972
views
1
recommends
+1 Recommend
1 collections
    4
    shares

      Interested in becoming an AMM published author?

      • Platinum Open Access with no APCs.
      • Fast peer review/Fast publication online after article acceptance.

      Check out the call for papers on our website https://amm-journal.org/index.php/2023/04/26/acta-materia-medica-call-for-papers-2/

      scite_
       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Molecular mechanisms of transporter regulation and their impairment in intrahepatic cholestasis

      review-article
      Bookmark

            Abstract

            Intrahepatic cholestasis (IC) is a liver disease caused by disorders in bile formation and excretion, owing to structural and functional abnormalities in hepatocytes and/or bile capillaries. IC is commonly caused by hepatitis virus, alcohol consumption, drug-induced liver damage, autoimmune liver disease and heredity. In the absence of effective treatment, IC can progress to liver fibrosis, cirrhosis and ultimately liver failure. However, the mechanisms underlying IC remain poorly understood. IC is believed to be closely associated with changes in the transcription, function and localization of hepatocellular transport proteins. To better understand the molecular mechanisms of transport proteins in IC, herein, we review the roles of these transport proteins and discuss their underlying regulatory mechanisms in IC. Our aim is to provide a reference for understanding IC pathogenesis and developing effective drug therapies.

            Main article text

            1. INTRODUCTION

            Intrahepatic cholestasis (IC) is characterized by damage to hepatocytes or intrahepatic bile ducts, and the accumulation of bile components in the serum [1]. IC, mainly including primary biliary cirrhosis (PBC), primary sclerosing cholangitis and intrahepatic cholestasis of pregnancy (ICP), can be caused by inflammatory disorders, drugs, heredity and the environment [2, 3]. Any functional perturbation in the bile secretory process may lead to IC, which is associated with intracellular accumulation of toxic bile constituents and consecutive cholestatic liver cell damage [4]. The main pathogenic mechanisms of IC may include deregulation of bile secretion, impaired cell-membrane fluidity, inflammatory responses, and changes in hepatocyte tight junctions and transporters [5].

            Many transporters expressed in hepatocytes and cholangiocytes are involved in bile formation and excretion. The secretion of bile is a hepatocellular transport processes occurring mainly across the canalicular membranes of hepatocytes. Perturbations in the function, expression and/or localization of transporters lead to the intracellular accumulation of toxic bile acids (BAs), thus promoting cholestatic liver injury [6]. Alterations in hepatobiliary transporter function are important risk factors for susceptibility to IC development [7]. Mutations in transporter genes can cause hereditary cholestatic liver disease [8]. Mutations in multidrug resistance 3 (MDR3) and bile salt export pump (BSEP) can cause an array of cholestatic syndromes, including progressive and benign forms of familial IC and ICP [9, 10], have been well established to cause inherited cholestatic syndromes [11, 12]. Furthermore, genetically determined functional changes in hepatobiliary transport systems have been demonstrated to cause acquired cholestatic syndromes, such as ICP and drug-induced cholestasis [7]. The transcription and expression of transporters are regulated by complex networks. Transporters are regulated by multiple nuclear receptors (NRs) at the transcriptional level [13]. After their transcription and translation, transporters can also be regulated by various protein kinases (PKs) such as protein kinase B (Akt) and protein kinase C family members (PKCs) [1416]. The first-line treatment for IC is ursodeoxycholic (UDCA). However, approximately 40% of patients have inadequate responses [17]. Therefore, herein, we review the molecular mechanisms of transporter dysregulation under IC, to provide a reference for understanding its pathogenesis and developing effective drug therapies.

            2. PHYSIOLOGY OF HEPATOBILIARY TRANSPORT AND BILE FORMATION

            Hepatic uptake and efflux processes involved in bile formation are maintained by distinct transport systems. After canalicular secretion, the bile composition undergoes further modification in the canaliculus, through reabsorption and secretion processes maintained by apical and basolateral transport systems in cholangiocytes. Figure 1 shows a scheme of the hepatocellular and bile duct transport proteins involved in the uptake and efflux of bile compounds (e.g., BAs).

            Figure 1 |

            The roles of transporters in enterohepatic circulation of BAs.

            BAs are synthesized from cholesterol. Subsequently, BAs are secreted into bile in the canaliculus by membrane transporters. Most BAs are reabsorbed into the portal vein by transporters on cholangiocytes and enterocytes. In the sinusoids of the liver, BAs are taken up by NTCP and OATPs and are recycled back to the liver.

            Bile, comprising mainly BAs, cholesterol, bilirubin, bile pigment, phosphatidylcholine, water and inorganic salts, plays crucial roles in the digestion and absorption of lipids and lipid-soluble drugs. Bile production begins at the canaliculus in hepatocytes, and bile is modified downstream by cholangiocytes [18, 19]. Bile formation is a fundamental physiological process comprising the active transport of BAs and other solutes across the canalicular membrane [20]. BAs, the most important components of bile, are synthesized from cholesterols [21]. BA synthesis requires 17 enzymatic reactions. In the classical synthetic pathway, metabolism of cholesterols into 7α-hydroxycholesterol via 7α-hydroxylase (CYP7A1) is a key step. Most primary BAs, such as cholic acid and deoxycholic acid, are metabolized immediately into taurine- and glycine-binding BAs. They are transported into the canaliculus by hepatic transporters, then mixed with other components to form bile [22].

            BAs in bile enter the intestine through the contraction of the gallbladder. Under the action of the intestinal microbiota, cholic acid and deoxycholic acid are transformed into UDCA and lithocholic acid (LCA), respectively, and all taurine- and glycine-binding BAs are deamidated in the terminal ileum and colon. In the entire process of enterohepatic circulation, 90% of BAs are reabsorbed by intestinal epithelium, and the rest is discharged from the human body through the feces [23]. In the small intestine, some free BAs and glycine-binding BAs can be reabsorbed passively, but most are actively absorbed in the terminal ileum by the apical sodium-dependent BA transporter (ASBT, SLC10A2) and organic solute transporter α/β (OSTα/β, SLC51a/b) on the basement membrane [24]. Most BAs in the hepatic portal vein, together with bilirubin and a variety of other organic anions, are reabsorbed from the blood into the liver through uptake transporters, and the rest are eliminated from the kidneys through the blood circulation. BAs can generally be eliminated after 20 enterohepatic circulations. Therefore, BA uptake disorders, obstructed BA efflux and bile duct injury in the liver can all lead to cholestasis [25].

            2.1 Liver transporters involved in bile formation and excretion

            The secretion and excretion of bile depend on complex hepatobiliary transport systems and cholangiocytes. Many transporters are expressed on the basolateral or canalicular membranes of hepatocyte, mainly including ATP-binding cassette (ABC) and solute carrier family (SLC) transporters. ABC transporters efflux substrates, whereas SLC transporters mediate the uptake of substrates into cells. These transporters play important roles in bile formation and the biliary excretion of xenobiotics. In bile flow obstruction, cholestasis has been attributed to the expression and functional abnormalities of various transporters [26]. The transporters in cholangiocytes take up electrolytes and water into the blood, thus forming a “biliary-liver” cycle, and excrete them into the bile duct, thereby further promoting bile flow ( Figure 1 ). Therefore, transporters play important roles in the secretion and excretion of bile in cholestasis.

            2.1.1 SLC transporters

            SLC transporters include Na+-taurocholate co-transport polypeptide (NTCP, SLC10A1), organic anion transporter polypeptides (OATPs, SLCO), OSTα/β and ASBT. On the basolateral membranes of hepatocytes, BAs are taken up primarily by NTCP and OATPs. Furthermore, NTCP also transports steroidal hormones and a variety of drugs. Repression and translocation of NTCP contribute to the etiopathogenesis of IC [27]. OATPs take up other cholephilic compounds, including glucuronidated bilirubin, exogenous organic anions, leukotrienes, estrogen conjugates (e.g., estrone-3-sulfate and estradiol-17-β-d-glucuronide), thyroid hormones, mycotoxins and numerous xenobiotics [2830]. Human OATP1A and rat OATP2 mediate the uptake of bulky organic cations, whereas small organic cations are taken up by organic cation transporter 1 (OCT1, SLC22A1) [31].

            2.1.2 ABC transporters

            In hepatocytes, bile excretion occurs mainly at the canalicular membrane, predominantly via ABC transporters. ABC transporters are a superfamily of membrane proteins that mediate diverse ATP-driven transport processes; main members include BSEP (ABCB11), multidrug resistance protein 2 (MRP2, ABCC2), P-glycoprotein (P-gp/MDR1, ABCB1), breast cancer drug resistance protein (BCRP, ABCG2) and MDR3 (ABCB4) [32, 33]. BSEP and MRP2, two main transporters on the canalicular membrane, excrete BAs into the bile duct. Monoanionic bile salts are excreted mainly into the canalicular pole by BSEP [34]. In contrast, canalicular efflux of divalent, sulfated or glucuronidated bile salts, glutathione or glucuronidated bilirubin is mediated by MRP2 [33]. In addition, phosphatidylcholine, cholesterol and other compounds are excreted into the canaliculus through ATP-binding cassette subfamily B member 4 (MDR2, ABCB4), ATP-binding cassette subfamily G member 5/8 (ABCG5/G8), ATP-binding cassette subfamily B member 1 (MDR1, ABCB1) and MRP2 [35].

            Cooperation among transporters is critical to maintaining bile homeostasis ( Figure 2 ). In early stages of acute and chronic cholestasis, the NTCP-BSEP axis is blocked, thus leading to accumulation of BAs in hepatocytes and spontaneously activating the OATP-MRP2 axis, thereby accelerating the excretion of BAs. It maintains bile homeostasis, delays rapid increases in intracellular BA concentrations, and alleviates hepatocyte structural and functional damage [21].

            Figure 2 |

            The roles of transporters in IC.

            IC results in intrahepatic accumulation of BAs, thus leading to a toxic hepatocellular bile acid burden. In addition, the uptake of BAs is restricted, owing to downregulation of NTCP and OATPs. Export of BAs is mediated by basolateral transporters, such as BSEP, MRP2, MRP3, MRP4 and OSTα/β. Decreased expression of these transporters results in diminished bile acid excretion, thus further increasing BA accumulation in the liver and triggering IC.

            2.2 Transporters in intrahepatic cholangiocytes

            During bile flow, formation and excretion, the intrahepatic bile duct secretes the electrolytes Cl−1 and HCO3 −1 into the bile via several transporters or channels expressed on cholangiocyte membranes, thus synergistically regulating the fluidity and pH of bile in the bile duct [36]. These transporters include cystic fibrosis transmembrane conductance regulator (CTFR), anion exchanger 2 (AE-2) and aquaporin-1 (AQP1). Their dysfunction directly leads to abnormal secretion of inorganic salts and water, and alterations in bile composition and flow [37]. Impairment of cholangiocyte transporters and aquaporin leads to “toxic” bile, owing to both a lack of the “HCO3 −1” and increased intraluminal levels of damaging Bas [38]. CTFR transports intracellular Cl−1 outside the plasma membrane [39]. Subsequently, Cl−1 on the plasma membrane secondarily drives the Cl−1-HCO3 −1 transporter AE-2, which actively secretes HCO3 −1 into the bile [40], whereas AQP1 transports water molecules into the bile [41]. Genetic abnormalities in CFTR result in attenuation of bile hydration, accumulation of toxic BAs, cholangiocyte damage and cholestasis, and ultimately progression to cystic fibrosis [42]. Abnormal function and expression of AE-2 are also associated with PBC [43].

            3. TRANSCRIPTIONAL REGULATION OF HEPATIC TRANSPORTERS BY NRS

            NRs, a family of 48 members, play important roles in BA homeostasis, lipid metabolism, and mechanisms involved in fibrosis and inflammation. Several of the adaptive changes in cholestasis are mediated by NRs, because biliary compounds retained during cholestasis (e.g., BAs, bilirubin, oxysterols, hormones and drugs) act as NR ligands and coordinately affect target-gene expression [44, 45]. NRs, mainly farnesoid X receptor (FXR, NR1H4), pregnane X receptor (PXR, NR1I2), constitutive androgen receptor (CAR, NR1I3), liver X receptor α (LXRa, NR1H3) and vitamin D receptor (VDR, NR1I1), are involved in the maintenance of BA homeostasis in IC [13]. In addition, other NRs, including liver receptor homolog-1 (LRH-1, NR5A2) and peroxisome proliferator-activated receptors (PPARs, NR1Cs) ( Table 1 ), play important roles in IC [46]. Changes in transporter regulation comprise a complex interaction network of several ligand-activated NRs as well as liver-enriched hepatocyte nuclear factors.

            Table 1 |

            Main nuclear receptors involved in transporter regulation.

            NRNameLigands
            FXR (NR1H4)Farnesoid X-activated receptorBile acids (CDCA, DCA, LCA and CA); possibly UDCA (weak ligand); synthetic: GW4064, 6α-ethyl-CDCA and fexaramines
            PXR (NR1I2)Pregnane X receptorBile acids, rifampicin in humans, phenobarbital, dexamethasone, statins, St. John’s wort and clotrimazole pregnenolone-16a-carbonitrile
            CAR (NR1I3)Constitutive androstane receptorBilirubin, phenobarbital, TCPOBOP, dimethoxycoumarin, xenobiotics, Yin Chin and CITCO in humans
            VDR (NR1I1)Vitamin D receptorVitamin D and LCA
            LRH-1 (NR5A2)Liver receptor homolog-1Phospholipids
            PPARα (NR1C1)Peroxisome proliferator-activated receptor αFatty acids, fibrates, statins, eicosanoids, leukotrienes, NSAIDs and WY-14643
            3.1 FXR

            FXR is a major NR regulating the expression of transporters and maintaining BA homeostasis during pathogenesis of cholestasis [47]. In early stages of chronic cholestasis, FXR is rapidly activated, thus forming FXR-RXR (retinoid X receptor, NR2B1) heterodimers. Subsequently, the dimers bind inverted repeat 1 (IR-1) elements in the target-gene promoter and significantly up-regulate the expression of BSEP and MRP2, thereby accelerating BA excretion. FXR also induces the expression of a nuclear orphan receptor small heterodimer partner (SHP), and consequently inhibits the functions of other NRs, such as liver X receptor (NR1H3) and hepatocyte nuclear factor 4α (HNF4α), and ultimately suppresses the expression of CYP7A1/CYP8B1 and NTCP in hepatocytes, thereby decreasing BA synthesis and uptake, and indirectly accelerating BA clearance [48]. Similarly to NTCP, OATP1B1 and ASBT are negatively regulated by FXR through the interaction of SHP with HNF4 [49]. In addition, FXR directly promotes cellular bile clearance via directly inducing canalicular BSEP and MRP2 [50, 51].

            In another regulatory pathway inhibiting BA synthesis, intestinal FXR induces the expression of an intestinal hormone-like peptide, fibroblast growth factor 15/19 (murine FGF15 or human FGF19). FXR induces the expression of FGF15/19 through the activation of hepatic FGF receptor 4 (FGFR4), then activates the intracellular stress-activated Jun N-terminal-kinase pathway and consequently inhibits CYP7A1 activity and decreases BA synthesis [52]. Thus, the FXR-FGF19 pathway, through a typical negative feedback regulation mechanism, plays a critical role in the pathogenesis of cholestatic diseases [53]. FXR may therefore be a promising therapeutic target for novel drug development in IC.

            3.2 PXR and CAR

            Recent studies have revealed that, beyond FXR, PXR and CAR are key NRs regulating many adaptive responses in IC. They coordinate protective hepatic responses to toxic stimuli, induced by endogenous compounds (BAs or bilirubin) and xenobiotics [54, 55]. As sensors of toxic byproducts, they are central in the detoxification pathways involving phase I/II detoxification and transporters [54, 56, 57]. Levels of PXR and CAR are diminished in IC [58], whereas PXR polymorphisms are associated with greater susceptibility to ICP [59]. MRP3/4 expression is upregulated via PXR and CAR, thereby alleviating cholestatic liver injury [60, 61]. In addition, FXR induces BA export and metabolism via transcriptional activation of PXR [62]. Targeting these NRs may provide therapeutic benefits for patients with cholestasis in the future.

            3.3 VDR

            VDR, expressed in the intestines, kidney and liver, is also activated by BAs. Recent reports have shown that VDR regulates BA transporters, and its polymorphic variants may affect individual susceptibility and quality of life in patients with IC, such as PBC or ICP [63, 64]. However, the direct effects of VDR polymorphisms on the pathogenesis of IC are unclear. Loss of VDR exacerbates cholestatic liver injury through the disruption of biliary epithelial cell junctions in mice [65]. VDR increases ASBT mRNA expression and promoter activity [66]. Moreover, VDR appears to play an indirect role in BA homeostasis. Furthermore, vitamin D intake has been suggested to relieve biliary fibrosis in ABCB-knockout mice and ameliorate cholestatic disease [67]. Therefore, VDR may serve as a therapeutic target in cholestatic diseases.

            3.4 LRH-1

            LRH-1, a transcription factor in bile salt synthesis, is expressed mainly in the liver, intestines, exocrine pancreas and reproductive tissue [68]. It binds DNA in its monomeric form and regulates other NRs and the transcription of genes involved in the biosynthesis and transport of BAs, including CYP7A1 [69], BSEP, MRP2, ASBT, NTCP, MRP3 and MDR2 [7073]. LRH-1 induces the expression of CYP7A1, BSEP [71] and ASBT [73]. In addition, deletion of LRH-1 significantly decreases the expression of FXP and SHP as well as multiple transporters (i.e., NTCP, BSEP, MRP3, MRP2 and MDR2) [70]. However, the effect of LRH-1 remains to be fully elucidated in IC.

            3.5 PPARα

            PPARα, a ligand-activated nuclear receptor, plays a central role in maintaining cholesterol, lipid and BA homeostasis by regulating genes involved in BA synthesis and transport. PPARα primarily down-regulates BA synthesis through inhibition of BA-synthesizing enzymes (i.e., CYP7A1 and CYP27A1) [74]. In addition, PPARα induces biliary phospholipid output by activating canalicular MDR3 [75]. PPARα activators directly induce canalicular MDR2, thereby inducing biliary phospholipid output [76]. In addition, ASBT expression in cholangiocytes and the intestines is induced by PPARα [77], thus increasing BA absorption from the intestines and bile ducts. Bezafibrate, a dual PPAR and PXR agonist, increases the expression of NTCP, MDR1, MDR3 and MRP2, thus protecting against cholestatic liver injury [78]. Agonists of PPARα are promising therapeutic approaches in IC.

            4. LOCALIZATION REGULATION OF HEPATIC TRANSPORTERS

            Accurate expression and localization of transporters on the plasma membrane require interactions among various proteins between the membrane and cytoskeleton—a complex process regulated by PKs. PKs contain serine (Ser), threonine (Thr) and tyrosine (Tyr) residues, or lysine (Lys), histidine (His) and arginine (Arg) residues. Various PKs, such as PKB (Akt) and PKC, regulate the localization of hepatic bile transporters after transcription, and the activation of phosphoinositide-3-kinase (PI3K)/Akt signaling causes sustained internalization of MRP2 and BSEP, thus eventually leading to cholestasis [79, 80]. Via second messengers, PKs initiate signaling cascades, regulate the phosphorylation and dephosphorylation of the hepatobiliary transport system and corresponding crosslinked proteins or scaffolding proteins, alter the membrane localization of transporters and rapidly adjust bile composition, thus subsequently promoting cholestasis and/or exerting choleretic effects.

            4.1 PKB (Akt)

            The Ser/Thr kinase PKB (Akt) has been widely studied as a cell growth factor regulating the functions of multiple downstream anti-apoptotic proteins [81]. Akt is considered the characteristic target protein and terminal effector of PI3K [82, 83]. Beuers et al. [84] have found that in a taurolithocholic acid (TLCA)-induced cholestasis model, wortmannin, a PI3K-specific inhibitor, decreases Akt activity and attenuates cholestasis, thus suggesting a causal link between these events. Furthermore, in an E217G- and TLCA-induced cholestasis model, the anti-cholestatic effect of an Akt inhibitor (Calbiochem 124005) is similar to that of a PI3K inhibitor (LY294002), and identical anti-cholestatic outcomes have been achieved by combining wortmannin with Calbiochem 124005 or a conventional PKC (cPKC) inhibitor (G6976) [80]. In addition, the activation of PI3K and Akt contributes to sustaining the internalization of transporters and the consequent impairment of their activity. Thus, the PI3K/Akt pathway is largely responsible for cholestasis.

            4.2 PKC

            PKCs are a group of PKs mediating the function of targeted proteins through phosphorylation of serine and threonine amino acid residues. Ten subtypes of PKC have been found in mammalian tissues, and can be divided into three groups: cPKC, including α, βI, βII and γ subtypes; novel PKC (nPKC), including δ, ε, η and θ subtypes; and atypical PKC (aPKC), including ι (also known as λ in mice) and ζ subtypes [85]. The activation of several PKC subtypes in the liver, such as aPKCζ and nPKCδ, depends on PI3K [8688]. However, the activation of cPKCs does not rely on PI3K [80, 89, 90], and oxidative stress can mediate the activation of cPKCs and nPKCs [91]. In the past, the activation of PKCs has been found to induce cholestasis [16], inhibit cAMP-induced intake of taurocholate and decrease MRP3- and OATP-mediated transport of organic solutes [9294]. Different PKCs are activated by various compounds and subsequently exert pro-cholestatic, anti-cholestatic and choleretic effects. Accordingly, different compounds appear to function differently by affecting various subtypes of PKCs and corresponding signaling pathways ( Figure 3 ).

            Figure 3 |

            Proposed model for the regulation of NTCP and MRP2 by PKC isoforms.

            Activation of nPKCδ and aPKCζ by cAMP leads to translocation of NTCP and MRP2 to the plasma membrane. Activation of nPKCδ by GCDCA facilitates MRP2 translocation to the plasma membrane. Activation of cPKCa by phorbol myristate acetate and taurochenodeoxycholate acid induces retrieval of NTCP from the plasma membrane. Activation of cPKCa and nPKCε has been implicated in MRP2 retrieval from the plasma membrane by cholestasis induced by E217G, ethacrynic acid and TLCA.

            4.2.1 The roles of PKC subtypes in IC

            Both cPKCα and nPKCδ participate in the pathogenesis of cholestasis. In an E217G-induced cholestasis model, cPKCα participates in pathogenesis by activating downstream estrogen receptor (ERα) signaling [95]. cPKCα mediates NTCP internalization induced by phorbol myristate acetate and taurochenodeoxycholate acid [90, 96]. Taurodeoxycholic acid exerts post-translational anticholestatic effects through a cooperative cPKCα-/PKA-dependent mechanism in an experimental model of TLCA-induced cholestasis [97]. A recent study has indicated that nPKCδ is activated by cAMP, and is involved in cAMP-mediated NTCP and MRP2 translocation in hepatocytes [87]. In contrast to those of cPKCα, the effects of nPKCδ are associated with its phosphorylation sites, which are activated by various signals. Activation of nPKCδ may lead to cholestatic effects via Tyr phosphorylation, whereas its activation may lead to anti-cholestatic effects via Thr phosphorylation [98, 99]. In agreement with this hypothesis, the activation of nPKCδ by cAMP and GCDCA is associated with Thr and not Tyr phosphorylation in rat hepatocytes [88, 100]. Nevertheless, the effects and underlying mechanism of differential phosphorylation of nPKCδ in IC still require validation.

            nPKCε and aPKCζ are two important PKC subtypes. In primary hepatocytes, TLCA activates nPKCε and induces MRP2 endocytosis. Knockdown of nPKCε reverses TLCA-induced internalization of MRP2 [101]. In addition, cAMP and taurodeoxycholic acid reverse TLCA-induced cholestasis and MRP2 retrieval by inhibiting nPKCε [84, 102, 103]. MRP2 retrieval induced by ethacrynic acid is also mediated via nPKCε in rats [91]. In contrast to nPKCε, cAMP promotes the delivery and localization of NTCP toward the basement membrane via the PI3K/aPKCζ pathway [104]. Because aPKCζ, BSEP and MRP2 are all expressed on the membranes of hepatocytes [105], aPKCζ may also be involved in the canalicular localization of the two transporters, a possibility that should be further studied.

            5. CONCLUSION

            Transporters participate in the transmembrane transport of bile, and their disorders play important roles in the pathogenesis of cholestasis. In past decades, the essential roles of hepatic transporters in the pathogenesis of cholestasis have been gradually revealed. However, studies on the molecular mechanism underlying cholestasis have been limited mainly to transcriptional, expression and functional abnormalities in individual or several transporters, thus yielding limited conclusions. Notably, abnormal transporter localization can also cause cholestasis. The localization of hepatocyte transporters has been studied, but a comprehensive understanding is lacking. In addition, few studies have assessed the location and function of cholangiocyte transporters. Overall, the pathogenesis of cholestasis induced by abnormal bile transport is a complex network comprising multiple transporters that synergistically secrete and excrete bile from the liver. Furthermore, given the compensatory protective mechanisms in the human body, transporters exert distinctly different effects on acute and chronic cholestasis, thus enabling the underlying molecular mechanisms to be unraveled.

            NRs, as transcription factors, regulate transporter genes required for hepatobiliary transport, as well as phase I and II metabolic enzymes involved in processing their substrates. Impaired NR signaling may affect the expression of transporters, and genetic variants of NR-encoding genes are associated with IC susceptibility and progression. In addition, altered localization of transporters participates in pathogenesis of IC. These changes in transporter localization are highly regulated post-translational events requiring various cellular signaling pathways, such as PKB (Akt) and PKC. Atypical PKCζ may mediate choleretic effects by inserting NTCP into the plasma membrane, and nPKCε may mediate cholestatic effects by retrieving MRP2 from the plasma membrane [91]. In contrast, cPKCα and nPKCδ may be involved in choleretic, cholestatic and anticholestatic effects by inserting, retrieving and inhibiting retrieval of transporters, respectively. Thus, we reviewed the molecular mechanisms through which transporters are regulated through various proteins such as NRs and PKCs in cholestasis, to provide a reference for understanding IC pathogenesis and developing effective drug therapies. Nevertheless, considerable in-depth studies remain necessary to comprehensively clarify the network of regulatory mechanisms of cholestasis-associated transporters.

            ABBREVIATIONS

            BA, bile acid; UDCA, ursodeoxycholic acid; LCA, lithocholic acid; TLCA, taurolithocholic acid; ABC, ATP-binding cassette; SLC, solute carrier; ABCB11, bile salt export pump, BSEP; CAR, constitutive androgen receptor; CYP7A1, cholesterol 7α-hydroxylase; FXR, farnesoid X receptor; ABCB4, multidrug resistance protein 3; ABCC2, multidrug resistance protein 2; ABCC3, multidrug resistance protein 3; ABCC4, multidrug resistance protein 4; NTCP, Na+-taurocholate co-transport polypeptide; OSTα/β, organic solute transporter α/β; PXR, pregnane X receptor; PPARα, peroxisome proliferator-activated receptor α; SHP, small heterodimer partner; PI3K, phosphoinositide-3-kinase; PKC, protein kinase C; CTFR, cystic fibrosis transmembrane conductance regulator; AE-2, Cl-1-HCO3 -1 transporter; APQ-1, aquaporin-1; VDR, Vitamin D receptor; LRH-1, Liver receptorhomolog-1; CDCA, Chenodeoxycholic acid; DCA, Deoxycholic acid; CA, Cholic acid; TCDCA, Taurochenodeoxycholic Acid; GCDCA, Glycochenodeoxycholic Acid; E217G, Estradiol-17beta-glucuronide.

            ACKNOWLEDGEMENTS

            This work was supported by the National Natural Science Foundation of China (No. 82073939).

            CONFLICTS OF INTERESTS

            The authors have no conflicts of interest associated with this publication.

            REFERENCES

            1. Amirneni S, Haep N, Gad MA, Soto-Gutierrez A, Squires JE, Florentino RM. Molecular Overview of Progressive Familial Intrahepatic Cholestasis. World Journal of Gastroenterology. 2020. Vol. 26:7470–7484. [Cross Ref]

            2. Xiao J, Li Z, Song Y, Sun Y, Shi H, Chen D, et al.. Molecular Pathogenesis of Intrahepatic Cholestasis of Pregnancy. Canadian Journal of Gastroenterology and Hepatology. 2021. Vol. 2021:6679322. [Cross Ref]

            3. Trauner M, Meier PJ, Boyer JL. Molecular Pathogenesis of Cholestasis. The New England Journal of Medicine. 1998. Vol. 339:1217–1227. [Cross Ref]

            4. Pauli-Magnus C, Meier PJ. Hepatocellular Transporters and Cholestasis. Journal of Clinical Gastroenterology. 2005. Vol. 39(4):Suppl 2:S103–S110. [Cross Ref]

            5. Zu Y, Yang J, Zhang C, Liu D. The Pathological Mechanisms of Estrogen-Induced Cholestasis: Current perspectives. Frontiers in Pharmacology. 2021. Vol. 12:761255. [Cross Ref]

            6. Piatek K, Kurzawinska G, Magielda J, Drews K, Barlik M, Malewski Z, et al.. The Role of ABC Transporters’ Gene Polymorphism in the Eetiology of Intrahepatic Cholestasis of Pregnancy. Ginekologia Polska. 2018. Vol. 89:393–397. [Cross Ref]

            7. Pauli-Magnus C, Meier PJ. Pharmacogenetics of Hepatocellular Transporters. Pharmacogenetics. 2003. Vol. 13:189–198. [Cross Ref]

            8. Thompson R, Jansen PL. Genetic Defects in Hepatocanalicular Transport. Seminars in Liver Disease. 2000. Vol. 20:365–372. [Cross Ref]

            9. Jacquemin E. Role of Multidrug Resistance 3 Deficiency in Pediatric and Adult Liver Disease: One Gene for Three Diseases. Seminars in Liver Disease. 2001. Vol. 21:551–562. [Cross Ref]

            10. Rosmorduc O, Hermelin B, Poupon R. MDR3 Gene Defect in Adults with Symptomatic Intrahepatic and Gallbladder Cholesterol Cholelithiasis. Gastroenterology. 2001. Vol. 120:1459–1467. [Cross Ref]

            11. Noe J, Kullak-Ublick GA, Jochum W, Stieger B, Kerb R, Haberl M, et al.. Impaired Expression and Function of the Bile Salt Export Pump Due to Three Novel ABCB11 Mutations in Intrahepatic Cholestasis. Journal of Hepatology. 2005. Vol. 43:536–543. [Cross Ref]

            12. Strautnieks SS, Bull LN, Knisely AS, Kocoshis SA, Dahl N, Arnell H, et al.. A Gene Encoding a Liver-Specific ABC Transporter is Mutated in Progressive Familial Intrahepatic Cholestasis. Nature Genetics. 1998. Vol. 20:233–238. [Cross Ref]

            13. Halilbasic E, Baghdasaryan A, Trauner M. Nuclear Receptors as Drug Targets in Cholestatic Liver Diseases. Clinical Liver Disease. 2013. Vol. 17:161–189. [Cross Ref]

            14. Abdullah M, Amélie M, Marc LV, Bruno S, Claire D, Yannick P, et al.. Protein Kinases C-Mediated Regulations of Drug Transporter Activity, Localization and Expression. International Journal of Molecular Sciences. 2017. Vol. 18:764

            15. Marcelo GR, Ismael RB, Gisel SM, Fernando AC, Enrique JSP. Dynamic Localization of Hepatocellular Transporters: Role in Biliary Excretion and Impairment in Cholestasis. Current Medicinal Chemistry. 2019. Vol. 26:1113–1154

            16. Anwer MS. Role of Protein Kinase C Isoforms in Bile Formation and Cholestasis. Hepatology. 2014. Vol. 60:1090–1097. [Cross Ref]

            17. Woolbright BL, Jaeschke H. Therapeutic Targets for Cholestatic Liver Injury. Expert Opinion on Therapeutic Targets. 2016. Vol. 20:463–475. [Cross Ref]

            18. Eshmuminov D, Schuler MJ, Becker D, Bautista BL, Mueller M, Hagedorn C, et al.. Bile Formation in Long-term Ex Situ Perfused Livers. Surgery. 2021. Vol. 169:894–902. [Cross Ref]

            19. Boyer JL, Soroka CJ. Bile Formation and Secretion: An Update. Journal of Hepatology. 2021. Vol. 75(1):190–201. [Cross Ref]

            20. Kullak-Ublick GA, Stieger B, Meier PJ. Enterohepatic Bile Salt Transporters in Normal Physiology and Liver Disease. Gastroenterology. 2004. Vol. 126:322–342. [Cross Ref]

            21. Cuperus FJ, Claudel T, Gautherot J, Halilbasic E, Trauner M. The Role of Canalicular ABC Transporters in Cholestasis. Drug Metabolism and Disposition. 2014. Vol. 42:546–560. [Cross Ref]

            22. Javitt NB. Hepatic Bile Formation: Bile Acid Transport and Water Flow into the Canalicular Conduit. American Journal of Physiology-Gastrointestinal and Liver Physiology. 2020. Vol. 319:G609–G618. [Cross Ref]

            23. Liu X, Wang Y. An Overview of Bile Acid Synthesis and Its Physiological and Pathological Functions. Yi Chuan. 2019. Vol. 41:365–374. [Cross Ref]

            24. Hofmann AF. The Enterohepatic Circulation of Bile Acids in Mammals: Form and Functions. Frontiers in Bioscience (Landmark Ed). 2009. Vol. 14:2584–2598. [Cross Ref]

            25. Rodrigues AD, Lai Y, Cvijic ME, Elkin LL, Zvyaga T, Soars MG. Drug-Induced Perturbations of the Bile Acid Pool, Cholestasis, and Hepatotoxicity: Mechanistic Considerations Beyond the Direct Inhibition of the Bile Salt Export Pump. Drug Metabolism and Disposition. 2014. Vol. 42:566–574. [Cross Ref]

            26. Strazzabosco M, Spirli C, Okolicsanyi L. Pathophysiology of the Intrahepatic Biliary Epithelium. Journal of Gastroenterology and Hepatology. 2000. Vol. 15:244–253. [Cross Ref]

            27. Irak K, Bayram M, Cifci S, Acar Z, Kazezoglu C, Yorulmaz E, et al.. Role of Serum Organic Solute Transporter Alpha/beta and Sodium Taurocholate Cotransporting Polypeptide in Intrahepatic Cholestasis of Pregnancy. Ginekologia Polska. 2021. Vol. 92:767–773. [Cross Ref]

            28. Bohan A, Boyer JL. Mechanisms of Hepatic Transport of Drugs: Implications for Cholestatic Drug Reactions. Seminars in Liver Disease. 2002. Vol. 22:123–136. [Cross Ref]

            29. Ito K, Suzuki H, Horie T, Sugiyama Y. Apical/basolateral Surface Expression of Drug Transporters and its Role in Vectorial Drug Transport. Pharmaceutical Research. 2005. Vol. 22:1559–1577. [Cross Ref]

            30. Beaudoin JJ, Bezencon J, Sjostedt N, Fallon JK, Brouwer K. Role of Organic Solute Transporter Alpha/Beta in Hepatotoxic Bile Acid Transport and Drug Interactions. Toxicological Sciences. 2020. Vol. 176:34–35. [Cross Ref]

            31. Van Montfoort JE, Muller M, Groothuis GM, Meijer DK, Koepsell H, Meier PJ. Comparison of “Type I” and “Type II” Organic Cation Transport by Organic Cation Transporters and Organic Anion-Transporting Polypeptides. Journal of Pharmacology and Experimental Therapeutics. 2001. Vol. 298:110–115

            32. Thoeni C, Waldherr R, Scheuerer J, Schmitteckert S, Roeth R, Niesler B, et al.. Expression Analysis of ATP-Binding Cassette Transporters ABCB11 and ABCB4 in Primary Sclerosing Cholangitis and Variety of Pediatric and Adult Cholestatic and Noncholestatic Liver Diseases. Canadian Journal of Gastroenterology and Hepatology. 2019. Vol. 2019:1085717. [Cross Ref]

            33. Akita H, Suzuki H, Ito K, Kinoshita S, Sato N, Takikawa H, et al.. Characterization of Bile Acid Transport Mediated by Multidrug Resistance Associated Protein 2 and Bile Salt Export Pump. Biochimica et Biophysica Acta. 2001. Vol. 1511:7–16. [Cross Ref]

            34. Suchy FJ, Ananthanarayanan M. Bile Salt Excretory Pump: Biology and Pathobiology. Journal of Pharmacology and Experimental Therapeutics. 2006. Vol. 43 Suppl 1:S10–S16. [Cross Ref]

            35. Ben SA, Bruneau A, Mareux E, Lapalus M, Delaunay JL, Gonzales E, et al.. Molecular Regulation of Canalicular ABC Transporters. International Journal of Molecular Sciences. 2021. Vol. 22:2113. [Cross Ref]

            36. Marinelli RA, Vore M, Javitt NB. Hepatic Bile Formation: Canalicular Osmolarity and Paracellular and Transcellular Water Flow. Journal of Pharmacology and Experimental Therapeutics. 2019. Vol. 371:713–717. [Cross Ref]

            37. Hatano R, Akiyama K, Tamura A, Hosogi S, Marunaka Y, Caplan MJ, et al.. Knockdown of Ezrin Causes Intrahepatic Cholestasis by the Dysregulation of Bile Fluidity in the Bile Duct Epithelium in Mice. Hepatology. 2015. Vol. 61:1660–1671. [Cross Ref]

            38. Trauner M, Fickert P, Halilbasic E, Moustafa T. Lessons from the Toxic Bile Concept for the Pathogenesis and Treatment of Cholestatic Liver Siseases. Wiener Medizinische Wochenschrift. 2008. Vol. 158:542–548. [Cross Ref]

            39. Wang S, Yue H, Derin RB, Guggino WB, Li M. Accessory Protein Facilitated CFTR-CFTR Interaction, A Molecular Mechanism to Potentiate the Chloride Channel Activity. Cell. 2000. Vol. 103:169–179. [Cross Ref]

            40. Thornell IM, Bevensee MO. Regulators of Slc4 Bicarbonate Transporter Activity. Frontiers in Physiology. 2015. Vol. 6:166. [Cross Ref]

            41. Banales JM, Prieto J, Medina JF. Cholangiocyte Anion Exchange and Biliary Bicarbonate Excretion. World Journal of Gastroenterology. 2006. Vol. 12:3496–3511. [Cross Ref]

            42. Herrmann U, Dockter G, Lammert F. Cystic Fibrosis-Associated Liver Disease. Best Practice and Research in Clinical Gastroenterology. 2010. Vol. 24:585–592. [Cross Ref]

            43. Salas JT, Banales JM, Sarvide S, Recalde S, Ferrer A, Uriarte I, et al.. Ae2a, b-deficient Mice Develop Antimitochondrial Antibodies and Other Features Resembling Primary Biliary Cirrhosis. Gastroenterology. 2008. Vol. 134:1482–1493. [Cross Ref]

            44. Wagner M, Zollner G, Trauner M. New Molecular Insights into the Mechanisms of Cholestasis. Journal of Hepatology. 2009. Vol. 51:565–580. [Cross Ref]

            45. Zollner G, Marschall HU, Wagner M, Trauner M. Role of Nuclear Receptors in the Adaptive Response to Bile Acids and Cholestasis: Pathogenetic and Therapeutic Considerations. Molecular Pharmaceutics. 2006. Vol. 3:231–251. [Cross Ref]

            46. Gonzalez-Sanchez E, Firrincieli D, Housset C, Chignard N. Nuclear Receptors in Acute and Chronic Cholestasis. Digestive Diseases. 2015. Vol. 33:357–366. [Cross Ref]

            47. Stofan M, Guo GL. Bile Acids and FXR: Novel Targets for Liver Diseases. Frontiers in Medicine (Lausanne). 2020. Vol. 7:544. [Cross Ref]

            48. Beuers U, Trauner M, Jansen P, Poupon R. New Paradigms in the Treatment of Hepatic Cholestasis: From UDCA to FXR, PXR and Beyond. Journal of Hepatology. 2015. Vol. 62 1 Suppl:S25–S37. [Cross Ref]

            49. Eloranta JJ, Kullak-Ublick GA. Coordinate Transcriptional Regulation of Bile Acid Homeostasis and Drug Metabolism. Archives of Biochemistry and Biophysics. 2005. Vol. 433:397–412. [Cross Ref]

            50. Ananthanarayanan M, Balasubramanian N, Makishima M, Mangelsdorf DJ, Suchy FJ. Human Bile Salt Export Pump Promoter is Transactivated by the Farnesoid X Receptor/Bile Acid Receptor. Journal of Biological Chemistry. 2001. Vol. 276:28857–28865. [Cross Ref]

            51. Kast HR, Goodwin B, Tarr PT, Jones SA, Anisfeld AM, Stoltz CM, et al.. Regulation of Multidrug Resistance-Associated Protein 2 (ABCC2) by the Nuclear Receptors Pregnane X Receptor, Farnesoid X-Activated Receptor, and Constitutive Androstane Receptor. Journal of Biological Chemistry. 2002. Vol. 277:2908–2915. [Cross Ref]

            52. Miyata M, Sakaida Y, Matsuzawa H, Yoshinari K, Yamazoe Y. Fibroblast Growth Factor 19 Treatment Ameliorates Disruption of Hepatic Lipid Metabolism in Farnesoid X Receptor (Fxr)-Null Mice. Biological and Pharmaceutical Bulletin. 2011. Vol. 34:1885–1889

            53. Zhou X, Cao L, Jiang C, Xie Y, Cheng X, Krausz KW, et al.. PPARalpha-UGT Axis Activation Represses Intestinal FXR-FGF15 Feedback Signalling and Exacerbates Experimental Colitis. Nature Communications. 2014. Vol. 5:4573[Cross Ref]

            54. Xu C, Huang M, Bi H. PXR- and CAR-Mediated Herbal Effect on Human Diseases. Biochimica et Biophysica Acta. 2016. Vol. 1859:1121–1129. [Cross Ref]

            55. Suh JI. [Role of PXR and CAR in Cholestasis]. Korean Journal of Hepatology. 2006. Vol. 12:5–15

            56. Staudinger JL, Goodwin B, Jones SA, Hawkins-Brown D, Mackenzie KI, Latour A, et al.. The Nuclear Receptor PXR is a Lithocholic Acid Sensor that Protects Against Liver Toxicity. Proceedings of the National Academy of Sciences of the United States of America. 2001. Vol. 98:3369–3374. [Cross Ref]

            57. Kakizaki S, Takizawa D, Tojima H, Horiguchi N, Yamazaki Y, Mori M. Nuclear Receptors CAR and PXR; Therapeutic Targets for Cholestatic Liver Disease. Frontiers in Bioscience (Landmark Ed). 2011. Vol. 16:2988–3005. [Cross Ref]

            58. Zollner G, Wagner M, Fickert P, Silbert D, Gumhold J, Zatloukal K, et al.. Expression of Bile Acid Synthesis and Detoxification Enzymes and the Alternative Bile Acid Efflux Pump MRP4 in Patients with Primary Biliary Cirrhosis. Liver International. 2007. Vol. 27:920–929. [Cross Ref]

            59. Castano G, Burgueno A, Fernandez GT, Pirola CJ, Sookoian S. The Influence of Common Gene Variants of the Xenobiotic Receptor (PXR) in Genetic Susceptibility to Intrahepatic Cholestasis of Pregnancy. Alimentary Pharmacology & Therapeutics. 2010. Vol. 31:583–592. [Cross Ref]

            60. Chai J, Luo D, Wu X, Wang H, He Y, Li Q, et al.. Changes of Organic Anion Transporter MRP4 and Related Nuclear Receptors in Human Obstructive Cholestasis. Journal of Gastrointestinal Surgery. 2011. Vol. 15:996–1004. [Cross Ref]

            61. Karpen SJ. Exercising the Nuclear Option to Treat Cholestasis: CAR and PXR Ligands. Hepatology. 2005. Vol. 42:266–269. [Cross Ref]

            62. Jung D, Mangelsdorf DJ, Meyer UA. Pregnane X Receptor is a Target of Farnesoid X Receptor. The Journal of Biological Chemistry. 2006. Vol. 281:19081–19091. [Cross Ref]

            63. Wolski H, Kurzawinska G, Ozarowski M, Drews K, Barlik M, Piatek K, et al.. FokI Vitamin D Receptor Polymorphism as a Protective Factor in Intrahepatic Cholestasis of Pregnancy. Ginekologia Polska. 2020. Vol. 1:719–725. [Cross Ref]

            64. Kempinska-Podhorecka A, Wunsch E, Jarowicz T, Raszeja-Wyszomirska J, Loniewska B, Kaczmarczyk M, et al.. Vitamin D Receptor Polymorphisms Predispose to Primary Biliary Cirrhosis and Severity of the Disease in Polish Population. Gastroenterology Research and Practice. 2012. Vol. 2012:408723. [Cross Ref]

            65. Firrincieli D, Zuniga S, Rey C, Wendum D, Lasnier E, Rainteau D, et al.. Vitamin D Nuclear Receptor Deficiency Promotes Cholestatic Liver Injury by Disruption of Biliary Epithelial cell Junctions in Mice. Hepatology. 2013. Vol. 58:1401–1412. [Cross Ref]

            66. Chen X, Chen F, Liu S, Glaeser H, Dawson PA, Hofmann AF, et al.. Transactivation of Rat Apical Sodium-dependent Bile Acid Transporter and Increased Bile Acid Transport by 1alpha, 25-dihydroxyvitamin D3 via the Vitamin D Receptor. Molecular Pharmacology. 2006. Vol. 69:1913–1923. [Cross Ref]

            67. Hochrath K, Stokes CS, Geisel J, Pollheimer MJ, Fickert P, Dooley S, et al.. Vitamin D Modulates Biliary Fibrosis in ABCB4-deficient Mice. Hepatology International. 2014. Vol. 8:443–452. [Cross Ref]

            68. Fayard E, Auwerx J, Schoonjans K. LRH-1: An Orphan Nuclear Receptor Involved in Development, Metabolism and Steroidogenesis. Trends in Cell Biology. 2004. Vol. 14:250–260. [Cross Ref]

            69. Out C, Hageman J, Bloks VW, Gerrits H, Sollewijn GM, Bos T, et al.. Liver Receptor Homolog-1 is Critical for Adequate Up-regulation of Cyp7a1 Gene Transcription and Bile Salt Synthesis During Bile Salt Sequestration. Hepatology. 2011. Vol. 53:2075–2085. [Cross Ref]

            70. Lee YK, Moore DD. Liver Receptor Homolog-1, an Emerging Metabolic Modulator. Frontiers in Bioscience. 2008. Vol. 13:5950–5958. [Cross Ref]

            71. Song X, Kaimal R, Yan B, Deng R. Liver Receptor Homolog 1 Transcriptionally Regulates Human Bile Salt Export Pump Expression. Journal of Lipid Research. 2008. Vol. 49:973–984. [Cross Ref]

            72. Bohan A, Chen WS, Denson LA, Held MA, Boyer JL. Tumor Necrosis Factor Alpha-dependent Up-regulation of Lrh-1 and Mrp3(Abcc3) Reduces Liver Injury in Obstructive Cholestasis. The Journal of Biological Chemistry. 2003. Vol. 278:36688–36698. [Cross Ref]

            73. Chen F, Ma L, Dawson PA, Sinal CJ, Sehayek E, Gonzalez FJ, et al.. Liver Receptor Homologue-1 Mediates Species- and Cell Line-specific Bile Acid-dependent Negative Feedback Regulation of the Apical Sodium-dependent Bile Acid Transporter. The Journal of Biological Chemistry. 2003. Vol. 278:19909–19916. [Cross Ref]

            74. Post SM, Duez H, Gervois PP, Staels B, Kuipers F, Princen HM. Fibrates Suppress Bile Acid Synthesis via Peroxisome Proliferator-activated Receptor-alpha-mediated Downregulation of Cholesterol 7alpha-hydroxylase and Sterol 27-hydroxylase Expression. Arteriosclerosis, Thrombosis, and Vascular Biology. 2001. Vol. 21:1840–1845. [Cross Ref]

            75. Ghonem NS, Ananthanarayanan M, Soroka CJ, Boyer JL. Peroxisome Proliferator-activated Receptor Alpha Activates Human Multidrug Resistance Transporter 3/ATP-binding Cassette Protein Subfamily B4 Transcription and Increases Rat Biliary Phosphatidylcholine Secretion. Hepatology. 2014. Vol. 59:1030–1042. [Cross Ref]

            76. Kok T, Bloks VW, Wolters H, Havinga R, Jansen PL, Staels B, et al.. Peroxisome Proliferator-activated Receptor Alpha (PPARalpha)-mediated Regulation of Multidrug Resistance 2 (Mdr2) Expression and Function in Mice. The Biochemical Journal. 2003. Vol. 369(Pt 3):539–547. [Cross Ref]

            77. Jung D, Fried M, Kullak-Ublick GA. Human Apical Sodium-dependent Bile Salt Transporter Gene (SLC10A2) is Regulated by the Peroxisome Proliferator-activated Receptor Alpha. The Journal of Biological Chemistry. 2002. Vol. 277:30559–30566. [Cross Ref]

            78. Honda A, Ikegami T, Nakamuta M, Miyazaki T, Iwamoto J, Hirayama T, et al.. Anticholestatic Effects of Bezafibrate in Patients with Primary Biliary Cirrhosis Treated with Ursodeoxycholic Acid. Hepatology. 2013. Vol. 57:1931–1941. [Cross Ref]

            79. Andermatten RB, Ciriaci N, Schuck VS, Di Siervi N, Razori MV, Miszczuk GS, et al.. Sphingosine 1-phosphate Receptor 2/adenylyl Cyclase/protein Kinase a Pathway is Involved in Taurolithocholate-induced Internalization of Abcc2 in Rats. Archives of Toxicology. 2019. Vol. 93:2279–2294. [Cross Ref]

            80. Boaglio AC, Zucchetti AE, Sanchez PE, Pellegrino JM, Ochoa JE, Mottino AD, et al.. Phosphoinositide 3-kinase/protein Kinase B Signaling Pathway is Involved in Estradiol 17beta-D-glucuronide-induced Cholestasis: Complementarity with Classical Protein Kinase C. Hepatology. 2010. Vol. 52:1465–1476. [Cross Ref]

            81. Manning BD, Toker A. AKT/PKB Signaling: Navigating the Network. Cell. 2017. Vol. 169:381–405. [Cross Ref]

            82. Webster CR, Anwer MS. Role of the PI3K/PKB Signaling Pathway in cAMP-mediated Translocation of Rat Liver Ntcp. The American Journal of Physiology. 1999. Vol. 277:G1165–G1172. [Cross Ref]

            83. Wu T, Zhang Q, Li J, Chen H, Wu J, Song H. Up-regulation of BSEP and MRP2 by Calculus Bovis Administration in 17alpha-ethynylestradiol-induced Cholestasis: Involvement of PI3K/Akt Signaling Pathway. Journal of Ethnopharmacology. 2016. Vol. 190:22–32. [Cross Ref]

            84. Beuers U, Denk GU, Soroka CJ, Wimmer R, Rust C, Paumgartner G, et al.. Taurolithocholic Acid Exerts Cholestatic Effects via Phosphatidylinositol 3-kinase-dependent Mechanisms in Perfused Rat Livers and Rat Hepatocyte Couplets. The Journal of Biological Chemistry. 2003. Vol. 278:17810–17818. [Cross Ref]

            85. Wang T, Liu C, Jia L. The Roles of PKCs in Regulating Autophagy. Journal of Cancer Research and Clinical Oncology. 2018. Vol. 144:2303–2311. [Cross Ref]

            86. Mcconkey M, Gillin H, Webster CR, Anwer MS. Cross-Talk Between Protein Kinases Czeta and B in Cyclic AMP-Mediated Sodium Taurocholate Co-Transporting Polypeptide Translocation in Hepatocytes. The Journal of Biological Chemistry. 2004. Vol. 279:20882–20888. [Cross Ref]

            87. Park SW, Schonhoff CM, Webster CR, Anwer MS. Protein Kinase Cdelta Differentially Regulates cAMP-Dependent Translocation of NTCP and MRP2 to the Plasma Membrane. American Journal of Physiology. Gastrointestinal and Liver Physiology. 2012. Vol. 303:G657–G665. [Cross Ref]

            88. Schonhoff CM, Gillin H, Webster CR, Anwer MS. Protein Kinase Cdelta Mediates Cyclic Adenosine Monophosphate-Stimulated Translocation of Sodium Taurocholate Cotransporting Polypeptide and Multidrug Resistant Associated Protein 2 in Rat Hepatocytes. Hepatology. 2008. Vol. 47:1309–1316. [Cross Ref]

            89. Schonhoff CM, Yamazaki A, Hohenester S, Webster CR, Bouscarel B, Anwer MS. PKC{epsilon}-Dependent and -Independent Effects of Taurolithocholate on PI3K/PKB Pathway and Taurocholate Uptake in HuH-NTCP Cell Line. American Journal of Physiology. Gastrointestinal and Liver Physiology. 2009. Vol. 297:G1259–G1267. [Cross Ref]

            90. Muhlfeld S, Domanova O, Berlage T, Stross C, Helmer A, Keitel V, et al.. Short-Term Feedback Regulation of Bile Salt Uptake by Bile Salts in Rodent Liver. Hepatology. 2012. Vol. 56:2387–2397. [Cross Ref]

            91. Sekine S, Ito K, Horie T. Oxidative Stress and Mrp2 Internalization. Free Radical Biology & Medicine. 2006. Vol. 40:2166–2174. [Cross Ref]

            92. Grune S, Engelking LR, Anwer MS. Role of Intracellular Calcium and Protein Kinases in the Activation of Hepatic Na+/taurocholate Cotransport by Cyclic AMP. The Journal of Biological Chemistry. 1993. Vol. 268:17734–17741

            93. Chandra P, Zhang P, Brouwer KL. Short-Term Regulation of Multidrug Resistance-Associated Protein 3 in Rat and Human Hepatocytes. American Journal of Physiology. Gastrointestinal and Liver Physiology. 2005. Vol. 288:G1252–G1258. [Cross Ref]

            94. Guo GL, Klaassen CD. Protein Kinase C Suppresses Rat Organic Anion Transporting Polypeptide 1- and 2-Mediated Uptake. The Journal of Pharmacology and Experimental Therapeutics. 2001. Vol. 299:551–557

            95. Barosso IR, Zucchetti AE, Boaglio AC, Larocca MC, Taborda DR, Luquita MG, et al.. Sequential Activation of Classic PKC and Estrogen Receptor Alpha is Involved in Estradiol 17ss-D-Glucuronide-Induced Cholestasis. PLoS One. 2012. Vol. 7:e50711. [Cross Ref]

            96. Kubitz R, Saha N, Kuhlkamp T, Dutta S, Vom DS, Wettstein M, et al.. Ca2+-Dependent Protein Kinase C Isoforms Induce Cholestasis in Rat Liver. The Journal of Biological Chemistry. 2004. Vol. 279:10323–10330. [Cross Ref]

            97. Wimmer R, Hohenester S, Pusl T, Denk GU, Rust C, Beuers U. Tauroursodeoxycholic Acid Exerts Anticholestatic Effects by a Cooperative cPKC Alpha-/PKA-Dependent Mechanism in Rat Liver. Gut. 2008. Vol. 57:1448–1454. [Cross Ref]

            98. Gliki G, Wheeler-Jones C, Zachary I. Vascular Endothelial Growth Factor Induces Protein Kinase C (PKC)-Dependent Akt/PKB Activation and Phosphatidylinositol 3’-Kinase-Mediates PKC Delta Phosphorylation: Role of PKC in Angiogenesis. Cell Biology International. 2002. Vol. 26:751–759. [Cross Ref]

            99. Reyland ME. Protein Kinase C Isoforms: Multi-functional Regulators of Cell Life and Death. Frontiers in Bioscience (Landmark Edition). 2009. Vol. 14:2386–2399. [Cross Ref]

            100. Webster CR, Johnston AN, Anwer MS. Protein Kinase Cdelta Protects Against Bile Acid Apoptosis by Suppressing Proapoptotic JNK and BIM Pathways in Human and Rat Hepatocytes. American Journal of Physiology. Gastrointestinal Liver Physiology. 2014. Vol. 307:G1207–G1215. [Cross Ref]

            101. Schonhoff CM, Webster CR, Anwer MS. Taurolithocholate-Induced MRP2 Retrieval Involves MARCKS Phosphorylation by Protein Kinase C in HUH-NTCP Cells. Hepatology. 2013. Vol. 58:284–292. [Cross Ref]

            102. Beuers U, Bilzer M, Chittattu A, Kullak-Ublick GA, Keppler D, Paumgartner G, et al.. Tauroursodeoxycholic Acid Inserts the Apical Conjugate Export Pump, Mrp2, into Canalicular Membranes and Stimulates Organic Anion Secretion by Protein Kinase C-Dependent Mechanisms in Cholestatic Rat Liver. Hepatology. 2001. Vol. 33:1206–1216. [Cross Ref]

            103. Scholmerich J, Baumgartner U, Miyai K, Gerok W. Tauroursodeoxycholate Prevents Taurolithocholate-Induced Cholestasis and Toxicity in Rat Liver. Journal of Hepatology. 1990. Vol. 10:280–283. [Cross Ref]

            104. Murray JW, Thosani AJ, Wang P, Wolkoff AW. Heterogeneous Accumulation of Fluorescent Bile Acids in Primary Rat Hepatocytes does not Correlate with their Homogenous Expression of ntcp. American Journal of Physiology. Gastrointestinal and Liver Physiology. 2011. Vol. 301:G60–G68. [Cross Ref]

            105. Stross C, Keitel V, Winands E, Haussinger D, Kubitz R. Expression and Localization of Atypical PKC Isoforms in Liver Parenchymal Cells. Biological Chemistry. 2009. Vol. 390:235–244. [Cross Ref]

            Author and article information

            Journal
            amm
            Acta Materia Medica
            Compuscript (Ireland )
            2737-7946
            06 October 2022
            : 1
            : 3
            : 381-391
            Affiliations
            [a ]Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
            Author notes
            *Correspondence: ld2069@ 123456outlook.com (Dong Liu); clzhang@ 123456tjh.tjmu.edu.cn (Chengliang Zhang)

            1These author contributed equally.

            Article
            10.15212/AMM-2022-0029
            01886b60-3df5-4959-ae51-65f9c3574207
            Copyright © 2022 The Authors.

            Creative Commons Attribution 4.0 International License

            History
            : 11 August 2022
            : 12 September 2022
            : 21 September 2022
            Page count
            Figures: 3, Tables: 1, References: 105, Pages: 11
            Categories
            Review Article

            Toxicology,Pathology,Biochemistry,Clinical chemistry,Pharmaceutical chemistry,Pharmacology & Pharmaceutical medicine
            regulation,localization,transport proteins,intrahepatic cholestasis

            Comments

            Comment on this article