Skip to main content
Cellular and Molecular Gastroenterology and Hepatology logoLink to Cellular and Molecular Gastroenterology and Hepatology
. 2024 Jan 10;17(4):553–565. doi: 10.1016/j.jcmgh.2024.01.005

Cellular Interactions and Crosstalk Facilitating Biliary Fibrosis in Cholestasis

Ludovica Ceci 1, Eugenio Gaudio 1, Lindsey Kennedy 2,3,
PMCID: PMC10883986  PMID: 38216052

Abstract

Biliary fibrosis is seen in cholangiopathies, including primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC). In PBC and PSC, biliary fibrosis is associated with worse outcomes and histologic scores. Within the liver, both hepatic stellate cells (HSCs) and portal fibroblasts (PFs) contribute to biliary fibrosis, but their roles can differ. PFs reside near the bile ducts and may be the first responders to biliary damage, whereas HSCs may be recruited later and initiate bridging fibrosis. Indeed, different models of biliary fibrosis can activate PFs and HSCs to varying degrees. The portal niche can be composed of cholangiocytes, HSCs, PFs, endothelial cells, and various immune cells, and interactions between these cell types drive biliary fibrosis. In this review, we discuss the mechanisms of biliary fibrosis and the roles of PFs and HSCs in this process. We will also evaluate cellular interactions and mechanisms that contribute to biliary fibrosis in different models and highlight future perspectives and potential therapeutics.

Keywords: biliary fibrosis, portal fibroblasts, hepatic stellate cells, ductular reaction, bile acids, angiogenesis, immune cells


Summary.

This comprehensive review highlights important research on biliary fibrosis. We discuss hepatic stellate cells and portal fibroblasts and outline the mechanisms and cellular niches influencing biliary fibrosis in different mouse models.

Cholangiopathies target the bile ducts, leading to fibroinflammatory responses that initiate biliary fibrosis and eventually bridging fibrosis and cirrhosis.1 Cholangiopathies are diagnosed in pediatric and adult patients, and the 6 most common cholangiopathies include primary sclerosing cholangitis (PSC), primary biliary cholangitis (PBC), cystic fibrosis–associated liver disease, polycystic liver disease, biliary atresia, and cholangiocarcinoma (CCA).1 PBC and PSC are the most common cholangiopathies, with prevalence reported as 4 in 100,000 and 35 in 100,00, respectively2,3; thus, these diseases will be the focus of this review.

During PBC and PSC, cholangiocytes undergo proliferative, senescent, apoptotic, and/or transdifferentiating events, which modulate the microenvironment through the secretion of growth factors, cholangiokines, senescent-associated secretory factors (SASPs), etc. Ductular reaction (DR) is the presence of atypical bile ducts that lack an apparent lumen, and DR may arise from cholangiocytes, hepatocytes, or hepatic progenitor cells (HPCs).4 It was proposed that senescent cholangiocytes remain at the bile duct and contribute to initial and sustained peribiliary fibrosis via activation of portal fibroblasts (PFs). Contrarily, reactive cholangiocytes undergo proliferation and recruit and activate hepatic stellate cells (HSCs), leading to bridging fibrosis.5 Cholangiocyte response to injury is multifaceted, and divergent fates differentially modulate fibrosis.

PBC and PSC injury follows similar steps: (1) persistent biliary damage leads to cholestasis, (2) bile acid (BA) buildup causes hepatocellular damage, (3) consequent inflammation and immune cell infiltration occur, and (4) the complex of biliary and hepatocellular damage and inflammation invoke fibrosis.6 However, there is nuance in fibrogenesis between PBC and PSC. In both PBC and PSC, biliary fibrosis starts from the portal tract and progresses with fibrotic septa formation leading to portal-portal pattern fibrosis. At later stages, fibrosis extends to the lobular area and continues into cirrhosis.7,8 Furthermore, in PBC, the bile ducts are infiltrated by immune/inflammatory cells that damage the portal tract structure and expand the peribiliary stroma. In PSC, patients are characterized by peribiliary inflammation, concentric fibrosis (onion-like structure) and atrophy, and/or disappearance of small bile ducts. Consequently, in both PSC and PBC, the continuous destruction of intrahepatic bile ducts promotes cholestasis.6

Fibrosis stage predicts survival in PBC patients independent of biochemical response.9 Similarly, fibrosis-related scores predict poorer outcomes for PSC patients.10 In PBC and PSC, biliary fibrosis is a shared pathway, an important prognostic factor, and precedes bridging fibrosis. Obviously, biliary fibrosis is an important pathologic process in PBC and PSC. In this review we will discuss mechanisms and cellular interactions regulating biliary fibrosis.

Hepatic Stellate Cells and Portal Fibroblasts: A Tale of Two Cells

HSCs and PFs are the main hepatic mesenchymal cells that synthesize extracellular matrix (ECM).11 Previous research hypothesized that fibrocytes and hepatocytes or cholangiocytes undergoing epithelial to mesenchymal transition (EMT) may contribute to fibrosis; however, in fibrotic livers fibrocytes are only 3%–6% of the collagen type I expressing cells,12 and neither hepatocytes nor cholangiocytes undergo EMT or contribute to ECM deposition.13,14 HSCs and PFs comprise >95% of collagen expressing cells in fibrosis models.7 Indeed, both PFs and HSCs are involved in collagen deposition around injured bile ducts.15

During normal states, HSCs and PFs are quiescent and may be transiently activated after acute exposure to pathogens or toxins to maintain homeostasis16; however, during PBC and PSC, myofibroblast activation is chronic and persistent.17 HSCs reside in the space of Disse and during quiescence express a neural phenotype; however, injury such as carbon tetrachloride (CCl4) treatment causes HSCs to release their vitamin A stores, synthesize and secrete collagen type I, proliferate, and obtain a myofibroblast phenotype.15,18,19 PFs reside near the portal vein,20 and in the biliary fibrosis model of bile duct ligation (BDL), activated PFs proliferate, take on a myofibroblast phenotype, and secrete collagen type I, collagen type IV, and procollagen III.21,22 Similarities and differences in morphology, features, and characteristics of PFs and HSCs during quiescence and activation are summarized in Table 1. One study suggests that early ECM deposition in biliary fibrosis is due to PF activation after 48 hours of BDL, with HSC contribution after 72 hours of BDL.22 Others have confirmed that PFs are primarily activated in BDL, with partial contribution to HSCs.23 In addition, portal mesenchymal cells (PMCs) in the 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) feeding model of biliary fibrosis display PF signatures, are highly proliferative, and contribute to ECM remodeling. Interestingly, HSC-derived myofibroblasts were non-proliferative but still increased Col1a1 expression in vitro, suggesting that PF-derived PMCs initiate fibrotic septa formation and later recruit HSCs.24 BDL surgery and DDC feeding induced PMC proliferation and up-regulation of fibrotic genes that were not seen in HSCs,25 suggesting that these cells are PFs. In Mdr2-/- mice, activated PFs accounted for 25%, 51%, and 54% of myofibroblasts at 4, 8, and 16 weeks of age, respectively, with the other percentage composed of HSCs,26 signifying that both cell types contribute to biliary fibrosis in Mdr2-/- mice. Although newer studies identify the importance of PFs in biliary fibrosis, many studies have revolved around HSCs. We will further discuss mechanisms and cellular interactions identified in models of biliary fibrosis that affect PFs and HSCs.

Table 1.

List of PF and HSC Features in Quiescence and Activation

Localization in liver PFs
HSCs
Portal vein Space of Disse
Morphology
 Quiescent state
  • Flat

  • Spindle-shaped cells

  • Dark, oval nucleus

  • Actively secrete ECM

  • Stellate-like shape

  • Reduced cellular body volume

  • Spindle-shaped

  • Voluminous oval or elongated nucleus
    • Often compressed by vitamin A and/or retinoid containing lipid drops placed in the perinuclear cytoplasm
  • Dilated, rough endoplasmic reticulum

  • Low number of mitochondria and lysosomes

 Activated state
  • Spindle-shaped cells

  • Smooth muscle-like features:
    • Contractile apparatus
    • Prominent rough endoplasmic reticulum
    • Golgi apparatus producing collagen
    • Peripheral myofilaments
    • Fibronexus (no lamina)
    • Gap junctions
  • Loss of retinoid/vitamin A lipid droplets

  • Flat-stellate shape

  • Expression of contractile fibers (myofibroblast differentiation – contractility) in cytoplasm

  • Matured rough endoplasmic reticulum associated with a well-formed Golgi apparatus

Functions
 Quiescent state
  • Maintain integrity of the biliary tree and portal tract

  • Balanced ECM turnover

  • Low proliferation

  • Storage of vitamin A and retinoids

 Activated state
  • Proliferative

  • Contribute to collagen type I deposition

  • Regulate cholangiocyte proliferation

  • Maintain cholangiocyte polarity

  • Support angiogenesis

  • Fibrogenesis

  • ECM synthesis and degradation

  • Increased cell proliferation

  • Contractility

  • Secretion of cytokines, chemokines, and immunomodulatory signals

  • Acts as antigen-presenting cells

  • Role in liver development and regeneration (hepatocyte proliferation)

  • Support angiogenesis and LSEC capillarization

Markers
 Quiescent state
  • Thy-1

  • Fibulin-2

  • Elastin

  • Gremlin-1

  • ENTPD2

  • IL-6

  • Vitamin A

  • Retinoid

  • Desmin

 Activated state
  • Thy-1

  • Mesothelin

  • Fibulin-2

  • Elastin

  • Gremlin-1

  • Asporin

  • IL-6

  • Collagen 15A1

  • Mucin 16

  • ENTPD2

  • Endoglin

  • CD73

  • CD143

  • ACTA2

  • Glial fibrillary acidic protein

  • Neurotrophin receptor p75

  • Synemin

  • Lecithin:retinol acyltransferase

  • ACTA2

  • Reelin

  • Endoglin

  • CD73

  • CD143

Injury response
  • Primary responders of cholangiocyte damage

  • Primary responders of hepatocyte damage

 ECM synthesis
  • Elastic fibers (elastin core surrounded by fibrillin-rich microfibrils)

  • Collagen XV

  • Collagen I and III

  • Fibronectin

  • Thrombospondin-1

  • Proteoglycans

ACTA2, alpha smooth muscle actin; CD, cluster of differentiation; ENTPD2, ectonucleotidase 2; LSEC, lymphatic sinusoidal endothelial cell; Thy-1, thymocyte differentiation antigen-1.

Ductular Reaction: A Catch-22

DR correlates with fibrosis stage in PBC, and higher DR inversely correlates with ursodeoxycholic acid (UDCA) response in PBC.27 In PSC, DR is associated with higher expression of ECM-related genes, including fibronectin-1 (FN-1), laminin subunit gamma-2, and collagens.28 After BDL for 24 hours, DR and portal ECM deposition occur and precede myofibroblast presence that appeared 48 hours after BDL,21 indicating that DR prompts myofibroblast activation. In turn, prolyl-4-hydroxylase, an enzyme that synthesizes collagen, promotes DR and biliary fibrosis in DDC-fed mice.29 Cytokeratin-19 (CK-19) is responsible for cell structure and in the liver is a bona fide cholangiocyte marker. Loss of CK-19 reduced DR and biliary fibrosis in DDC-fed mice,30 indicating cholangiocyte involvement in biliary fibrosis. Cholangiocytes have enhanced transforming growth factor (TGF)-β, which induced transcription of FN-1, and blocking this transcriptional regulation in cholangiocytes blunted biliary fibrosis in Mdr2-/- mice.31,32 Similarly, blocking hepatic TGF-β2 decreased biliary fibrosis in Mdr2-/- mice.33 Other studies have found that TgfB2-directed antisense oligonucleotides reduced parenchymal liver fibrosis in CCl4-treated mice but had limited effect on biliary fibrosis in Mdr2-/- mice.34 Models of liver or biliary fibrosis activate different mesenchymal cells, and thus local cellular niches need to be accounted for when designing fibrosis experiments.

Lineage tracing studies have revealed that DR pattern/morphology varies on the basis of the cell compartment that is injured. Indeed, it has been hypothesized that DR could arise from the biliary system, but also from hepatocytes. One study analyzed R26tomHnf1b-CreER mice, which label bile ducts and periportal ductules (but not hepatocytes) with HNF1β, that were subjected to BDL, methylene dianiline (MDA), DDC, or choline-deficient, ethionine-supplemented (CDE) to evaluate DR in acute and chronic models. It was observed that DR arose from HNF1β+ biliary comportments in short-term (2–3 weeks) liver injury (BDL, MDA, and DDC); however, CDE feeding for 16 weeks had a small fraction of hepatocytes that co-expressed HNF1β.35 Another study found that DR invading the parenchyma expressed biliary markers and formed de novo junctions that maintained the canaliculi after CDE feeding. Contrarily, in BDL and DDC models, DR was confined to the portal mesenchyme and formed pseudo-ductular structures.36 Others found that thioacetamide (TAA) and DDC fed mice have a collapse of bile canaliculi that precedes DR; however, during recovery, reduced DR is paralleled with reconstruction of the bile canaliculi.37 Therefore, DR may have a beneficial role regarding regeneration.

In 2 models of HPC activation, portal HPC expansion was associated with myofibroblast activation and laminin deposition; however, the source of laminin was not evaluated.38 Others have found that PFs express laminin that supports HPC proliferation in mouse embryonic and neonatal liver.39 Interestingly, glioma-associated oncogene (Gli1)+ PMCs surrounded intrahepatic bile ducts in BDL and DDC fed mice, but the DR in these models was extended into the parenchyma and surrounded by HSCs. However, the PMCs did not express HSC markers, indicating that they may be PF derived.25 Interestingly, Gli1 facilitates cholangiocyte maturation and FN1 transcription, leading to ECM deposition in vitro.40 In the dominant-negative TGF-β receptor II (dnTGFβRII) model of PBC, cholangiocytes had reduced expression of mature cholangiocyte markers, including secretin and its receptor, and restoration of mature cholangiocyte markers via secretin administration reduced biliary fibrosis and HSC presence.41 In vitro, HPCs differentiated to “mature” biliary-like cells through taurocholate (TCA) stimulation secreted chemokines that drive HSC chemotaxis.42 The origin and location of the DR are important for demarcating fibrosis patterns. DR in the parenchyma may interact predominantly with HSCs, whereas portal DR may interact with PFs and HSCs.

Biliary Senescence and TGF-β1: Lord of the SASPs

One characteristic of PSC and PBC is the presence of senescent cholangiocytes.5 K19-Mdm2flox/floxtdTomLSL mice are a model of tamoxifen-inducible cholangiocyte senescence, and after tamoxifen injection, these mice have elevated collagen deposition surrounding senescent bile ducts. Furthermore, cholangiocytes in these mice release SASPs, specifically TGF-β1, that recruit myofibroblasts.43 Cholangiocytes from human PSC samples and cholestatic models exhibit telomerase damage, which was not seen in hepatocytes. The telomerase reverse transcriptase (TERT) was epigenetically repressed in senescent cholangiocytes, and overexpression of TERT decreased senescent and TGF-β1–mediated phenotypes of cholangiocytes in vitro. Therefore, selective genetic deletion of TERT from cholangiocytes may exacerbate biliary fibrosis.44 The secretin/secretin receptor axis is enhanced in cholangiocytes in BDL mice, and silencing the secretin/secretin receptor axis reduced biliary senescence, which in turn reduced TGF-β1 levels and biliary fibrosis.45 This phenotype was also observed in double knockout SR-/-Mdr2−/− mice.46 Finally, blocking cholangiocyte senescence by p16 Vivo-Morpholino or senolytic (Fisetin) treatment in Mdr2-/- mice reduced biliary fibrosis.47 Overall, these studies underline the relationship between biliary senescence, SASPs secretion (eg, TGF-β1), and biliary fibrosis during cholestasis.

Bile Acids: Chasing the Great White Whale

Bile stasis in PBC and PSC can lead to changes in BA composition and accumulation that perpetuate liver damage.48 Early studies found that all BAs at physiological concentrations induce HSC proliferation and activation in vitro,49 and taurodeoxycholate (TDCA) and TCA induce PF proliferation and activation in precision cut liver slices from normal rats.50 Hydrophobic BAs (eg, TCA, TDCA and lithocholate [LCA]) are considered damaging, whereas hydrophilic BAs (eg, UDCA) are considered protective.51 Supporting this, administration of hydrophilic BAs protects against immune cell infiltration and biliary fibrosis in Mdr2-/- mice.52,53 In addition, the hydrophobic BA chenodeoxycholate (CDCA) induces HSC activation and proliferation in vitro.54 These studies suggest that alterations in BA hydrophobicity modulate myofibroblast activation; however, studies have suggested that dampening the overall BA pool may be key.55 Inhibition of ileal apical sodium-dependent BA transporter (ASBT) reduced biliary BA output and increased fecal BA excretion, and this corresponded with reduced inflammation and biliary fibrosis in Mdr2-/- mice.56,57 Similarly, in dnTGFβRII mice secretin treatment enhances choleresis, thereby reducing hepatic BA levels, immune cell presence, and fibrosis.41 Reducing BA levels or hydrophobicity may be important for reversing biliary fibrosis.

Bile salt export pump (BSEP) is the main hepatic BA efflux transporter, but Bsep-/- mice show a hydrophilic BA composition, and Mdr2-/- mice crossed with Bsep-/- mice show reduced immune cell infiltration and biliary fibrosis.52,53 Na+-taurocholate co-transporting polypeptide (NTCP) is the major hepatic BA uptake transporter, and inhibition of NTCP reduces biliary damage, inflammation, and fibrosis in DDC fed and BDL mice; however, inhibition of NTCP aggravated inflammation and fibrosis in Mdr2-/- mice,58 indicating that targeting BA homeostasis differs between models. Introduction of human MDR3 (homolog to murine Mdr2), which regulates phospholipid secretion into bile, ameliorates inflammation and biliary fibrosis in Mdr2-/- mice by reducing toxic BA levels.59,60 Farnesoid X receptor (FXR) is a nuclear BA receptor that mediates BA synthesis and transporter expression, as well as inflammation. Tropifexor, a non-BA FXR agonist, reduces biliary fibrosis in piglets subjected to BDL for 14 days.61 A similar non-BA FXR agonist, EDP-305, decreased inflammation and fibrosis in Mdr2-/- mice.62 Rat HSCs do not express BSEP,63 primary human HSCs from fibrotic livers have increased NTCP expression,64 and the role of FXR on human and mouse HSC activation is controversial.65,66 Modulation of BA homeostasis may have direct effects on HSC and PF activation but also indirectly influences biliary fibrosis via modulation of inflammation.

BA-based drugs are currently used for PBC and PSC treatment. UDCA and obeticholic acid (OCA, ie, Ocaliva) are first- and second-line treatments, respectively, in PBC67; however, only OCA shows beneficial effects on liver enzymes in PSC.67,68 Cytochrome P450 family 2 subfamily c polypeptide 70 (Cyp2c70) knockout mice lack hydrophilic muricholic acids and display a more human-like BA pool. Cyp2c70-/- mice showed spontaneous cholangiopathic features at 8 months of age, and UDCA treatment reduced inflammation and biliary fibrosis in Cyp2c70-/- mice.69 24-norursodeoxycholic acid (norUDCA) is a side chain-shortened C23 UDCA homologue that reduced inflammation and biliary fibrosis in Mdr2-/- mice.70,71 Importantly, norUDCA has shown potential therapeutic benefit in PSC patients.72 BA-based therapies have important functions in reducing biliary fibrosis.

Endothelial Cells: The Angiogenesis Also Rises

Angiogenesis is the formation of new blood vessels and is noted in PBC and PSC samples.73,74 In BDL rats, neovascularization is associated with biliary fibrosis.75 Furthermore, hepatic vascular endothelial growth factor (VEGF) A (Vegfa/VEGFA) expression increases during fibrosis progression in CCl4 treated, BDL, and Mdr2-/- mice.74,76,77 However, neutralizing antibody against VEGFA impairs fibrosis resolution after CCl4 treatment through enhanced sinusoidal permeability.78 Importantly, biliary fibrosis in cholangiopathies is associated with portal angiogenesis that may involve vascular endothelial cells as opposed to sinusoidal endothelial cells.73,74 In line with this, DR in PSC is associated with enhanced fibrogenic gene expression and lies in close vicinity to angiogenic vessels.28 It was observed that liver blood vessels (portal and central vessels and liver sinusoid) act differently during liver fibrogenesis. Blocking portal angiogenesis alleviates fibrosis, but the inhibition of both central angiogenesis and sinusoidal capillarization promotes liver fibrosis in CCl4 treated mice.79 This underlines the strong correlation between portal angiogenesis and liver fibrosis.

HSCs release angiogenic factors, including angiopoietin-1 (Angpt-1) and platelet-derived growth factor (PDGF), that drive angiogenesis.75,80 Similarly, activated PFs secrete VEGFA, which promotes angiogenesis.81,82 In turn, VEGFA and Angpt-1 promote HSC activation,83 indicating a feedback loop between angiogenic and fibrogenic cells. Placental growth factor (PlGF) promotes angiogenesis in CCl4 mice, and HSCs treated with PlGF in vitro have enhanced ECM synthesis.84 Also, angiotensin enzyme 2 therapy directly blocks HSC activation and biliary fibrosis in Mdr2-/- mice.85 Activated PFs display both fibrogenic and angiogenic gene signatures, specifically split guidance ligand 2 (Slit2),24 and Slit2 drives angiogenesis in DDC fed mice.86 Portal myofibroblasts also drive angiogenesis through the release of VEGFA-enriched microparticles that signal to nearby endothelial cells.82 HSCs and PFs interact with endothelial cells to enhance angiogenesis, which, in turn, drives scar formation.

Aside from direct communication between endothelial and fibrogenic cells, angiocrine factors can regulate HSCs and PFs. Endothelin (ET) regulates the synthesis and secretion of TGF-β1 and VEGFA, and cholangiocytes with enhanced ET-A signaling initiate angiogenesis and fibrosis in Mdr2-/- mice.74 Vasohibin-1 (VASH-1) regulates VEGF through a negative feedback loop, and overexpression of VASH-1 reduced VEGF levels, pathologic angiogenesis, and biliary fibrosis in BDL mice.87 These findings are significant because VEGF promotes pathologic, but not physiological, angiogenesis that is associated with fibrosis.88 PSC is a risk factor for CCA,1 and fibroblast-derived VEGFA/C promotes intravasation and early metastasis in CCA89; therefore, VEGFA/C may be an important link between PSC and CCA development. It is apparent that angiogenic signaling can influence biliary fibrosis.

Immune Cells: For Whom the Fibrosis Tolls

Innate immunity is one of the first lines of defense against infection and broadly involves 3 cell types: granulocytes (ie, neutrophils, eosinophils, basophils, and mast cells [MCs]), monocytes/macrophages (eg, Kupffer cells [KCs, liver-resident macrophages]), and dendritic cells. If the innate immune response is unable to combat infection, the adaptive immune system is initiated. On first infection, the adaptive immune cells, T and B lymphocytes, perform cell- and antibody-mediated responses, and these signatures are memorized to allow for quicker responses if reinfection occurs.90 In chronic liver diseases, both innate and adaptive immunity have been identified as drivers of fibrosis.91 Indeed, initial biliary fibrosis is characterized by portal inflammation (stage 1) but can expand into the periportal area along with inflammatory interface (stage 2). Next, portal-portal fibrosis bridges form with moderate/marked portal inflammation (stage 3), with eventual cirrhosis formation occurring (stage 4).92,93 Here, we will describe how innate and adaptive immune cells can contribute to biliary fibrosis.

Monocytes and macrophages (MΦ) are phagocytes that maintain homeostasis through the clearance of cell debris and microbes. Hepatic MΦ, consisting of KCs and monocyte-derived macrophages (MoMΦ), exhibit heterogeneity and can contribute to fibrosis progression or resolution.94 MoMΦ accumulate in PBC and PSC and are associated with DR, and MoMΦ infiltration adjacent to DR predicts advanced cirrhosis in PSC.95,96 MΦ isolated from human liver fibrosis samples are pro-fibrotic and have overlapping pathways with HSCs, alluding to MΦ induction of HSC activation.97 In BDL and DDC models, MoMΦ and KCs were pro-inflammatory, but elimination of KCs had no impact on biliary fibrosis.98 Other research found KCs indispensable in BDL-induced biliary fibrosis.99 MoMΦ are recruited by cholangiocyte released monocyte chemoattractant protein-1 (MCP-1), and blocking monocyte recruitment attenuated fibrosis in Mdr2-/- mice.100 MCP-1 further promotes pro-inflammatory phenotypes in KCs and recruits MoMΦ in BDL and Mdr2-/- mice.101 HSCs, in turn, imprint pro-inflammatory signatures on MoMΦ.102 These studies suggest differential roles of infiltrating and resident MΦ on biliary fibrosis.

MCs release histamine and other mediators after their activation. In PSC livers, MCs reside in fibrotic areas and correlate with portal fibrosis.103,104 Interestingly, HSCs produce stem cell factor, a MC chemoattractant, that induces MC homing in vitro.105 One study found that in vitro co-culture of MCs with HSCs reduced HSC fibrogenesis106; however, the HSC:MC ratio was 1:2.25, which is not appropriate for physiological standards. Selectively inhibiting MC-dependent H2 histamine receptor (HR) signaling reduces biliary fibrosis induced by MC injection,107 and liver-specific down-regulation of H2HR reduces hepatic MC infiltration and biliary fibrosis in Mdr2-/- mice.108 Similarly, H1HR and H2HR antagonists diminish portal MC presence, as well as biliary fibrosis in Mdr2-/- mice.109 Last, when MC recruitment or activation is blocked, HSC number and biliary fibrosis are repressed in Mdr2-/- mice and BDL models.110, 111, 112 Targeting MCs or MC-derived mediators, such as histamine, may be useful for regression of biliary fibrosis.

Neutrophils infiltrate the liver in acute infection or injury and act in a protective manner, but chronic liver disease leads to aberrant neutrophil signaling that may perturb damage.113 Human PSC samples have increased neutrophils in the portal region,114 and neutrophils are recruited early (3 hours after BDL).115 In DDC fed and CCl4 treated mice, DR-associated neutrophils increase, and loss of neutrophils mitigates liver fibrosis.116 Early in vitro studies found that neutrophil production of reactive oxygen species promoted collagen synthesis in HSCs.117 In addition, neutrophils are a main source of interleukin 17 (IL-17) and IL-22, which show pro-fibrogenic effects on HSCs.118,119 However, depletion of neutrophils after CCl4 administration impairs fibrosis regression.120 Also, neutrophils promote fibrosis resolution through matrix metalloproteinase secretion in BDL rats.121 Others have found that neutrophil attenuation had no effect on biliary fibrosis in BDL rats.122 Neutrophil involvement in biliary fibrosis is nuanced and may rely on the timing of infiltration and exposure to acute versus chronic damage.

In the spleen or lymph node, B cells, while in contact with cluster differentiation (CD)4 T helper (Th) cells, recognize antigen that causes them to differentiate to germinal center B cells or plasmablasts that can give rise to memory B cells or plasma cells, respectively. Plasma cells secrete antibodies in response to antigen, and B cells can produce cytokines through interaction with other immune cells.123 PBC and PSC patients have increased numbers of B cells and plasma cells in the portal region,124, 125, 126, 127 and memory B cells are found in the fibrotic septa in PBC and PSC.128 Mdr2-/- mice have enhanced portal infiltration of pro-inflammatory B cells, which is attributed to increased expression of myeloid cell leukemia 1 (MCL1) in cholangiocytes. When Mdr2-/- mice were treated with S63845, an MCL1 inhibitor, hepatic B cell number decreased along with biliary fibrosis.129 Pro-inflammatory B cells increase in CCl4 treated mice, and activated HSCs augmented B cell activity. Conversely, anti-B cell activating factor (BAFF) treatment had no effect on fibrosis in Mdr2-/- mice; however, total depletion of B cells using anti-CD20 antibody treatment reduced hepatic fibrosis in Mdr2-/- mice.130 Genetic loss of B cells in dnTGFβRII mice led to enhanced pro-inflammatory markers, including IL-6 and tumor necrosis factor (TNF)-α,131 that are known to activate HSCs.132 Similarly, anti-CD20 antibody depletion of B cells in dnTGFβRII mice increased inflammation and IL-1α levels,133 which are known to promote HSC proliferation.132 Others found that anti-CD20 antibody reduced hepatic inflammation in dnTGFβRII mice.134,135 B cell modulation of biliary fibrosis is controversial and requires further investigation.

Naive T cells survey lymphoid organs and, after contact with an antigen presenting cell (APC), travel to the lymph nodes to (1) undergo clonal expansion, (2) become effector T cells (ie, Th cells, cytotoxic T lymphocytes [CTLs], regulatory T cells [Tregs]), and (3) traffic to the infected organ. Infection can produce memory T cells that recognize antigen on reinfection to become effector T cells.136 T cells can promote HSC activation,137 and HSCs cause apoptosis of T cells in fibrosis.138 Interestingly, HSCs counteract APC initiated naive T-cell activation.139 PBC patients have an increased number of Th cells and tissue-resident memory T cells (TRM), whereas Tregs are decreased.126,140,141 Additionally, PSC patients have increased Th cells and CTLs near the bile ducts,142 and enhanced interferon γ signaling in PSC and Mdr2-/- mice enhances CTL cytotoxicity and biliary fibrosis.143 Th cell-derived IL-21, but not IL-17, enhanced fibrosis and CTL activity in dnTGFβRII mice.144 In Mdr2-/- and DDC fed mice, intrahepatic TRM cells expanded, and inhibiting TRM cell activity reduced biliary fibrosis.145 NorUDCA attenuates CTL activity and biliary fibrosis in Mdr2-/- mice.70 Interestingly, loss of γδ T cells, a primitive T-cell subset, blocks liver fibrosis by inducing apoptosis in HSCs and inhibiting Th cell activity.146 T cell heterogeneity is complex, and understanding how different T cells interact with PFs and HSCs will be essential for defining immunomodulation of biliary fibrosis.

Conclusion

In relation to biliary fibrosis, HSCs have historically been studied; however, research has identified PFs as an important contributor. Numerous studies have identified that PFs are the first fibrogenic cells to activate in cholestatic damage, thereby initiating biliary fibrosis, with recruitment of HSCs coming later. Indeed, the recruitment of HSCs later may be important in initiating bridging fibrosis. It is also key to understand that variations in damage initiation between BDL, DDC, Mdr2-/-, and other models differentially impact PFs and HSCs. Discernable differences in fibrosis between models should be taken into consideration when designing experiments and analyzing samples. Additionally, variations in DR and its localization (eg, portal or parenchymal) can variably impact PFs and HSCs. This area of study can be further complicated when considering the different cellular sources that can give rise to DR and the role of senescent cholangiocytes and secreted SASPs on the activation/modulation of PFs and HSCs.

The buildup of BAs initiates both biliary and hepatocellular damage, which may variably induce fibrosis. The role of BA accumulation has been well-studied in HSCs; however, more work is needed in PFs. Unfortunately, BA receptor and transporter expression are controversial or unknown in HSCs and PFs. Portal angiogenesis is a pathologic feature known to promote fibrosis, and better demarcation of angiogenesis and how it interacts with HSCs and PFs is needed. Last, the portal niche can be inundated with infiltrating immune cells that show diversity. This complex immune response shapes cellular interactions, but the plethora of immune subsets makes teasing out mechanisms difficult.

Unsurprisingly, complex cellular interactions and diverse cell populations in cholangiopathies have varying impacts on biliary fibrosis (Figure 1). Studies thus far have identified potential therapeutics for PBC and PSC (Table 2); however, additional research will uncover novel therapeutic avenues. Although biliary fibrosis is multifarious, appreciating the nuance will be beneficial for furthering this area of study.

Figure 1.

Figure 1

Graphical representation of cellular crosstalk supporting biliary fibrosis. DR can occur at the portal and parenchymal area, differentially facilitating biliary and bridging fibrosis. Different factors secreted from DR cells can contribute to fibrosis progression. Cholestasis leads to hepatic BA buildup, and various changes in BA excretion/secretion, transport, homeostasis, and interactions with PFs and HSCs can lead to biliary fibrosis. Senescent cholangiocytes have increased p16 expression, temolerase damage, and SASPs secretion, including TGF-β1, which stimulate activation of both PFs and HSCs during cholestasis. Angiogenesis is closely associated with biliary fibrosis, and different angiocrine signals between PFs, HSCs, and endothelial cells/LSECs can give rise to these pathologies. Last, immune cells infiltrate the liver during cholestasis and cholangiopathies, and the different immune subtypes have varying roles on PFs, HSCs, and biliary fibrosis. BA, bile acid; DR, ductular reaction; HSC, hepatic stellate cell; LSEC, lymphatic sinusoidal endothelial cell; PF, portal fibroblast; SASP, senescence-associated secretory phenotype; TGF-β, transforming growth factor β. Image made with BioRender.com.

Table 2.

Potential Therapeutic Agents for PBC and PSC

Drug class Drug name/agent Target/mode of action Experimental model Reference(s)
Anti-neoplastic Bortezomib Proteosome inhibition Mdr2-/- mice 32
Senolytic Sec 5-27 SR antagonist Mdr2-/- mice 45
Fisetin Anti-oxidation Mdr2-/- mice 47
Antifibrotic FAP-specific inhibitor (FAPi) (S)-N-(2-cyano-4,4-difluoropyrrolidin-1-yl)-2-oxoethyl)-quinoline-4-carboxamide Fibroblast activation protein (FAP) CCl4 and Mdr2-/- mice 34
Bile acid–based Tetrahydroxylated bile acids (THBAs) Less toxic bile acid pool Mdr2-/- mice 52,53
SC-435 Ileal ASBT inhibition Cyp2c70-/- and Mdr2-/- mice 55,57
A4250 Ileal ASBT inhibition Mdr2-/- mice 56
Myrcludex B NTCP inhibition DDC, BDL, and Mdr2-/- mice 58
Synthetic human ABCB4 gene therapy Restore ABCB4/MDR3 Mdr2-/- mice 59,60
Tropifexor FXR agonist BDL piglets 61
EDP-305 FXR agonist Mdr2-/- mice 62
Nor-UDCA Anti-cholestatic and anti-inflammatory Mdr2-/- mice 70,71
Anti-angiogenic Ambrisentan ET-A antagonist Mdr2-/- mice 74
Sorafenib Kinase inhibitor BDL rats 75
MCR84 VEGF-neutralizing antibody BDL mice 78
αPlGF PlGF-neutralizing antibody CCl4 mice 84
AdVASH1 (synthetic human VASH1 gene therapy) Restore vasohibin-1 BDL rats 87
Immunomodulatory Cenicriviroc CCR2/5 antagonist BV6 injected mice 100
Cromolyn sodium Mast cell stabilizer Mdr2-/- mice and BDL rats 110,112
Mepyramine and ranitidine H1 and H2 histamine receptor antagonists Mdr2-/- mice 109
SCH-527123 CXCR1/2 inhibitor DDC mice 116
S63845 MCL1 inhibitor Mdr2-/- mice 129
Anti-CD20 antibody B cell depletion dnTGFβRII mice 134,135
Anti-IFNγ antibody IFNγ neutralization Mdr2-/- mice 143
4-OI Itaconate derivative DDC and Mdr2-/- mice 145

CD20, cluster of differentiation 20; CXCR1/2, C-X-C chemokine receptor type 1/2; Cyp2c70, cytochrome P450 family 2 subfamily c polypeptide 70; FAP, fibroblast activation protein; IFNγ, interferon γ; Mdr2/MDR3, multidrug resistance 2/3; SR, secretin receptor; THBA, tetrahydroxylated bile acid; VASH1, vasohibin 1.

Acknowledgments

CRediT Authorship Contributions

Ludovica Ceci (Writing – original draft, equal; Visualization, equal)

Eugenio Gaudio (Writing – review and editing, equal; Resources, equal; Funding acquisition, equal)

Lindsey Kennedy (Writing – original draft, lead; Visualization, lead; Resources, lead; Conceptualization, lead)

Footnotes

Conflicts of interest The authors disclose no conflicts.

Funding Supported by a Career Development Award-2 to LK (1IK2BX005306) from the United States Department of Veteran’s Affairs, Biomedical Laboratory Research and Development Service, and research project grants to EG from Sapienza, University of Rome. Portions of these studies were supported by resources at the Richard L. Roudebush VA Medical Center, Indianapolis, IN. The views expressed in this article are those of the authors and do not necessarily represent the views of the Department of Veterans Affairs.

References

  • 1.Lazaridis K.N., LaRusso N.F. The cholangiopathies. Mayo Clin Proc. 2015;90:791–800. doi: 10.1016/j.mayocp.2015.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Toy E., Balasubramanian S., Selmi C., et al. The prevalence, incidence and natural history of primary sclerosing cholangitis in an ethnically diverse population. BMC Gastroenterol. 2011;11:83. doi: 10.1186/1471-230X-11-83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Boberg K.M. In: Biliary disease: from science to clinic. Hirschfield G., Adams D., Liaskou E., editors. Springer International Publishing; New York: 2017. The clinical burden of biliary disease: a global perspective; pp. 1–15. [Google Scholar]
  • 4.Roskams T.A., Theise N.D., Balabaud C., et al. Nomenclature of the finer branches of the biliary tree: canals, ductules, and ductular reactions in human livers. Hepatology. 2004;39:1739–1745. doi: 10.1002/hep.20130. [DOI] [PubMed] [Google Scholar]
  • 5.Guicciardi M.E., Trussoni C.E., LaRusso N.F., et al. The spectrum of reactive cholangiocytes in primary sclerosing cholangitis. Hepatology. 2020;71:741–748. doi: 10.1002/hep.31067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Pinzani M., Luong T.V. Pathogenesis of biliary fibrosis. Biochim Biophys Acta Mol Basis Dis. 2018;1864(Pt B):1279–1283. doi: 10.1016/j.bbadis.2017.07.026. [DOI] [PubMed] [Google Scholar]
  • 7.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:E3297–E3305. doi: 10.1073/pnas.1400062111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Kinnman N., Housset C. Peribiliary myofibroblasts in biliary type liver fibrosis. Front Biosci. 2002;7:d496–d503. doi: 10.2741/A790. [DOI] [PubMed] [Google Scholar]
  • 9.Murillo Perez C.F., Hirschfield G.M., Corpechot C., et al. Fibrosis stage is an independent predictor of outcome in primary biliary cholangitis despite biochemical treatment response. Aliment Pharmacol Ther. 2019;50:1127–1136. doi: 10.1111/apt.15533. [DOI] [PubMed] [Google Scholar]
  • 10.Oyama A., Takaki A., Adachi T., et al. Oxidative stress-related markers as prognostic factors for patients with primary sclerosing cholangitis in Japan. Hepatol Int. 2023;17:1215–1224. doi: 10.1007/s12072-023-10557-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Fabris L., Spirli C., Cadamuro M., et al. Emerging concepts in biliary repair and fibrosis. Am J Physiol Gastrointest Liver Physiol. 2017;313:G102–G116. doi: 10.1152/ajpgi.00452.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kisseleva T., Uchinami H., Feirt N., et al. Bone marrow-derived fibrocytes participate in pathogenesis of liver fibrosis. J Hepatol. 2006;45:429–438. doi: 10.1016/j.jhep.2006.04.014. [DOI] [PubMed] [Google Scholar]
  • 13.Scholten D., Osterreicher C.H., Scholten A., et al. Genetic labeling does not detect epithelial-to-mesenchymal transition of cholangiocytes in liver fibrosis in mice. Gastroenterology. 2010;139:987–998. doi: 10.1053/j.gastro.2010.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Taura K., Miura K., Iwaisako K., et al. Hepatocytes do not undergo epithelial-mesenchymal transition in liver fibrosis in mice. Hepatology. 2010;51:1027–1036. doi: 10.1002/hep.23368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Geerts A. History, heterogeneity, developmental biology, and functions of quiescent hepatic stellate cells. Semin Liver Dis. 2001;21:311–335. doi: 10.1055/s-2001-17550. [DOI] [PubMed] [Google Scholar]
  • 16.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;109:9448–9453. doi: 10.1073/pnas.1201840109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Karin D., Koyama Y., Brenner D., et al. The characteristics of activated portal fibroblasts/myofibroblasts in liver fibrosis. Differentiation. 2016;92:84–92. doi: 10.1016/j.diff.2016.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lepreux S., Desmouliere A. Human liver myofibroblasts during development and diseases with a focus on portal (myo)fibroblasts. Front Physiol. 2015;6:173. doi: 10.3389/fphys.2015.00173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Seifert W.F., Roholl P.J., Blauw B., et al. Fat-storing cells and myofibroblasts are involved in the initial phase of carbon tetrachloride-induced hepatic fibrosis in BN/BiRij rats. Int J Exp Pathol. 1994;75:131–146. [PMC free article] [PubMed] [Google Scholar]
  • 20.Lorena D., Darby I.A., Reinhardt D.P., et al. 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;84:203–212. doi: 10.1038/labinvest.3700023. [DOI] [PubMed] [Google Scholar]
  • 21.Desmouliere A., Darby I., Costa A.M., et al. Extracellular matrix deposition, lysyl oxidase expression, and myofibroblastic differentiation during the initial stages of cholestatic fibrosis in the rat. Lab Invest. 1997;76:765–778. [PubMed] [Google Scholar]
  • 22.Tuchweber B., Desmouliere A., Bochaton-Piallat M.L., et al. Proliferation and phenotypic modulation of portal fibroblasts in the early stages of cholestatic fibrosis in the rat. Lab Invest. 1996;74:265–278. [PubMed] [Google Scholar]
  • 23.Yang W., He H., Wang T., et al. Single-cell transcriptomic analysis reveals a hepatic stellate cell-activation roadmap and myofibroblast origin during liver fibrosis in mice. Hepatology. 2021;74:2774–2790. doi: 10.1002/hep.31987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Lei L., Bruneau A., El Mourabit H., et al. Portal fibroblasts with mesenchymal stem cell features form a reservoir of proliferative myofibroblasts in liver fibrosis. Hepatology. 2022;76:1360–1375. doi: 10.1002/hep.32456. [DOI] [PubMed] [Google Scholar]
  • 25.Gupta V., Gupta I., Park J., et al. Hedgehog signaling demarcates a niche of fibrogenic peribiliary mesenchymal cells. Gastroenterology. 2020;159:624–638 e9. doi: 10.1053/j.gastro.2020.03.075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Nishio T., Hu R., Koyama Y., et al. Activated hepatic stellate cells and portal fibroblasts contribute to cholestatic liver fibrosis in MDR2 knockout mice. J Hepatol. 2019;71:573–585. doi: 10.1016/j.jhep.2019.04.012. [DOI] [PubMed] [Google Scholar]
  • 27.Overi D., Carpino G., Cristoferi L., et al. Role of ductular reaction and ductular-canalicular junctions in identifying severe primary biliary cholangitis. JHEP Rep. 2022;4 doi: 10.1016/j.jhepr.2022.100556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Govaere O., Cockell S., Van Haele M., et al. High-throughput sequencing identifies aetiology-dependent differences in ductular reaction in human chronic liver disease. J Pathol. 2019;248:66–76. doi: 10.1002/path.5228. [DOI] [PubMed] [Google Scholar]
  • 29.Zhang J., Lyu Z., Li B., et al. P4HA2 induces hepatic ductular reaction and biliary fibrosis in chronic cholestatic liver diseases. Hepatology. 2023;78:10–25. doi: 10.1097/HEP.0000000000000317. [DOI] [PubMed] [Google Scholar]
  • 30.Chen Y., Guldiken N., Spurny M., et al. Loss of keratin 19 favours the development of cholestatic liver disease through decreased ductular reaction. J Pathol. 2015;237:343–354. doi: 10.1002/path.4580. [DOI] [PubMed] [Google Scholar]
  • 31.Aseem S.O., Jalan-Sakrikar N., Chi C., et al. Epigenomic evaluation of cholangiocyte transforming growth factor-beta signaling identifies a selective role for histone 3 lysine 9 acetylation in biliary fibrosis. Gastroenterology. 2021;160:889–905 e10. doi: 10.1053/j.gastro.2020.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Jalan-Sakrikar N., De Assuncao T.M., Shi G., et al. Proteasomal degradation of enhancer of zeste homologue 2 in cholangiocytes promotes biliary fibrosis. Hepatology. 2019;70:1674–1689. doi: 10.1002/hep.30706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Dropmann A., Dooley S., Dewidar B., et al. TGF-beta2 silencing to target biliary-derived liver diseases. Gut. 2020;69:1677–1690. doi: 10.1136/gutjnl-2019-319091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Yang A.T., Kim Y.O., Yan X.Z., et al. Fibroblast activation protein activates macrophages and promotes parenchymal liver inflammation and fibrosis. Cell Mol Gastroenterol Hepatol. 2023;15:841–867. doi: 10.1016/j.jcmgh.2022.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Jors S., Jeliazkova P., Ringelhan M., et al. Lineage fate of ductular reactions in liver injury and carcinogenesis. J Clin Invest. 2015;125:2445–2457. doi: 10.1172/JCI78585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Clerbaux L.A., Manco R., Van Hul N., et al. Invasive ductular reaction operates hepatobiliary junctions upon hepatocellular injury in rodents and humans. Am J Pathol. 2019;189:1569–1581. doi: 10.1016/j.ajpath.2019.04.011. [DOI] [PubMed] [Google Scholar]
  • 37.Kamimoto K., Nakano Y., Kaneko K., et al. Multidimensional imaging of liver injury repair in mice reveals fundamental role of the ductular reaction. Commun Biol. 2020;3:289. doi: 10.1038/s42003-020-1006-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Lorenzini S., Bird T.G., Boulter L., et al. Characterisation of a stereotypical cellular and extracellular adult liver progenitor cell niche in rodents and diseased human liver. Gut. 2010;59:645–654. doi: 10.1136/gut.2009.182345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Tanimizu N., Kikkawa Y., Mitaka T., et al. alpha1- and alpha5-containing laminins regulate the development of bile ducts via beta1 integrin signals. J Biol Chem. 2012;287:28586–28597. doi: 10.1074/jbc.M112.350488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Jalan-Sakrikar N., De Assuncao T.M., Lu J., et al. Hedgehog signaling overcomes an EZH2-dependent epigenetic barrier to promote cholangiocyte expansion. PLoS One. 2016;11 doi: 10.1371/journal.pone.0168266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Kennedy L., Carpino G., Owen T., et al. Secretin alleviates biliary and liver injury during late-stage primary biliary cholangitis via restoration of secretory processes. J Hepatol. 2023;78:99–113. doi: 10.1016/j.jhep.2022.07.034. [DOI] [PubMed] [Google Scholar]
  • 42.Pozniak K.N., Pearen M.A., Pereira T.N., et al. Taurocholate induces biliary differentiation of liver progenitor cells causing hepatic stellate cell chemotaxis in the ductular reaction: role in pediatric cystic fibrosis liver disease. Am J Pathol. 2017;187:2744–2757. doi: 10.1016/j.ajpath.2017.08.024. [DOI] [PubMed] [Google Scholar]
  • 43.Ferreira-Gonzalez S., Lu W.Y., Raven A., et al. Paracrine cellular senescence exacerbates biliary injury and impairs regeneration. Nat Commun. 2018;9:1020. doi: 10.1038/s41467-018-03299-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Jalan-Sakrikar N., Anwar A., Yaqoob U., et al. Telomere dysfunction promotes cholangiocyte senescence and biliary fibrosis in primary sclerosing cholangitis. JCI Insight. 2023;8 doi: 10.1172/jci.insight.170320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Wu N., Meng F., Zhou T., et al. The secretin/secretin receptor axis modulates ductular reaction and liver fibrosis through changes in transforming growth factor-beta1-mediated biliary senescence. Am J Pathol. 2018;188:2264–2280. doi: 10.1016/j.ajpath.2018.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Zhou T., Wu N., Meng F., et al. Knockout of secretin receptor reduces biliary damage and liver fibrosis in Mdr2(-/-) mice by diminishing senescence of cholangiocytes. Lab Invest. 2018;98:1449–1464. doi: 10.1038/s41374-018-0093-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Alsuraih M., O’Hara S.P., Woodrum J.E., et al. Genetic or pharmacological reduction of cholangiocyte senescence improves inflammation and fibrosis in the Mdr2 (-/-) mouse. JHEP Rep. 2021;3 doi: 10.1016/j.jhepr.2021.100250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Zeng J., Fan J., Zhou H. Bile acid-mediated signaling in cholestatic liver diseases. Cell Biosci. 2023;13:77. doi: 10.1186/s13578-023-01035-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Svegliati-Baroni G., Ridolfi F., Hannivoort R., et al. Bile acids induce hepatic stellate cell proliferation via activation of the epidermal growth factor receptor. Gastroenterology. 2005;128:1042–1055. doi: 10.1053/j.gastro.2005.01.007. [DOI] [PubMed] [Google Scholar]
  • 50.Wood M.J., Sjoblom P., Lindenberg S., et al. Effect of slow and ultra-rapid freezing on cell surface antigens of 8-cell mouse embryos. J Exp Zool. 1992;262:330–339. doi: 10.1002/jez.1402620313. [DOI] [PubMed] [Google Scholar]
  • 51.Fickert P., Fuchsbichler A., Marschall H.U., et al. Lithocholic acid feeding induces segmental bile duct obstruction and destructive cholangitis in mice. Am J Pathol. 2006;168:410–422. doi: 10.2353/ajpath.2006.050404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Wang R., Sheps J.A., Liu L., et al. Hydrophilic bile acids prevent liver damage caused by lack of biliary phospholipid in Mdr2(-/-) mice. J Lipid Res. 2019;60:85–97. doi: 10.1194/jlr.M088070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Fuchs C.D., Dixon E.D., Hendrikx T., et al. Tetrahydroxylated bile acids improve cholestatic liver and bile duct injury in the Mdr2(-/-) mouse model of sclerosing cholangitis via immunomodulatory effects. Hepatol Commun. 2022;6:2368–2378. doi: 10.1002/hep4.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Zimny S., Koob D., Li J., et al. Hydrophobic bile salts induce pro-fibrogenic proliferation of hepatic stellate cells through PI3K p110 alpha signaling. Cells. 2022:11. doi: 10.3390/cells11152344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Truong J.K., Bennett A.L., Klindt C., et al. Ileal bile acid transporter inhibition in Cyp2c70 KO mice ameliorates cholestatic liver injury. J Lipid Res. 2022;63 doi: 10.1016/j.jlr.2022.100261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Baghdasaryan A., Fuchs C.D., Osterreicher C.H., 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;64:674–681. doi: 10.1016/j.jhep.2015.10.024. [DOI] [PubMed] [Google Scholar]
  • 57.Miethke A.G., 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;63:512–523. doi: 10.1002/hep.27973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Slijepcevic D., Roscam Abbing R.L.P., Fuchs C.D., et al. Na(+) -taurocholate cotransporting polypeptide inhibition has hepatoprotective effects in cholestasis in mice. Hepatology. 2018;68:1057–1069. doi: 10.1002/hep.29888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Aronson S.J., Bakker R.S., Shi X., et al. Liver-directed gene therapy results in long-term correction of progressive familial intrahepatic cholestasis type 3 in mice. J Hepatol. 2019;71:153–162. doi: 10.1016/j.jhep.2019.03.021. [DOI] [PubMed] [Google Scholar]
  • 60.Wei G., Cao J., Huang P., et al. Synthetic human ABCB4 mRNA therapy rescues severe liver disease phenotype in a BALB/c.Abcb4(-/-) mouse model of PFIC3. J Hepatol. 2021;74:1416–1428. doi: 10.1016/j.jhep.2020.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Xiao Y., Wang Y., Liu Y., et al. A nonbile acid farnesoid X receptor agonist tropifexor potently inhibits cholestatic liver injury and fibrosis by modulating the gut-liver axis. Liver Int. 2021;41:2117–2131. doi: 10.1111/liv.14906. [DOI] [PubMed] [Google Scholar]
  • 62.An P., Wei G., Huang P., Li W., et al. A novel non-bile acid FXR agonist EDP-305 potently suppresses liver injury and fibrosis without worsening of ductular reaction. Liver Int. 2020;40:1655–1669. doi: 10.1111/liv.14490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Hannivoort R.A., Dunning S., Vander Borght S., et al. Multidrug resistance-associated proteins are crucial for the viability of activated rat hepatic stellate cells. Hepatology. 2008;48:624–634. doi: 10.1002/hep.22346. [DOI] [PubMed] [Google Scholar]
  • 64.Salhab A., Amer J., Lu Y., et al. Sodium(+)/taurocholate cotransporting polypeptide as target therapy for liver fibrosis. Gut. 2022;71:1373–1385. doi: 10.1136/gutjnl-2020-323345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Fickert P., Fuchsbichler A., Moustafa T., et al. Farnesoid X receptor critically determines the fibrotic response in mice but is expressed to a low extent in human hepatic stellate cells and periductal myofibroblasts. Am J Pathol. 2009;175:2392–2405. doi: 10.2353/ajpath.2009.090114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Fiorucci S., Antonelli E., Rizzo G., et al. The nuclear receptor SHP mediates inhibition of hepatic stellate cells by FXR and protects against liver fibrosis. Gastroenterology. 2004;127:1497–1512. doi: 10.1053/j.gastro.2004.08.001. [DOI] [PubMed] [Google Scholar]
  • 67.Carbone M., Mells G.F., Pells G., et al. Sex and age are determinants of the clinical phenotype of primary biliary cirrhosis and response to ursodeoxycholic acid. Gastroenterology. 2013;144:560–5609 e7. doi: 10.1053/j.gastro.2012.12.005. quiz e13–e14. [DOI] [PubMed] [Google Scholar]
  • 68.Lindor K.D., Kowdley K.V., Luketic V.A., et al. High-dose ursodeoxycholic acid for the treatment of primary sclerosing cholangitis. Hepatology. 2009;50:808–814. doi: 10.1002/hep.23082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.de Boer J.F., de Vries H.D., Palmiotti A., et al. Cholangiopathy and biliary fibrosis in Cyp2c70-deficient mice are fully reversed by ursodeoxycholic acid. Cell Mol Gastroenterol Hepatol. 2021;11:1045–1069. doi: 10.1016/j.jcmgh.2020.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Zhu C., Boucheron N., Muller A.C., et al. 24-Norursodeoxycholic acid reshapes immunometabolism in CD8(+) T cells and alleviates hepatic inflammation. J Hepatol. 2021;75:1164–1176. doi: 10.1016/j.jhep.2021.06.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Halilbasic E., Fiorotto R., Fickert P., et al. Side chain structure determines unique physiologic and therapeutic properties of norursodeoxycholic acid in Mdr2-/- mice. Hepatology. 2009;49:1972–1981. doi: 10.1002/hep.22891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Fickert P., Hirschfield G.M., Denk G., et al. Norursodeoxycholic acid improves cholestasis in primary sclerosing cholangitis. J Hepatol. 2017;67:549–558. doi: 10.1016/j.jhep.2017.05.009. [DOI] [PubMed] [Google Scholar]
  • 73.Medina J., Sanz-Cameno P., Garcia-Buey L., et al. Evidence of angiogenesis in primary biliary cirrhosis: an immunohistochemical descriptive study. J Hepatol. 2005;42:124–131. doi: 10.1016/j.jhep.2004.09.024. [DOI] [PubMed] [Google Scholar]
  • 74.Owen T., Carpino G., Chen L., et al. Endothelin receptor-A inhibition decreases ductular reaction, liver fibrosis, and angiogenesis in a model of cholangitis. Cell Mol Gastroenterol Hepatol. 2023;16:513–540. doi: 10.1016/j.jcmgh.2023.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Thabut D., Routray C., Lomberk G., et al. Complementary vascular and matrix regulatory pathways underlie the beneficial mechanism of action of sorafenib in liver fibrosis. Hepatology. 2011;54:573–585. doi: 10.1002/hep.24427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Yoshiji H., Kuriyama S., Yoshii J., et al. Vascular endothelial growth factor and receptor interaction is a prerequisite for murine hepatic fibrogenesis. Gut. 2003;52:1347–1354. doi: 10.1136/gut.52.9.1347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Tanaka A., Tsuneyama K., Mikami M., et al. Gene expression profiling in whole liver of bile duct ligated rats: VEGF-A expression is up-regulated in hepatocytes adjacent to the portal tracts. J Gastroenterol Hepatol. 2007;22:1993–2000. doi: 10.1111/j.1440-1746.2006.04629.x. [DOI] [PubMed] [Google Scholar]
  • 78.Yang L., Kwon J., Popov Y., et al. Vascular endothelial growth factor promotes fibrosis resolution and repair in mice. Gastroenterology. 2014;146:1339–1350 e1. doi: 10.1053/j.gastro.2014.01.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Lin Y., Dong M.Q., Liu Z.M., et al. A strategy of vascular-targeted therapy for liver fibrosis. Hepatology. 2022;76:660–675. doi: 10.1002/hep.32299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Semela D., Das A., Langer D., et al. Platelet-derived growth factor signaling through ephrin-b2 regulates hepatic vascular structure and function. Gastroenterology. 2008;135:671–679. doi: 10.1053/j.gastro.2008.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Loeuillard E., El Mourabit H., Lei L., et al. Endoplasmic reticulum stress induces inverse regulations of major functions in portal myofibroblasts during liver fibrosis progression. Biochim Biophys Acta Mol Basis Dis. 2018;1864:3688–3696. doi: 10.1016/j.bbadis.2018.10.008. [DOI] [PubMed] [Google Scholar]
  • 82.Lemoinne S., Cadoret A., Rautou P.E., et al. Portal myofibroblasts promote vascular remodeling underlying cirrhosis formation through the release of microparticles. Hepatology. 2015;61:1041–1055. doi: 10.1002/hep.27318. [DOI] [PubMed] [Google Scholar]
  • 83.Novo E., Cannito S., Zamara E., et al. Proangiogenic cytokines as hypoxia-dependent factors stimulating migration of human hepatic stellate cells. Am J Pathol. 2007;170:1942–1953. doi: 10.2353/ajpath.2007.060887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Van Steenkiste C., Ribera J., Geerts A., et al. Inhibition of placental growth factor activity reduces the severity of fibrosis, inflammation, and portal hypertension in cirrhotic mice. Hepatology. 2011;53:1629–1640. doi: 10.1002/hep.24238. [DOI] [PubMed] [Google Scholar]
  • 85.Rajapaksha I.G., Gunarathne L.S., Asadi K., et al. Liver-targeted angiotensin converting enzyme 2 therapy inhibits chronic biliary fibrosis in multiple drug-resistant gene 2-knockout mice. Hepatol Commun. 2019;3:1656–1673. doi: 10.1002/hep4.1434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Coll M., Arino S., Martinez-Sanchez C., et al. Ductular reaction promotes intrahepatic angiogenesis through Slit2-Roundabout 1 signaling. Hepatology. 2022;75:353–368. doi: 10.1002/hep.32140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Coch L., Mejias M., Berzigotti A., et al. Disruption of negative feedback loop between vasohibin-1 and vascular endothelial growth factor decreases portal pressure, angiogenesis, and fibrosis in cirrhotic rats. Hepatology. 2014;60:633–647. doi: 10.1002/hep.26995. [DOI] [PubMed] [Google Scholar]
  • 88.Calderone V., Gallego J., Fernandez-Miranda G., et al. Sequential functions of CPEB1 and CPEB4 regulate pathologic expression of vascular endothelial growth factor and angiogenesis in chronic liver disease. Gastroenterology. 2016;150:982–997 e30. doi: 10.1053/j.gastro.2015.11.038. [DOI] [PubMed] [Google Scholar]
  • 89.Cadamuro M., Brivio S., Mertens J., et al. Platelet-derived growth factor-D enables liver myofibroblasts to promote tumor lymphangiogenesis in cholangiocarcinoma. J Hepatol. 2019;70:700–709. doi: 10.1016/j.jhep.2018.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.The innate and adaptive immune systems. 2006. https://www.ncbi.nlm.nih.gov/books/NBK279396 Available from:
  • 91.Tilg H., Adolph T.E., Tacke F. Therapeutic modulation of the liver immune microenvironment. Hepatology. 2023;78:1581–1601. doi: 10.1097/HEP.0000000000000386. [DOI] [PubMed] [Google Scholar]
  • 92.Lewis J. Pathological patterns of biliary disease. Clin Liver Dis (Hoboken) 2017;10:107–110. doi: 10.1002/cld.667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Nakanuma Y., Zen Y., Harada K., et al. Application of a new histological staging and grading system for primary biliary cirrhosis to liver biopsy specimens: interobserver agreement. Pathol Int. 2010;60:167–174. doi: 10.1111/j.1440-1827.2009.02500.x. [DOI] [PubMed] [Google Scholar]
  • 94.Wen Y., Lambrecht J., Ju C., et al. Hepatic macrophages in liver homeostasis and diseases-diversity, plasticity and therapeutic opportunities. Cell Mol Immunol. 2021;18:45–56. doi: 10.1038/s41423-020-00558-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Guillot A., Winkler M., Silva Afonso M., et al. Mapping the hepatic immune landscape identifies monocytic macrophages as key drivers of steatohepatitis and cholangiopathy progression. Hepatology. 2023;78:150–166. doi: 10.1097/HEP.0000000000000270. [DOI] [PubMed] [Google Scholar]
  • 96.Chen Y.Y., Arndtz K., Webb G., et al. Intrahepatic macrophage populations in the pathophysiology of primary sclerosing cholangitis. JHEP Rep. 2019;1:369–376. doi: 10.1016/j.jhepr.2019.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Cheng S., Zou Y., Zhang M., et al. Single-cell RNA sequencing reveals the heterogeneity and intercellular communication of hepatic stellate cells and macrophages during liver fibrosis. MedComm (2020) 2023;4:e378. doi: 10.1002/mco2.378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.De Muynck K., Vanderborght B., De Ponti F.F., et al. Kupffer cells contested as early drivers in the pathogenesis of primary sclerosing cholangitis. Am J Pathol. 2023;193:366–379. doi: 10.1016/j.ajpath.2022.12.008. [DOI] [PubMed] [Google Scholar]
  • 99.Cai S.Y., Ge M., Mennone A., et al. Inflammasome is activated in the liver of cholestatic patients and aggravates hepatic injury in bile duct-ligated mouse. Cell Mol Gastroenterol Hepatol. 2020;9:679–688. doi: 10.1016/j.jcmgh.2019.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Guicciardi M.E., Trussoni C.E., Krishnan A., et al. Macrophages contribute to the pathogenesis of sclerosing cholangitis in mice. J Hepatol. 2018;69:676–686. doi: 10.1016/j.jhep.2018.05.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Li X., Liu R., Wang Y., et al. Cholangiocyte-derived exosomal lncRNA H19 promotes macrophage activation and hepatic inflammation under cholestatic conditions. Cells. 2020;9 doi: 10.3390/cells9010190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Bonnardel J., T’Jonck W., Gaublomme D., et al. Stellate cells, hepatocytes, and endothelial cells imprint the Kupffer cell identity on monocytes colonizing the liver macrophage niche. Immunity. 2019;51:638–654 e9. doi: 10.1016/j.immuni.2019.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Ishii M., Iwai M., Harada Y., et al. A role of mast cells for hepatic fibrosis in primary sclerosing cholangitis. Hepatol Res. 2005;31:127–131. doi: 10.1016/j.hepres.2005.01.007. [DOI] [PubMed] [Google Scholar]
  • 104.Tsuneyama K., Kono N., Yamashiro M., et al. Aberrant expression of stem cell factor on biliary epithelial cells and peribiliary infiltration of c-kit-expressing mast cells in hepatolithiasis and primary sclerosing cholangitis: a possible contribution to bile duct fibrosis. J Pathol. 1999;189:609–614. doi: 10.1002/(SICI)1096-9896(199912)189:4<609::AID-PATH474>3.0.CO;2-2. [DOI] [PubMed] [Google Scholar]
  • 105.Gaca M.D., Pickering J.A., Arthur M.J., et al. Human and rat hepatic stellate cells produce stem cell factor: a possible mechanism for mast cell recruitment in liver fibrosis. J Hepatol. 1999;30:850–858. doi: 10.1016/s0168-8278(99)80139-1. [DOI] [PubMed] [Google Scholar]
  • 106.Amiot L., Vu N., Drenou B., et al. The anti-fibrotic role of mast cells in the liver is mediated by HLA-G and interaction with hepatic stellate cells. Cytokine. 2019;117:50–58. doi: 10.1016/j.cyto.2019.02.002. [DOI] [PubMed] [Google Scholar]
  • 107.Zhou T., Meadows V., Kundu D., et al. Mast cells selectively target large cholangiocytes during biliary injury via H2HR-mediated cAMP/pERK1/2 signaling. Hepatol Commun. 2022;6:2715–2731. doi: 10.1002/hep4.2026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Kennedy L., Meadows V., Kyritsi K., et al. Amelioration of large bile duct damage by histamine-2 receptor vivo-morpholino treatment. Am J Pathol. 2020;190:1018–1029. doi: 10.1016/j.ajpath.2020.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Kennedy L., Hargrove L., Demieville J., et al. Blocking H1/H2 histamine receptors inhibits damage/fibrosis in Mdr2(-/-) mice and human cholangiocarcinoma tumorigenesis. Hepatology. 2018;68:1042–1056. doi: 10.1002/hep.29898. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 110.Jones H., Hargrove L., Kennedy L., et al. Inhibition of mast cell-secreted histamine decreases biliary proliferation and fibrosis in primary sclerosing cholangitis Mdr2(-/-) mice. Hepatology. 2016;64:1202–1216. doi: 10.1002/hep.28704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Hargrove L., Kennedy L., Demieville J., et al. Bile duct ligation-induced biliary hyperplasia, hepatic injury, and fibrosis are reduced in mast cell-deficient Kit(W-sh) mice. Hepatology. 2017;65:1991–2004. doi: 10.1002/hep.29079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Kennedy L.L., Hargrove L.A., Graf A.B., et al. Inhibition of mast cell-derived histamine secretion by cromolyn sodium treatment decreases biliary hyperplasia in cholestatic rodents. Lab Invest. 2014;94:1406–1418. doi: 10.1038/labinvest.2014.129. [DOI] [PubMed] [Google Scholar]
  • 113.Tang J., Yan Z., Feng Q., Yu L., et al. The roles of neutrophils in the pathogenesis of liver diseases. Front Immunol. 2021;12 doi: 10.3389/fimmu.2021.625472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Zimmer C.L., von Seth E., Buggert M., et al. A biliary immune landscape map of primary sclerosing cholangitis reveals a dominant network of neutrophils and tissue-resident T cells. Sci Transl Med. 2021;13 doi: 10.1126/scitranslmed.abb3107. [DOI] [PubMed] [Google Scholar]
  • 115.Saito J.M., Maher J.J. Bile duct ligation in rats induces biliary expression of cytokine-induced neutrophil chemoattractant. Gastroenterology. 2000;118:1157–1168. doi: 10.1016/s0016-5085(00)70369-6. [DOI] [PubMed] [Google Scholar]
  • 116.Arino S., Aguilar-Bravo B., Coll M., et al. Ductular reaction-associated neutrophils promote biliary epithelium proliferation in chronic liver disease. J Hepatol. 2023;79:1025–1036. doi: 10.1016/j.jhep.2023.05.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Casini A., Ceni E., Salzano R., et al. Neutrophil-derived superoxide anion induces lipid peroxidation and stimulates collagen synthesis in human hepatic stellate cells: role of nitric oxide. Hepatology. 1997;25:361–367. doi: 10.1053/jhep.1997.v25.pm0009021948. [DOI] [PubMed] [Google Scholar]
  • 118.Fabre T., Molina M.F., Soucy G., et al. Type 3 cytokines IL-17A and IL-22 drive TGF-beta-dependent liver fibrosis. Sci Immunol. 2018;3 doi: 10.1126/sciimmunol.aar7754. [DOI] [PubMed] [Google Scholar]
  • 119.O’Brien K.M., Allen K.M., Rockwell C.E., et al. IL-17A synergistically enhances bile acid-induced inflammation during obstructive cholestasis. Am J Pathol. 2013;183:1498–1507. doi: 10.1016/j.ajpath.2013.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Calvente C.J., Tameda M., Johnson C.D., et al. Neutrophils contribute to spontaneous resolution of liver inflammation and fibrosis via microRNA-223. J Clin Invest. 2019;129:4091–4109. doi: 10.1172/JCI122258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Harty M.W., Muratore C.S., Papa E.F., et al. Neutrophil depletion blocks early collagen degradation in repairing cholestatic rat livers. Am J Pathol. 2010;176:1271–1281. doi: 10.2353/ajpath.2010.090527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Saito J.M., Bostick M.K., Campe C.B., et al. Infiltrating neutrophils in bile duct-ligated livers do not promote hepatic fibrosis. Hepatol Res. 2003;25:180–191. doi: 10.1016/s1386-6346(02)00247-4. [DOI] [PubMed] [Google Scholar]
  • 123.Taylor S.A., Assis D.N., Mack C.L. The contribution of B cells in autoimmune liver diseases. Semin Liver Dis. 2019;39:422–431. doi: 10.1055/s-0039-1688751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Takahashi T., Miura T., Nakamura J., et al. Plasma cells and the chronic nonsuppurative destructive cholangitis of primary biliary cirrhosis. Hepatology. 2012;55:846–855. doi: 10.1002/hep.24757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Li Y., Wang W., Tang L., et al. Chemokine (C-X-C motif) ligand 13 promotes intrahepatic chemokine (C-X-C motif) receptor 5+ lymphocyte homing and aberrant B-cell immune responses in primary biliary cirrhosis. Hepatology. 2015;61:1998–2007. doi: 10.1002/hep.27725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Cichoz-Lach H., Grywalska E., Michalak A., et al. Deviations in peripheral blood cell populations are associated with the stage of primary biliary cholangitis and presence of itching. Arch Immunol Ther Exp (Warsz) 2018;66:443–452. doi: 10.1007/s00005-018-0515-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Chung B.K., Henriksen E.K.K., Jorgensen K.K., et al. Gut and liver B cells of common clonal origin in primary sclerosing cholangitis-inflammatory bowel disease. Hepatol Commun. 2018;2:956–967. doi: 10.1002/hep4.1200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Chung B.K., Ogaard J., Reims H.M., et al. Spatial transcriptomics identifies enriched gene expression and cell types in human liver fibrosis. Hepatol Commun. 2022;6:2538–2550. doi: 10.1002/hep4.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Azad A.I., Krishnan A., Troop L., et al. Targeted apoptosis of ductular reactive cells reduces hepatic fibrosis in a mouse model of cholestasis. Hepatology. 2020;72:1013–1028. doi: 10.1002/hep.31211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Thapa M., Tedesco D., Gumber S., et al. Blockade of BAFF reshapes the hepatic B cell receptor repertoire and attenuates autoantibody production in cholestatic liver disease. J Immunol. 2020;204:3117–3128. doi: 10.4049/jimmunol.1900391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Moritoki Y., Zhang W., Tsuneyama K., et al. B cells suppress the inflammatory response in a mouse model of primary biliary cirrhosis. Gastroenterology. 2009;136:1037–1047. doi: 10.1053/j.gastro.2008.11.035. [DOI] [PubMed] [Google Scholar]
  • 132.Tsukamoto H. Cytokine regulation of hepatic stellate cells in liver fibrosis. Alcohol Clin Exp Res. 1999;23:911–916. [PubMed] [Google Scholar]
  • 133.Dhirapong A., Lleo A., Yang G.X., et al. B cell depletion therapy exacerbates murine primary biliary cirrhosis. Hepatology. 2011;53:527–535. doi: 10.1002/hep.24044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Moritoki Y., Tsuneyama K., Nakamura Y., et al. Anti-drug antibodies against a novel humanized anti-CD20 antibody impair its therapeutic effect on primary biliary cholangitis in human CD20- and FcgammaR-expressing mice. Front Immunol. 2018;9:2534. doi: 10.3389/fimmu.2018.02534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Moritoki Y., Lian Z.X., Lindor K., et al. B-cell depletion with anti-CD20 ameliorates autoimmune cholangitis but exacerbates colitis in transforming growth factor-beta receptor II dominant negative mice. Hepatology. 2009;50:1893–1903. doi: 10.1002/hep.23238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Wang Y., Zhang C. The roles of liver-resident lymphocytes in liver diseases. Front Immunol. 2019;10:1582. doi: 10.3389/fimmu.2019.01582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Yang Y., Sheng Y., Wang J., et al. Double-negative T cells regulate hepatic stellate cell activation to promote liver fibrosis progression via NLRP3. Front Immunol. 2022;13 doi: 10.3389/fimmu.2022.857116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Kobayashi S., Seki S., Kawada N., et al. Apoptosis of T cells in the hepatic fibrotic tissue of the rat: a possible inducing role of hepatic myofibroblast-like cells. Cell Tissue Res. 2003;311:353–364. doi: 10.1007/s00441-002-0670-4. [DOI] [PubMed] [Google Scholar]
  • 139.Schildberg F.A., Wojtalla A., Siegmund S.V., et al. Murine hepatic stellate cells veto CD8 T cell activation by a CD54-dependent mechanism. Hepatology. 2011;54:262–272. doi: 10.1002/hep.24352. [DOI] [PubMed] [Google Scholar]
  • 140.Brinkmann H., Cerff R., Salomon M., et al. Cloning and sequence analysis of cDNAs encoding the cytosolic precursors of subunits GapA and GapB of chloroplast glyceraldehyde-3-phosphate dehydrogenase from pea and spinach. Plant Mol Biol. 1989;13:81–94. doi: 10.1007/BF00027337. [DOI] [PubMed] [Google Scholar]
  • 141.Huang B., Lyu Z., Qian Q., et al. NUDT1 promotes the accumulation and longevity of CD103(+) T(RM) cells in primary biliary cholangitis. J Hepatol. 2022;77:1311–1324. doi: 10.1016/j.jhep.2022.06.014. [DOI] [PubMed] [Google Scholar]
  • 142.Liaskou E., Jeffery L.E., Trivedi P.J., et al. Loss of CD28 expression by liver-infiltrating T cells contributes to pathogenesis of primary sclerosing cholangitis. Gastroenterology. 2014;147:221–232 e7. doi: 10.1053/j.gastro.2014.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Ravichandran G., Neumann K., Berkhout L.K., et al. Interferon-gamma-dependent immune responses contribute to the pathogenesis of sclerosing cholangitis in mice. J Hepatol. 2019;71:773–782. doi: 10.1016/j.jhep.2019.05.023. [DOI] [PubMed] [Google Scholar]
  • 144.Chan C.W., Chen H.W., Wang Y.W., et al. IL-21, not IL-17A, exacerbates murine primary biliary cholangitis. Clin Exp Immunol. 2023 doi: 10.1093/cei/uxad107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Li Y., Li B., Xiao X., et al. Itaconate inhibits CD103+ TRM cells and alleviates hepatobiliary injury in mouse models of primary sclerosing cholangitis. Hepatology. 2023 doi: 10.1097/HEP.0000000000000549. [DOI] [PubMed] [Google Scholar]
  • 146.Liu Q., Yang Q., Wu Z., et al. IL-1beta-activated mTORC2 promotes accumulation of IFN-gamma(+) gammadelta T cells by upregulating CXCR3 to restrict hepatic fibrosis. Cell Death Dis. 2022;13:289. doi: 10.1038/s41419-022-04739-3. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cellular and Molecular Gastroenterology and Hepatology are provided here courtesy of Elsevier

RESOURCES