Intercellular crosstalk of hepatic stellate cells in liver fibrosis: New insights into therapy

Intercellular crosstalk of hepatic stellate cells in liver fibrosis: New insights into therapy

Journal Pre-proof Intercellular crosstalk of hepatic stellate cells in liver fibrosis: new insights into therapy Xuanyan Cai, Jiajia Wang, Jincheng Wan...

4MB Sizes 0 Downloads 53 Views

Journal Pre-proof Intercellular crosstalk of hepatic stellate cells in liver fibrosis: new insights into therapy Xuanyan Cai, Jiajia Wang, Jincheng Wang, Qian Zhou, Bo Yang, Qiaojun He, Qinjie Weng

PII:

S1043-6618(19)32419-3

DOI:

https://doi.org/10.1016/j.phrs.2020.104720

Reference:

YPHRS 104720

To appear in:

Pharmacological Research

Received Date:

29 October 2019

Revised Date:

8 January 2020

Accepted Date:

20 February 2020

Please cite this article as: Cai X, Wang J, Wang J, Zhou Q, Yang B, He Q, Weng Q, Intercellular crosstalk of hepatic stellate cells in liver fibrosis: new insights into therapy, Pharmacological Research (2020), doi: https://doi.org/10.1016/j.phrs.2020.104720

This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. 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. © 2020 Published by Elsevier.

Title: Intercellular crosstalk of hepatic stellate cells in liver fibrosis: new insights into therapy

Author: Xuanyan Cai a, Jiajia Wang a, Jincheng Wang a, Qian Zhou c, Bo Yang a, Qiaojun He a, b, Qinjie Weng a, b, *.

a

ro of

Affiliation:

Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of

Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China

Center for Drug Safety Evaluation and Research, Zhejiang University, Hangzhou

-p

b

310058, PR China

Department of Pharmacy, Hangzhou Medical College, Hangzhou 310053, PR China

re

c

Qinjie Weng, PhD.

lP

Corresponding author:

Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of

na

Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China Tel: +86-13819494249; Fax: +86-0571-8820-6087

Jo

ur

E-mail: [email protected]

1

re

-p

ro of

Graphical abstract

lP

Abstract

Liver fibrosis is a dynamic wound-healing process characterized by the net

na

accumulation of extracellular matrix. There is no efficient antifibrotic therapy other than liver transplantation to date. Activated hepatic stellate cells (HSCs) are the major

ur

cellular source of matrix-producing myofibroblasts, playing a central role in the

Jo

initiation and progression of liver fibrosis. Paracrine signals from resident and inflammatory cells such as hepatocytes, liver sinusoidal endothelial cells, hepatic macrophages, natural killer/natural killer T cells, biliary epithelial cells, hepatic progenitor cells, and platelets can directly or indirectly regulate HSC differentiation and activation. Intercellular crosstalk between HSCs and those “responded” cells has been 2

a critical event involved in HSC activation and fibrogenesis. This review summarizes recent advancement regarding intercellular communication between HSCs and other “responded cells” during liver fibrosis and experimental models of intercellular crosstalk systems, and provides novel ideas for potential antifibrotic therapeutic strategy.

ro of

Key words: liver fibrosis, hepatic stellate cells, intercellular crosstalk, antifibrotic

Jo

ur

na

lP

re

-p

therapy

3

1. Introduction Liver fibrosis is a wound-healing response to the continuous action of various injury factors, characterized by the net accumulation of extracellular matrix (ECM), or scar [1]. A variety of chronic stimuli can cause liver fibrosis, including chronic viral infection (hepatitis B virus or hepatitis C virus), toxic damage, alcohol abuse (longstanding excessive alcohol consumption), non-alcoholic fatty liver disease /non-

ro of

alcoholic steatohepatitis (NASH), autoimmune liver diseases, and metabolic and

genetic diseases [2-5]. If left untreated, liver fibrosis can progress to cirrhosis associated

with a series of lethal complications, such as functional liver failure, portal hypertension,

-p

ascites, hepatic encephalopathy, renal and cardiac disturbances, and even hepatocellular

re

carcinoma (HCC) [6-8]. Cirrhosis is a critical risk factor for liver cancer and a challenge for human health, resulting in 1.16 million deaths annually worldwide [9]. The direct

lP

and indirect costs for treating cirrhosis exceed $12 billion each year in the United States

na

alone [10]. Although some studies have indicated that liver fibrosis is reversible and cirrhosis may regress in some cases, there is no effective antifibrotic therapy [11, 12].

ur

Currently, the only available therapeutic approaches entail eliminating chronic stress and liver transplantation [13, 14]. Therefore, it is urgent to identify the vital mechanism

Jo

underlying liver fibrosis and develop more efficient antifibrotic therapies to improve the quality of life in patients with liver fibrosis. HSCs, nonparenchymal cells (NPCs) localized in the perisinusoidal space of Disse, are publicly regarded as the major cellular source of matrix-producing myofibroblasts following their transdifferentiation into proliferative and fibrogenic myofibroblast-like 4

cells in a process termed “activation” [15]. Preventing the activation of HSCs can reduce deposition of ECM proteins, and slow down or even reverse liver fibrosis. Therefore, HSCs represent a primary target for antifibrotic therapy. In fact, HSC activation is a dynamic process that mainly depends on the interaction with other “responded” cells, which are mainly composed of hepatocytes, liver sinusoidal endothelial cells (LSECs), hepatic macrophages, natural killer/natural killer T

ro of

(NK/NKT) cells, biliary epithelial cells (BECs), hepatic progenitor cells (HPCs) and platelets, which can precisely “talk” to each other [16]. In the interactive network,

chemokines and molecular signals released from those cells directly or indirectly affect

-p

the state of HSCs, namely, “quiescent” or “activated” HSCs regulate the progression,

re

aggravation, and resolution of liver fibrosis. Understanding the intercellular crosstalk between HSCs and those “responded” cells and clarifying the role of their interaction

lP

at different stages of fibrosis progression are critical for discovering novel antifibrotic

na

options.

This review mainly summarizes recent advancement in intercellular crosstalk

ur

between HSCs and other “responded cells” in the pathogenesis of liver fibrosis, focusing on the role of their interaction on HSC activation. It also outlines the common

Jo

experimental models of intercellular crosstalk systems used in this field, which provides a deeper understanding of the molecular mechanisms underlying liver fibrosis and novel ideas for effective antifibrotic therapy. 2. HSCs as precursors of myofibroblasts HSCs are derived from embryonic mesothelial cells and reside in the virtual 5

subendothelial space between hepatocytes and endothelial cells (ECs), where small or soluble molecules exchange in bidirectional transfer. In the normal liver, HSCs are quiescent and represent 5–8% of the total number of liver cells; they are known for vitamin A (retinoid) storage in their cytoplasmic droplets [17, 18]. The vitamin A lipid droplet is a conspicuous feature for identification during HSC isolation. Desmin and some neural markers such as glial fibrillary acidic protein and synaptophysin, are also

ro of

common identification markers [19]. Following liver fibrosis from any cause, quiescent

HSCs become activated, gradually reducing the vitamin A storage capacity, proliferating, developing contractile function, secreting excessive ECM proteins, and

-p

releasing a series of pro-inflammatory (e.g., interleukin 6 [IL-6]) and profibrogenic

re

factors (e.g. transforming growth factor beta [TGF-β]) [20]. Activated HSCs transdifferentiate into myofibroblast-like cells, which are key sources of excess ECM

lP

in liver fibrosis and can be identified by a set of proteins including α-smooth muscle

na

actin (α-SMA), collagens (1a1, 3a1), vimentin, osteopontin, lysyl oxidases (LOX) and tissue inhibitor of metalloproteinase 1 (TIMP1) [21, 22]. It is now widely believed that

ur

activated HSCs are the main contributors to the pool of fibrogenic myofibroblasts, contributing 82–96% of myofibroblasts in various types of chronic liver diseases, and

Jo

are crucial components of the fibrotic response during fibrogenesis [23]. Generally, HSC activation comprises two phases: initiation and perpetuation (Fig. 1). If fibrosis begins to regress, activated HSCs may progress to a potential third stage, resolution, where they may undergo apoptosis, become senescent, or revert to a quiescent phenotype [4, 24]. In the initiation phase, HSCs receive molecular signals from 6

neighboring cells that secrete profibrogenic cytokines and growth factors, and then they manifest profibrogenic transcriptional and secretory properties. When activated, HSCs proliferate and constantly secrete ECM proteins, leading to ECM accumulation and scar tissue formation. They are in a process of perpetuation. The perpetuation of HSCs is a closely orchestrated process that comprises a number of cell-cell interactions and wound-healing responses.

ro of

3. Intercellular crosstalk in the activation of HSCs

Studies have elucidated the complex dynamic process of HSC activation through

multiple signaling pathways from other “responded” cells during liver fibrosis as

-p

summarized below (Fig. 2).

re

3.1. Hepatocytes

Hepatocytes are the most abundant cell type in liver and are predisposed to damage

lP

to the liver, such as damage from alcohol, viral infection, and toxic compounds, leading

na

to acute or chronic liver disease. In response to liver injury, hepatocytes release a series of key signals such as reactive oxygen species (ROS) [25], hedgehog (Hh) [26, 27],

ur

nucleotides [28], fatty accumulation [29], damage-associated molecular patterns (DAMPs) [30], eliciting inflammation and fibrosis. Damage-induced hepatocyte cell

Jo

death responses in various forms including apoptosis, necrosis, necroptosis and autophagic cell death can directly affect HSC activation [31]. It has been reported that HSCs

are

able

to

phagocytose

hepatocyte

apoptotic

bodies,

inducing

transdifferentiation of HSCs into myofibroblasts along with a series of profibrogenic responses [32, 33]. Blocking the “phagocytosis” of HSCs, or building a “screen” 7

between HSCs and hepatocytes may be an effective strategy. Galectin-3 can regulate phagocytosis-mediated HSC activation, and inhibiting galectin-3 in animal models has shown to significantly decrease fibrosis [34]. GR-MD-02 (Galectin Therapeutics), a galectin-3 inhibitor, is currently under evaluation in phase IIB trials for patients with compensated NASH cirrhosis (NCT02462967) [35]. Additionally, hepatocytes are also capable of pyroptosis, a newly discovered programmed death. The NOD-like receptor

ro of

family pyrin domain containing 3 (NLRP3) inflammasome activation triggers hepatocyte pyroptosis, resulting in activation of HSCs with collagen deposition in the liver [30].

-p

Some important inflammatory mediators released from damaged hepatocytes

re

might directly or indirectly promote HSC activation and fibrosis. Inactivation of hepatocyte nuclear factor 1α (HNF1α), an important transcription factor in hepatocytes,

lP

enlarges liver injury and results in hepatic dysfunction [36, 37]. Inhibition of HNF1α

na

in hepatocytes leads to the elevation of IL-6 and TGF-1, inducing the activation of HSCs; However, activated HSCs further suppress the expression of HNF1α in

ur

hepatocytes by releasing TNF-α and IL-6 [38]. Exosomes from damaged hepatocytes can mediate toll-like receptor 3 (TLR3) activation and enhance expression of IL-17A,

Jo

IL-1 and IL-23 in HSCs, triggering IL-17A production by T cells and exacerbating liver fibrosis [39]. Abnormal expression of proteins in hepatocytes is related to HSC activation during liver fibrosis. Hepatocyte-derived high-mobility group box-1 (HMGB1) participates in fibrosis progression by activating phosphorylated MAPK/ERK kinase 8

1/2 (p-MEK1/2), phosphorylated extracellular signal-related protein kinases 1 and 2 (pERK1/2) and phosphorylated c-Jun, and signal receptor advanced glycation end products (RAGE) in HSCs, ultimately increasing collagen type I production [40]. Based on the important impact of HMGB1 on HSC activation, researchers have found some promising treatments for liver fibrosis related to the RAGE/HMGB1 axis or HMGBrelated signaling pathways, such as nilotinib and quercetin [41-44]. Furthermore, the

ro of

upregulation of spleen tyrosine kinase (SYK) and platelet-derived growth factor receptor α (PDGFRα) in hepatocytes can directly shift its expression in HSCs,

regulating HSC activation and proliferation [45-48]. In light of current research, key

-p

proteins involved in the interaction between hepatocytes and HSCs may be potent

re

therapeutic targets in the future. 3.2. LSECs

lP

LSECs form the wall of liver sinusoids and represent approximately 15–20% of

na

liver cells, but only 3% of liver volume [49]. In physiological conditions, LSECs represent a permeable barrier for the bidirectional transfer of small or soluble molecules

ur

between the blood and the space of Disse [22]. Meanwhile, LSECs also interact with hepatocytes and HSCs for protein, lipid, and glucose metabolism [50]. In a normal liver,

Jo

paracrine production of vascular endothelial growth factor (VEGF) by hepatocytes and HSCs stimulates LSECs to produce nitric oxide (NO), which is essential for maintaining the LSEC phenotype [51]. Following liver injury, LSECs release different angiocrine signals to balance liver regeneration and fibrosis, ultimately maintaining liver homeostasis. When stimulated by acute liver injury or partial hepatectomy, 9

upregulated VEGF promotes the expression of hepatocyte growth factor (HGF) by LSECs and resident sinusoidal progenitor cells, which in turn elicits liver regeneration [52]. In contrast, activation of fibroblast growth factor receptor 1 (FGFR1) in LSECs by chronic injury drives a profibrotic angiocrine response to HSCs, thereby provoking fibrosis [53]. In the progress of liver fibrosis, damaged LSECs gradually lose fenestration with

ro of

the basement membrane, becoming capillarized, which is one of the main pathological

changes of liver fibrosis. Capillarized LSECs lose their hepatoprotective property and the capacity to inactivate HSCs, thus promoting intrahepatic vasoconstriction and liver

-p

fibrosis [50]. Activated HSCs contract and deposit excessive ECM in the space of Disse,

re

leading to the loss of endothelial fenestration and dysfunction of LSECs. Thus, a self-

hepatic fibrosis.

lP

perpetuating cycle connects and stimulates HSCs and LSECs, further contributing to

na

Current research indicates that normally differentiated LSECs are gatekeepers of fibrosis through maintaining HSC quiescence and promoting reversion of activated

ur

HSCs to quiescence via a VEGF-stimulated-NO-dependent/independent pathway [54, 55]. Upregulation of the transcription factor kruppel-like factor 2 (KLF2) induced by

Jo

statins in LSECs increases endothelial NO synthase expression, conferring vasoprotection and HSC deactivation [56]. Interestingly, recent data support the fact that LSECs promote HSC activation through the liberation of cellular microvesicles. Exosome-packaged sphingosine kinase 1 (SK1) derived from ECs promotes HSC migration and AKT phosphorylation through fibronectin/integrin-dependent exosome 10

adherence and dynamin-dependent exosome internalization [57]. This research opens a new avenue to understand the sinusoidal communications between LSECs and HSCs. Considering the cellular role of LSECs in HSCs, LSEC protection may be an effective strategy for attenuating fibrosis progression. Currently, some compounds have shown good ability to maintain the typical phenotype of LSECs and promising results for fibrosis [58-61]. BAY 60-2770, an activator of soluble guanylate cyclase, can restore

ro of

the differentiation of LSECs and lead to quiescence of HSCs without directly affecting HSCs, preventing progression of thioacetamide-induced cirrhosis in rats [61].

Amusingly, Liu and colleagues [62] utilized fibrotic microniches (FμNs), recapitulating

-p

the interaction of HSCs and LSECs, to understand the response to antiangiogenic drugs

re

at different stages of liver fibrosis. These drugs were only found to be effective for early-stage fibrosis, and the exact mechanism is unclear. Therefore, future studies are

lP

needed to discover the key event in the crosstalk of HSCs and LSECs at different stages

na

of fibrosis, which will assist in drug designed and precise intervention strategies targeting stage-specific disease progression.

ur

3.3. Immune cells

Immune cells have diverse functions to ensure homeostasis, antimicrobial defense

Jo

and proper metabolism in the liver. Ample studies have shown that immune signaling pathways play important roles in hepatic fibrogenesis. Immune cells secrete inflammatory cytokines or chemokines in response to inflammatory or metabolic stimuli, which directly or indirectly affects hepatic injury. Importantly, bidirectional interaction between immune cells and HSCs regulates positively or negatively the 11

pathogenesis of liver fibrosis. 3.3.1. Hepatic macrophages Hepatic macrophages hold dual functions in initiating, perpetuating and even restricting inflammation in the pathogenesis of liver fibrosis. Traditionally, according to their ontogeny, hepatic macrophages can be broadly divided into liver-resident macrophages, termed Kupffer cells (KCs), and blood/bone marrow-derived

ro of

macrophages, termed monocyte derived macrophages [63].

KCs, attached to the sinusoidal endothelial layer, can be activated by circulating

diverse stimuli of blood and secreting various cytokines including TGF-1, TNF-,

-p

MCP-1, chemokines (e.g., CCL3 and CCL5), and other soluble mediators, eliciting a

re

physiological response to other liver cells [64, 65]. Currently, there is clear evidence that KCs bilaterally regulate the activation of HSCs. Pradere and colleagues [66]

lP

demonstrated that KCs contribute to HSC survival in an NF-κB-dependent manner.

na

Consistently, inhibition of NF-κB by sulfasalazine stimulates HSC apoptosis, exerting a potential antifibrotic role [67]. Leptin possesses profibrogenic activity in the liver,

ur

which is related to the direct effect on KCs with the release of TGF-1 and connective tissue growth factor (CTGF) to mediate HSC activation [68]. Suppression of

Jo

macrophage infiltration can also directly inhibit activation of HSCs and liver fibrogenesis[69]. On the other hand, KCs can negatively mediate the fibrogenic response of HSCs on an H2O2-IL-6-dependent mechanism [70]. Based on current studies, it is not known why KCs have two different responses to HSCs. It is necessary to elucidate the key triggers between KC activity and HSC activation in future works, 12

which will benefit precision intervention of KCs on liver fibrosis. Researchers have found that some important proteins can regulate the cooperation between KCs and HSCs [71-73], which can be promising therapeutic targets for liver fibrosis. Oncostatin M (OSM), a member of the IL-6 family cytokines, induces collagen production in HSCs depending on the cellular source of fibrogenic cytokines from hepatic macrophages [72]. Blocking macrophage regulation on OSM inhibits collagen

ro of

secretion of HSCs, leading to decreased fibrosis.

Massive monocyte-derived macrophages can be recruited to the injured liver, and a part of them can differentiate into resident macrophages, contributing to liver injury

-p

progression. Generally, infiltrating monocytes are divided into two major subsets: Ly-

re

6Chi monocytes and Ly-6Clo monocytes. Ly-6Chi monocytes mainly express inflammatory cytokines or chemokine receptors, such as C-C motif chemokine

lP

receptors (e.g., CCR1, CCR2 and CCR5), exhibiting a pro-inflammatory phenotype.

na

Whereas the Ly-6Clo monocytes express more scavenging receptors, such as CX3CR1, and the high expression of matrix degrading metalloproteinases (MMPs) (e.g., MMP-

ur

9, MMP-12, MMP-13), representing a fibrolytic subset in resolving fibrosis [74]. Karlmark and colleagues [75] claimed that Ly-6Chi monocytes, acting as precursors of

Jo

CD11b+F4/80+ intrahepatic macrophages, directly activate HSCs by producing TGF1. CCR1 and CCR5 are upregulated in experimental mouse models of fibrogenesis. CCR1 and CCR5-deficient mice displayed reduced macrophage infiltration in the injured liver, thereby inhibiting HSC activation and fibrogenesis [76]. Additionally, the increased recruitment of Ly-6Chi monocytes after CCl4 treatment is CCR213

dependent. Inhibiting CCRs has been a drug research hotspot in liver fibrosis. The dual CCR2/CCR5 inhibitor, Cenicriviroc (Allergan), is being evaluated for its antifibrotic efficacy in phase III clinical trials (NCT 03028740) [74, 77, 78]. It is commonly believed that CX3CR1 controls the differentiation of Ly-6Clo monocytes [79, 80]. CX3CR1-deficient mice display prolonged inflammatory monocyte infiltration by activating anti-apoptotic BCL-2 expression, promoting HSC activation and resulting

ro of

in enhanced liver fibrosis [79, 81]. Interestingly, a phenotypic shift between LyC6hi and LyC6lo can determine the development of fibrosis. Bárcena and colleagues [82] found that CD5L can promote a phenotypic shift in monocytes from LyC6hi to LyC6lo,

-p

regulating HSC activation and attenuating CCl4-induced fibrosis. Furthermore, a

re

recent study uncovered a novel scar-associated TREM2+CD9+ macrophage sub (SAMФ), differentiating from circulating monocytes, can promote primary human

lP

HSC proliferation, which may be a potential therapeutic target cell in the future [9].

na

Bone marrow-derived macrophages (BMDMs) have a nonnegligible role in modulating the immune microenvironment in the fibrotic liver [83-85]. M1-polarized

ur

BMDMs can recruit significantly more F4/80+ endogenous macrophages and NK cells to promote HSC apoptosis through their paracrine function. Therefore, M1-polarized

Jo

BMDMs have a better output in the therapy of hepatic fibrosis compared to the M0 or M2 BMDMs [85]. As a bridge of inflammation to HSC activation, macrophages have been potential effective targets for fibrosis treatment. Researchers are working tirelessly to develop effective drugs targeting macrophages, and some are making their way into clinical 14

trials. Curcumin, an anti-inflammatory and anti-oxidant natural compound, has been widely reported to inhibit KC activation and Ly6Chi monocyte infiltration, thereby reducing HSC activation and protecting against liver fibrosis [86, 87]. Amine oxidase copper-containing 3 (AOC3) positively regulates the lymphocyte recruitment signal in the liver [88]. A pharmacological inhibitor of AOC3, BI 1467335 (Boehringer), has been tested in phase II trials for patients with NASH (NCT 03166735) [35].

ro of

Additionally, autologous macrophage therapy, a potential effective antifibrotic cell

therapy, has shown safety and feasibility for cirrhosis patients in the first-in-human study. This macrophage therapy is currently under evaluation in a phase II randomized

-p

controlled trial (ISRCTN 10368050) for an antifibrotic ability test [9].

re

3.3.2. NK/NKT cells

NK cells reside in the liver sinusoid and represent the major liver lymphocyte

lP

population [89]. Many studies have indicated that NK cells have an antifibrotic function

na

through killing activated HSCs and producing interferon-γ (IFN-γ) in mice and humans [90-92]. NKp46 is one of the major NK activating receptors expressed by NK cells.

ur

Absence of NKp46 can exacerbate liver fibrosis directly through the killing of primary murine and human HSCs [93]. The upregulated retinoic acid (RA) in activated HSCs

Jo

can sensitize early activated HSCs to NK cell death [94]. However, RA can also suppress NK cell activity via induction of TGF-β and suppression of cytokine signaling 1 to inhibit IFN-γ signaling during advanced liver injury [95]. Notably, NK cells preferentially kill senescent-activated HSCs, facilitating the resolution of fibrosis [96]. Generally, NKT cells can be divided into two distinct subpopulations: type I NKT 15

cells (express a semi-invariant T cell receptors [TCRs], comprising 95% of liver NKT cells) and type II NKT cells (express more diverse TCRs) [97]. NKT cells share similar phenotypic and functional characteristics with NK cells, but have both antifibrotic and profibrotic properties in regulating liver fibrosis. Similar to NK cells, NKT cells can directly kill activated HSCs and produce IFN-γ to have inhibitory effects on liver fibrosis [98]. Despite this, NKT cells promote HSC activation and liver fibrosis by

ro of

producing inflammatory cytokines such as IL-4 and IL-13 [99]. NKT cell-conditioned medium stimulates primary HSCs to become myofibroblastic, and CD1d-deficient mice

lacking NKT cells develop less fibrosis in NASH-related fibrogenesis [100]. Different

-p

from the regulation models mentioned above, IL-30 can recruit NKT cells to the injured

re

liver and selectively remove activated HSCs, indirectly modulating intercellular communication in NKT cells and HSCs [101-103].

lP

3.4. BECs

na

BECs, also known as cholangiocytes, are epithelial cells arranged along the biliary tract. They are comprised of a complex three-dimensional conduction network

ur

extending from the small intrahepatic bile duct to the extrahepatic bile duct. Generally, BECs are primary sites of damage in liver for primary biliary cirrhosis and primary

Jo

sclerosing cholangitis [104]. Moreover, cholangiocyte senescence has been a hallmark in the progression of liver fibrosis, where cholangiocytes secrete a number of profibrogenic and chemotactic factors including CTGF, PDGF, TGF-β1, MCP-1, and IL-6 to attract HSCs [105]. Proliferating cholangiocytes are a major source of the profibrogenic CTGF during biliary fibrosis [106] and directly attract lobular 16

myofibroblastic HSCs into the portal tracts through releasing PDGF. Additionally, cholangiocytes modulate mesenchymal-epithelial interactions through producing Hh ligands, directly promoting the growth of myofibroblast cells [107]. As we understand that cholangiocyte proliferation and paracrine have stimulating effects on HSC activation, the targeting proliferating cholangiocytes or the disruption of cholangiocyte secretion of profibrogenic factors will be potential anti-fibrosis therapies. Over and

ro of

above these interference strategies, targeting some key regulators (e.g., substance P,

osteopontin) that indirectly promote collagen secretion and inhibit HSC senescence by regulating cholangiocytes function may be another hotspot for further research [108,

-p

109].

re

3.5. HPCs

HPCs are positioned within the terminal bile ductile and are quiescent in the

lP

healthy liver. With a strong potential for a heterogeneous multipotent transient

na

amplifying cell population (oval cells), HPCs play an irreplaceable role in regeneration of the damaged liver [110, 111]. Kordes and colleagues [112] found that CD133+ HSCs

ur

could differentiate into endothelial or hepatocyte lineages, and recognize them as progenitor cells. Subsequently, the research team found that transplanted HSCs could

Jo

form progenitor cells in rats, and cultured HSCs transiently adopted a similar expression profile to that of HPCs during differentiation, showing the inseparable relationship between HPCs and HSCs [113]. Combined with the present research, HPCs represent an intermediate stage for differentiating HSCs in liver regeneration, where HSCs can differentiate into HPCs through mesenchymal-to-epithelial transition (MET) [113-115]. 17

Along with their differentiation into HPCs, HSCs can further regulate HPC differentiation via a series of signaling axes such as the TGF-β1/Jagged1 pathway [116], NF-κB-inducible NO synthase signaling [117]. There is currently little research on fibrogenesis and regression; further investigation is required. 3.6. Platelets Platelets are derived from mature megakaryocytes (MKs) through cytoplasmic

ro of

fragmentation consisting of long pseudopodial elongations that break in blood flow

[118]. It has been reported that platelets can accumulate at the site of injury liver in

pathological conditions, such as liver cirrhosis [119], viral hepatitis [120], cholestasis

-p

[121], ischemia/reperfusion [122, 123], and the residual liver after hepatectomy [124],

re

playing important roles in hemostasis [125], wound healing [126], and liver regeneration [127-129]. In fibrotic liver, platelets can be directly adjacent to collagen

lP

fibrils [119, 129]. Platelets and platelet-derived ATP can suppress HSC activation and

na

reduce type I collagen production via the adenosine-cAMP signaling pathway [130]. In addition, platelets release many important moderators including PDGF [131], TGF-β1

ur

[132], HGF [133, 134], VEGF [135], and epidermal growth factor (EGF) [136], which are intrinsically involved in fibrogenesis. Inhibiting the production of these moderators

Jo

from platelets, such as MOR8457, a high-affinity monoclonal antibody against PDGFB, can reduce the level of HSC activation and collagen deposition in liver fibrosis [137]. Therefore, antiplatelet therapy, such as anti-PDGF-B treatment, has been a potential strategy to treat fibrosis in patients.

18

4. Experimental models of intercellular crosstalk system To better decipher the key signaling network of intercellular crosstalk, a series of established experimental systems (in vitro, ex vivo and in vivo ) are used to try to mimic the complex cell-cell interactions involved in HSC activation and liver fibrosis (Table 1). Moreover, these models are comprehensive and precise to screen putative antifibrotic compounds, evaluate potential drug efficacy and toxicity, and greatly

ro of

accelerate the development of antifibrotic strategies. Co-culture systems, joining at least two cell types, are the main in vitro models for exploring the role of intercellular

crosstalk. Ex vivo models, represented by PCLSs and liver organoids, attempt to

-p

simulate the hepatic microcirculatory architecture, and have shown great appeal to

re

researchers in recent years. Animal models can dynamically display the pathological process and partly reflect the major pathological features in humans, which is an

na

4.1. In vitro models

lP

indispensable tool for researchers to study liver fibrosis.

For in-depth investigation of the mechanisms driving HSC activation and liver

ur

fibrosis, in vitro models are powerful and indispensable. Generally, primary HSCs and a variety of HSC cell lines from rodents and humans are abundantly used. Primary

Jo

HSCs can be freshly isolated based on a density gradient centrifugation method [138140], and then cultivated in a mono-layer culture where HSCs would be activated over a 7–10 day culture period to acquire a myofibroblast-like phenotype [22]. Besides, a series of well-established cell lines are important for investigating the signaling pathway in HSC activation, such as human cell lines (LX-2, TWNT-4, GREF-X), rat 19

cell lines (HSC-T6, CFSC, PAV-1, T-HSC/Cl6, RGF), mouse cell lines (GRX, A640IS, JS1, Col-GFP) and rat portal myofibroblast cell lines (RGF-N2 and RGF) [141-145]. Monoculture systems are useful but inadequate to resemble the complicated progress in the development of fibrosis where HSCs interact with other “responded” cells. Co-culture systems gain more popularity for researching intercellular crosstalk of HSCs in subsequent studies. A multicellular in vitro system usually contains functional

ro of

hepatocytes that could respond to external injury stimulation and HSCs that could be activated towards a myofibroblast phenotype. Human hepatoma cells, such as HepG2 [146] and HepaRG [147, 148], are the current options as sources of hepatocytes. HSC

-p

cell lines and primary HSCs from rodents or humans are available and represent the

re

activated HSC phenotypes as well [149]. Hepatocyte-HSC co-culture system have been a potent system to study hepatocyte damage-dependent HSC activation. Giraudi and

lP

colleagues [29] demonstrated that cell-to-cell proximity between hepatocytes and HSCs

na

is of importance to initiate the fibrotic process by a well-established co-culture system. To determine the role of immune cells in regulating HSC activation, researchers

ur

establish co-culture based on HSCs and KCs or BMDMs [73, 150]. By contrast, cocultures consisting of HSCs and ECs reflect the importance of ECs on regulating HSC

Jo

activation [151]. Historically, in vitro co-cultures have been developed with the help of robust techniques including seeding between two layers of ECM compounds or by culturing in spheroids, micromolds of non-adherent materials, gravitational aggregation using hanging drop cultures, or the use of rotation of concave 96 well cell-repellent plates, making the systems more stable and controllable [22, 141]. 20

4.2 Ex vivo models Although co-cultures are good alternatives for liver fibrosis research and drug screening, they still exist with great differences to actual pathological mechanisms. In this sense, precision cut liver slices (PCLSs) are the best representation for the in vitro study of liver fibrosis, with all the cells including all cell-matrix interactions maintained in their original environment. PCLSs are fresh liver slices with a thickness of 100 to

ro of

250 µm from rodents or humans, which can subsequently be cultured in cell culture

dishes, perfectly mimicking the multicellular characteristics of organs in vivo [152]. PCLSs have been extensively used to examine the potency of putative antifibrotic

-p

compounds [153-155]. Unfortunately, the limited viability of PCLSs has restricted their

re

use to short-term study. In addition to PCLSs, liver organoids derived from stem cells or organ progenitors can also mimic the process in vivo and have been used to test anti-

lP

fibrotic compounds, which are important tools to transform the basic research to

na

therapeutic applications [156, 157]. Leite and colleagues [158] developed a 3D microrganoids where functionality of both hepatocytes and HSCs can be maintained for

ur

at least 21 days, which optimizes the study of hepatocyte-dependent HSC activation and allows potential compounds testing.

Jo

4.3. In vivo models

In vivo models are the most popular assessment models to study liver fibrosis for

researchers. A series of experiments in vivo are widely employed to mimic the complex pathology of liver fibrosis and hepatic cell-cell interactions. Traditionally, CCl4 toxicity model [159] and the common bile duct ligation model (CBDL) [160] are the “work21

horse” to study liver fibrosis in vivo , which can be used in multiple genetic backgrounds (transgenic, constitutive, or inducible knockout mice) and are easy to set up for both rats and mice [161]. Other rodent models, such as alcohol-induced alcoholic liver disease (ALD) [162, 163], diet-based models (MCD/HF diet-induced liver steatosis/NASH) [164], genetically modified models (Multidrug resistance-associated protein 2-deficient mice (Mdr2-/-)) [165], and hepatitis virus infection-based models

ro of

[166], are available tools to study liver fibrosis induced by specific etiologies.

Considering the high prevalence of hepatitis virus infections worldwide, hepatitis virus infection-based models have been increasingly crucial. In fact, hepatitis virus infection

-p

induces liver fibrosis in humans but not in rodents. Nevertheless, humanized animal

re

models with human immune and liver cells have allowed researchers to closely study liver fibrosis with HCV and long-term HBV infection in recent years, which are

lP

valuable tools to elucidate the contribution of immune system to HSC activation in

na

humans [167, 168]. Chimeras with BM cells from other genetically modified mice are also powerful research models to study the specific proteins or signaling involved in

ur

the interaction between immune cells and HSCs [169]. For the significant roles of hepatocytes on activated HSCs, hepatocyte-specific gene knockout models have been

Jo

widely used to explore the key molecular mechanisms existing in the process of HSC activation [48]. 5. Conclusion and perspective Liver fibrosis is a complex progress involving multiple events, in which HSC activation is publicly recognized as the central event. In responding to stimuli from the 22

intracellular and extracellular microenvironment, activated HSCs interact with other “responded” cells, triggering the wound-healing response. These “responded” cells play dual roles in HSC activation through diverse molecular mechanisms and cell signaling during different stages of fibrosis. Paracrine profibrogenic chemokines and molecular signals activate HSCs or promote their activation; inhibiting HSC activation and eliminating activated HSCs by reverting, transdifferentiating, or inducing HSC

ro of

death and senescence. Deciphering the specific cellular mechanisms of cell-cell

interaction in HSC activation is essential to provide new insights into therapeutic interventions in liver fibrosis. Further investigation should focus on the key factors

-p

determining the dual regulation of HSC activation by these “responded” cells in order

re

to discover precise intervention strategies for liver fibrosis. Furthermore, the intercellular crosstalk in regression of liver fibrosis is poorly understood. We still do

lP

not know what plays the leading role in regulating HSC activation in the process.

na

Improved knowledge of the crucial process would develop more efficient strategies to promote fibrosis resolution and liver regeneration.

ur

Of equal importance, there is a lack of complete system building in the field of intercellular crosstalk to study the interaction between HSCs and other “responded”

Jo

cells. Monolayer HSC culture is inadequate to unearth the complexity of HSC activation. As valuable alternatives, co-culture systems can resemble the multicellular interaction process in liver fibrosis and have been widely used in research labs. Nevertheless, we must be aware that the in vitro cell conditions may differ greatly from actual pathology in vivo . Most cells lose their characteristics in vitro, which may 23

change their function on HSC activation. In this regard, ex vivo models like PCLSs are needed and will be more popular in the upcoming years. Additionally, it has been confirmed that the latest liver-on-a-chip can recapitulate the hepatic microcirculatory architecture, allowing for more credible intercellular crosstalk research and drug

Jo

ur

na

lP

re

-p

ro of

screening in the future [170].

24

References [1] S.L. Friedman, Liver fibrosis - from bench to bedside, J Hepatol, 38 (2003) S38-S53. [2] T. Higashi, S.L. Friedman, Y. Hoshida, Hepatic stellate cells as key target in liver fibrosis, Adv Drug Deliver Rev, 121 (2017) 27-42. [3] G.O. Elpek, Cellular and molecular mechanisms in the pathogenesis of liver fibrosis: An update, World J Gastroenterol, 20 (2014) 7260-7276.

ro of

[4] J.E. Puche, Y. Saiman, S.L. Friedman, Hepatic stellate cells and liver fibrosis, Compr Physiol, 3 (2013) 1473-1492.

[5] V. Hernandez-Gea, S.L. Friedman, Pathogenesis of liver fibrosis, Annu Rev Pathol, 6 (2011)

-p

425-456.

re

[6] M. Pinzani, Pathophysiology of Liver Fibrosis, Dig Dis, 33 (2015) 492-497. [7] K. Bottcher, M. Pinzani, Pathophysiology of liver fibrosis and the methodological barriers to

lP

the development of anti-fibrogenic agents, Adv Drug Deliv Rev, 121 (2017) 3-8. [8] E.A. Tsochatzis, J. Bosch, A.K. Burroughs, Liver cirrhosis, Lancet, 383 (2014) 1749-1761.

na

[9] F. Moroni, B.J. Dwyer, C. Graham, C. Pass, L. Bailey, L. Ritchie, D. Mitchell, A. Glover, A.

ur

Laurie, S. Doig, E. Hargreaves, A.R. Fraser, M.L. Turner, J.D.M. Campbell, N.W.A. McGowan, J. Barry, J.K. Moore, P.C. Hayes, D.J. Leeming, M.J. Nielsen, K. Musa, J.A. Fallowfield, S.J.

Jo

Forbes, Safety profile of autologous macrophage therapy for liver cirrhosis, Nat Med, (2019). [10] P.S. Ge, B.A. Runyon, Treatment of Patients with Cirrhosis, N Engl J Med, 375 (2016) 2104-2105. [11] Y.A. Lee, M.C. Wallace, S.L. Friedman, Pathobiology of liver fibrosis: a translational success story, Gut, 64 (2015) 830-841. 25

[12] S.L. Friedman, D. Sheppard, J.S. Duffield, S. Violette, Therapy for fibrotic diseases: nearing the starting line, Sci Transl Med, 5 (2013) 167sr161. [13] D. Schuppan, Y.O. Kim, Evolving therapies for liver fibrosis, J Clin Invest, 123 (2013) 18871901. [14] L. Campana, J.P. Iredale, Regression of Liver Fibrosis, Semin Liver Dis, 37 (2017) 1-10. [15] T. Tsuchida, S.L. Friedman, Mechanisms of hepatic stellate cell activation, Nat Rev

ro of

Gastroenterol Hepatol, 14 (2017) 397-411.

[16] G. Marrone, V.H. Shah, J. Gracia-Sancho, Sinusoidal communication in liver fibrosis and regeneration, J Hepatol, 65 (2016) 608-617.

-p

[17] A. Geerts, History, heterogeneity, developmental biology, and functions of quiescent

re

hepatic stellate cells, Semin Liver Dis, 21 (2001) 311-335.

[18] N. Higashi, H. Senoo, Distribution of vitamin A storing lipid droplets in hepatic stellate cells

lP

in liver lobules - A comparative study, Anat Rec Part A, 271a (2003) 240-248. [19] A. Pellicoro, P. Ramachandran, J.P. Iredale, J.A. Fallowfield, Liver fibrosis and repair:

na

immune regulation of wound healing in a solid organ, Nat Rev Immunol, 14 (2014) 181-194.

ur

[20] S.L. Friedman, Hepatic stellate cells: protean, multifunctional, and enigmatic cells of the liver, Physiol Rev, 88 (2008) 125-172.

Jo

[21] X. Liu, J. Xu, D.A. Brenner, T. Kisseleva, Reversibility of Liver Fibrosis and Inactivation of Fibrogenic Myofibroblasts, Curr Pathobiol Rep, 1 (2013) 209-214. [22] L.A. van Grunsven, 3D in vitro models of liver fibrosis, Adv Drug Deliv Rev, 121 (2017) 133-146. [23] I. Mederacke, C.C. Hsu, J.S. Troeger, P. Huebener, X.R. Mu, D.H. Dapito, J.P. Pradere, 26

R.F. Schwabe, Fate tracing reveals hepatic stellate cells as dominant contributors to liver fibrosis independent of its aetiology, Nat Commun, 4 (2013). [24] J.A. Fallowfield, J.P. Iredale, Reversal of liver fibrosis and cirrhosis--an emerging reality, Scott Med J, 49 (2004) 3-6. [25] E. Novo, S. Cannito, C. Paternostro, C. Bocca, A. Miglietta, M. Parola, Cellular and molecular mechanisms in liver fibrogenesis, Arch Biochem Biophys, 548 (2014) 20-37.

ro of

[26] M.V. Machado, G.A. Michelotti, A. Pereira Tde, J. Boursier, L. Kruger, M. Swiderska-Syn, G. Karaca, G. Xie, C.D. Guy, B. Bohinc, K.R. Lindblom, E. Johnson, S. Kornbluth, A.M. Diehl,

Reduced lipoapoptosis, hedgehog pathway activation and fibrosis in caspase-2 deficient mice

-p

with non-alcoholic steatohepatitis, Gut, 64 (2015) 1148-1157.

re

[27] S.I. Chung, H. Moon, H.L. Ju, K.J. Cho, D.Y. Kim, K.H. Han, J.W. Eun, S.W. Nam, S. Ribback, F. Dombrowski, D.F. Calvisi, S.W. Ro, Hepatic expression of Sonic Hedgehog induces

lP

liver fibrosis and promotes hepatocarcinogenesis in a transgenic mouse model, J Hepatol, 64 (2016) 618-627.

ur

(2014) 516-524.

na

[28] B.P. Vaughn, S.C. Robson, M.S. Longhi, Purinergic signaling in liver disease, Dig Dis, 32

[29] P.J. Giraudi, V.J.B. Becerra, V. Marin, N.C. Chavez-Tapia, C. Tiribelli, N. Rosso, The

Jo

importance of the interaction between hepatocyte and hepatic stellate cells in fibrogenesis induced by fatty accumulation, Exp Mol Pathol, 98 (2015) 85-92. [30] A. Wree, A. Eguchi, M.D. McGeough, C.A. Pena, C.D. Johnson, A. Canbay, H.M. Hoffman, A.E. Feldstein, NLRP3 inflammasome activation results in hepatocyte pyroptosis, liver inflammation, and fibrosis in mice, Hepatology, 59 (2014) 898-910. 27

[31] T. Luedde, N. Kaplowitz, R.F. Schwabe, Cell death and cell death responses in liver disease: mechanisms and clinical relevance, Gastroenterology, 147 (2014) 765-783 e764. [32] A. Canbay, P. Taimr, N. Torok, H. Higuchi, S. Friedman, G.J. Gores, Apoptotic body engulfment by a human stellate cell line is profibrogenic, Lab Invest, 83 (2003) 655-663. [33] J.X. Jiang, K. Mikami, S. Venugopal, Y. Li, N.J. Toroek, Apoptotic body engulfment by hepatic stellate cells promotes their survival by the JAK/STAT and Akt/NF-kappa B-dependent

ro of

pathways, J Hepatol, 51 (2009) 139-148.

[34] J.X. Jiang, X.L. Chen, D.K. Hsu, K. Baghy, N. Serizawa, F. Scott, Y. Takada, Y. Takada,

H. Fukada, J. Chen, S. Devaraj, R. Adamson, F.T. Liu, N.J. Torok, Galectin-3 modulates

-p

phagocytosis-induced stellate cell activation and liver fibrosis in vivo , Am J Physiol-Gastr L,

re

302 (2012) G439-G446.

[35] M.A. Konerman, J.C. Jones, S.A. Harrison, Pharmacotherapy for NASH: Current and

lP

emerging, J Hepatol, 68 (2018) 362-375.

[36] M. Pontoglio, J. Barra, M. Hadchouel, A. Doyen, C. Kress, J.P. Bach, C. Babinet, M. Yaniv,

na

Hepatocyte nuclear factor 1 inactivation results in hepatic dysfunction, phenylketonuria, and

ur

renal Fanconi syndrome, Cell, 84 (1996) 575-585. [37] Y.H. Lee, B. Sauer, F.J. Gonzalez, Laron dwarfism and non-insulin-dependent diabetes

Jo

mellitus in the Hnf-1alpha knockout mouse, Mol Cell Biol, 18 (1998) 3059-3068. [38] H. Qian, X. Deng, Z.W. Huang, J. Wei, C.H. Ding, R.X. Feng, X. Zeng, Y.X. Chen, J. Ding, L. Qiu, Z.L. Hu, X. Zhang, H.Y. Wang, J.P. Zhang, W.F. Xie, An HNF1 alpha-regulated feedback circuit modulates hepatic fibrogenesis via the crosstalk between hepatocytes and hepatic stellate cells, Cell Res, 25 (2015) 930-945. 28

[39] W. Seo, H.S. Eun, S.Y. Kim, H.S. Yi, Y.S. Lee, S.H. Park, M.J. Jang, E. Jo, S.C. Kim, Y.M. Han, K.G. Park, W.I. Jeong, Exosome-Mediated Activation of Toll-Like Receptor 3 in Stellate Cells Stimulates Interleukin-17 Production by gamma delta T Cells in Liver Fibrosis, Hepatology, 64 (2016) 616-631. [40] X.D. Ge, E. Arriazu, F. Magdaleno, D.J. Antoine, R. dela Cruz, N. Theise, N. Nieto, High Mobility Group Box-1 Drives Fibrosis Progression Signaling via the Receptor for Advanced

ro of

Glycation End Products in Mice, Hepatology, 68 (2018) 2380-2404.

[41] V. Khanjarsim, J. Karimi, I. Khodadadi, A. Mohammadalipour, M.T. Goodarzi, G. Solgi, M. Hashemnia, Ameliorative Effects of Nilotinib on CCl4 Induced Liver Fibrosis Via Attenuation of

-p

RAGE/HMGB1 Gene Expression and Oxidative Stress in Rat, Chonnam Med J, 53 (2017) 118-

re

126.

[42] X. Li, Q.W. Jin, Q.Y. Yao, B.L. Xu, Z. Li, C.T. Tu, Quercetin attenuates the activation of

lP

hepatic stellate cells and liver fibrosis in mice through modulation of HMGB1-TLR2/4-NF-kappa B signaling pathways, Toxicol Lett, 261 (2016) 1-12.

na

[43] S. Zhang, Z.C. Wang, J. Zhu, T. Xu, Y. Zhao, H.Y. Zhao, F. Tang, Z.L. Li, J.J. Zhou, D.Y.

ur

Gao, X.F. Tian, J.H. Yao, Carnosic Acid Alleviates BDL-Induced Liver Fibrosis through miR29b-3p-Mediated Inhibition of the High-Mobility Group Box 1/Toll-Like Receptor 4 Signaling

Jo

Pathway in Rats, Front Pharmacol, 8 (2018). [44] S.Z. Wei, S.Q. Luo, J. Wang, J.B. Wang, R.S. Li, X.M. Zhang, Y.L. Guo, C. Chen, X. Ma, Z. Chen, H.H. Liu, Z.R. Yang, J.Y. Li, R.L. Wang, Y.M. Zhang, H.Y. Yang, X.H. Xiao, Y.L. Zhao, San-Cao Granule () Ameliorates Hepatic Fibrosis through High Mobility Group Box-1 Protein/Smad Signaling Pathway, Chin J Integr Med, 24 (2018) 502-511. 29

[45] C. Qu, D.D. Zheng, S. Li, Y.J. Liu, A. Lidofsky, J.A. Holmes, J.N. Chen, L. He, L. Wei, Y.D. Liao, H. Yuan, Q.M. Jin, Z.L. Lin, Q.T. Hu, Y.C. Jiang, M.X. Tu, X.J. Chen, W.M. Li, W.Y. Lin, B.C. Fuchs, R.T. Chung, J. Hong, Tyrosine kinase SYK is a potential therapeutic target for liver fibrosis, Hepatology, 68 (2018) 1125-1139. [46] P. Kocabayoglu, A. Lade, Y.A. Lee, A.C. Dragomir, X. Sun, M.I. Fiel, S. Thung, C. Aloman, P. Soriano, Y. Hoshida, S.L. Friedman, beta-PDGF receptor expressed by hepatic stellate cells

ro of

regulates fibrosis in murine liver injury, but not carcinogenesis, J Hepatol, 63 (2015) 141-147.

[47] A. Kikuchi, T. Pradhan-Sundd, S. Singh, S. Nagarajan, N. Loizos, S.P. Monga, PlateletDerived Growth Factor Receptor alpha Contributes to Human Hepatic Stellate Cell Proliferation

-p

and Migration, American Journal of Pathology, 187 (2017) 2273-2287.

re

[48] B.J. Lim, W.K. Lee, H.W. Lee, K.S. Lee, J.K. Kim, H.Y. Chang, J.I. Lee, Selective deletion of hepatocyte platelet-derived growth factor receptor and development of liver fibrosis in mice,

lP

Cell Commun Signal, 16 (2018).

[49] E. Maslak, A. Gregorius, S. Chlopicki, Liver sinusoidal endothelial cells (LSECs) function

na

and NAFLD; NO-based therapy targeted to the liver, Pharmacol Rep, 67 (2015) 689-694.

ur

[50] J. Poisson, S. Lemoinne, C. Boulanger, F. Durand, R. Moreau, D. Valla, P.E. Rautou, Liver sinusoidal endothelial cells: Physiology and role in liver diseases, J Hepatol, 66 (2017) 212-

Jo

227.

[51] L.D. DeLeve, X.D. Wang, L.P. Hu, M.K. McCuskey, R.S. McCuskey, Rat liver sinusoidal endothelial cell phenotype is maintained by paracrine and autocrine regulation, Am J PhysiolGastr L, 287 (2004) G757-G763. [52] B.S. Ding, D.J. Nolan, J.M. Butler, D. James, A.O. Babazadeh, Z. Rosenwaks, V. Mittal, H. 30

Kobayashi, K. Shido, D. Lyden, T.N. Sato, S.Y. Rabbany, S. Rafii, Inductive angiocrine signals from sinusoidal endothelium are required for liver regeneration, Nature, 468 (2010) 310-315. [53] B.S. Ding, Z.W. Cao, R. Lis, D.J. Nolan, P.P. Guo, M. Simons, M.E. Penfold, K. Shido, S.Y. Rabbany, S. Rafii, Divergent angiocrine signals from vascular niche balance liver regeneration and fibrosis, Nature, 505 (2014) 97-+. [54] L.D. DeLeve, X.D. Wang, Y.M. Guo, Sinusoidal endothelial cells prevent rat stellate cell

ro of

activation and promote reversion to quiescence, Hepatology, 48 (2008) 920-930.

[55] G.H. Xie, X.D. Wang, L. Wang, L. Wang, R.D. Atkinson, G.C. Kanel, W.A. Gaarde, L.D. DeLeve, Role of Differentiation of Liver Sinusoidal Endothelial Cells in Progression and

-p

Regression of Hepatic Fibrosis in Rats, Gastroenterology, 142 (2012) 918-U392.

re

[56] G. Marrone, L. Russo, E. Rosado, D. Hide, G. Garcia-Cardena, J.C. Garcia-Pagan, J. Bosch, J. Gracia-Sancho, The transcription factor KLF2 mediates hepatic endothelial protection

lP

and paracrine endothelial-stellate cell deactivation induced by statins, J Hepatol, 58 (2013) 98103.

na

[57] R.S. Wang, Q. Ding, U. Yaqoob, T.M. de Assuncao, V.K. Verma, P. Hirsova, S. Cao, D.

ur

Mukhopadhyay, R.C. Huebert, V.H. Shah, Exosome Adherence and Internalization by Hepatic Stellate Cells Triggers Sphingosine 1-Phosphate-dependent Migration, J Biol Chem, 290 (2015)

Jo

30684-U30651.

[58] M. Di Pascoli, M. Divi, A. Rodriguez-Vilarrupla, E. Rosado, J. Gracia-Sancho, M. Vilaseca, J. Bosch, J.C. Garcia-Pagan, Resveratrol improves intrahepatic endothelial dysfunction and reduces hepatic fibrosis and portal pressure in cirrhotic rats, J Hepatol, 58 (2013) 904-910. [59] M. Guillaume, A. Rodriguez-Vilarrupla, J. Gracia-Sancho, E. Rosado, A. Mancini, J. Bosch, 31

J.C. Garcia-Pagan, Recombinant human manganese superoxide dismutase reduces liver fibrosis and portal pressure in CCl4-cirrhotic rats, J Hepatol, 58 (2013) 240-246. [60] C. Zhang, M. Bian, X. Chen, H. Jin, S. Zhao, X. Yang, J. Shao, A. Chen, Q. Guo, F. Zhang, S. Zheng, Oroxylin A prevents angiogenesis of LSECs in liver fibrosis via inhibition of YAP/HIF1alpha signaling, J Cell Biochem, 119 (2018) 2258-2268. [61] G. Xie, X. Wang, L. Wang, L. Wang, R.D. Atkinson, G.C. Kanel, W.A. Gaarde, L.D. Deleve,

ro of

Role of differentiation of liver sinusoidal endothelial cells in progression and regression of hepatic fibrosis in rats, Gastroenterology, 142 (2012) 918-927 e916.

[62] L. Liu, Z. You, H. Yu, L. Zhou, H. Zhao, X. Yan, D. Li, B. Wang, L. Zhu, Y. Xu, T. Xia, Y.

-p

Shi, C. Huang, W. Hou, Y. Du, Mechanotransduction-modulated fibrotic microniches reveal the

re

contribution of angiogenesis in liver fibrosis, Nat Mater, 16 (2017) 1252-1261. [63] M.P. Holt, L. Cheng, C. Ju, Identification and characterization of infiltrating macrophages

lP

in acetaminophen-induced liver injury, J Leukoc Biol, 84 (2008) 1410-1421. [64] F. Tacke, H.W. Zimmermann, Macrophage heterogeneity in liver injury and fibrosis, J

na

Hepatol, 60 (2014) 1090-1096.

ur

[65] R. Sasaki, P.B. Devhare, R. Steele, R. Ray, R.B. Ray, Hepatitis C virus-induced CCL5 secretion from macrophages activates hepatic stellate cells, Hepatology, 66 (2017) 746-757.

Jo

[66] J.P. Pradere, J. Kluwe, S. De Minicis, J.J. Jiao, G.Y. Gwak, D.H. Dapito, M.K. Jang, N.D. Guenther, I. Mederacke, R. Friedman, A.C. Dragomir, C. Aloman, R.F. Schwabe, Hepatic Macrophages But Not Dendritic Cells Contribute to Liver Fibrosis by Promoting the Survival of Activated Hepatic Stellate Cells in Mice, Hepatology, 58 (2013) 1461-1473. [67] F. Oakley, M. Meso, J.P. Iredale, K. Green, C.J. Marek, X. Zhou, M.J. May, H. Millward32

Sadler, M.C. Wright, D.A. Mann, Inhibition of inhibitor of kappaB kinases stimulates hepatic stellate cell apoptosis and accelerated recovery from rat liver fibrosis, Gastroenterology, 128 (2005) 108-120. [68] J.H. Wang, I. Leclercq, J.M. Brymora, N. Xu, M. Ramezani-Moghadam, R.M. London, D. Brigstock, J. George, Kupffer Cells Mediate Leptin-Induced Liver Fibrosis, Gastroenterology, 137 (2009) 713-723.

ro of

[69] M. Imamura, T. Ogawa, Y. Sasaguri, K. Chayama, H. Ueno, Suppression of macrophage infiltration inhibits activation of hepatic stellate cells and liver fibrogenesis in rats, Gastroenterology, 128 (2005) 138-146.

-p

[70] N. Nieto, Oxidative-stress and IL-6 mediate the fibrogenic effects of [corrected] Kupffer

re

cells on stellate cells, Hepatology, 44 (2006) 1487-1501.

[71] P.S. Chu, N. Nakamoto, H. Ebinuma, S. Usui, K. Saeki, A. Matsumoto, Y. Mikami, K.

lP

Sugiyama, K. Tomita, T. Kanai, H. Saito, T. Hibi, C-C motif chemokine receptor 9 positive

(2013) 337-350.

na

macrophages activate hepatic stellate cells and promote liver fibrosis in mice, Hepatology, 58

ur

[72] M. Matsuda, S. Tsurusaki, N. Miyata, E. Saijou, H. Okochi, A. Miyajima, M. Tanaka, Oncostatin M causes liver fibrosis by regulating cooperation between hepatic stellate cells and

Jo

macrophages in mice, Hepatology, 67 (2018) 296-312. [73] X. Ge, E. Arriazu, F. Magdaleno, D.J. Antoine, R. Dela Cruz, N. Theise, N. Nieto, High Mobility Group Box-1 Drives Fibrosis Progression Signaling via the Receptor for Advanced Glycation End Products in Mice, Hepatology, 68 (2018) 2380-2404. [74] F. Tacke, Targeting hepatic macrophages to treat liver diseases, J Hepatol, 66 (2017) 33

1300-1312. [75] K.R. Karlmark, R. Weiskirchen, H.W. Zimmermann, N. Gasssler, F. Ginhoux, C. Weber, M. Merad, T. Luedde, C. Trautwein, F. Tacke, Hepatic Recruitment of the Inflammatory Gr1(+) Monocyte Subset Upon Liver Injury Promotes Hepatic Fibrosis, Hepatology, 50 (2009) 261-274. [76] E. Seki, S. De Minicis, G.Y. Gwak, J. Kluwe, S. Inokuchi, C.A. Bursill, J.M. Llovet, D.A. Brenner, R.F. Schwabe, CCR1 and CCR5 promote hepatic fibrosis in mice, J Clin Invest, 119

ro of

(2009) 1858-1870.

[77] V.R. A.J. Sanyal, S. Harrison, M.F. Abdelmalek, G.P. Aithal, J. Caballeria, et al.,

Cenicriviroc placebo for the treatment of nonalcoholic steatohepatitis with liver fibrosis Results

-p

from the Year 1 primary analysis of the Phase 2b CENTAUR study, Hepatology, 64 (2016) pp.

re

1118A-1119A.

[78] S.L. Friedman, V. Ratziu, S.A. Harrison, M.F. Abdelmalek, G.P. Aithal, J. Caballeria, S.

lP

Francque, G. Farrell, K.V. Kowdley, A. Craxi, K. Simon, L. Fischer, L. Melchor-Khan, J. Vest, B.L. Wiens, P. Vig, S. Seyedkazemi, Z. Goodman, V.W.S. Wong, R. Loomba, F. Tacke, A.

na

Sanyal, E. Lefebvre, A randomized, placebo-controlled trial of cenicriviroc for treatment of

ur

nonalcoholic steatohepatitis with fibrosis, Hepatology, 67 (2018) 1754-1767. [79] K.R. Karlmark, H.W. Zimmermann, C. Roderburg, N. Gassler, H.E. Wasmuth, T. Luedde,

Jo

C. Trautwein, F. Tacke, The fractalkine receptor CX(3)CR1 protects against liver fibrosis by controlling differentiation and survival of infiltrating hepatic monocytes, Hepatology, 52 (2010) 1769-1782. [80] A. Pellicoro, R.L. Aucott, P. Ramachandran, A.J. Robson, J.A. Fallowfield, V.K. Snowdon, S.N. Hartland, M. Vernon, J.S. Duffield, R.C. Benyon, S.J. Forbes, J.P. Iredale, Elastin 34

accumulation is regulated at the level of degradation by macrophage metalloelastase (MMP12) during experimental liver fibrosis, Hepatology, 55 (2012) 1965-1975. [81] T. Aoyama, S. Inokuchi, D.A. Brenner, E. Seki, CX3CL1-CX3CR1 interaction prevents CCl4-induced liver inflammation and fibrosis, Hepatology, 52 (2010) 447a-447a. [82] C. Barcena, G. Aran, L. Perea, L. Sanjurjo, E. Tellez, A. Oncins, H. Masnou, I. Serra, M. Garcia-Gallo, L. Kremer, M. Sala, C. Armengol, P. Sancho-Bru, M.R. Sarrias, CD5L is a

ro of

pleiotropic player in liver fibrosis controlling damage, fibrosis and immune cell content, EBioMedicine, (2019).

[83] X.J. Luo, H.G. Li, L.Q. Ma, J. Zhou, X. Guo, S.L. Woo, Y. Pei, L.R. Knight, M. Deveau, Y.M.

-p

Chen, X.X. Qian, X.Q. Xiao, Q.F. Li, X.B. Chen, Y.Q. Huo, K. McDaniel, H. Francis, S. Glaser,

re

F.Y. Meng, G. Alpini, C.D. Wu, Expression of STING Is Increased in Liver Tissues From Patients With NAFLD and Promotes Macrophage-Mediated Hepatic Inflammation and Fibrosis

lP

in Mice, Gastroenterology, 155 (2018) 1971-+.

[84] J. Han, X. Zhang, J.K. Lau, K. Fu, H.C. Lau, W. Xu, E.S. Chu, H. Lan, J. Yu, Bone marrow-

na

derived macrophage contributes to fibrosing steatohepatitis through activating hepatic stellate

ur

cells, J Pathol, (2019).

[85] P.F. Ma, C.C. Gao, J. Yi, J.L. Zhao, S.Q. Liang, Y. Zhao, Y.C. Ye, J. Bai, Q.J. Zheng, K.F.

Jo

Dou, H. Han, H.Y. Qin, Cytotherapy with M1-polarized macrophages ameliorates liver fibrosis by modulating immune microenvironment in mice, J Hepatol, 67 (2017) 770-779. [86] X.A. Zhao, G. Chen, Y. Liu, Y. Chen, H. Wu, Y. Xiong, G. Wang, B. Jia, Y. Li, J. Xia, J. Wang, X. Yan, Z. Zhang, R. Huang, C. Wu, Curcumin reduces Ly6C(hi) monocyte infiltration to protect against liver fibrosis by inhibiting Kupffer cells activation to reduce chemokines secretion, 35

Biomed Pharmacother, 106 (2018) 868-878. [87] R. Huang, Y. Liu, Y. Xiong, H. Wu, G. Wang, Z. Sun, J. Chen, X. Yan, Z. Pan, J. Xia, Z. Zhang, J. Wang, C. Wu, Curcumin protects against liver fibrosis by attenuating infiltration of Gr1hi monocytes through inhibition of monocyte chemoattractant protein-1, Discov Med, 21 (2016) 447-457. [88] C.J. Weston, E.L. Shepherd, L.C. Claridge, P. Rantakari, S.M. Curbishley, J.W. Tomlinson,

ro of

S.G. Hubscher, G.M. Reynolds, K. Aalto, Q.M. Anstee, S. Jalkanen, M. Salmi, D.J. Smith, C.P.

Day, D.H. Adams, Vascular adhesion protein-1 promotes liver inflammation and drives hepatic fibrosis, J Clin Invest, 125 (2015) 501-520.

-p

[89] K. Hudspeth, E. Pontarini, P. Tentorio, M. Cimino, M. Donadon, G. Torzilli, E. Lugli, S. Della

re

Bella, M.E. Gershwin, D. Mavilio, The role of natural killer cells in autoimmune liver disease: a comprehensive review, J Autoimmun, 46 (2013) 55-65.

lP

[90] S. Radaeva, R. Sun, B. Jaruga, V.T. Nguyen, Z. Tian, B. Gao, Natural killer cells ameliorate liver fibrosis by killing activated stellate cells in NKG2D-dependent and tumor necrosis factor-

na

related apoptosis-inducing ligand-dependent manners, Gastroenterology, 130 (2006) 435-452.

ur

[91] A. Melhem, N. Muhanna, A. Bishara, C.E. Alvarez, Y. Ilan, T. Bishara, A. Horani, M. Nassar, S.L. Friedman, R. Safadi, Anti-fibrotic activity of NK cells in experimental liver injury through

Jo

killing of activated HSC, J Hepatol, 45 (2006) 60-71. [92] N. Muhanna, L. Abu Tair, S. Doron, J. Amer, M. Azzeh, M. Mahamid, S. Friedman, R. Safadi, Amelioration of hepatic fibrosis by NK cell activation, Gut, 60 (2011) 90-98. [93] C. Gur, S. Doron, S. Kfir-Erenfeld, E. Horwitz, L. Abu-Tair, R. Safadi, O. Mandelboim, NKp46-mediated killing of human and mouse hepatic stellate cells attenuates liver fibrosis, Gut, 36

61 (2012) 885-893. [94] S. Radaeva, L. Wang, S. Radaev, W.I. Jeong, O. Park, B. Gao, Retinoic acid signaling sensitizes hepatic stellate cells to NK cell killing via upregulation of NK cell activating ligand RAE1, Am J Physiol Gastrointest Liver Physiol, 293 (2007) G809-816. [95] W.I. Jeong, O. Park, Y.G. Suh, J.S. Byun, S.Y. Park, E. Choi, J.K. Kim, H. Ko, H. Wang, A.M. Miller, B. Gao, Suppression of innate immunity (natural killer cell/interferon-gamma) in the

ro of

advanced stages of liver fibrosis in mice, Hepatology, 53 (2011) 1342-1351.

[96] V. Krizhanovsky, M. Yon, R.A. Dickins, S. Hearn, J. Simon, C. Miething, H. Yee, L. Zender,

S.W. Lowe, Senescence of activated stellate cells limits liver fibrosis, Cell, 134 (2008) 657-667.

-p

[97] H. Wang, S. Yin, Natural killer T cells in liver injury, inflammation and cancer, Expert Rev

re

Gastroenterol Hepatol, 9 (2015) 1077-1085.

[98] O. Park, W.I. Jeong, L. Wang, H. Wang, Z.X. Lian, M.E. Gershwin, B. Gao, Diverse roles

lP

of invariant natural killer T cells in liver injury and fibrosis induced by carbon tetrachloride, Hepatology, 49 (2009) 1683-1694.

na

[99] Z. Jin, R. Sun, H. Wei, X. Gao, Y. Chen, Z. Tian, Accelerated liver fibrosis in hepatitis B

ur

virus transgenic mice: involvement of natural killer T cells, Hepatology, 53 (2011) 219-229. [100] W.K. Syn, Y.H. Oo, T.A. Pereira, G.F. Karaca, Y. Jung, A. Omenetti, R.P. Witek, S.S.

Jo

Choi, C.D. Guy, C.M. Fearing, V. Teaberry, F.E. Pereira, D.H. Adams, A.M. Diehl, Accumulation of natural killer T cells in progressive nonalcoholic fatty liver disease, Hepatology, 51 (2010) 1998-2007. [101] R.X. Wang, C.R. Yu, R.M. Mahdi, C.E. Egwuagu, Novel IL27p28/IL12p40 Cytokine Suppressed Experimental Autoimmune Uveitis by Inhibiting Autoreactive Th1/Th17 Cells and 37

Promoting Expansion of Regulatory T Cells, J Biol Chem, 287 (2012) 36012-36021. [102] D. Dibra, J. Cutrera, X.Q. Xia, B. Kallakury, L. Mishra, S.L. Li, Interleukin-30: A novel antiinflammatory cytokine candidate for prevention and treatment of inflammatory cytokineinduced liver injury, Hepatology, 55 (2012) 1204-1214. [103] A. Mitra, A. Satelli, J. Yan, X.Q. Xia, M. Gagea, C.A. Hunter, L. Mishra, S.L. Li, IL-30 (IL27p28) Attenuates Liver Fibrosis Through Inducing NKG2D-Rae1 Interaction Between NKT

ro of

and Activated Hepatic Stellate Cells in Mice, Hepatology, 60 (2014) 2027-2039.

[104] R. Li, J. Dong, X.Q. Bu, Y. Huang, J.Y. Yang, X. Dong, J. Liu, Interleukin-6 promotes the migration and cellular senescence and inhibits apoptosis of human intrahepatic biliary epithelial

-p

cells, J Cell Biochem, 119 (2018) 2135-2143.

re

[105] L. Meng, M. Quezada, P. Levine, Y. Han, K. McDaniel, T. Zhou, E. Lin, S. Glaser, F.

185 (2015) 602-609.

lP

Meng, H. Francis, G. Alpini, Functional role of cellular senescence in biliary injury, Am J Pathol,

[106] N. Sedlaczek, J.D. Jia, M. Bauer, H. Herbst, M. Ruehl, E.G. Hahn, D. Schuppan,

na

Proliferating bile duct epithelial cells are a major source of connective tissue growth factor in

ur

rat biliary fibrosis, Am J Pathol, 158 (2001) 1239-1244. [107] A. Omenetti, L. Yang, Y.X. Li, S.J. McCall, Y. Jung, J.K. Sicklick, J. Huang, S. Choi, A.

Jo

Suzuki, A.M. Diehl, Hedgehog-mediated mesenchymal-epithelial interactions modulate hepatic response to bile duct ligation, Lab Invest, 87 (2007) 499-514. [108] Y. Wan, F. Meng, N. Wu, T. Zhou, J. Venter, H. Francis, L. Kennedy, T. Glaser, F. Bernuzzi, P. Invernizzi, S. Glaser, Q. Huang, G. Alpini, Substance P increases liver fibrosis by differential changes in senescence of cholangiocytes and hepatic stellate cells, Hepatology, 66 38

(2017) 528-541. [109] X. Wang, A. Lopategi, X. Ge, Y. Lu, N. Kitamura, R. Urtasun, T.M. Leung, M.I. Fiel, N. Nieto, Osteopontin induces ductular reaction contributing to liver fibrosis, Gut, 63 (2014) 18051818. [110] A. Miyajima, M. Tanaka, T. Itoh, Stem/Progenitor Cells in Liver Development, Homeostasis, Regeneration, and Reprogramming, Cell Stem Cell, 14 (2014) 561-574.

ro of

[111] M. Kitade, K. Kaji, H. Yoshiji, Relationship between hepatic progenitor cell-mediated liver regeneration and non-parenchymal cells, Hepatol Res, 46 (2016) 1187-1193.

[112] C. Kordes, I. Sawitza, A. Muller-Marbach, N. Ale-Agha, V. Keitel, H. Klonowski-Stumpe,

-p

D. Haussinger, CD133(+) hepatic stellate cells are progenitor cells, Biochem Bioph Res Co,

re

352 (2007) 410-417.

[113] C. Kordes, I. Sawitza, S. Gotze, D. Herebian, D. Haussinger, Hepatic stellate cells

lP

contribute to progenitor cells and liver regeneration, J Clin Invest, 124 (2014) 5503-5515. [114] G.A. Michelotti, G.H. Xie, M. Swiderska, S.S. Choi, G. Karaca, L. Kruger, R. Premont, L.

na

Yang, W.K. Syn, D. Metzger, A.M. Diehl, Smoothened is a master regulator of adult liver repair,

ur

J Clin Invest, 123 (2013) 2380-2394.

[115] M.J. Williams, A.D. Clouston, S.J. Forbes, Links Between Hepatic Fibrosis, Ductular

Jo

Reaction, and Progenitor Cell Expansion, Gastroenterology, 146 (2014) 349-356. [116] Y. Aimaiti, X. Jin, Y. Shao, W. Wang, D. Li, Hepatic stellate cells regulate hepatic progenitor cells differentiation via the TGF-beta1/Jagged1 signaling axis, J Cell Physiol, 234 (2019) 9283-9296. [117] P. Gajalakshmi, S. Majumder, C.S. Viebahn, A. Swaminathan, G.C. Yeoh, S. Chatterjee, 39

Interleukin-6 secreted by bipotential murine oval liver stem cells induces apoptosis of activated hepatic stellate cells by activating NF-kappa B-inducible nitric oxide synthase signaling, Biochem Cell Biol, 95 (2017) 263-272. [118] Y. Chang, D. Bluteau, N. Debili, W. Vainchenker, From hematopoietic stem cells to platelets, J Thromb Haemost, 5 (2007) 318-327. [119] M.M. Zaldivar, K. Pauels, P. von Hundelshausen, M.L. Berres, P. Schmitz, J. Bornemann,

ro of

M.A. Kowalska, N. Gassler, K.L. Streetz, R. Weiskirchen, C. Trautwein, C. Weber, H.E.

Wasmuth, CXC Chemokine Ligand 4 (CXCL4) Is a Platelet-Derived Mediator of Experimental Liver Fibrosis, Hepatology, 51 (2010) 1345-1353.

-p

[120] P.A. Lang, C. Contaldo, P. Georgiev, A.M. El-Badry, M. Recher, M. Kurrer, L. Cervantes-

re

Barragan, B. Ludewig, T. Calzascia, B. Bolinger, D. Merkler, B. Odermatt, M. Bader, R. Graf, P.A. Clavien, A.N. Hegazy, M. Loehning, N.L. Harris, P.S. Ohashi, H. Hengartner, R.M.

14 (2008) 756-761.

lP

Zinkernagel, K.S. Lang, Aggravation of viral hepatitis by platelet-derived serotonin, Nat Med,

na

[121] M.W. Laschke, S. Dold, M.D. Menger, B. Jeppsson, H. Thorlacius, Platelet-dependent

ur

accumulation of leukocytes in sinusoids mediates hepatocellular damage in bile duct ligationinduced cholestasis, Brit J Pharmacol, 153 (2008) 148-156.

Jo

[122] A. Khandoga, P. Biberthaler, K. Messmer, F. Krombach, Platelet-endothelial cell interactions during hepatic ischemia-reperfusion in vivo: a systematic analysis, Microvasc Res, 65 (2003) 71-77. [123] S. Pak, T. Kondo, Y. Nakano, S. Murata, K. Fukunaga, T. Oda, R. Sasaki, N. Ohkohchi, Platelet adhesion in the sinusoid caused hepatic injury by neutrophils after hepatic ischemia 40

reperfusion, Platelets, 21 (2010) 282-288. [124] S. Murata, N. Ohkohchi, R. Matsuo, O. Ikeda, A. Myronovych, R. Hoshi, Platelets promote liver regeneration in early period after hepatectomy in mice, World J Surg, 31 (2007) 808-816. [125] H. Holmsen, Physiological Functions of Platelets, Ann Med, 21 (1989) 23-30. [126] L. Mazzucco, P. Borzini, R. Gope, Platelet-derived factors involved in tissue repair-from signal to function, Transfus Med Rev, 24 (2010) 218-234.

ro of

[127] M. Lesurtel, R. Graf, B. Aleil, D.J. Walther, Y.H. Tian, W. Jochum, C. Gachet, M. Bader,

P.A. Clavien, Platelet-derived serotonin mediates liver regeneration, Science, 312 (2006) 104107.

-p

[128] H. Kono, H. Fujii, K. Suzuki-Inoue, O. Inoue, S. Furuya, K. Hirayama, Y. Akazawa, Y.

re

Nakata, C. Sun, N. Tsukiji, T. Shirai, Y. Ozaki, The platelet-activating receptor C-type lectin receptor-2 plays an essential role in liver regeneration after partial hepatectomy in mice, J

lP

Thromb Haemost, 15 (2017) 998-1008.

[129] S. Murata, I. Hashimoto, Y. Nakano, A. Myronovch, M. Watanabe, N. Hkohchi, Single

na

Administration of Thrombopoietin Prevents Progression of Liver Fibrosis and Promotes Liver

ur

Regeneration After Partial Hepatectomy in Cirrhotic Rats, Ann Surg, 248 (2008) 821-828. [130] N. Ikeda, S. Murata, T. Maruyama, T. Tamura, R. Nozaki, T. Kawasaki, K. Fukunaga, T.

Jo

Oda, R. Sasaki, M. Homma, N. Ohkohchi, Platelet-derived adenosine 5 '-triphosphate suppresses activation of human hepatic stellate cell: In vitro study, Hepatol Res, 42 (2012) 91102. [131] A.A. Tanikawa, R.M.T. Grotto, G.F. Silva, A.C. Ferrasi, V.C.R. Sarnighausen, M.I.D.C. Pardini, Platelet-derived growth factor A mRNA in platelets is associated with the degree of 41

hepatic fibrosis in chronic hepatitis C, Rev Soc Bras Med Tro, 50 (2017) 113-116. [132] S. Ghafoory, R. Varshney, T. Robison, K. Kouzbari, S. Woolington, B. Murphy, L.J. Xia, J. Ahamed, Platelet TGF-beta 1 deficiency decreases liver fibrosis in a mouse model of liver injury, Blood Adv, 2 (2018) 470-480. [133] J.L. Xia, C.S. Dai, G.K. Michalopoulos, Y.H. Liu, Hepatocyte growth factor attenuates liver fibrosis induced by bile duct ligation, American Journal of Pathology, 168 (2006) 1500-1512.

ro of

[134] T. Kodama, T. Takehara, H. Hikita, S. Shimizu, W. Li, T. Miyagi, A. Hosui, T. Tatsumi, H. Ishida, S. Tadokoro, A. Ido, H. Tsubouchi, N. Hayashi, Thrombocytopenia Exacerbates Cholestasis-Induced Liver Fibrosis in Mice, Gastroenterology, 138 (2010) 2487-U2370.

-p

[135] P. Starlinger, S. Haegele, F. Offensperger, L. Oehlberger, D. Pereyra, J.B. Kral, W.C.

re

Schrottmaier, S. Badrnya, T. Reiberger, A. Ferlitsch, J. Stift, F. Luf, C. Brostjan, T. Gruenberger, A. Assinger, The profile of platelet alpha-granule released molecules affects postoperative liver

lP

regeneration, Hepatology, 63 (2016) 1675-1688.

[136] L.Y. King, K.B. Johnson, H. Zheng, L. Wei, T. Gudewicz, Y. Hoshida, K.E. Corey, T. Ajayi,

na

N. Ufere, T.F. Baumert, A.T. Chan, K.K. Tanabe, B.C. Fuchs, R.T. Chung, Host Genetics

ur

Predict Clinical Deterioration in HCV-Related Cirrhosis, Plos One, 9 (2014). [137] S. Yoshida, N. Ikenaga, S.B. Liu, Z.W. Peng, J. Chung, D.Y. Sverdlov, M. Miyamoto, Y.O.

Jo

Kim, S. Ogawa, R.H. Arch, D. Schuppan, Y. Popov, Extrahepatic platelet-derived growth factorbeta, delivered by platelets, promotes activation of hepatic stellate cells and biliary fibrosis in mice, Gastroenterology, 147 (2014) 1378-1392. [138] S. Schafer, O. Zerbe, A.M. Gressner, The synthesis of proteoglycans in fat-storing cells of rat liver, Hepatology, 7 (1987) 680-687. 42

[139] A.M. Elsharkawy, M.C. Wright, R.T. Hay, M.J. Arthur, T. Hughes, M.J. Bahr, K. Degitz, D.A. Mann, Persistent activation of nuclear factor-kappaB in cultured rat hepatic stellate cells involves the induction of potentially novel Rel-like factors and prolonged changes in the expression of IkappaB family proteins, Hepatology, 30 (1999) 761-769. [140] S.L. Friedman, F.J. Roll, Isolation and culture of hepatic lipocytes, Kupffer cells, and sinusoidal endothelial cells by density gradient centrifugation with Stractan, Anal Biochem, 161

ro of

(1987) 207-218.

[141] S.C. Yanguas, B. Cogliati, J. Willebrords, M. Maes, I. Colle, B. van den Bossche, C.P.M.S.

de Oliveira, W. Andraus, V.A. Alves, I. Leclercq, M. Vinken, Experimental models of liver fibrosis,

-p

Arch Toxicol, 90 (2016) 1025-1048.

re

[142] L. Xu, A.Y. Hui, E. Albanis, M.J. Arthur, S.M. O'Byrne, W.S. Blaner, P. Mukherjee, S.L. Friedman, F.J. Eng, Human hepatic stellate cell lines, LX-1 and LX-2: new tools for analysis of

lP

hepatic fibrosis, Gut, 54 (2005) 142-151.

[143] N. Shibata, T. Watanabe, T. Okitsu, M. Sakaguchi, M. Takesue, T. Kunieda, K. Omoto,

na

S. Yamamoto, N. Tanaka, N. Kobayashi, Establishment of an immortalized human hepatic

ur

stellate cell line to develop antifibrotic therapies, Cell Transplant, 12 (2003) 499-507. [144] R. Borojevic, A.N. Monteiro, S.A. Vinhas, G.B. Domont, P.A. Mourao, H. Emonard, G.

Jo

Grimaldi, Jr., J.A. Grimaud, Establishment of a continuous cell line from fibrotic schistosomal granulomas in mice livers, In vitro Cell Dev Biol, 21 (1985) 382-390. [145] M. Fausther, J.R. Goree, E.G. Lavoie, A.L. Graham, J. Sevigny, J.A. Dranoff, Establishment and characterization of rat portal myofibroblast cell lines, PLoS One, 10 (2015) e0121161. 43

[146] P.J. O'Brien, W. Irwin, D. Diaz, E. Howard-Cofield, C.M. Krejsa, M.R. Slaughter, B. Gao, N. Kaludercic, A. Angeline, P. Bernardi, P. Brain, C. Hougham, High concordance of druginduced human hepatotoxicity with in vitro cytotoxicity measured in a novel cell-based model using high content screening, Arch Toxicol, 80 (2006) 580-604. [147] C. Aninat, A. Piton, D. Glaise, T. Le Charpentier, S. Langouet, F. Morel, C. GuguenGuillouzo, A. Guillouzo, Expression of cytochromes P450, conjugating enzymes and nuclear

ro of

receptors in human hepatoma HepaRG cells, Drug Metab Dispos, 34 (2006) 75-83.

[148] A. Guillouzo, A. Corlu, C. Aninat, D. Glaise, F. Morel, C. Guguen-Guillouzo, The human hepatoma HepaRG cells: A highly differentiated model for studies of liver metabolism and

-p

toxicity of xenobiotics, Chem-Biol Interact, 168 (2007) 66-73.

re

[149] J. Herrmann, A.M. Gressner, R. Weiskirchen, Immortal hepatic stellate cell lines: useful tools to study hepatic stellate cell biology and function?, J Cell Mol Med, 11 (2007) 704-722.

lP

[150] Y. Li, J.I. Sun, J. Wang, W.H. Lu, L.Y. Xie, J. Lv, H.X. Li, S.F. Yang, Expression of Vsig4 attenuates macrophage-mediated hepatic inflammation and fibrosis in high fat diet (HFD)-

na

induced mice, Biochem Bioph Res Co, 516 (2019) 858-865.

ur

[151] W. Wirz, M. Antoine, C.G. Tag, A.M. Gressner, T. Kor, C. Hellerbrand, P. Kiefer, Hepatic stellate cells display a functional vascular smooth muscle cell phenotype in a three-dimensional

Jo

co-culture model with endothelial cells, Differentiation, 76 (2008) 784-794. [152] P. Olinga, D. Schuppan, Precision-cut liver slices: a tool to model the liver ex vivo, J Hepatol, 58 (2013) 1252-1253. [153] S. Vatakuti, W.G. Schoonen, M.L. Elferink, G.M. Groothuis, P. Olinga, Acute toxicity of CCl4 but not of paracetamol induces a transcriptomic signature of fibrosis in precision-cut liver 44

slices, Toxicol In vitro, 29 (2015) 1012-1020. [154] I.M. Westra, H.A.M. Mutsaers, T. Luangmonkong, M. Hadi, D. Oosterhuis, K.P. de Jong, G.M.M. Groothuis, P. Olinga, Human precision-cut liver slices as a model to test antifibrotic drugs in the early onset of liver fibrosis, Toxicology in vitro, 35 (2016) 77-85. [155] T. Luangmonkong, S. Suriguga, E. Bigaeva, M. Boersema, D. Oosterhuis, K.P. de Jong, D. Schuppan, H.A.M. Mutsaers, P. Olinga, Evaluating the antifibrotic potency of galunisertib in

ro of

a human ex vivo model of liver fibrosis, Br J Pharmacol, 174 (2017) 3107-3117.

[156] M.A. Lancaster, J.A. Knoblich, Organogenesis in a dish: Modeling development and disease using organoid technologies, Science, 345 (2014).

-p

[157] N. Prior, P. Inacio, M. Huch, Liver organoids: from basic research to therapeutic

re

applications, Gut, 68 (2019) 2228-2237.

[158] S.B. Leite, T. Roosens, A. El Taghdouini, I. Mannaerts, A.J. Smout, M. Najimi, E. Sokal,

lP

F. Noor, C. Chesne, L.A. van Grunsven, Novel human hepatic organoid model enables testing of drug-induced liver fibrosis in vitro, Biomaterials, 78 (2016) 1-10.

na

[159] T.F. Slater, K.H. Cheeseman, K.U. Ingold, Carbon-Tetrachloride Toxicity as a Model for

ur

Studying Free-Radical Mediated Liver-Injury, Philos T R Soc B, 311 (1985) 633-645. [160] A. Symeonidis, E.G. Trams, Morphologic and functional changes in the livers of rats after

Jo

ligation or excision of the common bile duct, Am J Pathol, 33 (1957) 13-27. [161] B. Delire, P. Starkel, I. Leclercq, Animal Models for Fibrotic Liver Diseases: What We Have, What We Need, and What Is under Development, J Clin Transl Hepato, 3 (2015) 53-66. [162] L.M. DeCarli, C.S. Lieber, Fatty liver in the rat after prolonged intake of ethanol with a nutritionally adequate new liquid diet, J Nutr, 91 (1967) 331-336. 45

[163] M. Setshedi, J.R. Wands, S.M. Monte, Acetaldehyde adducts in alcoholic liver disease, Oxid Med Cell Longev, 3 (2010) 178-185. [164] Q.M. Anstee, R.D. Goldin, Mouse models in non-alcoholic fatty liver disease and steatohepatitis research, Int J Exp Pathol, 87 (2006) 1-16. [165] P. Fickert, G. Zollner, A. Fuchsbichler, C. Stumptner, A.H. Weiglein, F. Lammert, H.U. Marschall, O. Tsybrovskyy, K. Zatloukal, H. Denk, M. Trauner, Ursodeoxycholic acid

cholangioles, Gastroenterology, 123 (2002) 1238-1251.

ro of

aggravates bile infarcts in bile duct-ligated and Mdr2 knockout mice via disruption of

[166] A.P. McCaffrey, H. Nakai, K. Pandey, Z. Huang, F.H. Salazar, H. Xu, S.F. Wieland, P.L.

-p

Marion, M.A. Kay, Inhibition of hepatitis B virus in mice by RNA interference, Nat Biotechnol,

re

21 (2003) 639-644.

[167] M.T. Bility, L.G. Zhang, M.L. Washburn, T.A. Curtis, G.I. Kovalev, L.S. Su, Generation of

lP

a humanized mouse model with both human immune system and liver cells to model hepatitis C virus infection and liver immunopathogenesis, Nat Protoc, 7 (2012) 1608-1617.

na

[168] M.T. Bility, L. Cheng, Z. Zhang, Y. Luan, F. Li, L.Q. Chi, L.G. Zhang, Z.K. Tu, Y.H. Gao,

ur

Y.X. Fu, J.Q. Niu, F.S. Wang, L.S. Su, Hepatitis B Virus Infection and Immunopathogenesis in a Humanized Mouse Model: Induction of Human-Specific Liver Fibrosis and M2-Like

Jo

Macrophages, Plos Pathog, 10 (2014). [169] T. McHedlidze, M. Waldner, S. Zopf, J. Walker, A.L. Rankin, M. Schuchmann, D. Voehringer, A.N. McKenzie, M.F. Neurath, S. Pflanz, S. Wirtz, Interleukin-33-dependent innate lymphoid cells mediate hepatic fibrosis, Immunity, 39 (2013) 357-371. [170] I. Maschmeyer, A.K. Lorenz, K. Schimek, T. Hasenberg, A.P. Ramme, J. Hubner, M. 46

Lindner, C. Drewell, S. Bauer, A. Thomas, N.S. Sambo, F. Sonntag, R. Lauster, U. Marx, A four-organ-chip for interconnected long-term co-culture of human intestine, liver, skin and

re

-p

ro of

kidney equivalents, Lab Chip, 15 (2015) 2688-2699.

Figure legends

lP

Fig. 1. Function and characteristic of activated HSCs.

na

Activation of HSCs comprises two phases: initiation and perpetuation. In the initiation period, quieted HSCs response to injury stimuli and begin to transdifferentiate into

ur

proliferative and fibrogenic myofibroblast-like cells. When activated HSCs proliferate and secrete ECM proteins constantly, leading to accumulation of ECM and formation

Jo

of scar tissue, HSCs are in the process of perpetuation, characterized by specific phenotypic changes including contractility, fibrogenesis, altered matrix degradation, chemotaxis and inflammatory signaling. If fibrosis begins to regress, activated HSCs may develop a potential third phase, resolution, where they may undergo apoptosis or become senescent, or revert to the quiescent phenotype. 47

Fig. 2. Intercellular crosstalk in the activation of HSCs. HSC activation is a dynamic process that mainly depends on the interaction with other “responded” cells, including hepatocytes, liver sinusoidal endothelial cells, hepatic macrophages, NK/NKT cells, biliary epithelial cells, hepatic progenitor cells, and platelets. Activation of HSCs is promoted (black arrows) or inhibited (red arrow) by

ro of

paracrine chemokines and molecular signals from those cells. Pharmacological

re

-p

intervention to each candidate target is shown in the green box.

Jo

ur

na

lP

Fig. 1. Function and characteristic of activated HSC

Fig. 2. Intercellular crosstalk in the activation of HSCs

48

49

ro of

-p

re

lP

na

ur

Jo

f Origin

Primary HSCs

Humans and rodent

LX-2, TWNT-4, GREF-X

Humans

HSC-T6, CFSC, PAV-1, T-HSC/Cl6, RGF

Rats

GRX, A640-IS, JS1, Col-GFP

Mice

e-

RGF-N2 and RGF

Pr

HSCs and hepatocytes

HSCs and KCs/BMDMs

Jo ur

Liver organoids Liver-on-a-chip

In vivo

HSCs and NKT cells

Close link with the in vivo situation

[138-140]

Ease of use; Supplied unlimitedly; Good reproducibility

[141-145]

Hepatocyte damage-dependent HSC activation

[146-148]

Roles of immune cells in the regulation of HSC activation

[73] [150]

Paracrine function of NKT cells on HSC activation

[100]

HSCs and ECs

Intercellular crosstalk between ECs and HSCs

[151]

HSCs and cholangiocytes

Effects of cholangiocytes on HSC activation

[107]

Fresh liver explants from rodents

Maintaining the complex cellular interactions that occur

and humans

in vivo; Limited human supply

Derived from stem cells or organ

Resembling an organ with self-organize through cell

progenitors

sorting

na l

Precision-cut liver slices Ex vivo

References

Rat portal myofibroblast

In vitro

Co-cultures

Characteristics

pr

Model

oo

Table 1 Experimental models of intercellular crosstalk systems

Containing cell culture equivalents of liver

[153-155] [156-158]

Recapitulate the hepatic microcirculatory architecture

[170]

CCl4 toxicity model

Rodent

High reproducibility; Showing the regression of fibrosis

[159]

Common bile duct ligation model

Rodent

Short time to develop; Close to human cholestatic injury

[160]

Humanized animal models

Mice

Similar to human viral infections

[167-168]

Chimeras with BM cells

Mice

Good model to study the specific proteins or signaling

[169]

Hepatocyte-specific gene knockout models

Mice

The role of hepatocyte in HSC activation

[40] [48]

50