Setanaxib

Activated Hepatic Stellate Cells and Portal Fibroblasts contribute to cholestatic liver fibrosis in MDR2 knockout mice

Authors: Takahiro Nishio1, Ronglin Hu1, Yukinori Koyama1,3, Shuang Liang1, Sara B. Rosenthal1, Gen Yamamoto1, Daniel Karin1, Jacopo Baglieri1, Hsiao-Yen Ma1, Jun Xu1, Xiao Liu1, Debanjan Dhar1, Keiko Iwaisako4, Kojiro Taura3, David A. Brenner1, Tatiana Kisseleva2,*

1Department of Medicine, and 2Department of Surgery, University of California San Diego, La Jolla, California, USA.
3Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan. 4Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Japan.
*Correspondence

Corresponding author: Tatiana Kisseleva

9500 Gilman Drive, #0063, La Jolla, California 92093, USA. Phone: 858.822.5339
E-mail: [email protected]

Key words: cholestatic fibrosis, activated Hepatic Stellate Cells, activated Portal Fibroblasts Electric word count: 5,647 words (including abstract, main text, and references)
Number of figures and tables: 7 Figures (10 Supplementary figures and 4 Supplementary tables)

Conflict of interests: nothing to declare

Grant support: Supported by the National Institutes of Health R01 DK101737-01A1, U01 AA022614-01A1, R01 DK099205-01A1, P50AA011999, AI043477 (T.K. and D.A.B.); and Herman Lopata Memorial Hepatitis Postdoctoral ALF Fellowship (J.X.).

Authors contributions:

T.N. performed the experiments, collected and analyzed the data, and wrote the manuscript. R.H., S.L., Y.K., G.Y., D.K., J.B., H-Y.M., J.X., X.L., and D.D. performed experiments. S.B.R. analyzed the data. K.I. and K.T. provided support with data collection. T.K. and D.A.B. provided support, designed the study, and wrote the manuscript.

Abstract
Background and Aims: Chronic liver injury often results in activation of hepatic myofibroblasts and development of liver fibrosis. Hepatic myofibroblasts may originate from three major sources; Hepatic Stellate Cells (HSCs), Portal Fibroblasts (PFs), or fibrocytes. Their contribution to liver fibrosis varies dependent on the etiology of liver injury. Here we assessed the composition of hepatic myofibroblasts in multidrug resistance gene 2 knockout (Mdr2-/-) mice, a genetic model that resembles primary sclerosing cholangitis in patients.
Methods: Mdr2-/- mice expressing a collagen-GFP reporter were analyzed at different ages.

Hepatic nonparenchymal cells isolated from collagen-GFP Mdr2-/- mice were sorted based on

collagen-GFP and vitamin A. A nicotinamide adenine dinucleotide phosphate oxidase (NOX) 1/4

inhibitor was administrated to Mdr2-/- mice during ages 12- to 16-weeks-old to assess the

therapeutic approach targeting oxidative stress in cholestatic injury.

Results: Thy1+ activated PFs (aPFs) accounted for 26%, 51%, and 54% of collagen-GFP+ myofibroblasts in Mdr2-/- mice at 4 weeks, 8 weeks, and 16 weeks of age, respectively. The remaining collagen-GFP+ myofibroblasts were composed of activated HSCs (aHSCs), suggesting
that PFs and HSCs are both activated in Mdr2-/- mice. BM-derived fibrocytes minimally

contributed to liver fibrosis in Mdr2-/- mice. Development of cholestatic liver fibrosis in Mdr2-/-

mice was associated with early recruitment of Gr1+ myeloid cells and upregulation of pro-inflammatory cytokines (4 weeks). Administration of a NOX inhibitor to 12-week-old Mdr2-/- mice suppressed activation of myofibroblasts and attenuated development of cholestatic fibrosis.
Conclusions: aPFs and aHSCs contribute to cholestatic fibrosis in Mdr2-/- mice, and serve as

targets for anti-fibrotic therapy.

Lay summary

Activated Portal Fibroblasts and Hepatic Stellate Cells, but not fibrocytes, contributed to the production of the fibrous scar in livers of Mdr2-/- mice, and these cells can serve as targets for anti-fibrotic therapy in cholestatic injury. Therapeutic inhibition of the enzyme nicotinamide
adenine dinucleotide phosphate oxidase (NOX) in Mdr2-/- mice reversed cholestatic fibrosis,

suggesting that targeting of NOXs can provide a novel strategy for treatment of cholestatic fibrosis.

Introduction

Chronic liver injury often results in liver fibrosis. Development of liver fibrosis is associated with migration and proliferation of Collagen Type I producing myofibroblasts, which are not present in the normal liver. Activated myofibroblasts originate from three major sources, hepatic stellate cells (HSCs), portal fibroblasts (PFs), or fibrocytes1-4. Activated HSCs (aHSCs) were implicated in the pathogenesis of experimental toxic liver fibrosis, such as chronic CCl4 administration and alcoholic liver disease1,5, while PFs are predominantly activated in response to cholestatic liver fibrosis, such as bile duct ligation (BDL)2.

Several experimental models of cholestatic liver injury have been developed6. BDL causes rapid activation of PFs, especially at the onset of injury. Although the pathology resulting from BDL resembles that seen in human chronic cholestatic disease, the surgical stress and the severity of cholestatic injury limits the utility of the BDL model. The multidrug resistance gene 2 knockout (Mdr2-/-, also known as Abcb4-/-) mouse is another well-established model of chronic cholestatic liver injury. Deficiency of Mdr2, a canalicular phospholipid flippase, disrupts biliary phospholipid secretion, leading to the increase of potentially toxic bile acid which induces hepatocyte damage and cholangiopathy7-9, which is characterized by pericholangitis and onion skin-type periductal fibrosis, resembling the pathological features of primary sclerosing cholangitis (PSC)10,11. Despite extensive studies12,13, the contribution of aHSCs and activated PFs (aPFs) to cholestatic fibrosis in Mdr2-/- mice has not been defined.

Under physiological conditions, quiescent HSCs (qHSCs) reside in the space of Disse (which is located between hepatocytes and sinusoidal endothelial cells)1 store Vitamin A, and serve as liver

pericytes. qHSCs express specific markers, such as glial fibrillary acid protein (GFAP), synaptophysin, nerve growth factor p75 (NGFR1), and lecithin retinol acyltransferase (Lrat)2,14,15. In response to toxic liver injury, HSCs downregulate expression of Vitamin A in lipid droplets, migrate to the pericentral areas, and transdifferentiate into Collagen Type I and  smooth muscle actin (SMA) expressing myofibroblasts.

PFs, which reside around the portal area and maintain the integrity of the biliary tree and portal tract16, comprise a small population of fibroblasts in the liver under physiological conditions. In response to cholestatic injury, PFs are activated, proliferate, and contribute to Collagen Type I deposition. aPFs can be distinguished from aHSCs by expression of Thy1, fibulin 2, elastin, Gremlin 1, ecto-ATPase nucleoside triphosphate diphosphohydrolase 2, mesothelin (Msln), and mucin 16 (Muc16)2,14,16,17, and the lack of HSC markers.

Therapeutic approach to cholestatic fibrosis remains challenging; hence liver transplantation is the only effective therapy for patients with late-stage PSC18. Activity of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX), an enzyme system that catalyzes the reduction of molecular oxygen to superoxide, plays an important role in activation of HSCs and development of hepatic fibrosis19-22, and treatment with a NOX1/4 dual inhibitor decreases both CCl4-induced hepatotoxic fibrosis and BDL-induced cholestatic fibrosis23.

Here we characterize the development of cholestatic fibrosis in Mdr2-/- mice. We determined that aPFs and aHSCs (but not fibrocytes) contribute to hepatic myofibroblasts in Mdr2-/- mice during progression of cholestatic fibrosis. Increased expression of fibrogenic genes was associated with

upregulation of Thy1 and CD34 in aPFs. Although activated PFs were in close proximity to proliferating cholangiocytes, cholangiocytes themselves did not express Collagen Type I. Hepatic expression of NOX was induced in Mdr2-/- mice only upon development of liver fibrosis. Therapeutic inhibition of NOX1/4 in Mdr2-/- mice reversed cholestatic fibrosis, suggesting that targeting of NOXs can provide a novel strategy for treatment of cholestatic fibrosis.

Materials and Methods

Mice

BALB/c-Mdr2-/- mice (gift of Dr. Frank Lammert)24 were crossed with Collagen-α1(I)-GFP

(ColGFP) mice25. LratCre mice (gift of Dr. Robert Schwabe)15 were crossed with

Rosa26flox-stop-flox-YFP reporter mice (Jackson Laboratory). Mice were housed and maintained

under specific pathogen-free conditions in a standard environment with a 12-hour light–dark cycle, and fed a diet of normal chow ad libitum, at the animal facilities of UC San Diego under protocol S07088, approved by the Institutional Animal Care and Use Committee.

Development of liver fibrosis and treatment with NOX1/4 inhibitor

Mdr2-/-ColGFP mice (male, BALB/c, n=5-7/group) were sacrificed at 3, 4, 8, 12, and 16 weeks of age. Mdr2-wild type (WT) ColGFP mice (male, BALB/c, n=5) were subjected to BDL (5days) or administration of CCl4 (1:4 in corn oil, 200µl, 2 x week, for 3 weeks) The NOX1/4 inhibitor (GKT137831, Genkyotex S.A., France)23 was administered in vivo (20mg/kg or 60mg/kg, or vehicle, 20 doses, oral gavage) to Mdr2-/- mice (male, BALB/c, n=6-8/group) from age 12- to 16-weeks-old.

Bone marrow transplantation

Mdr2-/- mice (male, BALB/c, 4 weeks old, n=6-8/group) were lethally irradiated (12 Gy), and i.v. reconstituted with the donor ColGFP bone marrow cells (1 x 107). The recipient Mdr2-/- mice were sacrificed at 16 weeks of age.

Histology and immunohistochemistry

Formalin-fixed livers were embedded in paraffin or OCT compound, and stained with Sirius Red or antibodies for immunohistochemistry. Positive area was quantified using Image J (see Supplementary materials).

Flow cytometry

Nonparenchymal cell fraction was isolated from Mdr2-/-ColGFP mice (4-, 8-, and 16 weeks old) using pronase/collagenase method as described26. GFP+VitaminA+ aHSCs and GFP+VitaminA- aPFs were quantified and sort purified using FACSAria II (BD Biosciences).

Quantitative real-time polymerase chain reaction (qPCR) and RNAseq analysis

Total RNA was extracted using RNeasy columns (Qiagen). Expression levels of selected genes (for primers, see Supplementary Table 1) were calculated after normalization to HPRT by using the ∆∆Ct method. RNAseq analysis was performed using the nonparenchymal cell fraction which was sort purified from 16-week-old Mdr2-/-ColGFP mice (see Supplementary materials).

Western blot

Western blot analysis was performed on liver tissue lysates that were separated by sodium dodecyl sulfate/polyacrylamide gel electrophoresis and transferred to nitrocellulose membranes (see Supplementary materials).

Statistics

Data are mean ± SD. Differences between groups were compared using ANOVA, followed by

the Tukey–Kramer test. A p value of less than 0.05 was considered significant. The JMP Pro 11 (SAS institute, Cary, NC, USA) software was used for all statistical analyses.

Results

Development of fibrosis in Mdr2-/- mice is associated with inflammation, oxidative stress

and ductular reaction.

Progression of cholestatic liver fibrosis was analyzed in 4-, 8-, 12-, and 16-week-old Mdr2-/-

mice. Extracellular matrix (ECM) deposition was observed in livers of Mdr2-/- mice as early as 4 weeks of age, and reached maximum at 12-16 weeks, as demonstrated by Sirius Red staining (Fig. 1A-B), which was accompanied by expression of fibrogenic genes (Col1a1, SMA, Desmin, TIMP1, and TGFR1 mRNA, Fig. 1C) and inflammatory genes (F4/80, IL-1, and IL-6
mRNA, Fig. 1D). Age-dependent ECM deposition in Mdr2-/- mice correlated with increased

upregulation of aHSC markers (Desmin, -SMA, and TIMP1 mRNA), and aPF markers (Thy1, Msln, Muc16, CD34, and Fibulin 2 mRNA, with the exception of Elastin, in which the expression peaked at 4 weeks of age, Fig. 1E). Expression of NOX genes (NOX1, NOX2 and NOX4 mRNA, Fig. 1F) and cholangiocyte markers (CK19 and Sox9 mRNA, Fig. 1G) followed
a similar trend, suggesting that activation of hepatic myofibroblasts in Mdr2-/- mice positively

correlates with the age-dependent development of oxidative stress, hepatic inflammation, and ductular proliferation.

Collagen Type I expressing myofibroblasts emerge in the livers of Mdr2-/- mice.

To visualize activated myofibroblasts, Mdr2-/- mice were crossed with Collagen1-(I)-GFP

reporter mice (in which all Collagen Type I expressing cells are labeled in real time with GFP)25 to generate Mdr2-/-ColGFP mice. Myofibroblasts first emerged in the livers of 4-week-old Mdr2-/-ColGFP mice (and contributed to 5.2% of total liver area), and were increased depending

on the progression of fibrosis (up to 10.3% area of total liver area at 8 weeks old, and 11.6% at 16 weeks old, Fig. 1H-I). Next, the composition of hepatic myofibroblasts was analyzed in Mdr2-/-ColGFP mice.

Thy1-GFP+ and Thy1+GFP+ myofibroblasts comprise the major populations of fibrogenic myofibroblasts in the livers of Mdr2-/-ColGFP mice.
Livers from 4-, 8-, and 16-week-old Mdr2-/-ColGFP mice were immunostained for the aPF marker Thy1. Compared to the total GFP+ myofibroblast area (considered as 100%), Thy1+ aPFs were progressively activated (ranging from 26% to 54% in the livers of 4 and 16-week-old Mdr2-/- mice, Fig. 1I-J). In turn, the number of Thy1-GFP+ cells increased with age in Mdr2-/- mice, but their relative contribution to myofibroblasts declined (from 73% to 45% of total GFP+
myofibroblasts in the livers of 4 and 16 week-old Mdr2-/- mice, respectively), suggesting that

aHSCs and aPFs are the major contributors to ECM producing myofibroblasts in Mdr2-/- mice.

Similar results were obtained using flow cytometry for Vitamin A (Vit.A) and Collagen1-(I)-GFP myofibroblasts2. aHSCs were identified by simultaneous expression of Vit.A and ColGFP, while GFP+ aPFs lacked expression of Vit.A. Vit.A+GFP+ aHSCs comprised 68% of GFP+ myofibroblasts (100%) in the livers of 4-week-old Mdr2-/- mice, but only 41% in livers of 16-week-old Mdr2-/- mice. This effect was attributed to rapid expansion of Vit.A-GFP+ aPFs (Fig. 1K-L), since their contribution to total GFP+ myofibroblasts was increased from 32%
in livers of 4-week-old Mdr2-/- mice to 57-59% in livers of 16-week-old Mdr2-/- mice. We

concluded that both aHSCs and aPFs critically contribute to cholestatic fibrosis. Age-dependent activation of aHSCs and aPFs was further evaluated using histological analysis of Mdr2-/- mice.

Thy1+ aPFs serve as a significant source of ECM in Mdr2-/- mice and human cholestatic

injury.

Pathological features of liver injury were first examined in 4-week-old Mdr2-/- mice. The onset of cholestatic fibrosis was associated with accumulation of Thy1+ and Desmin+ cells around the portal area, and displayed typical “onion skin-like” fibrosis surrounding PanCK+ and Sox9+ cholangiocytes (Fig. 2A). We also observed proliferation of small Sox9+ bile ducts around Thy1+ aPFs. Immunostaining of serial sections revealed that the area of ECM deposition overlapped mostly with the Thy1+ area, suggesting that aPFs significantly contribute to the fibrous scar at 4 weeks of age.

At 8 weeks of age, areas positive for Thy1, Desmin, and -SMA were markedly enlarged and correlated with increased ECM deposition in portal areas of these mice (Fig. 2B). At this age, expression of PanCK and Sox9 mostly overlapped, and was indicative of maturation of proliferating bile ducts. Similar, but more severe pathology was observed in livers of
16-week-old Mdr2-/- mice, and was associated with formation of a more compact fibrous scar

and further activation of Thy1+ and Desmin+ myofibroblasts, whereas the immunoreactivity for αSMA was observed in the portal area, e.g. in fibrotic regions and in basal membranes underlying biliary epithelium (Fig. 2C).

To support our findings in Mdr2-/- mice, we validated the contribution of aPFs to myofibroblasts in human cholestatic disease using immunostaining of human liver sections (Supplementary Fig. 1). In human livers with cholestatic injury, Thy1+ cells accumulated in the area corresponding to

ECM deposition and surrounding the CK19+ cholangiocytes. Notably, the Thy1+ area mostly overlapped with Msln+ area, indicating that Thy1+Msln+ aPFs contribute to human cholestatic fibrosis.

aPFs surround the bile duct and express Desmin and Thy1.

Previous reports suggested that injured cholangiocytes can produce Collagen Type I27. Here we examined if PanCK+ and/or Sox9+ bile ducts upregulate Collagen Type I in 8-week-old Mdr2-/-ColGFP mice. Despite close proximity to Thy1+ aPFs, neither PanCK+ nor Sox9+ cholangiocytes expressed collagen1-(I)-GFP (Fig. 2D-E), indicating that cholangiocytes do not contribute to ECM deposition. The majority of Thy1+ aPFs accumulated in the portal area, while Desmin+GFP+ aHSCs were mostly located in the hepatic acini or capsular areas (Fig. 2F). Surprisingly, Desmin positive staining was also observed in the portal areas populated with aPFs (Fig. 2G). Furthermore, immunoreactivity for Desmin and Thy1 was co-localized in 30-40% of GFP+ portal myofibroblasts, while parenchymal and sinusoidal GFP+ HSCs were stained positive only for Desmin. (Fig. 2H). Similar results were obtained in 4 and 16-week-old Mdr2-/- mice, suggesting that anti-Desmin antibodies might not discriminate between aHSCs and aPFs in livers of Mdr2-/- mice.

To address this question, cholestatic injury was induced in HSC-specific LratYFP mice15 (generated by crossing LratCre and Rosa26YFP mice) via ligation of the common bile duct (BDL, 5 days). Similarly, LratYFP-labelled cells were stained positive not only for Desmin+Thy1- HSCs, but also for Desmin-Thy1+ aPFs (Supplementary Fig. 2), and exhibited a distribution pattern similar to that observed in Mdr2-/- mice. Our data imply that LratCre mice are suited for lineage

tracing of both aHSCs and aPFs (due to Lrat expression during embryonic development). We also concluded that a subset of Mdr2-deficient aPFs upregulate Desmin. Therefore, additional markers that discriminate aPFs from aHSCs are needed.

CD34+ aPFs contribute to cholestatic fibrosis in Mdr2-/- mice.

In an attempt to identify additional markers of aPFs that can be used for immunohistochemical studies, expression of CD34 on aPFs and aHSCs was evaluated. Transmembrane phosphoglycoprotein CD34, a member of the sialomucin family, is expressed by a variety of cells, including hematopoietic and endothelial cells, as well as mesenchymal progenitors and aPFs2. Here we demonstrate that CD34+ was strongly expressed in ColGFP+ aPFs upon
development of cholestatic fibrosis in Mdr2-/- mice (Fig. 3A-B, Supplementary Fig. 3A).

Upregulation of CD34 mRNA also positively correlated with increased expression of other aPF-specific markers (Thy1, Msln, Muc16, Fibulin 2, Fig. 1D). CD34+ cells accumulated predominantly in the portal (57%) and sinusoidal (24%) areas, and were scattered throughout the capsular (17%) area (Fig. 3C, Supplementary Fig. 4A). CD34+ staining did not overlay with PanCK+ and/or Sox9+ cells (Fig. 3D-E), suggesting that CD34 marks a population of aPFs in the
livers of Mdr2-/- mice. In support, a population of CD34+ aPFs was detected in the livers of

BDL-injured (but not CCl4-injured) ColGFP mice (Fig. 3F-G). Our data are in concordance with previous reports demonstrating that aPFs minimally contribute to CCl4-induced liver fibrosis2 (Supplementary Fig. 4B). The majority of CD34+ cells co-expressed Thy1 (63%) and Desmin
(48%), and exhibited a fibroblast-like spindle shape in the livers of Mdr2-/- mice (Fig. 3H-I,

Supplementary Fig. 3A). Hence, a small fraction of CD34+GFP+ cells (4%) exhibited a fibrocyte-like shape (Fig. 3J), and co-expressed CD45 and F4/80 (Fig. 3K-L), suggesting that

these cells might derive from BM fibrocytes.

aHSCs exhibit a more fibrogenic phenotype than aPFs.

To further dissect the contribution of aHSCs and aPFs to fibrosis, Vit.A+GFP+ aHSCs and Vit.A-GFP+ aPFs (with high [hi] and low expression of GFP, for comparison based on the degree
of activation) were sort purified (Fig. 4A) from livers of 16-week-old Mdr2-/- mice, and their

gene expression profile was analyzed by RNA-Seq. As expected, Vit.A+GFP+ aHSCs expressed typical HSC markers (Lrat, Pdgfrb, Desmin, GFAP), and lacked expression of aPFs (Msln, Glipican 3, Fibulin 2, Uroplakin 1b, Fig. 4B). In turn, Vit.A-GFPhi aPFs expressed markers of aPFs (but lacked Desmin, Lrat, and GFAP). Slight upregulation of Thy1, Wt1 and CD34 mRNA was observed in aHSCs compared to qHSCs, while these markers were downregulated in Vit.A-GFPlow aPFs compared to Vit.A-GFPhi aPFs. In addition, expression of Msln, Glipican 3, Fibulin 2, Uroplakin, Vimentin, Col6a1, Col5a2 mRNA and other genes was also reduced in Vit.A-GFPlow aPFs (Fig. 4C), suggesting that Vit.A-GFPlow aPFs exhibit a less activated phenotype compared to Vit.A-GFPhi aPFs, consistent with their higher Collagen GFP expression. Despite these differences, the majority of genes were similarly expressed in Vit.A-GFPhi and Vit.A-GFPlow aPFs (Fig. 4D-E), which suggests that these two populations originate from the same cell type at different stages of activation. In support, expression of CD34 has been linked to maturation of tissue fibroblasts28.

Furthermore, high expression of Early Growth Response 1 (Egr1), intermediate expression of Upk1bm Fbln2, and Msln, and low expression of CD34 and Thy1 mRNA distinguished Vit.A-GFPlow aPFs from Vit.A-GFPhi aPFs and Vit.A+GFP+ aHSCs (Fig. 4F). When

REACTOME pathways were compared, aHSCs and both populations of aPFs expressed genes associated with ECM deposition (see Supplementary Table 2-4), which is consistent with their myofibroblast phenotypes. Hi expression of Col1a1, Col1a2, and other fibrogenic genes was observed mostly in aHSCs. Unlike BDL-activated aPFs2, Vit.A-GFPhi aPFs in MDr2-deficient mice expressed less Col1a1 than aHSCs (Fig. 4H), but almost exclusively upregulated Bmp1, Col27a1, Col11a2, Col4a3, and Tll1 mRNA (Fig. 4G). Based on the gene expression profile, we concluded that aHSCs serve as a major source of ECM in the livers of 16-week-old Mdr2-/- mice.

Minor contribution of bone marrow-derived fibrocytes in Mdr2-/- mice.

CD34+ fibrocytes were implicated in the pathogenesis of cholestatic fibrosis in Mdr2-/- mice29. In

accord, a small number of ColGFP+ cells co-expressing CD34+, CD45, and F4/80 (2-4%),

designated as fibrocytes, were scattered throughout portal, sinusoidal, and capsular areas in the livers of Mdr2-/- mice (Fig. 5A-C, Supplementary Fig. 3B). To directly assess the contribution of bone marrow-derived fibrocytes to Collagen Type I producing cells/myofibroblasts in livers of
Mdr2-/- mice, bone marrow (BM) from donor ColGFP mice was transplanted into lethally

irradiated recipient 4-week-old Mdr2-/- mice. Recipient Mdr2-/- mice were sacrificed at 16 weeks of age, and livers were analyzed for the presence of GFP+ and CD45+ and F4/80+ cells (Fig. 5D). Although BM-derived ColGFP+ fibrocytes migrated to the livers of recipient Mdr2-/- mice, they retained a fibrocyte-like round shape, and contributed to neither Desmin+ nor CD34+ myofibroblasts (Fig. 5E-F, Supplementary Fig.5), suggesting that ColGFP+ fibrocytes minimally contribute to fibrogenic myofibroblasts in livers of Mdr2-/- mice. Meanwhile, the composition of CD45+ cells in the livers of Mdr2-/- mice was further analyzed.

CD45+Gr1+ neutrophils migrate into portal area in early stage of Mdr2-/- cholestatic injury. Immunostaining with anti-CD45 antibody detected a flux of BM-derived cells into the livers of Mdr2-/- mice as early as 3 weeks of age, which persisted throughout the 16 weeks of age (Fig. 6A-B). Based on the timeline, we concluded that recruitment of CD45+Gr1+ cells (at 3 weeks of age, Supplementary Fig. 6A) precedes activation of PFs (4 weeks of age), and development of
fibrogenic responses in Mdr2-/- mice. Remarkably, at 4 weeks of age, expression pattern of

CD45+ and Gr1+ cells overlapped. CD45+ and Gr1+ cells were located between ColGFP+ myofibroblasts, and were incorporated into the fibrous scar (Fig. 6C). CD45+ or Gr1+ cells did not express ColGFP (except a 2-4% population of BM-derived fibrocytes, Fig. 5), or CD3 in livers of 4-week-old Mdr2-/- mice (Supplementary Fig. 6B), suggesting that majority of CD45+ cells were neutrophils/granulocytes. Notably, some CD45+ and Gr1+ cells also infiltrated the structures of the bile ducts and some Gr1+ cells were scattered between cholangiocytes (Fig. 6D-E).

Progression of cholestatic fibrosis is associated with upregulation of NOX1 and NOX4 in livers of Mdr2-/- mice.
Therapeutic approaches to treat cholestatic fibrosis remain limited. Activity of NOX, an enzyme system that catalyzes the reduction of molecular oxygen to superoxide, plays an important role in activation of HSCs and macrophages, and promotes development of hepatic fibrosis19,22. In support, strong induction of NOX genes was associated with activation of myeloid cells and myofibroblasts, especially in 12- to 16-week-old Mdr2-/- mice (Fig. 1F). The RNAseq analysis also confirmed the expression of NOX4, Cyba (p22phox), Cybb (NOX2), and Noxo1 in aPFs; and Cyba, Ncf2 (p67phox), Cybb, and Ncf1 (p47phox) in aHSCs (Supplementary Fig. 7).
When livers of 16-week-old Mdr2-/- mice were examined, expression of 4-HNE (a marker of

lipid peroxidation, which reflects oxidative stress) was highly upregulated in hepatocytes (in centrilobular area, Supplementary Fig. 8A), and cholangiocytes, Thy1+ cells, CD34+ cells, and CD45+ cells (in portal area, Supplementary Fig. 8B). We hypothesized that blocking of NOX pathway might attenuate or reverse cholestatic fibrosis in 12- to 16-week-old Mdr2-/- mice.

Administration of NOX1/4 inhibitor attenuates cholestatic fibrosis in Mdr2-/- mice.

In an attempt to identify a novel therapeutic strategy to suppress cholestatic injury of hepatocytes and cholangiocytes, and prevent activation of BM-derived neutrophils and fibrogenic myofibroblasts in Mdr2-/- mice, 12-week-old Mdr2-/- mice were treated with a NOX1/4 inhibitor for 4 weeks (GKT137831: 20 or 60 mg/kg, oral gavage, 5x week, total 20 doses, Fig. 7A). Livers of Mdr2-/- mice were analyzed at 16 weeks of age. Administration of NOX1/4 inhibitor improved liver function (1.5 fold ALT, and bilirubin, Fig. 7B-C), effectively downregulated histological markers 4-HNE, Desmin, Thy1, CD34, CD45, PanCK and Sox9 expression, and reversed peri-central fibrosis (Fig. 7D-H, Supplementary Fig. 9). This effect was accompanied by reduced (2 fold) expression of all major fibrogenic markers (Col1a1, -SMA, PAI1, and TIMP1), aPF-specific markers (Thy1, Msln, Muc-16, Fibulin, Elastin, and CD34), and cholangiocyte markers (PanCK and Sox9) as detected by qPCR (Fig. 7I-L). In a protein assay using western blot, administration of the NOX1/4 inhibitor significantly attenuated the αSMA and Thy1 upregulation with reduced activation of Smad2 and Erk1/2 pathway (Fig. 7M). To further address the specific effect of the NOX inhibitor on aHSCs and aPFs, we additionally performed in vitro studies, in which HSCs and PFs stimulated by TGFβ1 were treated with the
NOX1/4 inhibitor. Administration of the NOX1/4 inhibitor significantly attenuated the expression of profibrogenic markers (Supplementary Fig. 10A) as well as NOX genes

(Supplementary Fig. 10B) in both HSCs and PFs.

Discussion

This study demonstrates that both aPFs and aHSCs contribute to the pathogenesis of cholestatic

fibrosis in Mdr2-/- mice. aPFs were mostly localized in the portal areas, while aHSCs were

located in the portal, sinusoidal and capsular areas. Consistent with a common etiology of

cholestatic injury, Thy1+ColGFP+ aPFs in Mdr2-/- mice exhibited a phenotype similar to that

observed in aPFs in the BDL-model14. aPFs were located in close proximity to proliferating bile ducts, but cholangiocytes themselves did not express Collagen Type I. Although activation of PFs was preceded by recruitment of CD45+ and GR1+ myeloid cells to the portal areas, ColGFP+CD45+ fibrocytes minimally contributed to ECM deposition in livers of Mdr2-/- mice. We concluded that aPFs and aHSCs are the primary targets for anti-fibrotic therapy. Activation of NOXs is a common mediator fibrosis in multiple organs and multiple models, and plays a critical role in activation of aHSCs. Here we demonstrate that NOXs are upregulates not only in aHSCs, but in aPFs in livers of Mdr2-/- mice. Therapeutic blocking of NOX1/4 reversed myofibroblasts activation and liver fibrosis in Mdr2-/- mice, suggesting that blocking of NOX1 and NOX4 might provide a novel target for treatment of cholestatic fibrosis.

Cholestatic injury in Mdr2-/- mice is caused by the damage to hepatocytes (which lack functional canaliculus)6, leading to toxic bile-related inflammation, and damage to biliary epithelium7. Activation of PFs is accompanied by progressive pericholangitis and cholangiocyte proliferation. Most PanCK+ and Sox9+ bile ducts were surrounded by Thy1+ aPFs, suggesting that aPFs may play a key role in the maintenance of the structure and integrity of the biliary tree, regulating cholangiocyte proliferation, and maintaining their polarity17,30,31. Although aHSCs were also detected in the portal areas, most aHSCs were located in the peri-central, sinusoidal, and capsular

areas in the livers of Mdr2-/- mice. In comparison, very few aPFs were scattered throughout

sinusoidal or capsular area, suggesting that specific distribution of different cell types is associated with their distinct roles during development of cholestatic fibrosis in Mdr2-/- mice16. In support, we identified a unique population of CD45+ and Gr1+ cells populating periductular areas at the very onset of cholestatic injury in Mdr2-/- mice. Some of these Gr1+ cells directly infiltrated into the bile duct structures, and were located underneath or between cholangiocytes, suggesting that they might play a critical role in regulation of ductular proliferation and/or activation of PFs. Mdr2-deficiency has been reported to induce the disruption of basement membrane of cholangial duct, leading to the leakage of bile acid into the periductal space7. We can speculate that the CD45+ and Gr1+ neutrophils migrated in response to early injury of cholangiocytes and bile acid leakage, causing acute inflammation and subsequent expansion of periportal myofibroblasts.

Our current study evaluated the dynamic age-dependent changes in histological and mRNA expression of cell-specific markers during development of cholestatic fibrosis in Mdr2-/- mice. To support our histological findings, a widely used reporter Collagen-1(1)-GFP mice were used to visualize Collagen Type I expressing myofibroblasts. In contrast to previous reports27, here we demonstrate that PanCK+ or Sox9+ cholangiocytes did not upregulate Collagen Type I in livers of
Mdr2-/- mice. Furthermore, Desmin has been previously considered as a HSC-specific marker.

Based on our analysis, Desmin positive staining was co-localized in approximately 36% of Thy1+ aPFs and Thy1- aHSCs. As this result could be attributed to a non-specific immunoreactivity of anti-Desmin Ab, we examined the livers from reporter LratYFP mice, which are used for lineage tracing of HSCs. We demonstrated that in mice expressing constitutive

LratYFP, 46% of Thy1+ aPFs were marked with LratYFP, and 73% of Desmin+ cells were marked with LratYFP, suggesting that LratCre does not discriminate between aPFs and HSCs. This fact would explain why Mederacke et al.15 did not identify aPFs as a population distinct from aHSCs that contributes to cholestatic fibrosis in LratYFP mice. In adult mice Lrat is expressed in HSCs and not PFs2,15. Given the mesenchymal origin of both aPFs and aHSCs, expression of Lrat during embryonic development might result in genetic labeling of both PF and HSC populations in LratYFP mice. Lrat is also expressed in extrahepatic tissues, including the lung and retina32. Such common markers as Desmin and Lrat may be expressed in myofibroblasts originating from either HSCs or PFs, suggesting that new markers are needed to discriminate between aPFs and
aHSCs. Thus, the contribution of aPFs in Mdr2-/- mice might have been masked by using the

conventional markers for HSCs including Desmin and LratYFP. In this study, the marker Thy1, expression of Type I collagen, absence of vitamin A, and the characteristic localization have revealed the significant role of aPFs in cholestatic fibrosis.

Furthermore, we identified that a subset of Thy1+ColGFP+ aPFs co-expressed CD34. CD34+ cells located in a portal area, exhibited a myofibroblast-like shape. Expansion of CD34+ColGFP+ aPFs was a unique feature of cholestatic liver fibrosis, as demonstrated by upregulation of
CD34+ColGFP+ aPFs in Mdr2-/- and BDL-injured mice, and was not observed in livers of

CCl4-injured mice. Our data suggest that CD34 is another marker of aPFs in adult mice. In support, expression of CD34 as a marker of hepatic mesenchymal progenitor cells was previously suggested28. Follow-up studies are needed to determine the functional properties of Thy1+CD34+ and Thy1+CD34- aPFs.

Bone marrow-derived fibrocytes are another population that can potentially give rise to myofibroblasts3,33. It has been reported that CD34+ fibrocytes of bone marrow origin are
involved in the fibrosis in Mdr2-/- mice29. In the present study, only few CD34+ fibrocytes

contributed to a population of ColGFP+ myofibroblasts.

The therapeutic properties of NOX1/4 inhibitor on liver fibrosis induced by CCl4, BDL, and NASH models have been previously reported23,34. This study demonstrated the effectiveness of
NOX1/4 inhibition on cholestatic fibrosis in Mdr2-/- mice. Notably, the administration of the

NOX1/4 inhibitor reduced oxidative stress, activation/proliferation of aHSCs and Thy1+desmin+CD34+ aPFs, portal infiltration with CD45+ cells, and cholangiocyte proliferation. Thus, we propose that the interaction between cholangiocyte injury, neutrophil migration, and PF activation are targets of NOX inhibition.

Acknowledgements

We thank Philippe Wiesel and Cedric Szyndralewiez (Genkyotex S.A., France) for providing GKT137831. We thank Dennis R. Petersen for providing us with antibody against 4-HNE. We thank Ryan McCubbin and Katrin Hochrath (UCSD, La Jolla, California, USA) for their help with in vivo experiments. We thank Karin Diggle (UCSD, La Jolla, California, USA) for her excellent management of the laboratory.

References

1.Bataller R, Brenner DA. Liver fibrosis. J Clin Invest. 2005;115(2):209-218.

2.Iwaisako K, Jiang C, Zhang M, et al. Origin of myofibroblasts in the fibrotic liver in mice.

Proc Natl Acad Sci U S A. 2014;111(32):E3297-3305.

3.Kisseleva T, Uchinami H, Feirt N, et al. Bone marrow-derived fibrocytes participate in pathogenesis of liver fibrosis. J Hepatol. 2006;45(3):429-438.
4.Wells RG, Schwabe RF, Schwabe R. Origin and function of myofibroblasts in the liver. Semin Liver Dis. 2015;35(2):97-106.
5.Kisseleva T, Cong M, Paik Y, et al. Myofibroblasts revert to an inactive phenotype during regression of liver fibrosis. Proc Natl Acad Sci U S A. 2012;109(24):9448-9453.
6.Mariotti V, Strazzabosco M, Fabris L, Calvisi DF. Animal models of biliary injury and altered bile acid metabolism. Biochim Biophys Acta. 2018;1864(4 Pt B):1254-1261.
7.Fickert P, Fuchsbichler A, Wagner M, et al. Regurgitation of bile acids from leaky bile ducts causes sclerosing cholangitis in Mdr2 (Abcb4) knockout mice. Gastroenterology. 2004;127(1):261-274.
8.Miethke AG, Zhang W, Simmons J, et al. Pharmacological inhibition of apical sodium-dependent bile acid transporter changes bile composition and blocks progression of sclerosing cholangitis in multidrug resistance 2 knockout mice. Hepatology.

2016;63(2):512-523.

9.Baghdasaryan A, Fuchs CD, Österreicher CH, et al. Inhibition of intestinal bile acid absorption improves cholestatic liver and bile duct injury in a mouse model of sclerosing cholangitis. J Hepatol. 2016;64(3):674-681.
10.Lazaridis KN, Strazzabosco M, Larusso NF. The cholangiopathies: disorders of biliary epithelia. Gastroenterology. 2004;127(5):1565-1577.
11.Fickert P, Pollheimer MJ, Beuers U, et al. Characterization of animal models for primary sclerosing cholangitis (PSC). J Hepatol. 2014;60(6):1290-1303.
12.Wan Y, Meng F, Wu N, et al. Substance P increases liver fibrosis by differential changes in senescence of cholangiocytes and hepatic stellate cells. Hepatology. 2017;66(2):528-541.
13.Meng F, Kennedy L, Hargrove L, et al. Ursodeoxycholate inhibits mast cell activation and reverses biliary injury and fibrosis in Mdr2. Lab Invest. 2018.
14.Koyama Y, Wang P, Liang S, et al. Mesothelin/mucin 16 signaling in activated portal fibroblasts regulates cholestatic liver fibrosis. J Clin Invest. 2017;127(4):1254-1270.
15.Mederacke I, Hsu CC, Troeger JS, et al. Fate tracing reveals hepatic stellate cells as dominant contributors to liver fibrosis independent of its aetiology. Nat Commun. 2013;4:2823.

16.Wells RG. The portal fibroblast: not just a poor man’s stellate cell. Gastroenterology.

2014;147(1):41-47.

17.Jhandier MN, Kruglov EA, Lavoie EG, Sévigny J, Dranoff JA. Portal fibroblasts regulate the proliferation of bile duct epithelia via expression of NTPDase2. J Biol Chem. 2005;280(24):22986-22992.
18.Liver EAftSot. EASL Clinical Practice Guidelines: management of cholestatic liver diseases. J Hepatol. 2009;51(2):237-267.
19.Paik YH, Iwaisako K, Seki E, et al. The nicotinamide adenine dinucleotide phosphate oxidase (NOX) homologues NOX1 and NOX2/gp91(phox) mediate hepatic fibrosis in mice. Hepatology. 2011;53(5):1730-1741.
20.Cui W, Matsuno K, Iwata K, et al. NOX1/nicotinamide adenine dinucleotide phosphate, reduced form (NADPH) oxidase promotes proliferation of stellate cells and aggravates liver fibrosis induced by bile duct ligation. Hepatology. 2011;54(3):949-958.
21.Bataller R, Schwabe RF, Choi YH, et al. NADPH oxidase signal transduces angiotensin II in hepatic stellate cells and is critical in hepatic fibrosis. J Clin Invest. 2003;112(9):1383-1394.
22.De Minicis S, Seki E, Oesterreicher C, Schnabl B, Schwabe RF, Brenner DA. Reduced

nicotinamide adenine dinucleotide phosphate oxidase mediates fibrotic and inflammatory

effects of leptin on hepatic stellate cells. Hepatology. 2008;48(6):2016-2026.

23.Aoyama T, Paik YH, Watanabe S, et al. Nicotinamide adenine dinucleotide phosphate oxidase in experimental liver fibrosis: GKT137831 as a novel potential therapeutic agent. Hepatology. 2012;56(6):2316-2327.
24.Hochrath K, Krawczyk M, Goebel R, et al. The hepatic phosphatidylcholine transporter ABCB4 as modulator of glucose homeostasis. FASEB J. 2012;26(12):5081-5091.
25.Yata Y, Scanga A, Gillan A, et al. DNase I-hypersensitive sites enhance alpha1(I) collagen gene expression in hepatic stellate cells. Hepatology. 2003;37(2):267-276.
26.Seki E, De Minicis S, Osterreicher CH, et al. TLR4 enhances TGF-beta signaling and hepatic fibrosis. Nat Med. 2007;13(11):1324-1332.
27.Lua I, Li Y, Zagory JA, et al. Characterization of hepatic stellate cells, portal fibroblasts, and mesothelial cells in normal and fibrotic livers. J Hepatol. 2016;64(5):1137-1146.
28.Chauhan H, Abraham A, Phillips JR, Pringle JH, Walker RA, Jones JL. There is more than one kind of myofibroblast: analysis of CD34 expression in benign, in situ, and invasive breast lesions. J Clin Pathol. 2003;56(4):271-276.
29.Roderfeld M, Rath T, Voswinckel R, et al. Bone marrow transplantation demonstrates medullar origin of CD34+ fibrocytes and ameliorates hepatic fibrosis in Abcb4-/- mice. Hepatology. 2010;51(1):267-276.

30.Lorena D, Darby IA, Reinhardt DP, Sapin V, Rosenbaum J, Desmoulière A. Fibrillin-1

expression in normal and fibrotic rat liver and in cultured hepatic fibroblastic cells: modulation by mechanical stress and role in cell adhesion. Lab Invest. 2004;84(2):203-212.
31.Li Z, Dranoff JA, Chan EP, Uemura M, Sévigny J, Wells RG. Transforming growth factor-beta and substrate stiffness regulate portal fibroblast activation in culture. Hepatology. 2007;46(4):1246-1256.
32.Liu L, Gudas LJ. Disruption of the lecithin:retinol acyltransferase gene makes mice more susceptible to vitamin A deficiency. J Biol Chem. 2005;280(48):40226-40234.
33.Forbes SJ, Russo FP, Rey V, et al. A significant proportion of myofibroblasts are of bone marrow origin in human liver fibrosis. Gastroenterology. 2004;126(4):955-963.
34.Bettaieb A, Jiang JX, Sasaki Y, et al. Hepatocyte Nicotinamide Adenine Dinucleotide Phosphate Reduced Oxidase 4 Regulates Stress Signaling, Fibrosis, and Insulin Sensitivity During Development of Steatohepatitis in Mice. Gastroenterology. 2015;149(2):468-480.e410.

Figure legend

Fig. 1. aPFs and HSCs contribute to the progression of cholestatic fibrosis in Mdr2-/- mice.

Livers from 4-, 8-, 12-, and 16-week-old Mdr2-/- mice and age-matched WT littermates were

analyzed. (A) Sirius Red staining, Scale bar: 200µm. (B) Positive area was calculated as percent. (C) Expression of fibrogenic markers, (D) inflammatory genes, (E) aPF markers, (F) NOX genes, and (G) cholangiocyte markers were analyzed by qPCR. Data are fold induction versus age-matched WT littermates. (H) Staining for ColGFP and Thy1. Scale bar: 100µm. (I-J) The composition of GFP+ myofibroblasts (100%) was calculated as percent of Thy1+GFP+ (yellow) and Thy1-GFP+ (green) area. (K-L) Nonparenchymal fraction was isolated from Mdr2-/-ColGFP mice. GFP+±Vitamin A+ cells were sort purified. (K) Representative dot plots and the percent of Vit.A+ and Vit.A- in GFP+ cells (100%). (L) Staining for Thy1. Scale bar: 100µm. n=5-7 mice per age group, mean ± SD, †p <0.05, ‡p <0.01, #p <0.001 vs WT; *p <0.05, **p <0.01, ***p <0.001, by ANOVA. Fig. 2. Pathological features of liver injury in 4-, 8-, and 16-week-old Mdr2-/- mice. Serial liver sections from (A) 4-week-old, (B) 8-week-old, and (C) 16-week-old Mdr2-/- mice were stained with Sirius Red, PanCK, Sox9, Thy1, Desmin, or αSMA. BD: bile duct. PV: portal vein. Asterisks: HSCs. (D) Livers from 8-week-old Mdr2-/-ColGFP mice were stained for PanCK, (E) Sox9, (F) Thy1, and (G) Desmin. (H) Livers from Mdr2-/- mice were stained for Desmin and Thy1. Asterisks: HSCs. PV, portal vein. BD, bile duct. Scale bar: 100µm. n=5-7 per age group, mean ± SD. Fig. 3. A subset of Thy1+ aPFs co-express CD34. (A) Livers from 4-, 8-, and 16-week-old Mdr2-/- mice were stained for CD34. Scale bar: 100µm. (B) Positive area was calculated as percent. (C) Livers from 8-week-old Mdr2-/- mice were stained for ColGFP and CD34, (D) CD34 and PanCK, and (E) CD34 and Sox9. Scale bar: 100µm. (F) Livers from BDL (5 days)-injured ColGFP mice, and (G) from CCl4-injured (3 weeks) ColGFP mice were stained for CD34. Scale bar: 200µm. (H-L) Livers from 8-week-old Mdr2-/- mice were stained. Scale bar: 100µm. (H) Staining for Desmin and CD34, and (I) CD34 and Thy1. Arrows: Desmin+CD34+ cells. Asterisks: HSCs. (J) Staining for ColGFP and CD34. Arrows: GFP+CD34+ fibrocytes. Asterisks: GFP+ CD34+ myofibroblasts. (K) Staining for CD34 and CD45, and (L) CD34 and F4/80. n=5-7 per age group, mean ± SD. †p <0.05, ‡p <0.01, #p <0.001 vs WT; *p <0.05, **p <0.01, ***p <0.001, by ANOVA. Fig. 4. Gene expression profiling of aHSCs and aPFs isolated from Mdr2-/- mice. (A) Non-parenchymal fraction was isolated from livers of 16-week-old Mdr2-/-ColGFP mice (n=5 mice combined), Vit.A+GFP+ aHSCs, Vit.A-GFPhi and Vit.A-GFPlow aPFs were sort purified, and analyzed by RNA-Seq for (B) expression of HSC- and aPF- signature genes (rows are grouped with hierarchical clustering); (C) 25 most upregulated and 25 most downregulated genes in Vit.A-GFPhi aPFs compared to Vit.A-GFPlow PFs; (D) 50 most similarly expressed genes in Vit.A-GFPhi and Vit.A-GFPlow PFs; (E) most upregulated and downregulated genes in Vit.A+GFP+ aHSCs compared to Vit.A-GFPhi and Vit.A-GFPlow PFs. An average expression filter of 4 log(counts/million) was applied before calculating the log fold change to filter out likely false positives. (F) Heatmaps displaying signature gene expression in Vit.A+GFP+ aHSCs compared to Vit.A-GFPhi and Vit.A-GFPlow PFs. (G) Using gene set enrichment analysis (GSEA) on the msigdb set of canonical pathways, gene expression in Reactome collagen formation pathway was significantly upregulated in HSCs compared to PF samples. Heatmap colors indicate the relative expression levels in each gene, false discover rate < 0.3, fdr = 0.04. (H) qPCR analysis of selected gene expression in isolated aHSCs and aPFs isolated from 4-, 8-, and 16-week-old Mdr2-/- mice (see Fig. 1K). Data are fold induction versus qHSCs from 4-week-old Mdr2-/-ColGFP mice. Fig. 5. Bone marrow-derived fibrocytes minimally contribute to myofibroblasts in Mdr2-/- mice. (A) Livers from 16-week-old Mdr2-/-ColGFP mice were stained for CD45. Arrows: GFP+ CD45+ fibrocytes. (B) Livers from 8-week-old Mdr2-/-ColGFP mice were stained for CD45 or (C) F4/80. (D) Bone Marrow Transplantation: lethally irradiated 4-week-old Mdr2-/- mice were transplanted with ColGFP bone marrow, and sacrificed at 16 weeks of age. (E) Livers were stained for ColGFP and Desmin, and (F) ColGFP and CD45, or F4/80. Scale bar: 100µm. n=6-8 per group, mean ± SD. Fig. 6. CD45+ Gr1+ neutrophils populate portal area in livers of Mdr2-/- mice. (A) Livers from 3-, 4-, 8-, and 16-week-old Mdr2-/- mice were stained for CD45. (B) Positive area was calculated as percent. (C) Livers from 8-week-old Mdr2-/- mice were stained for ColGFP and CD45, or Gr1 (serial sections), and (D) PanCK and Gr1. Scale bar: 100µm. n=5-7 per age group, mean ± SD. †p <0.05, ‡p <0.01, #p <0.001 vs WT; NS, not significant, by ANOVA. (E) Schematic summary: Cells contributing to cholestatic fibrosis in Mdr2-/- mice. Fig. 7. Administration of NOX1/4 inhibitor attenuates cholestatic fibrosis in Mdr2-/- mice. (A) 12-week-old male Mdr2-/- mice were treated with 20 doses of GKT137831 (20mg/kg [GKT20] or 60mg/kg [GKT60]) or vehicle. Mice were sacrificed at 16 weeks old. (B) Serum levels of ALT and (C) total bilirubin. (D) Livers were stained with Sirius red. Scale bar: 200µm. (E) Positive area was calculated as percent. (F) Livers were stained for 4-HNE. Scale bar: 200µm. (G) Positive area was calculated as percent. (H) Serial liver sections were stained for Desmin, Thy1, CD34, or CD45. (I) Expression of fibrogenic genes, (J) aPF markers, (K) inflammatory genes, (L) cholangiocyte markers were analyzed by qPCR. Data are fold induction compared with 16-week-old WT mice. (M) Expression of αSMA, Smad2, Erk1/2, and Thy1 in liver was analyzed using western blot. n=6 per group, mean ± SD. †p <0.05, ‡p <0.01, #p <0.001 vs WT; *p <0.05, **p <0.01, ***p <0.001, by ANOVA. Highlights  Hepatic myofibroblasts progressively accumulate in the livers of Mdr2-/- mice.  Myofibroblasts mostly originate from Hepatic Stellate Cells or Portal Fibroblasts. Meanwhile, fibrocytes minimally contribute to myofibroblasts in Mdr2-/- mice.  In addition to collagen production, myofibroblasts serve as a source of NOX.  Therapeutic blocking of NOX1/4 ameliorates cholestatic fibrosis in Mdr2-/- mice.Setanaxib