Open Access

Liver sinusoidal endothelial cells as potential drivers of liver fibrosis (Review)

  • Authors:
    • Jiaqin Gao
    • Bin Zuo
    • Yang He
  • View Affiliations

  • Published online on: January 16, 2024     https://doi.org/10.3892/mmr.2024.13164
  • Article Number: 40
  • Copyright: © Gao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Liver fibrosis due to viral or metabolic chronic liver diseases is a major challenge of global health. It is a critical pre‑stage condition of severe hepatopathy, characterized by excessive accumulation of extracellular matrix components and ongoing chronic inflammation. To date, early prevention of liver fibrosis remains challenging. As the most abundant non‑parenchymal hepatic cell population, liver sinusoidal endothelial cells (LSECs) are stabilizers that maintain the intrahepatic environment. Notably, LSECs dysfunction appears to be implicated in the progression of liver fibrosis via numerous mechanisms. Following sustained liver injury, they lose their fenestrae (cytoplasmic pores) and change their crosstalk with other cellular interactions in the hepatic blood environment. LSEC‑targeted therapy has shown promising effects on fibrosis resolution, opening up new opportunities for anti‑fibrotic therapy. In light of this, the present study summarized changes in LSECs during liver fibrosis and their interactions with hepatic milieu, as well as possible therapeutic approaches that specially target LSECs.

Introduction

As it has long been linked to severe morbidity and mortality, liver fibrosis is one of the most significant contributors to the worldwide disease burden (1). The liver, the most important detoxification organ, maintains a stable metabolic balance within the body. After exposure to toxic metabolites, viruses and alcohol that interfere with the normal physiological functions, the liver develops drug-induced liver injury, viral hepatitis and alcoholic liver disease (2), eventually progressing to fibrosis. In addition, nonalcoholic fatty liver disease (NAFLD) is a continuum of liver diseases, starting from simple steatosis (NAFL) and gradually progressing to nonalcoholic steatohepatitis (NASH), which can progress to liver fibrosis and cirrhosis. As NAFLD is a disease caused by excessive fat accumulation in the liver closely related to metabolic dysfunction, which excludes alcohol and other diseases, a recent consensus among international experts has proposed NAFLD as a change of nomenclature for metabolic dysfunction-associated fatty liver disease (3). Fibrosis is a risk factor for cirrhosis and hepatocellular carcinoma (4). In addition, liver fibrosis can result in chronic portal hypertension, a common clinical complication (5).

Liver fibrosis is characterized by excessive deposition of extracellular matrix (ECM) and its reduced degradation. The activation of hepatic stellate cells (HSCs) is a well-known event in liver fibrosis. HSCs are primarily activated by stimulated Kupffer cells (KCs) and liver sinusoidal endothelial cells (LSECs) via paracrine production of pro-inflammatory and pro-fibrotic cytokines such as TGF-β, connective tissue growth factor and platelet-derived growth factor (PDGF) and then transform into resilient myofibroblast which produces ECM to form fibrous scars around the damaged area (6). If liver injury is acute or self-limiting, the liver returns to normal due to the specific destruction of collagen and ECM by matrix metalloproteinases (MMPs). Otherwise, persistent injury can cause chronic inflammation and ECM deposition, eventually leading to replacement of parenchymal cells by scar tissues and ultimately cirrhosis. How to balance the synthesis and degradation of ECM is still challenging. Although regression of liver fibrosis in patients with chronic liver disease has been recognized for decades, there are still no FDA-approved anti-fibrosis drugs. Although the exact definition of fibrotic regression has not been properly established (7), the success of antiviral therapy has given us hope to elucidate the cellular and molecular mechanisms and develop therapeutic agents that mimic the endogenous capabilities of the liver (8).

Dysfunction of LSECs allows initiation and progression of liver fibrosis and is responsible for its clinical complication, portal hypertension. LSECs create a bridge between hepatocytes and hepatic sinusoids, acting as protective gatekeepers of the microvascular environment (9). Although HSC activation is the primary event in liver fibrosis, it is still important and necessary to seek other new directions to reverse the condition.

The present review summarized the structural and functional alterations of LSECs and their crosstalk with hepatic microenvironment during liver fibrosis and highlighted the role of LSECs in portal hypertension. Several novel therapeutic strategies targeting LSECs are also reviewed.

Physiological characteristics of LSECs

Compared to other endothelial cells, LSECs are morphologically and functionally specialized. LSECs are the most abundant non-parenchymal cells in the liver and form the sinusoidal wall (10). LSECs represent a gatekeeper between hepatocytes and hepatic sinusoids during liver injury. They are highly specialized as nondiaphragmatic fenestrae (cytoplasmic pores) and lack a basal membrane, facilitating high-efficient material exchange from the bloodstream into the space of Disse and regulate hepatic stellate cell activation status (11). The fenestrae are 100–150 nm in size and are organized in clusters termed sieve plates (12). Their size and number varies across liver lobules with larger but fewer fenestrae per sieve plate in the periportal region and smaller but more numerous fenestrae per sieve plate in the centrilobular region (13,14), which may be related to the need for oxygen exchange along the lobules. Notably, heterogeneities between periportal and centrilobular LSECs exist in morphology (15), antigen expression (16) and lectin affinity (17). Di Martino et al (18) established an approach of stimulated emission depletion (STED) microscopy to visualize and analyze LSEC fenestrae and revealed that cytoskeleton, especially actin, is closely associated with fenestrae formation. Herrnberger et al (19) found that plasmalemma vesicle-associated protein (PLVAP) is required for the formation of endothelial diaphragms and endothelial fenestrae and opening fenestrae in LSECs are critical for the passage of lipoproteins.

In addition to their unique structure, LSECs contain various receptors to perform multiple functions. A total of three endocytosis receptors have been identified on LSECs, collagen-α-chain/mannose receptor, hyaluronan/scavenger receptor and FcγIIb2 receptor (20). Among them, mannose receptor is the main candidate for endocytosis of denatured collagen on LSECs and functions as MMPs, making it a promising target in the study of hepatic fibrosis (21). Through the combined effect of endocytic vesicles and receptor-mediated endocytosis, LSECs can eliminate antigens, cell debris and immune complexes. SR-H/stabillin-1 and SR-H/stabillin2, the main scavenger receptors of LSECs, mediate endocytosis of polyanionic molecules, including oxidized low-density lipoproteins, hyaluronan, chondroitin sulfate, formaldehyde treated serum albumin, procollagen type I and III N-terminal peptides and advanced glycation end products (22). FcγIIb2 receptor on LSECs mediates vascular immunity mainly through cleaning circulating IgG immune complexes (23). Under homeostatic conditions, LSECs are not merely endocytic fenestrated endothelial cells, they also exhibit vasodilatory, anti-inflammatory, anti-thrombotic and anti-fibrotic phenotypes, which are important for orchestrating liver microenvironment response and regulating intrahepatic vascular tone and immune cell function (11).

Changes of LSECs in liver fibrosis

Capillarization

Maintenance of the highly specialized phenotype of LSECs is crucial for liver homeostasis. During liver fibrosis and cirrhosis, loss of LSEC phenotype and functions amplify liver damage by undergoing a process called ‘capillarization’, which is clarified by the loss of fenestrae (cytoplasmic pores) and the manifestation of a basement membrane, leading to LSEC dysfunction (20).

Su et al (24) mapped the spatial distribution of heterogeneous liver endothelial cells in normal and cirrhotic mouse liver and identified zone-specific transcriptomic changes of LSECs associated with liver cirrhosis using small conditional RNA-seq technology, finding that zone 3 LSECs are more susceptible to capillarization and that CD34 is more useful than conventional CD31 as a marker of capillarization in LSECs. In 2008, Deleve et al (25) employed cell co-culture to demonstrate that capillarized LSECs are unable to maintain HSC quiescence due to loss of VEGF-stimulated release of nitric oxide (NO), while differentiated LSECs can prevent HSC activation and promote reversion of activated HSCs to quiescent HSCs. This study highlighted the critical role of LSECs in regulating HSC status. In addition, capillarization of LSECs occurs in the early stage of NASH. An in vivo study showed that loss of the unique fenestrated phenotype is an initial step for fibrogenesis, preceding the activation of HSCs and onset of fibrosis in alcoholic liver injury (26), suggesting that signals from LSECs may be one of the earliest triggers of HSC activation.

Regulation of the LSEC phenotype

The exact mechanisms regulating the loss of fenestrae have not been fully elucidated, but several molecules and pathways have been identified. Maintenance of the fenestrated LSEC phenotype requires VEGF, which is derived from hepatocytes and HSCs. VEGF is a key regulator for the LSEC phenotype (27). It has been established from various in vitro and in vivo studies that VEGF operates in two pathways, the NO-dependent and NO-independent (28). In the NO-dependent pathway, endothelial nitric oxide synthase (eNOS)-NO-cyclic guanosine monophosphate signaling is critical for regulating the formation of fenestrae in LSECs (29). Endothelial Notch activation leads to LSEC dedifferentiation and promotes liver fibrogenesis through eNOS-soluble guanylate cyclase (sGC) signaling. Regarding Notch ligands, Delta-like ligand 4 (DLL4) is predominantly expressed in endothelial cells under hypoxic conditions during liver fibrosis (30). It is reported that DLL4 overexpression accelerated defenestration of LSECs and production of basement membrane in both human fibrotic livers and carbon tetrachloride (CCl4)-induced mouse livers (31). In addition to Notch signaling, the Hedgehog (Hh) pathway also induces LSEC capillarization and formation of vascular tubes in vitro by Hh ligands binding to their transmembrane receptors [Patched (Ptc)] to activate Smoothened (32). Besides, miR-511-3p was found to positively regulate Hh signaling by targeting Ptch1 in hepatic sinusoidal obstruction syndrome (33). Long noncoding RNA (lncRNA) Airn was identified to interact with enhancer of zeste homolog 2 to maintain LSEC differentiation through Krüppel-like factor 2 (KLF2)-eNOS-sGC pathway, thus indirectly inhibiting HSC activation (34). Studies have found that several transcription factors in liver endothelial cells also play key roles in regulating the characteristics and function of LSECs. Genetic endothelial GATA4 deletion leads to liver fibrosis and hepatopathy by GATA4-MYC-PDGFB axis (35). In 2015, transcriptional factor KLF2 was found to upregulate eNOS expression, which is essential for maintaining a functional endothelial phenotype and also explains the molecular mechanisms of statins (36). In 2022, de Haan et al (37) found that LSEC-enriched zinc-finger E-box-binding homeobox2 (Zeb2) can preserve the liver angioarchitecture and prevent liver fibrosis.

On the other hand, capillarization is thought to be the repair of damaged LSECs by bone marrow endothelial progenitors that engraft but fail to fully mature through cell autonomous pathways that inhibit the NO-dependent pathway. Maretti-Mira et al (38) identified heparin-binding EGF-like growth factor (HB-EGF) as a signal to maintain HSCs quiescence while the immature LSECs from bone marrow were unable to shed HB-EGF from the cytosolic membrane. These data demonstrate that capillarization is an early event in the fibrotic process and is a perfect target for the treatment of liver fibrosis. The number of fenestrae was shown to be significantly decreased in LSECs from BMP9-knockout mice and addition of BMP9 to the LSECs culture recovers fenestrae and elevates the expression of GATA4 and PLVAP, suggesting that BMP9 is a key paracrine regulator of LSEC fenestration (39). Conversely, Haristi et al found that high BMP9 in combination with LPS stimulation induced the expression of certain capillarization markers in LSEC (40). Desroches-Castan et al (41) found that the role of BMP9 in LSEC differentiation depends on genetic background of C57BL/6, BALB/c and 129/Ola mice. Further studies may warrant the exact roles of BMP9 in LSEC capillarization.

In addition, cell-specific junctional adhesion molecule A (JAM-A) is reported to exert crucial functions in hepatic fibrogenesis (10). Loss of endothelial JAM-A induces LSEC capillarization and HSC activation by triggering Hh signaling and loss of VEGFR1/2 with no alterations in myeloid recruitment (10). Elevated portal blood lipopolysaccharide (LPS) due to leaky gut induces LSEC capillarization in mice and in vitro, while antimicrobial treatment lowers portal blood LPS concentration and inhibits LSEC capillarization (42). Overall, the signaling pathways that regulate LSEC fenestration varies under physiological and pathological conditions and the mechanisms involved by various factors are summarized in Table I.

Table I.

Involved regulators for the LSEC phenotype.

Table I.

Involved regulators for the LSEC phenotype.

TypeMoleculeMechanism(s)(Refs.)
Fenestration factorsVEGFeNOS-NO-cGMP signaling pathway(29)
KLF2Upregulates eNOS expression(36)
lncRNA Airnthrough KLF2-eNOS-sGC pathway and interacts with EZH2(34)
GATA4Prevents a pathogenic switch in angiocrine signaling(35)
Zeb2Preserves liver angioarchitecture and protects against liver fibrosis(37)
BMP9Promotes GATA4 and PLVAP expression(39)
HB-EGFInterferes in the nitric oxide pathway(38)
Defenestration factorsNotch signalingBinds to DLL4 and induces LSEC dedifferentiation through eNOS-sGC signaling(30,31)
Hh signalingInduces formation of vascular tubes by binding to Ptc and activating Smo(32)
miR-511-3pActivates Hh signaling via targeting Ptch1(33)
endothelial JAM-ATriggers Hh signaling and loss of VEGFR1/2(10)

[i] eNOS, endothelial nitric oxide synthase; NO, nitric oxide; cGMP, cyclic guanosine monophosphate; KLF2, Krüppel-like factor 2; lncRNA, long noncoding RNA; sGC, soluble guanylate cyclase; EZH2, enhancer of zeste homolog 2; Zeb2, zinc-finger E-box-binding homeobox2; PLVAP, plasmalemma vesicle-associated protein; HB-EGF, heparin-binding EGF-like growth factor; DLL4, Delta-like ligand 4; LSEC, liver sinusoidal endothelial cell; Hh, Hedgehog; Ptc, Patched; Smo, Smoothened; miR, microRNA; JAM-A, junctional adhesion molecule A; Ptch1, Patched homolog 1.

Changes in crosstalk of LSECs in hepatic microenvironment

Increasing evidence has indicated that fibroblast-like cells in liver fibrosis are mainly derived from activated hepatic stellate cells (aHSCs). Therefore, HSC activation is considered a pivotal event leading to fibrosis. HSC activation is fine-tuned by the angiocrine signaling from LSECs and is also tightly modulated by dynamic interactions between LSECs and other cell types in the liver. Understanding these crosstalk is expected to improve liver disease therapy (Fig. 1).

LSECs and HSCs

HSCs are the main source of ECM and thus have been the central to the development of liver fibrosis. The interaction between LSECs and HSCs forms a positive feedback loop. The starting point of the loop is VEGF. As described above, VEGF from hepatocytes and HSCs maintains the phenotype of fenestrated LSECs and, in turn, NO from VEGF-stimulated LSECs prevents HSC activation. Therefore, VEGF may have a dual role in fibrosis. It induces fibrosis-associated angiogenesis while recruiting monocytes mainly through the CXC chemokine (CXCL)9-MMP13 axis to control the secretion of multiple MMPs and tissue inhibitor of MMPs during fibrosis resolution (43). Therefore, as a key mediator of angiocrine signaling, the regulatory role of VEGF in liver diseases remains to be elucidated. Endothelin-1 from HSCs also plays a key role in the regulation of eNOS activation in LSECs (44). In addition, capillarized LSECs activate HSCs through secreting PDGF, TGF-β and lowering a transcription factor KLF2, which acts as a vasoprotective molecule of the liver endothelium, subsequently activating HSCs (45). Activated HSCs further act on quiescent HSCs and LSECs by autocrine TGF-β1 (46). Thrombospondin-1 can be induced by HSC activation, enhancing LSEC capillarization by blocking the NO-dependent pathway (47). In addition, LSECs stimulate HSC migration by producing CXCL12/stromal-derived factor-1. CXCL12 binds to CXC chemokine receptor (CXCR)4 on HSCs, thereby inducing HSC proliferation and collagen production perpetuating fibrosis, while CXCR7-expressing LSECs activated by CXCL12 initiate liver regeneration (48). Enhancement of intracellular adipocyte fatty acid-binding protein (A-FABP) potentiates the LSEC capillarization by inducing Hh signaling, leading to impairment of the gatekeeper function of LSECs on HSC activation. On the other hand, LSEC derived A-FABP releases and acts on HSCs in a paracrine manner to potentiate the transactivation of TGF-β1 by activating JNK/c-Jun signaling. Elevated TGF-β1 subsequently promotes the expression of ECM in both paracrine and autocrine manners. These findings define that A-FABP is a novel therapeutic target of liver fibrosis (49). The contribution of exosomes and their cargoes has been proposed as a new model of intercellular communication between LSECs and HSCs. LSEC-derived sphingosine kinase 1-containing exosomes regulate HSC signaling and migration through fibronectin-integrin-dependent exosome adherence and dynamin-dependent exosome internalization (50).

LSECs and hepatocytes

Loss of fenestration of LSECS protects the liver from ongoing damage by limiting toxins to specific areas (19). However, the formation of a continuous basement membrane lining the sinusoids disrupts the bidirectional exchange of oxygen and nutrients between hepatocytes and sinusoids (51), as well as the lipoprotein secretion of hepatocytes and the de novo lipogenesis in hepatocytes which aggravates hepatic steatosis in NASH (52). LSECs can also secrete Wnt and hepatocyte growth factor (HGF), which together act as key hepatocyte mitogens with a potent liver regenerative capacity that is readily switched to a pro-fibrotic phenotype, in which the Erk1/2/Akt axis in LSECs acts as a switch (53). Growth factors from LSECs, such as bone morphogenetic protein (BMP)2, BMP6 and TGFβ1 act on hepatocytes and HSCs to control systemic iron homeostasis and fibrotic processes in a paracrine manner, respectively (54). Notch activation can alter the angiocrine profile of LSECs to compromise hepatocyte proliferation and liver regeneration through downregulating critical hepatocyte mitogens, including Wnt2a, Wnt9b and HGF (31). A recent study found that lncRNA Airn, a nuclear localized and highly unstable in the form of non-splicing 108 kb ncRNA from LSECs, promoted hepatocyte proliferation by increasing paracrine secretion of Wnt2a and HGF from LSECs (34). lncRNA Airn is inherent and might be a serum biomarker for liver fibrogenesis. Indeed, after acute injury, endothelial cells determine if the liver undergoes regeneration or fibrosis by angiocrine signaling (55). LSECs and hepatocytes also communicate with each other through the VEGF-A/VEGFR-2 signaling which is regarding to angiogenesis. Yan et al (56) investigated the biological roles of cluster of differentiation (CD)147 in a CCl4-induced liver fibrosis mouse model and found that CD147 promoted liver fibrosis progression via VEGF-A/VRGFR-2 signaling, mediating crosstalk between hepatocytes and LSECs. A combination of leukocyte derived chemokine2 expressed by hepatocytes and Tie1 produced by LSECs can promote LSECs capillarization and reduce portal angiogenesis (57).

LSECs and macrophages

Hepatic macrophages are important to the pathogenesis of chronic liver injury. Activated KCs can secrete a wide variety of proinflammatory activating HSCs. KCs and infiltrating macrophages can usually be divided into two subtypes, including M1 macrophages, which produce proinflammatory cytokines such as TNF-α, IL-1b, CCL2 and CCL5, and M2 macrophages which secrete a distinct set of mediators including IL-13, IL-10, IL-4 and TGF-b (58,59). Imbalance of M1/M2 ratio may be the key to the progression of NASH to liver fibrosis (52). Pro-inflammatory KCs may also produce cytokines and chemokines which increase the expression of adhesion molecules by LSECs, leading to leukocyte infiltration and activation (60). In a murine model of acute liver injury caused by overdose of acetaminophen, You et al (61) found that liver resident macrophages and infiltrating macrophages express an array of angiogenic factors and induce LSEC proliferation and migration to repair liver blood vessels, while in NASH, the capillarization of LSECs is necessary for activating KCs in that LSECs injury appeared during the simple steatosis phase and preceded the appearance of activate KCs and HSCs (62). Ford et al (63) investigated the effects of KCs on human liver sinusoidal endothelial cells (hLSECs) phenotype on soft materials, which indicate that TNF-α secreted by KCs undermines the effects of a soft matrix that is representative of healthy tissues and leads to loss of LSEC fenestrae. The relationship between LSECs and macrophages in liver fibrosis needs further investigation.

LSECs and lymphocytes

LSECs belong to nonprofessional antigen presenting cells, plasmacytoid DCs and are primary immunologic mediators underlying hepatic tolerance. They express major histocompatibility complex (MHC) class I, MHC class II and the co-stimulatory molecules CD80, CD86 and CD40 (11). The LSECs mediate hepatic immune tolerance toward self or foreign antigens by expression of anti-inflammatory mediators under physiological conditions, but they gain pro-inflammatory functions upon viral infection (64). LSECs release cytokines via toll-like receptors and contribute to viral infection of the liver, such as hepatitis B or hepatitis C virus (65). The physiologic consequences of higher antigen-presenting cell-related immunogenicity is associated with elevated intrahepatic inflammatory milieu in fibrosis. These LSECs gain enhanced capacity to capture antigens and induce the immunogenic T cell to sensitize endogenous cytotoxic T lymphocytes (66). During liver fibrosis, Th1 and Th2 cells recruit and adhere to liver sinusoids using α4β1-integrin and vascular adhesion protein-1 (VAP-1), respectively (67). However, the specific mechanisms by which T cells and LSECs crosstalk regulate sinusoidal capillarization remain largely unknown. Emerging evidences suggested B-cell activation is involved in NAFLD (68), while crosstalk between LSECs and B-cell remains to be elucidated. In addition, chemokine receptor CXCR6 on LSECs and its ligand CXCL16 on natural killer T cells (NKTs) control NKT cell migration (69).

LSECs and leucocytes

Inflammation is an essential part involved in the process of liver fibrosis. Hepatitis and NAFLD, especially NASH, have recently received increasing attention (70). LSECs interact with portal circulation via expression of surface receptors and release of paracrine factors, such as intercellular adhesion molecule, vascular cell adhesion molecule-1, VAP-1, chemokines and cytokines (71), recruiting the circulating leucocytes to enter the liver parenchyma. Exploring these inflammatory mediators may help to discover new therapeutic targets for the treatment of liver fibrosis. Gola et al (72) considered LSECs as a microbiome sensor, sustaining commensal-induced MyD88-dependent signaling to regulate the composition of the peri-cellular matrix involved in chemokine gradient formation. In vitro and in vivo findings demonstrate that specific focal adhesion proteins recruited by phosphofructokinase can parlay LSEC mechanotransduction into stiffness-induced angiocrine signaling and mediate neutrophil infiltration and promote liver fibrosis (73). By contrast, Brozat et al found that LSEC-derived JAM-A had no effect on cell migration from sinusoids or adhesion to LSECs (10), suggesting a complex role of leukocyte-endothelial cell interactions in promoting liver fibrosis.

LSECs and platelets

At present, an increasing number of individuals realize that the platelets are no longer bystanders in liver diseases and anti-platelet therapy has been proven effective in treating chronic liver injury (74,75). In vitro studies show that platelet adhesion is partly mediated by integrin (glycoprotein IIb/IIIa and αvβ3) with human hepatic sinusoidal endothelial cells (76), leading to platelet and endothelial activation and leukocyte recruitment. On the other hand, in wild-type mice after chronic CCl4 challenge plasma Von Willebrand factor levels are increased (77), which could be associated with the interaction between platelets and LSECs. However, it is not clear whether platelet adhesion is associated with capillarization. Platelets have well appreciated roles in physiological and pathological processes including inflammation, tissue repair, angiogenesis and tumor growth (78). A few studies have focused on the release of platelet granule content or platelet RNA transfer to activated HSCs (79,80), but the influence of platelets on LSECs needs more research. Although platelets have been shown to simulate proliferation of cultured hepatocytes (81), this is a long way to elucidating the role of modulators of platelets in chronic liver diseases.

LSECs and ECM

While most researchers focused on intercellular crosstalk of LSECs, Brougham-Cook et al (82) redirected attention to identify the unique phenotypes that LSECs exhibit due to ECM composition, stiffness and soluble factor alterations in fibrotic models by a high-throughput cellular microarray. LSECs play a crucial role in hepatic fibrogenesis by participating in ECM deposition in the form of collagen and fibronectin synthesis and regulating ECM metabolism. Extra domain A from injured LSECs promotes stellate cell motility and parenchymal liver fibrosis (83). Liu et al (84) used the principles of mechanical mechanics to explain capillarization of LSECs, which might contribute to early-stage collagen contraction, then collagen fibers function as mechanical transducer to activate HSCs. Notably, researchers found that capillarized LSECs undergo a partial endothelial-mesenchymal transition characterized by increased ECM production without activating cell mobility, leading to perisinusoidal ECM deposition preferentially in liver sinusoids but not septal/portal scars (85) (Fig. 1)

Dysfunction of LSECs and portal hypertension

Portal hypertension, defined as increased pressure in the portal vein, develops as a consequence of increased intrahepatic vascular resistance and is the initial step towards complications of chronic liver disease (86). It can cause ascites and gastroesophageal varices, as well as hepatic encephalopathy due to portosystemic shunting, hepatorenal syndrome and hypersplenism (87). To date, a number of researchers have considered that the effect of increased intrahepatic vascular resistance in liver fibrosis is closely related to LSECs, ultimately leading to portal hypertension and its complications (8890). Decreased NO levels and capillarization can increase vascular resistance (91). In addition, endothelial autophagy (92) and aging (93) are also involved in the occurrence of portal hypertension and liver fibrosis.

Recently, an increasing number of studies have suggested that thrombosis is one of the key pathological factors mediating portal hypertension and anticoagulation therapy seems to be beneficial in the treatment of liver fibrosis and portal hypertension in rats, although it depends on the stage of cirrhosis (86,94). The current consensus is that most portosystemic collateral vessels are formed through angiogenesis, the formation of new blood vessels from pre-existing vasculature especially within fibrous scar (95). In addition, mechanical stretch increases expression of CXCL1 in LSECs to recruit neutrophils, generate sinusoidal microthrombi and promote portal hypertension (96). Therefore, reversing LSEC dysregulation maybe an attractive strategy for portal hypertension.

LSEC targeting: Potential for therapy

It is well-known that statins have beneficial effects on dysfunctional sinusoidal cells by selectively NO bioavailability and inhibiting RhoA/Rho-kinase in the liver. This was confirmed in portal hypertension of rats with NASH (88). In addition, simvastatin administration for 15 days in aged cirrhotic rats improves the hepatic sinusoidal milieu, demonstrating its therapeutic potential in advanced chronic liver disease (93). Simvastatin restores the quiescence of aHSCs via stimulation of KLF2-NO signaling in LSECs (97). Treatment with the pan-peroxisome proliferator-activated receptor (pan-PPAR) agonist lanifibranor demonstrates phenotypic improvement in a rat model of cBDL as well as in human hepatocytes from patients with cirrhosis (98). Although further validation is needed in human trials, it paves a way to develop novel drugs that target both HSCs and LSECs to treat liver fibrosis.

The property of containing numerous receptors to endocytose soluble macromolecules and small particles (20) makes LSECs suitable for specific drug delivery. To date, several modified polymers targeting LSECs have been developed to delivery drugs. Hide et al (99) designed simvastatin-loaded polymeric micelles to treat chronic liver disease and showed promising results, thus suggesting that nanoparticles are a promising therapeutic approach. On the other hand, liver sinusoidal capillarization and ECM deposition are dual pathological barriers to drug delivery (20), Zhang et al (100) constructed an efficient nanodrug delivery system with LSEC-targeting and fenestrae-repairing nanoparticles (named HA-NPs/SMV) on the basis of the modification with hyaluronic acid.

Based on a specific route of drug delivery targeting LSECs, strategies that reverse endothelial dysfunction by inhibiting Notch signaling (30,101) and Hh signaling (102,103), downregulating VEGF-R2 (104), or suppressing KLF5-mediated LSEC angiogenesis (105), inhibiting hypoxia-inducible factor-1α (HIF-1α) (106) or enhancing endothelial barrier function through the cyclic adenosine monophosphate/Rac/cortactin pathway (107), can be applied to improve liver fibrosis. Besides special targeting of LSECs, there are other methods of liver fibrosis treatment that act via an indirect effect on LSECs, such as simultaneous consumption of LSECs and aHSCs (108), artificial control of the switch of the Erk1/2-Akt axis in LSECs to reverse LSECs from a proregenerative to a profibrotic phenotype (53), prevention of interstellar interaction of LSECs (109,110), selective enhancement of autophagy in LSEC in the early stages (111) and treatment of liver fibrosis with anti-angiogenic agents (112). Finally, a more detailed understanding of cytoskeletal structure and function of LSECs may help with the design of drugs that control fenestrae opening, which seems to be a simple and straightforward anti-liver fibrosis approach.

Conclusion and future perspectives

The present study reviewed the capillarization of LSECs and involved mechanisms and sinusoidal communication mediated by LSECs in liver fibrosis. LSECs play a central role in the maintenance of HSC quiescence and modulating the crosstalk among various types of liver cells and dysfunction of LSEC characterized by loss of fenestration is widely considered to be an early event in the development of liver fibrosis. In recent years, a number of anti-fibrotic drug candidates have shown potent effects in experimental animal models; however, an urgent question remains that they have limited or no anti-fibrotic effects in clinical trials. At present, there are no approved drugs for liver fibrosis. The studies on LSECs suggest that protection of LSECs may be an effective strategy to prevent fibrosis initiation and progression. LSEC-targeted drugs or technologies will provide some novel strategies for the treatment of liver fibrosis in the future. Meanwhile, exploring the regulatory mechanisms of LSEC development and crosstalk with other liver cells will help understand the initiation progress of liver fibrosis and develop improved therapeutic targets.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

JG prepared and wrote the original draft of the manuscript. BZ and YH wrote, reviewed and edited the manuscript. All authors read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Parola M and Pinzani M: Liver fibrosis: Pathophysiology, pathogenetic targets and clinical issues. Mol Aspects Med. 65:37–55. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Mallat A and Lotersztajn S: Cellular mechanisms of tissue fibrosis. 5. Novel insights into liver fibrosis. Am J Physiol Cell Physiol. 305:C789–C799. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Gofton C, Upendran Y, Zheng MH and George J: MAFLD: How is it different from NAFLD? Clin Mol Hepatol. 29 (Suppl):S17–S31. 2023. View Article : Google Scholar : PubMed/NCBI

4 

Asrani SK, Devarbhavi H, Eaton J and Kamath PS: Burden of liver diseases in the world. J Hepatol. 70:151–171. 2019. View Article : Google Scholar : PubMed/NCBI

5 

D'Amico G, Morabito A, D'Amico M, Pasta L, Malizia G, Rebora P and Valsecchi MG: New concepts on the clinical course and stratification of compensated and decompensated cirrhosis. Hepatol Int. 12 (Suppl 1):S34–S43. 2018. View Article : Google Scholar

6 

Dewidar B, Meyer C, Dooley S and Meindl-Beinker AN: TGF-β in hepatic stellate cell activation and liver fibrogenesis-updated 2019. Cells. 8:14192019. View Article : Google Scholar : PubMed/NCBI

7 

Rockey DC and Friedman SL: Fibrosis regression after eradication of hepatitis C virus: From bench to bedside. Gastroenterology. 160:1502–1520.e1. 2021. View Article : Google Scholar : PubMed/NCBI

8 

Friedman SL and Pinzani M: Hepatic fibrosis 2022: Unmet needs and a blueprint for the future. Hepatology. 75:473–488. 2022. View Article : Google Scholar : PubMed/NCBI

9 

Xu M, Wang X, Zou Y and Zhong Y: Key role of liver sinusoidal endothelial cells in liver fibrosis. Biosci Trends. 11:163–168. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Brozat JF, Brandt EF, Stark M, Fischer P, Wirtz TH, Flaßhove A, Rodenhausen AN, Vajen T, Heinzmann ACA, Schmitz SM, et al: JAM-A is a multifaceted regulator in hepatic fibrogenesis, supporting LSEC integrity and stellate cell quiescence. Liver Int. 42:1185–1203. 2022. View Article : Google Scholar : PubMed/NCBI

11 

Lafoz E, Ruart M, Anton A, Oncins A and Hernández-Gea V: The endothelium as a driver of liver fibrosis and regeneration. Cells. 9:9292020. View Article : Google Scholar : PubMed/NCBI

12 

Poisson J, Lemoinne S, Boulanger C, Durand F, Moreau R, Valla D and Rautou PE: Liver sinusoidal endothelial cells: Physiology and role in liver diseases. J Hepatol. 66:212–227. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Xie G, Wang L, Wang X, Wang L and DeLeve LD: Isolation of periportal, midlobular, and centrilobular rat liver sinusoidal endothelial cells enables study of zonated drug toxicity. Am J Physiol Gastrointest Liver Physiol. 299:G1204–G1210. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Mönkemöller V, Øie C, Hübner W, Huser T and McCourt P: Multimodal super-resolution optical microscopy visualizes the close connection between membrane and the cytoskeleton in liver sinusoidal endothelial cell fenestrations. Sci Rep. 5:162792015. View Article : Google Scholar : PubMed/NCBI

15 

Wack KE, Ross MA, Zegarra V, Sysko LR, Watkins SC and Stolz DB: Sinusoidal ultrastructure evaluated during the revascularization of regenerating rat liver. Hepatology. 33:363–378. 2001. View Article : Google Scholar : PubMed/NCBI

16 

Scoazec JY, Racine L, Couvelard A, Flejou JF and Feldmann G: Endothelial cell heterogeneity in the normal human liver acinus: In situ immunohistochemical demonstration. Liver. 14:113–123. 1994. View Article : Google Scholar : PubMed/NCBI

17 

Barberá-Guillem E, Rocha M, Alvarez A and Vidal-Vanaclocha F: Differences in the lectin-binding patterns of the periportal and perivenous endothelial domains in the liver sinusoids. Hepatology. 14:131–139. 1991. View Article : Google Scholar : PubMed/NCBI

18 

Di Martino J, Mascalchi P, Legros P, Lacomme S, Gontier E, Bioulac-Sage P, Balabaud C, Moreau V and Saltel F: Actin depolymerization in dedifferentiated liver sinusoidal endothelial cells promotes fenestrae re-formation. Hepatol Commun. 3:213–219. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Herrnberger L, Hennig R, Kremer W, Hellerbrand C, Goepferich A, Kalbitzer HR and Tamm ER: Formation of fenestrae in murine liver sinusoids depends on plasmalemma vesicle-associated protein and is required for lipoprotein passage. PLoS One. 9:e1150052014. View Article : Google Scholar : PubMed/NCBI

20 

DeLeve LD: Liver sinusoidal endothelial cells in hepatic fibrosis. Hepatology. 61:1740–1746. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Malovic I, Sørensen KK, Elvevold KH, Nedredal GI, Paulsen S, Erofeev AV, Smedsrød BH and McCourt PA: The mannose receptor on murine liver sinusoidal endothelial cells is the main denatured collagen clearance receptor. Hepatology. 45:1454–1461. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Verwilligen RAF, Mulder L, Rodenburg FJ, Van Dijke A, Hoekstra M, Bussmann J and Van Eck M: Stabilin 1 and 2 are important regulators for cellular uptake of apolipoprotein B-containing lipoproteins in zebrafish. Atherosclerosis. 346:18–25. 2022. View Article : Google Scholar : PubMed/NCBI

23 

Sørensen KK, McCourt P, Berg T, Crossley C, Le Couteur D, Wake K and Smedsrød B: The scavenger endothelial cell: A new player in homeostasis and immunity. Am J Physiol Regul Integr Comp Physiol. 303:R1217–R1230. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Su T, Yang Y, Lai S, Jeong J, Jung Y, McConnell M, Utsumi T and Iwakiri Y: Single-cell transcriptomics reveals zone-specific alterations of liver sinusoidal endothelial cells in cirrhosis. Cell Mol Gastroenterol Hepatol. 11:1139–1161. 2021. View Article : Google Scholar : PubMed/NCBI

25 

Deleve LD, Wang X and Guo Y: Sinusoidal endothelial cells prevent rat stellate cell activation and promote reversion to quiescence. Hepatology. 48:920–930. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Horn T, Christoffersen P and Henriksen JH: Alcoholic liver injury: Defenestration in noncirrhotic livers-a scanning electron microscopic study. Hepatology. 7:77–82. 1987. View Article : Google Scholar : PubMed/NCBI

27 

Jin Y, Guo YH, Li JC, Li Q, Ye D, Zhang XX and Li JT: Vascular endothelial growth factor protein and gene delivery by novel nanomaterials for promoting liver regeneration after partial hepatectomy. World J Gastroenterol. 29:3748–3757. 2023. View Article : Google Scholar : PubMed/NCBI

28 

Xie G, Wang X, Wang L, Wang L, Atkinson RD, Kanel GC, Gaarde WA and Deleve LD: Role of differentiation of liver sinusoidal endothelial cells in progression and regression of hepatic fibrosis in rats. Gastroenterology. 142:918–927.e6. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Carpenter B, Lin Y, Stoll S, Raffai RL, McCuskey R and Wang R: VEGF is crucial for the hepatic vascular development required for lipoprotein uptake. Development. 132:3293–3303. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Chen L, Gu T, Li B, Li F, Ma Z, Zhang Q, Cai X and Lu L: Delta-like ligand 4/DLL4 regulates the capillarization of liver sinusoidal endothelial cell and liver fibrogenesis. Biochim Biophys Acta Mol Cell Res. 1866:1663–1675. 2019. View Article : Google Scholar : PubMed/NCBI

31 

Duan JL, Ruan B, Yan XC, Liang L, Song P, Yang ZY, Liu Y, Dou KF, Han H and Wang L: Endothelial Notch activation reshapes the angiocrine of sinusoidal endothelia to aggravate liver fibrosis and blunt regeneration in mice. Hepatology. 68:677–690. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Francis H, Bohanan J and Alpini G: Hedgehog signalling and LSEC capillarisation: Stopping this one in its tracks. Gut. 61:1243–1244. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Yang L, Xu X, Chen Z, Zhang Y, Chen H and Wang X: miR-511-3p promotes hepatic sinusoidal obstruction syndrome by activating hedgehog pathway via targeting Ptch1. Am J Physiol Gastrointest Liver Physiol. 321:G344–G354. 2021. View Article : Google Scholar : PubMed/NCBI

34 

Chen T, Shi Z, Zhao Y, Meng X, Zhao S, Zheng L, Han X, Hu Z, Yao Q, Lin H, et al: LncRNA Airn maintains LSEC differentiation to alleviate liver fibrosis via the KLF2-eNOS-sGC pathway. BMC Med. 20:3352022. View Article : Google Scholar : PubMed/NCBI

35 

Winkler M, Staniczek T, Kürschner SW, Schmid CD, Schönhaber H, Cordero J, Kessler L, Mathes A, Sticht C, Neßling M, et al: Endothelial GATA4 controls liver fibrosis and regeneration by preventing a pathogenic switch in angiocrine signaling. J Hepatol. 74:380–393. 2021. View Article : Google Scholar : PubMed/NCBI

36 

Marrone G, Maeso-Díaz R, García-Cardena G, Abraldes JG, García-Pagán JC, Bosch J and Gracia-Sancho J: KLF2 exerts antifibrotic and vasoprotective effects in cirrhotic rat livers: Behind the molecular mechanisms of statins. Gut. 64:1434–1443. 2015. View Article : Google Scholar : PubMed/NCBI

37 

de Haan W, Dheedene W, Apelt K, Décombas-Deschamps S, Vinckier S, Verhulst S, Conidi A, Deffieux T, Staring MW, Vandervoort P, et al: Endothelial Zeb2 preserves the hepatic angioarchitecture and protects against liver fibrosis. Cardiovasc Res. 118:1262–1275. 2022. View Article : Google Scholar : PubMed/NCBI

38 

Maretti-Mira AC, Wang X, Wang L and DeLeve LD: Incomplete differentiation of engrafted bone marrow endothelial progenitor cells initiates hepatic fibrosis in the rat. Hepatology. 69:1259–1272. 2019. View Article : Google Scholar : PubMed/NCBI

39 

Desroches-Castan A, Tillet E, Ricard N, Ouarné M, Mallet C, Belmudes L, Couté Y, Boillot O, Scoazec JY, Bailly S and Feige JJ: Bone morphogenetic protein 9 is a paracrine factor controlling liver sinusoidal endothelial cell fenestration and protecting against hepatic fibrosis. Hepatology. 70:1392–1408. 2019. View Article : Google Scholar : PubMed/NCBI

40 

Gaitantzi H, Karch J, Germann L, Cai C, Rausch V, Birgin E, Rahbari N, Seitz T, Hellerbrand C, König C, et al: BMP-9 modulates the hepatic responses to LPS. Cells. 9:6172020. View Article : Google Scholar : PubMed/NCBI

41 

Desroches-Castan A, Tillet E, Ricard N, Ouarné M, Mallet C, Feige JJ and Bailly S: Differential Consequences of Bmp9 deletion on sinusoidal endothelial cell differentiation and liver fibrosis in 129/Ola and C57BL/6 Mice. Cells. 8:10792019. View Article : Google Scholar : PubMed/NCBI

42 

Kawai H, Osawa Y, Matsuda M, Tsunoda T, Yanagida K, Hishikawa D, Okawara M, Sakamoto Y, Shimagaki T, Tsutsui Y, et al: Sphingosine-1-phosphate promotes tumor development and liver fibrosis in mouse model of congestive hepatopathy. Hepatology. 76:112–125. 2022. View Article : Google Scholar : PubMed/NCBI

43 

Kantari-Mimoun C, Krzywinska E, Castells M, Milien C, Klose R, Meinecke AK, Lemberger U, Mathivet T, Gojkovic M, Schrödter K, et al: Boosting the hypoxic response in myeloid cells accelerates resolution of fibrosis and regeneration of the liver in mice. Oncotarget. 8:15085–15100. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Kwok W, Lee SH, Culberson C, Korneszczuk K and Clemens MG: Caveolin-1 mediates endotoxin inhibition of endothelin-1-induced endothelial nitric oxide synthase activity in liver sinusoidal endothelial cells. Am J Physiol Gastrointest Liver Physiol. 297:G930–G939. 2009. View Article : Google Scholar : PubMed/NCBI

45 

Li H: Intercellular crosstalk of liver sinusoidal endothelial cells in liver fibrosis, cirrhosis and hepatocellular carcinoma. Dig Liver Dis. 54:598–613. 2022. View Article : Google Scholar : PubMed/NCBI

46 

Trautwein C, Friedman SL, Schuppan D and Pinzani M: Hepatic fibrosis: Concept to treatment. J Hepatol. 62 (1 Suppl):S15–S24. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Venkatraman L and Tucker-Kellogg L: The CD47-binding peptide of thrombospondin-1 induces defenestration of liver sinusoidal endothelial cells. Liver Int. 33:1386–1397. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Liepelt A and Tacke F: Stromal cell-derived factor-1 (SDF-1) as a target in liver diseases. Am J Physiol Gastrointest Liver Physiol. 311:G203–G209. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Wu X, Shu L, Zhang Z, Li J, Zong J, Cheong LY, Ye D, Lam KSL, Song E, Wang C, Xu A and Hoo RLC: Adipocyte fatty acid binding protein promotes the onset and progression of liver fibrosis via mediating the crosstalk between liver sinusoidal endothelial cells and hepatic stellate cells. Adv Sci (Weinh). 8:e20037212021. View Article : Google Scholar : PubMed/NCBI

50 

Wang R, Ding Q, Yaqoob U, de Assuncao TM, Verma VK, Hirsova P, Cao S, Mukhopadhyay D, Huebert RC and Shah VH: Exosome adherence and internalization by hepatic stellate cells triggers Sphingosine 1-Phosphate-dependent Migration. J Biol Chem. 290:30684–30696. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Zapotoczny B, Szafranska K, Kus E, Braet F, Wisse E, Chlopicki S and Szymonski M: Tracking fenestrae dynamics in live murine liver sinusoidal endothelial cells. Hepatology. 69:876–888. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Du W and Wang L: The crosstalk between liver sinusoidal endothelial cells and hepatic microenvironment in NASH related liver fibrosis. Front Immunol. 13:9361962022. View Article : Google Scholar : PubMed/NCBI

53 

Lao Y, Li Y, Zhang P, Shao Q, Lin W, Qiu B, Lv Y, Tang L, Su S, Zhang H, et al: Targeting Endothelial Erk1/2-Akt axis as a regeneration strategy to bypass fibrosis during chronic liver injury in mice. Mol Ther. 26:2779–2797. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Colucci S, Altamura S, Marques O, Dropmann A, Horvat NK, Müdder K, Hammad S, Dooley S and Muckenthaler MU: Liver sinusoidal endothelial cells suppress bone morphogenetic protein 2 production in response to TGFβ pathway activation. Hepatology. 74:2186–2200. 2021. View Article : Google Scholar : PubMed/NCBI

55 

Ding BS, Cao Z, Lis R, Nolan DJ, Guo P, Simons M, Penfold ME, Shido K, Rabbany SY and Rafii S: Divergent angiocrine signals from vascular niche balance liver regeneration and fibrosis. Nature. 505:97–102. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Yan Z, Qu K, Zhang J, Huang Q, Qu P, Xu X, Yuan P, Huang X, Shao Y, Liu C, et al: CD147 promotes liver fibrosis progression via VEGF-A/VEGFR2 signalling-mediated cross-talk between hepatocytes and sinusoidal endothelial cells. Clin Sci (Lond). 129:699–710. 2015. View Article : Google Scholar : PubMed/NCBI

57 

Su T and Iwakiri Y: Endothelial Leukocyte Cell-Derived Chemotaxin 2/Tyrosine Kinase with immunoglobulin-like and epidermal growth factor-like domains 1 signaling in liver fibrosis. Hepatology. 72:347–349. 2020. View Article : Google Scholar : PubMed/NCBI

58 

Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, Gordon S, Hamilton JA, Ivashkiv LB, Lawrence T, et al: Macrophage activation and polarization: Nomenclature and experimental guidelines. Immunity. 41:14–20. 2014. View Article : Google Scholar : PubMed/NCBI

59 

Arrese M, Cabrera D, Kalergis AM and Feldstein AE: Innate Immunity and Inflammation in NAFLD/NASH. Dig Dis Sci. 61:1294–1303. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Shetty S, Lalor PF and Adams DH: Liver sinusoidal endothelial cells-gatekeepers of hepatic immunity. Nat Rev Gastroenterol Hepatol. 15:555–567. 2018. View Article : Google Scholar : PubMed/NCBI

61 

You Q, Holt M, Yin H, Li G, Hu CJ and Ju C: Role of hepatic resident and infiltrating macrophages in liver repair after acute injury. Biochem Pharmacol. 86:836–843. 2013. View Article : Google Scholar : PubMed/NCBI

62 

Miyao M, Kotani H, Ishida T, Kawai C, Manabe S, Abiru H and Tamaki K: Pivotal role of liver sinusoidal endothelial cells in NAFLD/NASH progression. Lab Invest. 95:1130–1144. 2015. View Article : Google Scholar : PubMed/NCBI

63 

Ford AJ, Jain G and Rajagopalan P: Designing a fibrotic microenvironment to investigate changes in human liver sinusoidal endothelial cell function. Acta Biomater. 24:220–227. 2015. View Article : Google Scholar : PubMed/NCBI

64 

Kamada Y, Sato M, Kida S, Akita M, Mizutani K, Fujii H, Sobajima T, Yoshida Y, Shinzaki S, Takamatsu S, et al: N-acetylglucosaminyltransferase V exacerbates concanavalin A-induced hepatitis in mice. Mol Med Rep. 11:3573–3584. 2015. View Article : Google Scholar : PubMed/NCBI

65 

Ni Y, Li JM, Liu MK, Zhang TT, Wang DP, Zhou WH, Hu LZ and Lv WL: Pathological process of liver sinusoidal endothelial cells in liver diseases. World J Gastroenterol. 23:7666–7677. 2017. View Article : Google Scholar : PubMed/NCBI

66 

Connolly MK, Bedrosian AS, Malhotra A, Henning JR, Ibrahim J, Vera V, Cieza-Rubio NE, Hassan BU, Pachter HL, Cohen S, et al: In hepatic fibrosis, liver sinusoidal endothelial cells acquire enhanced immunogenicity. J Immunol. 185:2200–2208. 2010. View Article : Google Scholar : PubMed/NCBI

67 

Bonder CS, Norman MU, Swain MG, Zbytnuik LD, Yamanouchi J, Santamaria P, Ajuebor M, Salmi M, Jalkanen S and Kubes P: Rules of recruitment for Th1 and Th2 lymphocytes in inflamed liver: A role for alpha-4 integrin and vascular adhesion protein-1. Immunity. 23:153–163. 2005. View Article : Google Scholar : PubMed/NCBI

68 

Barrow F, Khan S, Wang H and Revelo XS: The Emerging Role of B Cells in the Pathogenesis of NAFLD. Hepatology. 74:2277–2286. 2021. View Article : Google Scholar : PubMed/NCBI

69 

Wehr A, Baeck C, Heymann F, Niemietz PM, Hammerich L, Martin C, Zimmermann HW, Pack O, Gassler N, Hittatiya K, et al: Chemokine receptor CXCR6-dependent hepatic NK T Cell accumulation promotes inflammation and liver fibrosis. J Immunol. 190:5226–5236. 2013. View Article : Google Scholar : PubMed/NCBI

70 

Neumann K, Rudolph C, Neumann C, Janke M, Amsen D and Scheffold A: Liver sinusoidal endothelial cells induce immunosuppressive IL-10-producing Th1 cells via the Notch pathway. Eur J Immunol. 45:2008–2016. 2015. View Article : Google Scholar : PubMed/NCBI

71 

Dauphinee SM and Karsan A: Lipopolysaccharide signaling in endothelial cells. Lab Invest. 86:9–22. 2006. View Article : Google Scholar : PubMed/NCBI

72 

Gola A, Dorrington MG, Speranza E, Sala C, Shih RM, Radtke AJ, Wong HS, Baptista AP, Hernandez JM, Castellani G, et al: Commensal-driven immune zonation of the liver promotes host defence. Nature. 589:131–136. 2021. View Article : Google Scholar : PubMed/NCBI

73 

Greuter T, Yaqoob U, Gan C, Jalan-Sakrikar N, Kostallari E, Lu J, Gao J, Sun L, Liu M, Sehrawat TS, et al: Mechanotransduction-induced glycolysis epigenetically regulates a CXCL1-dominant angiocrine signaling program in liver sinusoidal endothelial cells in vitro and in vivo. J Hepatol. 77:723–734. 2022. View Article : Google Scholar : PubMed/NCBI

74 

Fujita K, Nozaki Y, Wada K, Yoneda M, Endo H, Takahashi H, Iwasaki T, Inamori M, Abe Y, Kobayashi N, et al: Effectiveness of antiplatelet drugs against experimental non-alcoholic fatty liver disease. Gut. 57:1583–1591. 2008. View Article : Google Scholar : PubMed/NCBI

75 

Malehmir M, Pfister D, Gallage S, Szydlowska M, Inverso D, Kotsiliti E, Leone V, Peiseler M, Surewaard BGJ, Rath D, et al: Platelet GPIbα is a mediator and potential interventional target for NASH and subsequent liver cancer. Nat Med. 25:641–655. 2019. View Article : Google Scholar : PubMed/NCBI

76 

Lalor PF, Herbert J, Bicknell R and Adams DH: Hepatic sinusoidal endothelium avidly binds platelets in an integrin-dependent manner, leading to platelet and endothelial activation and leukocyte recruitment. Am J Physiol Gastrointest Liver Physiol. 304:G469–G478. 2013. View Article : Google Scholar : PubMed/NCBI

77 

Joshi N, Kopec AK, Ray JL, Cline-Fedewa H, Groeneveld DJ, Lisman T and Luyendyk JP: Von Willebrand factor deficiency reduces liver fibrosis in mice. Toxicol Appl Pharmacol. 328:540592017. View Article : Google Scholar : PubMed/NCBI

78 

Weyrich AS: Platelets: More than a sack of glue. Hematology Am Soc Hematol Educ Program. 2014:400–403. 2014. View Article : Google Scholar : PubMed/NCBI

79 

Chauhan A, Adams DH, Watson SP and Lalor PF: Platelets: No longer bystanders in liver disease. Hepatology. 64:1774–1784. 2016. View Article : Google Scholar : PubMed/NCBI

80 

Ripoche J: Blood platelets and inflammation: Their relationship with liver and digestive diseases. Clin Res Hepatol Gastroenterol. 35:353–357. 2011. View Article : Google Scholar : PubMed/NCBI

81 

Lisman T and Luyendyk JP: Platelets as modulators of liver diseases. Semin Thromb Hemost. 44:114–125. 2018. View Article : Google Scholar : PubMed/NCBI

82 

Brougham-Cook A, Kimmel HRC, Monckton CP, Owen D, Khetani SR and Underhill GH: Engineered matrix microenvironments reveal the heterogeneity of liver sinusoidal endothelial cell phenotypic responses. APL Bioeng. 6:0461022022. View Article : Google Scholar : PubMed/NCBI

83 

Olsen AL, Sackey BK, Marcinkiewicz C, Boettiger D and Wells RG: Fibronectin extra domain-A promotes hepatic stellate cell motility but not differentiation into myofibroblasts. Gastroenterology. 142:928–937.e3. 2012. View Article : Google Scholar : PubMed/NCBI

84 

Liu L, You Z, Yu H, Zhou L, Zhao H, Yan X, Li D, Wang B, Zhu L, Xu Y, et al: Mechanotransduction-modulated fibrotic microniches reveal the contribution of angiogenesis in liver fibrosis. Nat Mater. 16:1252–1261. 2017. View Article : Google Scholar : PubMed/NCBI

85 

Ruan B, Duan JL, Xu H, Tao KS, Han H, Dou GR and Wang L: Capillarized Liver sinusoidal endothelial cells undergo partial endothelial-mesenchymal transition to actively deposit sinusoidal ECM in liver fibrosis. Front Cell Dev Biol. 9:6710812021. View Article : Google Scholar : PubMed/NCBI

86 

Iwakiri Y and Trebicka J: Portal hypertension in cirrhosis: Pathophysiological mechanisms and therapy. JHEP Rep. 3:1003162021. View Article : Google Scholar : PubMed/NCBI

87 

Trebicka J, Reiberger T and Laleman W: Gut-Liver Axis Links Portal Hypertension to Acute-on-Chronic Liver Failure. Visc Med. 34:270–275. 2018. View Article : Google Scholar : PubMed/NCBI

88 

Bravo M, Raurell I, Hide D, Fernández-Iglesias A, Gil M, Barberá A, Salcedo MT, Augustin S, Genescà J and Martell M: Restoration of liver sinusoidal cell phenotypes by statins improves portal hypertension and histology in rats with NASH. Sci Rep. 9:201832019. View Article : Google Scholar : PubMed/NCBI

89 

Iwakiri Y: Endothelial dysfunction in the regulation of portal hypertension. Liver Int. 32:199–213. 2012. View Article : Google Scholar : PubMed/NCBI

90 

Rockey DC and Chung JJ: Reduced nitric oxide production by endothelial cells in cirrhotic rat liver: Endothelial dysfunction in portal hypertension. Gastroenterology. 114:344–351. 1998. View Article : Google Scholar : PubMed/NCBI

91 

Gracia-Sancho J, Marrone G and Fernández-Iglesias A: Hepatic microcirculation and mechanisms of portal hypertension. Nat Rev Gastroenterol Hepatol. 16:221–234. 2019. View Article : Google Scholar : PubMed/NCBI

92 

Hammoutene A, Biquard L, Lasselin J, Kheloufi M, Tanguy M, Vion AC, Mérian J, Colnot N, Loyer X, Tedgui A, et al: A defect in endothelial autophagy occurs in patients with non-alcoholic steatohepatitis and promotes inflammation and fibrosis. J Hepatol. 72:528–538. 2020. View Article : Google Scholar : PubMed/NCBI

93 

Maeso-Díaz R, Ortega-Ribera M, Lafoz E, Lozano JJ, Baiges A, Francés R, Albillos A, Peralta C, García-Pagán JC, Bosch J, et al: Aging influences hepatic microvascular biology and liver fibrosis in advanced chronic liver disease. Aging Dis. 10:684–698. 2019. View Article : Google Scholar : PubMed/NCBI

94 

Bosch J, Groszmann RJ and Shah VH: Evolution in the understanding of the pathophysiological basis of portal hypertension: How changes in paradigm are leading to successful new treatments. J Hepatol. 62 (1 Suppl):S121–S130. 2015. View Article : Google Scholar : PubMed/NCBI

95 

Medina J, Arroyo AG, Sánchez-Madrid F and Moreno-Otero R: Angiogenesis in chronic inflammatory liver disease. Hepatology. 39:1185–1195. 2004. View Article : Google Scholar : PubMed/NCBI

96 

Hilscher MB, Sehrawat T, Arab JP, Zeng Z, Gao J, Liu M, Kostallari E, Gao Y, Simonetto DA, Yaqoob U, et al: Mechanical stretch increases expression of CXCL1 in liver sinusoidal endothelial cells to recruit neutrophils, generate sinusoidal microthombi, and promote portal hypertension. Gastroenterology. 157:193–209.e9. 2019. View Article : Google Scholar : PubMed/NCBI

97 

Yu Z, Guo J, Liu Y, Wang M, Liu Z, Gao Y and Huang L: Nano delivery of simvastatin targets liver sinusoidal endothelial cells to remodel tumor microenvironment for hepatocellular carcinoma. J Nanobiotechnology. 20:92022. View Article : Google Scholar : PubMed/NCBI

98 

Boyer-Diaz Z, Aristu-Zabalza P, Andrés-Rozas M, Robert C, Ortega-Ribera M, Fernández-Iglesias A, Broqua P, Junien JL, Wettstein G, Bosch J and Gracia-Sancho J: Pan-PPAR agonist lanifibranor improves portal hypertension and hepatic fibrosis in experimental advanced chronic liver disease. J Hepatol. 74:1188–1199. 2021. View Article : Google Scholar : PubMed/NCBI

99 

Hide D, Gil M, Andrade F, Rafael D, Raurell I, Bravo M, Barberá A, Gracia-Sancho J, Vargas V, Augustin S, et al: Simvastatin-loaded polymeric micelles are more effective and less toxic than conventional statins in a pre-clinical model of advanced chronic liver disease. Nanomedicine. 29:1022672020. View Article : Google Scholar : PubMed/NCBI

100 

Zhang LF, Wang XH, Zhang CL, Lee J, Duan BW, Xing L, Li L, Oh YK and Jiang HL: Sequential nano-penetrators of capillarized liver sinusoids and extracellular matrix barriers for liver fibrosis therapy. ACS Nano. 16:14029–14042. 2022. View Article : Google Scholar : PubMed/NCBI

101 

Gu T, Shen B, Li B, Guo Y, Li F, Ma Z, Chen L, Zhang Q, Qu Y, Dong H, et al: miR-30c inhibits angiogenesis by targeting delta-like ligand 4 in liver sinusoidal endothelial cell to attenuate liver fibrosis. FASEB J. 35:e215712021. View Article : Google Scholar : PubMed/NCBI

102 

Mookerjee RP, Mehta G, Balasubramaniyan V, Mohamed Fel Z, Davies N, Sharma V, Iwakiri Y and Jalan R: Hepatic dimethylarginine-dimethylaminohydrolase1 is reduced in cirrhosis and is a target for therapy in portal hypertension. J Hepatol. 62:325–331. 2015. View Article : Google Scholar : PubMed/NCBI

103 

Kumar K, Dong Y, Kumar V, Almawash S and Mahato RI: The use of micelles to deliver potential hedgehog pathway inhibitor for the treatment of liver fibrosis. Theranostics. 9:7537–7555. 2019. View Article : Google Scholar : PubMed/NCBI

104 

Zhao S, Zhang Z, Qian L, Lin Q, Zhang C, Shao J, Zhang F and Zheng S: Tetramethylpyrazine attenuates carbon tetrachloride-caused liver injury and fibrogenesis and reduces hepatic angiogenesis in rats. Biomed Pharmacother. 86:521–530. 2017. View Article : Google Scholar : PubMed/NCBI

105 

Gao L, Yang X, Li Y, Wang Z, Wang S, Tan S, Chen A, Cao P, Shao J, Zhang Z, et al: Curcumol inhibits KLF5-dependent angiogenesis by blocking the ROS/ERK signaling in liver sinusoidal endothelial cells. Life Sci. 264:1186962021. View Article : Google Scholar : PubMed/NCBI

106 

Yang X, Wang Z, Kai J, Wang F, Jia Y, Wang S, Tan S, Shen X, Chen A, Shao J, et al: Curcumol attenuates liver sinusoidal endothelial cell angiogenesis via regulating Glis-PROX1-HIF-1α in liver fibrosis. Cell Prolif. 53:e127622020. View Article : Google Scholar : PubMed/NCBI

107 

Bae CR, Zhang H and Kwon YG: The endothelial dysfunction blocker CU06-1004 ameliorates choline-deficient L-amino acid diet-induced non-alcoholic steatohepatitis in mice. PLoS One. 15:e02434972020. View Article : Google Scholar : PubMed/NCBI

108 

Turaga RC, Satyanarayana G, Sharma M, Yang JJ, Wang S, Liu C, Li S, Yang H, Grossniklaus H, Farris AB, et al: Targeting integrin αvβ3 by a rationally designed protein for chronic liver disease treatment. Commun Biol. 4:10872021. View Article : Google Scholar : PubMed/NCBI

109 

Ye Q, Zhou Y, Zhao C, Xu L and Ping J: Salidroside Inhibits CCl4-Induced Liver fibrosis in mice by reducing activation and migration of HSC induced by liver sinusoidal endothelial cell-derived exosomal SphK1. Front Pharmacol. 12:6778102021. View Article : Google Scholar : PubMed/NCBI

110 

Guo Q, Furuta K, Islam S, Caporarello N, Kostallari E, Dielis K, Tschumperlin DJ, Hirsova P and Ibrahim SH: Liver sinusoidal endothelial cell expressed vascular cell adhesion molecule 1 promotes liver fibrosis. Front Immunol. 13:9832552022. View Article : Google Scholar : PubMed/NCBI

111 

Ruart M, Chavarria L, Campreciós G, Suárez-Herrera N, Montironi C, Guixé-Muntet S, Bosch J, Friedman SL, Garcia-Pagán JC and Hernández-Gea V: Impaired endothelial autophagy promotes liver fibrosis by aggravating the oxidative stress response during acute liver injury. J Hepatol. 70:458–469. 2019. View Article : Google Scholar : PubMed/NCBI

112 

Lin Y, Dong MQ, Liu ZM, Xu M, Huang ZH, Liu HJ, Gao Y and Zhou WJ: A strategy of vascular-targeted therapy for liver fibrosis. Hepatology. 76:660–675. 2022. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2024
Volume 29 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gao J, Zuo B and He Y: Liver sinusoidal endothelial cells as potential drivers of liver fibrosis (Review). Mol Med Rep 29: 40, 2024
APA
Gao, J., Zuo, B., & He, Y. (2024). Liver sinusoidal endothelial cells as potential drivers of liver fibrosis (Review). Molecular Medicine Reports, 29, 40. https://doi.org/10.3892/mmr.2024.13164
MLA
Gao, J., Zuo, B., He, Y."Liver sinusoidal endothelial cells as potential drivers of liver fibrosis (Review)". Molecular Medicine Reports 29.3 (2024): 40.
Chicago
Gao, J., Zuo, B., He, Y."Liver sinusoidal endothelial cells as potential drivers of liver fibrosis (Review)". Molecular Medicine Reports 29, no. 3 (2024): 40. https://doi.org/10.3892/mmr.2024.13164