizzled-related protein (Sfrp) 1 and Dickkopf (Dkk) 2, was decreased. White or black bars represent
the sham-operated or splenectomy-performed mice, respectively. Black bars represent mean + SD of three distinct experiments. B: Quantitative RT-PCR for genes included in the Notch signaling
pathway revealed the down-regulated mRNA expression of Dll4, Jag2, Notch3, Notch4, Hes5, and Hey1 after splenectomy. C: Double fluorescence immunostaining for Wnt2 and F4/80 of the liver in
35
sham-operated or splenectomy-performed mice is shown. Splenectomy increased the protein
expression of Wnt2, and most of the F4/80-positive cells participated in the expression. The original
magnification was x 400 or x 1200, as indicated. The area outlined with a square is enlarged on the right side. D: Double fluorescence immunostaining for CK-19 and β-catenin indicated that a significant amount of β-catenin accumulated in the CK-19-positive cells after splenectomy. The original magnification was x 800. Figure 7. Splenectomy stimulated the proliferation of relatively small hepatocytes. A:
Immunostaining for Ki67 of the liver is shown. The number of Ki67-positive nuclei was markedly
increased in the liver after splenectomy. The original magnification was x 200 or x 400, as indicated.
Perioperative changes in the mean number of Ki67-positive nuclei in high power fields (HPFs x200) are summarized in B. White or black bars represent the sham-operated or splenectomy-performed
mice, respectively. Each experimental group contains 6-10 animals. * P < 0.05 compared with
untreated normal mice. # P < 0.05, ## P < 0.01 compared with mice treated with thiocetamide (TAA) for 32 weeks (TAA32W). C: Double immunostaining for β-catenin and Ki67 indicated that cells with Ki67-positive nuclei merely overlapped with β-catenin-expressing cells. White triangles indicate relatively small hepatocytes with Ki67-positive nuclei. The original magnification was x 400. D: Double immunostaining for EpCAM and Ki67 indicated that some of the cells with
relatively small and round Ki67-positive nuclei expressed EpCAM at day 7 after splenectomy (Fig.
36
7D, white triangle). The area indicated by rectangle in x 400 (left) was enlarged in X 1000 (right).
The original magnification was x 400 or x100, as indicated. Figure 8.
Splenectomy up-regulated MMP-9 protein expression in the liver. A: Immunostainings
for MMP-9 of the liver are shown. The original magnification was x 200 or x 400, as indicated. Semi-quantitative image analysis of MMP-9 protein expression (%) is summarized in B. White or
black bars represent the sham-operated or splenectomy-performed mice, respectively. Each group
contains 6-10 animals. ** P< 0.01 compared with mice treated with thiocetamide (TAA) for 32
weeks (TAA32W). ## P < 0.01 compared with sham-operated mice. Double fluorescence
immunostaining for MMP-9 and F4/80 (C) or MMP-9 and Ly-6G (D) revealed that Ly-6G-positive
neutrophils participated in the MMP-9 expression after splenectomy. The original magnification was
x 400.
37
Percentage of liver fibrosis area αSMA % % Sirius-red 30
14
*
*
12
Sham op
25
10
20
8
X40
15
6
*
*
4
splenectomy
10 *
2
0
×103/㎜3
1200
250
1000 200 **
**
30000 **
*
**
0.07
20000
0.06
800
50
200
0
0 Day7
day 28
day 7
day 1
3.5 3 **
2.5 **
0.05
*
0.04
1
0.02 5000
0.5
0.01
0
0
0
2 1.5
0.03
10000
400
*
0.09
25000
600
100
g/dl
4
0.08
15000
150
0.1 **
**
1400
300
㎎/dl
/㎕
35000
1600
350
hydroxyplorin
TAA 32w
D
μg/g
*
5
0
C
*<0.05
day28
B
Day28
day7
Day7
day1
Day1
TAA32W
A
Day28
Plt
WBC
T.Bil
S-Alb
Yada et al. Figure 1
A
splenectomy
sham
%
B
blood
splenectomy
sham
%
*
12
30
10
25
8
20
6
15
4
10
2
5
0
0
% T cell - blood
25 20 15 10
5 0
% T cell - liver
25 20
15 10 5 0
%
B cell - blood
*
*<0.05
*<0.05
C
liver
% B cell - liver
% Other cells - blood
% Other cells - liver
50
50
50
45
45
45
40
40
40
35
35
35
30
30
30
25
25
25
20
20
20
15
15
15
15
10
10
10
10
5
5
5
0
0
0
35 30 25 20
5 0
Yada et al. Figure 2
A
B Day1
TAA32W
*<0.05 **<0.01
%
4.5
TAA32W
**
4 3.5
* **
3 X200 Day7
X400
X200
2
Day7
Day28
2.5
1.5 1 0.5 X200
X400
X200
C Day1
TAA32W
0 normal TAA32W Day1
Day7
D 3%
TAA32W
*<0.05
*
2.5
*
2 X200 Day7
X200
X400
Day28
1.5
Day7
Day28
1 0.5
X200
X200
X400
0 normal TAA32W Day1
Day7
Day28
Yada et al. Figure 3
A
normal
TAA32W
Day1
Day7
Day28
CK19
x100
x100
x100
x100
x100
x100
x100
x100
x100
x100
x100
x100
x100
x100
x100
EpCAM
CD133
%
6 *
5 ##
##
**
**
4 ##
3
**
2
## **
1
EpCAM
Day28
Day7
Day1
Day28
0 TAA32W
CK-19
**
7
*
Day7
Day7
Day1
**
##
Day28
##
%
Day1
*
*
9 8 7 6 5 4 3 2 1 0
TAA32W
%
TAA32W
B
18 16 14 12 10 8 6 4 2 0
* <0.05
**<0.01
## <0.01
CD-133
Yada et al. Figure 4
A
CK19
F4/80
CK19
F4/80
DAPI
MERGE
DAPI
MERGE
X400
TAA32w + sham (day 7)
B
Ly6C
DAPI
X400
TAA32w + splenectomy (day 7) F4/80
MERGE
X400
TAA32w + sham; liver (day 7)
Ly6C
F4/80
Ly6C
F4/80
DAPI
MERGE
DAPI
MERGE
X400
TAA32w + splenectomy; liver (day 7)
X400
TAA32W; spleen
Yada et al. Figure 5
A
C Wnt9b 30
Wnt4
TAA32w + sham (day 7)
TAA32w + splenectomy (day 7)
Wnt2
F4/80
Wnt2
F4/80
DAPI
MERGE
DAPI
MERGE
1.0
20 0.5 10 0.0
0
Wnt2 20 16 12 8 4 0
Sfrp1 1.0
0.5
X400
X400
X1200
0.0
Wnt16 15
MERGE
Dkk2 1.0
D
TAA32w + sham (day 7) CK19
TAA32w + splenectomy (day 7) CTNNB1
CK19
CTNNB1
10 0.5 5 0
0.0
B 1
DAPI
MERGE
DAPI
MERGE
0.5
0
X800
X800
Yada et al. Figure 6
A
B TAA32W
normal
35
Day1
* ##
30
X200
X200
Day7
Day28
X200 Day7
/ x200 HPFs
25
*#
20
*
*
* *
*
15 10 5 0
X200
C
X200
D
Sham Day7
X400
Sham Day7
CTNNB1 (orange), Ki67(brown)
Day28 # <0.05
## <0.01
Sham Day7
X1000
X400 Splenectomy Day7
X400
Day7
* <0.05
X400 Splenectomy Day7
normal TAA32W Day1
Splenectomy Day7
X400 EpCAM (orange), Ki67(brown)
X1000
Yada et al. Figure 7
A
B
MMP-9 TAA32W
normal
Day1
MMP-9
% 2
** ## ** ##
1.5
X200
X200
Day7
X200
Day2 8
** ##
1
Day7 0.5
0 X200
X200
C
X400
normal TAA32W Day1
Day28
**<0.01, ## <0.01
MMP-9
F4/80
DAPI
MERGE
D
MMP-9
Ly-6G
DAPI
MERGE
X400
TAA32w + splenectomy (day 7)
Day7
X400
TAA32w + splenectomy (day 7)
Yada et al. Figure 8