Abstract
Liver sinusoidal endothelial cells (LSECs) are key players in maintaining hepatic homeostasis. They also play crucial roles during liver injury by communicating with liver cell types as well as immune cells and promoting portal hypertension, fibrosis, and inflammation. Cutting-edge technology, such as single cell and spatial transcriptomics, have revealed the existence of distinct LSEC subpopulations with a clear zonation in the liver. The signals released by LSECs are commonly called “angiocrine signaling.” In this review, we summarize the role of angiocrine signaling in health and disease, including zonation in healthy liver, regeneration, fibrosis, portal hypertension, nonalcoholic fatty liver disease, alcohol-associated liver disease, aging, drug-induced liver injury, and ischemia/reperfusion, as well as potential therapeutic advances. In conclusion, sinusoidal endotheliopathy is recognized in liver disease and promising preclinical studies are paving the path toward LSEC-specific pharmacotherapies.
Keywords: angiocrine signalling, liver sinusoidal endothelial cells, liver health, liver disease, therapeutics
Graphical Abstract
Lay Summary
Liver sinusoidal endothelial cells (LSECs) are key players in maintaining hepatic homeostasis. They also play crucial roles during liver injury by sending signals, referred to as angiocrine signaling, to liver cell types as well as immune cells. In this review, we will summarize angiocrine signaling in health and disease.
Hepatic microvessels are called sinusoids due to their unique sinusoid alanatomical structure. These sinusoids are composed of liver sinusoidal endothelial cells (LSECs), which represent a subpopulation of endothelial cells in the liver with distinct features, such as lack of a basal membrane and the presence of fenestrae.1–3 The fusion of the luminal and abluminal plasma membrane to form the sinusoids occurs at the level of LSEC fenestrae, differently from other endothelial cells where it usually happens at the level of cell junctions.4 The fenestrated cellular walls of LSECs are highly permeable enabling the rapid transport of nutrients, metabolites, and soluble angiocrine signaling (Asig) molecules between the circulation and the space of Disse for access to hepatocytes and resident non-parenchymal cells such as hepatic stellate cells (HSCs) and immune cells.5–7 Recent technological advances, such as single cell RNA sequencing (scRNAseq) and spatial transcriptomics, have revealed LSEC angiocrine phenotype heterogeneity6 and allow studies of the angioimmune niche at unprecedented depth. LSEC contributions to liver health and disease were very recently extensively reviewed.6 Therefore, in this article, we will give an overview of the recent advances regarding Asig in liver health, regeneration, and disease, specifically.
Asig in Hepatic Health and Regeneration
The importance of hepatic zonation during health and injury has received significant attention recently, due in part to advances in scRNAseq and spatial transcriptomics/multiomics.8,9 Furthermore, web-browsable versions of published datasets and species/organ atlases are increasingly available—a powerful resource to explore newly appreciated Asig molecules, cellular subtypes, and interaction partners (► Table 1).
Table 1.
Browsable datasets for angiocrine signaling
First author and year | Article title | Web site | Modality | Species |
---|---|---|---|---|
Inverso 202110 | A spatial vascular transcriptomic, proteomic, and phosphoproteomic atlas unveils an angiocrine Tie-Wnt signaling axis in the liver | http://pproteomedb.dkfz.de | Transcriptome, proteome, phosphoproteome | Mouse |
Kalucka 2020150 | Single-cell transcriptome atlas of murine endothelial cells | https://carmelietlab.sites.vib.be/en/software-tools | Multiple | Mouse |
Aizarani 2019151 | A human liver cell atlas reveals heterogeneity and epithelial progenitors | http://human-liver-cellatlas.ie-freiburg.mpg.de | scRNAseq | Human |
Drzewiecki 202178 | GIMAP5 maintains liver endothelial cell homeostasis and prevents portal hypertension | https://cellbrowser.yalespace.org/gimap5_res0-3/?ds=gimap5 | scRNAseq | Mouse |
Ramachandran 201936 | Resolving the fibrotic niche of human liver cirrhosis at single-cell level | http://www.livercellatlas.mvm.ed.ac.uk | scRNAseq | Human, mouse |
McParland 201823 | Single cell RNA sequencing of human liver reveals distinct intrahepatic macrophage populations | http://shiny.baderlab.org/HumanLiverAtlas/ | scRNAseq | Human |
Dobie 201934 | Single-cell transcriptomics uncovers zonation of function in the mesenchyme during liver fibrosis | http://livermesenchyme.hendersonlab.mvm.ed.ac.uk | scRNAseq | Mouse |
Pita-Juarez 2022152 | A single-nucleus and spatial transcriptomic atlas of the COVID-19 liver reveals topological, functional, and regenerative organ disruption in patients | Supplemental data | snRNAseq and nanostring DSP WTA | Human |
Holland 2022153 | Transcriptomic cross-species analysis of chronic liver disease reveals consistent regulation between humans and mice | https://saezlab.shinyapps.io/liverdiseaseatlas/ | scRNAseq | Human, mouse |
Bondareva 2022154 | Single-cell profiling of vascular endothelial cells reveals progressive organ-specific vulnerabilities during obesity | https://obesity-ecatlas.helmholtz-muenchen.de/ | scRNAseq | Mouse |
Betsholtz 2022155 | Human cell atlas: contains links to multiple browsable portals and datasets | https://www.humancellatlas.org/portals/ | Multiple | Human |
The Tabula Muris Consortium 2018156 | Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris | http://tabula-muris.ds.czbiohub.org/ | Multiple | Mouse |
Paik 2020157 | Single-cell RNA sequencing unveils unique transcriptomic signatures of organ-specific endothelial cells | Supplemental data | Multiple | Mouse |
Massalha 2020158 | A single cell atlas of the human liver tumor microenvironment | https://itzkovitzwebapps.weizmann.ac.il/webapps/home/ | Multiple | Human |
Cavalli 2020159 | A multi-omics approach to liver diseases: integration of single nuclei transcriptomics with proteomics and HiCap bulk data in human liver | Supplemental data | Multiple | Human |
Gainullina 202327 | Network analysis of large-scale ImmGen and Tabula Muris datasets highlights metabolic diversity of tissue mononuclear phagocytes | https://artyomovlab.wustl.edu/immgen-met/ | Multiple | Mouse |
Guilliams 202228 | Spatial proteogenomics reveals distinct and evolutionarily conserved hepatic macrophage niches | https://www.livercellatlas.org/index.php | Multiple | Human, mouse |
Andrews 202233 | Single-cell, single-nucleus, and spatial RNA sequencing of the human liver identifies cholangiocyte and mesenchymal heterogeneity | https://macparlandlab.shinyapps.io/healthylivermapspatialgui/ | Visium 10× Spatial | Human |
Ben-Moshe 202238 | The spatiotemporal program of zonal liver regeneration following acute injury | https://itzkovitzapapp.weizmann.ac.il/apap/ | Multiple | Mouse |
Kim 202147 | Gene deconvolution reveals aberrant liver regeneration and immune cell infiltration in alcohol-associated hepatitis | Supplemental data | RNAseq | Human |
Kostallari 202135 | Stiffness is associated with hepatic stellate cell heterogeneity during liver fibrosis | Supplemental data | scRNAseq | Mouse |
Furuta 202197 | Lipid-induced endothelial vascular cell adhesion molecule 1 promotes nonalcoholic steatohepatitis pathogenesis | Supplemental data | Multiple | Mouse |
Su 202124 | Single-cell transcriptomics reveals zone-specific alterations of liver sinusoidal endothelial cells in cirrhosis | Supplemental data | scRNAseq | Mouse |
Zonation in Healthy Liver by Asig
In addition to central vein-associated angiocrine Wnt signaling, endothelial tyrosine kinase with Ig-like and EGF-like domains 1/2 (Tie1/2),10 Notch,11,12 c-Maf,13–16 Heg,17 and periportal Dll4 and Esm116,18 also participate to hepatic and endothelial zonation.5,6,19–22 Briefly, paired endothelial cell/hepatocyte sequencing was originally utilized to develop a panel of pericentral to periportal endothelial functional landmark genes (70 each).8 Subsequent studies have demonstrated that Stab2, Cd32b, Flt4, and Lyve1 are primarily expressed by Zone2/3 mid-central LSECs, while CD36 and Adam23 are highly expressed in the periportal zone 1 LSECs. This contrasts abundant expression of Wnt2, Wnt9b, Lhx6, and cKit in pericentral LSECs and central venous endothelial cells.6,23,24 The field has integrated these findings into cell type-specific zonation marker panels critical to the analysis of single cell and spatial transcriptomic platforms.24 However, zonation of protein phosphorylation, despite ubiquitous expression, was demonstrated as a significant mechanism for regulation of pericentral Wnt signaling by Tie1/2. The authors further demonstrated a global increase in tyrosine kinase activity pericentrally versus a periportal serine-threonine kinase activity, highlighting the importance of moving toward integrated multiomics studies, including posttranslational protein modification analyses,10 an important consideration for bioinformatic data interpretation.
Liver immune zonation is not developmentally determined, but rather driven by LSEC interaction with commensal gut-derived microbes and microbial products.7 Periportal LSECs increase C-X-C chemokine receptor (CXCR) CXCL9 expression and utilize sustained MyD88 signaling to alter pericellular ECM binding of CXCL9 to generate a chemokine gradient.7 Chemokine gradients are critical for regulating the spatial distribution of immune cells. During injury, LSEC-derived CXCL9 interaction with macrophage CXCR3 is a key example.25 Other studies have further delved into the zonation of macrophages in health and disease, demonstrating a macrophage niche that is regulated by complex angiocrine interactions with neighboring LSEC and HSCs26–31 and is recently reviewed.32 Furthermore, scRNAseq increasingly reveals that HSCs are far more heterogeneous than previously appreciated and zonated in both health and disease.33–36
Asig in Liver Regeneration
LSECs, macrophages, and HSCs significantly contribute in the process of liver regeneration,5,6,9,10,12,18,22,28,32,33,37–54 supported by scRNAseq and spatial transcriptomics studies.55 Regeneration is studied via acute injury models such as the two-thirds partial hepatectomy model (PHx)10 resulting in rapid regeneration of liver mass by 96 hours postinjury or the centrilobular necrosis with acetaminophen overdose (APAP)38 (further discussed later). In the APAP model, Hgf expression and hepatocyte proliferation did not show zonation.38 However, the pericentral endothelial cell-specific-secreted Wnt9b, Wnt2, and Rspo3 were shown to reprogram new hepatocytes in pericentral areas.38,45 This was confirmed with single cell “omics,” further demonstrating that Wnt agonist administration late in APAP toxicity can support rodent liver regeneration by Wnt2 and Wnt9b induction.45 In the PHx model, regeneration is a well-organized, LSEC-driven series of events that includes the inductive phase of hepatocyte proliferation, angiogenic phase, and terminal phase.5,6 During the inductive phase, hepatocyte proliferation is stimulated via LSEC-derived nitric oxide (NO) signaling, which can be stimulated via multiple mechanisms including flow-induced sheer stress and LSEC-derived HGF.6,56,57 Indeed, increased HGF secretion activates c/MET, Deptor, liver kinase B1, and subsequent adenosine monophosphate-activated protein kinase (AMPK) phosphorylation to increase NO release via phosphorylated endothelial nitric oxide synthase (eNOS).54,58 In addition, Wnt2 release for hepatocyte proliferation can be regulated downstream of the vascular endothelial growth factor receptor (VEGFR)-2/inhibitor of the DNA binding 1 (ID1) axis.42 During the switch between the inductive and angiogenic phases, LSEC-derived angiopoietin-2 (Angpt2) is reduced initially, which lessens transforming growth factor-β (TGF-β) production and is permissive for hepatocyte proliferation. This is reversed at a later stage via an autocrine mechanism, as well as increased VEGFR-2 to begin the angiogenic phase.59 Animal models have demonstrated that, as the angiogenic phase progresses, hepatocytes secrete hypoxia-induced angiogenic factors such as VEGF and angiopoietins that bolster LSEC proliferation until the population reaches its peak at around 3 to 4 days post PHx.42,60 Finally, LSECs play a key role during the termination phase of liver regeneration by secreting TGF-β, which stimulates HSCs, contributes to reconstruction of the extracellular matrix (ECM) scaffold, and terminates hepatocyte regeneration.61 Asig is a critical driver of the regenerative response and thus, the focus of developing potential therapeutic targets for liver regeneration.
Diseases
Cirrhosis and Fibrosis
Dysregulated liver regeneration can result in fibrosis and ultimately cirrhosis, characterized by (1) HSC activation secreting profibrotic molecules and extracellular vesicles62–67 and depositing primarily collagen 1 (COL I) into the Space of Disse; (2) LSECs depositing COL IV. These events are potentiated by the profibrotic angioimmune niche, which we define as the microenvironment where LSECs interact with immune cells to promote fibrosis.5,68 Examples of the angioimmune niche in the liver include the sinusoidal microenvironment where interactions between LSEC and macrophages or neutrophils promote fibrosis.68,69 LSEC NO signaling, critical for normal sinusoidal function, is impaired during injury (►Fig. 1).70,71 Activated serine/threonine protein kinase B (PKB/AKT) promotes binding of endothelial NO synthase (eNOS) to G-protein–coupled receptor kinase-interacting protein-1 (GIT1) scaffold protein, leading to NO release.72 While important for vascular tone, LSEC-produced NO also protects the liver from fibrosis by preventing HSC activation.73 HSCs are activated into myofibroblasts with injury and characterized by increased proliferation, migration, and ECM deposition. The stimuli, including stimuli from LSECs, responsible for myofibroblastic phenotypes, such as platelet-derived growth factor (PDGF) and TGF-β, are known, but the mechanisms leading to activated HSC myofibroblast persistence and subsequent fibrosis are not fully understood.74,75 However, HSCs are not the only cell type depositing collagen into the ECM. Capillarized LSECs also deposit COL IV and significantly contribute to fibrosis.76 Loss of the LSEC master regulator GATA4 permits sinusoidal capillarization and promotes perisinusoidal liver fibrosis via de novo expression of profibrotic angiocrine factors including PDGF receptor β (PDGFRB), secreted protein acidic and cysteine rich like 1 (SPARCL1), endothelial cell specific molecule 1 (ESM1), and insulin-like growth factor binding protein 5 (IGFBP5).77 Furthermore, loss of GATA4 upstream regulator GTPase, IMAP family member 5 (GIMAP5), increases capillarization and vascular resistance during human portal hypertension (PH).78 Immune cells can contribute to LSEC capillarization via leukocyte cell-derived chemotaxin 2 (LECT2) binding to the endothelial Tie1 to upregulate MAPK-dependent PPAR signaling and results in the release of matrix proteins such as collagen IV and fibronectin.10,79 Modulation of LECT2 ameliorated the effect in CCl4-induced liver fibrosis.79
Fig. 1.
Role of LSECs and angiocrine signaling during fibrosis. LSEC NO signaling is critical for normal sinusoidal function. In a healthy environment, AKT activation induces eNOS binding to GIT1 leading to NO release, which in turn is involved in maintaining HSC quiescence. Injury and stress promote LSEC capillarization, dysfunction, and ultimately fibrosis. Injured LSECs produce profibrogenic molecules, such as PDGF, IGFBP5, and ESM1. During injury, eNOS is inhibited, which is accompanied by HSC activation and matrix deposition. Finally, immune cell–derived LECT2 binds to the endothelial Tie1 leading to the release of matrix proteins such as collagen IV and fibronectin. eNOS, endothelial nitric oxide synthase; LSEC, liver sinusoidal endothelial cell; NO, nitric oxide; PDGF, platelet-derived growth factor. (Created with BioRender.)
Portal Hypertension
Clinically significant PH is defined by an increased hepatic venous pressure gradient of at least 10 mm Hg.80–83 PH stems from increased hepatic vascular resistance. This mechanism has two primary components: architectural due to maladaptive tissue remodeling and dynamic due to endothelial dysfunction and increased hepatic microcirculatory tone. Increased vasoconstrictors such as endothelin-1, cyclooxygenase 1 (COX1), and thromboxane A2 (TXA2) exaggerate contractile responses of LSEC, HSC, and sinusoidal vascular smooth muscle cells along with impaired synthesis of vasodilators like eNOS.57,71 eNOS-derived NO inhibits the release of P-selectin containing Weibel-Palade bodies, which can contribute to leukocyte recruitment and increase vascular inflammation.84 NO also blocks platelet adhesion and aggregation on LSECs to reduce microvascular thrombosis.85 Mechanical stretch and stretch-induced glycolysis can induce LSEC expression of C-X-C chemokine ligand 1 (CXCL1), which recruits neutrophils to generate microthrombi and neutrophil extracellular traps (NETs) that contribute to vascular resistance.69,86 In addition, LSEC-derived CCL2 is increased via tumor necrosis factor-α (TNF-α)-induced p300 epigenetic modification of the CCL2 enhancer and promoter regions and specific interactions with BRD4 and nuclear factor kappa B (NF-κB) to recruit CCR2+ macrophages.68 Additionally, p300 activity in HSCs is critical for epigenetic regulation of IGFBP-3, which has been demonstrated to be critical for migration in vitro and promotes PH in vivo.67
Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis
Nonalcoholic fatty liver disease (NAFLD) has the highest prevalence among chronic liver diseases and refers to the accumulation of steatosis in the absence of alcohol.87 A subset of patients with NAFLD develop nonalcoholic steatohepatitis (NASH), which is characterized by the release of toxic lipids, increase of inflammation, and development of liver fibrosis.6 LSEC phenotype can be affected in the lipotoxic hepatic microenvironment during the development of NAFLD toward NASH (►Fig. 2). Although one study reports that LSEC fenestrae are preserved at 20 weeks of high-fat diet,88 several other studies confirm a loss of the fenestrae due to high-fat high-carbohydrate diet, choline-deficient high-fat diet, choline-deficient L-amino-acid–defined high-fat diet with 0.1% methionine, or 22 to 24 weeks of high-fat diet.89–91 The loss of fenestration is also observed in human livers at the stage of steatosis as compared with steatohepatitis stage,92 suggesting that the change in fenestrae likely happens during the early development of NAFLD. During the progression of NAFLD, the mechanical stress induced by ballooned hepatocytes impairs NO endothelin 1 balance leading to LSEC dysfunction.93 The association of NAFLD to decreased eNOS expression, NO release, and increased endothelin1expression93,94suggests that blocking endothelin 1 receptor could be potential strategy to ameliorate LSEC dysfunction in NASH.95 Notch, expressed on LSECs, is also involved in LSEC dysfunction and NASH. eNOS activator and NOTCH inhibitor ameliorate LSEC capillarization, steatosis, inflammation, and fibrosis.96 During NASH progression, LSECs become proinflammatory and are characterized by an increased release of proinflammatory cytokines and chemokines and upregulation of adhesion molecule expression.97–99 This change in phenotype and function is called endotheliopathy.100 LSECs, which represent the interface between sinusoidal circulation and liver parenchyma, respond to toll-like receptors (TLRs) ligands by releasing TNF-α, IL-1β, IL-6, and IFN-β and, thus, participate in the host innate immune response.101,102 ScRNA-seq and spatial mapping show that LSECs in the fibrotic niche of human liver with NASH cirrhosis express atypical chemokine receptor 1 (ACKR1), also known as Duffy antigen receptor for chemokines (DARC), which is involved in transendothelial migration of leukocytes.36,103 The immunomodulatory abilities of LSECs seem to be regulated by autophagy as well. Indeed, patients with NASH present with reduced autophagic vacuoles in LSECs. In line with this, LSECs from mice with defective endothelial autophagy increase the expression of proinflammatory genes.104 All these studies suggest that LSECs are the main immunomodulatory cells during NAFLD and NASH and restoring their homeostasis is beneficial to disease progression.
Fig. 2.
Role of LSECs and angiocrine signaling during NAFLD and NASH. During NAFLD progression, LSECs become capillarized and increase the expression of endothelin-1. Ballooned hepatocytes exert a mechanical stress toward capillarized LSECs, which increase Notch expression and decrease eNOS activity. Notch inhibitor and eNOS activator ameliorate LSEC capillarization, liver steatosis, inflammation, and fibrosis. Injured LSEC release proinflammatory molecules such as TNF-α, IL-6, IL-1β, IFN-β, and express ACKR1, which in turn promotes leukocyte transendothelial migration. eNOS, endothelial nitric oxide synthase; LSEC, liver sinusoidal endothelial cell; NAFLD, nonalcoholic fatty liver disease; NASH, nonalcoholic steatohepatitis; NO, nitric oxide. (Created with BioRender.)
Aging
Aging impacts endothelial dysfunction and liver disease.40,59,105–108 Aging amplifies pathogenic injury mechanisms, increases inflammation, and impairs sinusoidal function. Age-related LSEC pseudocapillarization results in fenestrae loss, increased basement membrane thickness, similar but less severe than fibrosis,105,109 and microvascular rarefication (MVR)110 which impedes the transport between the sinusoidal circulation and space of Disse.111 VEGF was recently shown to ameliorate age-related MVR,112 which contrasts with increases in VEGF signaling that induces angiogenesis and aggravate PH (see section “Portal Hypertension”). Paradoxically, aging increases the accumulation of steatosis-associated soluble VEGFR (sFLT1) in rodent liver vasculature and major vessels, but blockade ameliorates age-related effects.112 LSEC cellular adhesion molecules such as ICAM-1 are increased, leading to neutrophils and macrophage recruitment, further increasing inflammatory cytokines like IL-6, TNF-α, and MPO.113 Prothrombotic (fibrinogen-2) and senescence-associated molecules (p16, IL-6, IL-10) are also increased in the aged liver, which are partially due to age-related impairment of angiocrine antioxidant signaling and NO production,59 and contribute to a proinflammatory environment in the aged liver. LSEC regulation of HSC activation and quiescence via VEGF-stimulated NO production is critical for sinusoidal homeostasis,73 but aging results in increased activation and transdifferentiation of HSCs into myofibroblasts, as well as HSC hyperplasia, lipid drop alterations, telomere attrition, impaired release of Asig proteins such as HGF, and finally impaired production of ECM components such as laminin.108,114,115 This failure of LSEC to maintain HSC quiescence with age was also associated with significantly increased macrophage infiltration in rodent models and altered gene expression of sinusoidal protective pathways in young and aged cirrhosis patients.109
Alcohol-Associated Liver Disease
Alcohol-associated liver disease (ALD) is a significant public health issue with extremely limited therapeutic options. Recent investigations into the role of Asig in ALD provide valuable insight for potential future therapeutics. While LSEC dysfunction has been previously known with ALD, the fact that LSECs are capable of metabolizing alcohol and express enzymes typically associated with hepatocytes including alcohol dehydrogenase 1 (ADH1) and cytochrome P450 2E1 (CYP2E1) has been more recently reported.116,117 This also is important because CYP2E1 has been highlighted as a zonation landmark gene for pericentral hepatocytes for single cell studies, and while it’s expression is highest in hepatocytes, approximately 3.3-fold higher than LSECs, using this gene to assess for hepatocyte mRNA contamination and zonation of mixed liver cell types may not be appropriate.118 It is not currently known if LSEC CYP2E1 expression is zonated, but studies have shown that ethanol not only increases CYP2E1 gene expression and protein stability in hepatocytes but also chronically induces LSEC CYP2E1.117,119 This CYP2E1 induction has been demonstrated to impair angiocrine NO signaling and potentiate liver injury via increasing the downstream production of the protein acetylation substrate acetyl-CoA, which promotes Hsp90 acetylation and decreases its activation of eNOS and subsequent NO production.117,120 Recruitment of infiltrating immune cells in alcohol-associated hepatitis (AH), the most severe manifestation of ALD, is another Asig-mediated mechanism of liver injury. Cytokine activation and LSEC-derived chemokine release have been demonstrated to be critical for neutrophil (CXCL 1, 6, 8), monocyte/macrophage (CXCL 9, 10, 11), and lymphocyte recruitment (CXCL 9, 12) during injury.47,121–123 Additionally, chemokine internalization by LSEC contributes to modulation of Asig and its importance in CD4+ T cell recruitment was demonstrated.123 Chemokine production is also potentiated by clinically relevant simultaneous insults such as alcohol plus high-fat diet and Chang et al reported that this paradigm increased production of neutrophil-recruiting CXCL1 from endothelial cells, hepatocytes, and HSCs.124 Finally, angiocrine-mediated neutrophil recruitment in AH is epigenetically regulated. A super-enhancer, which is a DNA element that activates the transcription of genes from a distance and independently of their orientation on DNA,125 upstream of CXCL8, regulates CXCL 1, 2, 3, 6, and 8 expression in a TNF-α/NF-κB-dependent manner.122
Drug-Induced Liver Injury
Drug-induced liver injury (DILI) is due to parenchymal cell, hepatocyte, or cholangiocyte injury. Acetaminophen toxicity has the highest occurrence among the DILIs, and it is characterized by a centrilobular necrosis. Acetaminophen-mediated DILI has been demonstrated to have a direct effect on LSECs. Indeed, mice treated with acetaminophen by oral gavage showed swollen LSECs starting from 30 minutes after injury, which precedes the hepatocellular injury.126 In addition, in this same model, TNF-related apoptosis-inducing ligand (TRAIL) signaling pathway significantly amplifies LSEC death through apoptosis.127 DILI is also promoted by the leukotriene pathway, from which cysteinyl leukotriene receptor 1 (CYSLTR1) is expressed in several liver cell types, including LSECs.128 Anti-Cysltr1 small interfering RNA pretreatment, in combination with a G-protein-coupled bile acid receptor 1 (GPBAR1) agonist, reversed LSEC/monocyte interactions and liver injury.128 The interaction of LSECs with Kupffer cells (KCs) is crucial to maintain liver health, as the depletion of KCs prior to acetaminophen insult aggravated DILI-mediated LSEC injury and enhanced the expression of cellular adhesion molecules in LSECs.129 In line with these studies, the depletion of both KCs and macrophages leads to a delayed liver repair due to a prolonged vascular leakage caused by acetaminophen-mediated LSEC injury.130 Finally, it has been shown that high cholesterol diet exacerbates acetaminophen-mediated hepatotoxicity through promoting free cholesterol accumulation in LSECs in a toll-like receptor 9 (TLR9) manner.131 These studies suggest that LSECs have a central role in DILI and acute liver injury.
Ischemia Reperfusion
Liver ischemia reperfusion (IR) injury is a blood circulation disorder following liver transplantation that involves the reestablishment of the hepatic circulation after the cold ischemia during organ preservation and warm ischemia period during the liver transplantation.6,132 LSEC damage following IR injury includes LSEC detachment and sinusoidal wall destruction, which is followed by extravasated platelet aggregation,133,134 most likely due to a change in Asig. LSECs and platelets produce platelet-activating factor which primes neutrophils for reactive oxygen species generation and leukotriene B4 release.135,136 Exposure of LSECs to cold ischemia increases intracellular calcium concentration, calpain activity, and induces actin disassembly.137 Reperfusion promotes autophagosome accumulation in LSECs and subsequent cell death and microvascular dysfunction.138 In addition, IR downregulates several metabolic pathways in LSECs, such as carbohydrate, lipid, as well as mitochondrial metabolism, and this effect can be attenuated by adenosine A2a receptor (A2aR) agonist.139 In aged rats, the effect of IR injury on LSEC damage are exacerbated. Indeed, warm IR increased LSEC capillarization and reduced vasodilation, which is associated with liver damage and cellular stress.133 However, these effects were reduced when rats were treated with KLF2-inducer simvastatin.133 In line with this study, simvastatin addition to the cold preservation solution ameliorated the hepatic IR injury in a rat experimental model.140 Utilizing spatial transcriptomics, a recent study demonstrates that IR injury affects mainly the pericentral zone (zone 3) in the liver where it induces an increase in endothelial cell proportion.141 However, it remains to be discovered what specific Asig molecules are altered in zone 3 LSECs and whether this could be a therapeutic strategy to alleviate hepatic IR injury.
Therapeutics
Although sinusoidal endotheliopathy is recognized in several liver diseases, no LSEC-specific therapy exists. LSECs immunomodulatory Asig is a promising therapeutic approach in inflammatory liver diseases.100,142 In this regard, vascular cell adhesion molecule 1 neutralizing antibody reduced monocyte/macrophage accumulation in the liver and ameliorated diet-induced liver damage in a mouse model of NASH.97 Another study demonstrates that NASH can be ameliorated in methionine choline deficient diet-fed mice via in vivo targeted silencing of the Runt-related transcription factor (RUNX1) gene in LSECs. This was achieved by administrating RUNX1 siRNA encapsulated in VEGFR-3 antibody tagged immuno-nano-lipocarriers.143 In a similar nanoparticle approach, LSEC-targeting and fenestrae-repairing nanoparticles (HA-NPs/SMV) containing hyaluronic acid were loaded with simvastatin. These HA-NPs/SMV repaired LSEC fenestrae and reduced liver fibrosis.144 Recent studies have attributed the effects of simvastatin on PH to Kruppel-like factor 2 (KLF2), a zinc finger transcription factor.145,146 In addition to nanoparticles, targeting Asig epigenetically by pharmacological inhibition of bromodomain and extraterminal (BET) proteins reduces chemokine production by LSECs in vitro, neutrophil infiltration in vivo, and ameliorates alcoholic hepatitis.122 Currently, the development of BET protein inhibitors is an active area of research147 with high potential for liver disease therapeutics. NO is a crucial molecule in maintaining LSEC homeostasis. Treatment with the NO signaling enhancer sildenafil reduced liver inflammation.148 A recent study suggests that chronic liver disease can be ameliorated by inducing capillarized LSEC apoptosis. The authors have developed a rationally designed protein (ProAgio), which is designed to target integrin αvβ3 at a novel site and induce HSC and LSEC apoptosis.149 As a potential treatment for DILI, the combinatorial G-protein-coupled bile acid receptor 1 (GPBAR1) agonist and cysteinyl leukotriene receptor 1 (CYSLTR1) antagonist, namely CHIN117, affected chemokine production and attenuated liver damage.128 All these studies testify of a quickly evolving field of LSEC biology. The recent technological advances, such as multiomics, single cell and spatial omics, as well as the development of CRISPR/Cas9 transgenic mouse models, will lead the field toward better therapeutic strategies.
Conclusion
Our knowledge of LSEC phenotypes, roles, and their Asig in liver diseases is rapidly evolving. LSECs damage during liver injury promotes disease progression through releasing proinflammatory, antiregeneration, and profibrotic molecules. Recent studies utilizing state-of-the-art technologies have brought granularity on zonation of LSEC subpopulations. However, specific Asig from each of these different LSEC subpopulations remains to be studied. LSECs are excellent candidates to target for treating liver disease as they are in the interface between circulation and liver parenchyma. Some promising options include exploring the immunomodulatory function of LSECs in the field of liver transplant, targeting LSEC luminal adhesion molecules in inflammatory liver diseases, and inhibiting shear-stress signaling pathways and subsequent proinflammatory molecule expression to ameliorate PH. With preclinical studies presenting encouraging results, we expect that LSEC-specific pharmacotherapies will be developed soon.
Funding
This paper received funding from the American Association for the Study of Liver Diseases Pinnacle Award, Gilead Scholar award to E.K.; NIDDK-supported 5T32DK124190 to S.A.C.; and P30DK084567 to the Mayo Clinic Center for Cell Signaling in Gastroenterology.
Footnotes
Conflict of Interest
None declared.
References
- 1.DeLeve LD. Liver sinusoidal endothelial cells in hepatic fibrosis. Hepatology 2015;61(05):1740–1746 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Grisham JW, Nopanitaya W, Compagno J, Nägel AE. Scanning electron microscopy of normal rat liver: the surface structure of its cells and tissue components. Am J Anat 1975;144(03):295–321 [DOI] [PubMed] [Google Scholar]
- 3.Livni N, Behar A, Brautbar N. Ultrastructure of hepatocellular carcinoma in a cirrhotic liver. Isr J Med Sci 1977;13(06):590–599 [PubMed] [Google Scholar]
- 4.Poisson J, Lemoinne S, Boulanger C, et al. Liver sinusoidal endothelial cells: Physiology and role in liver diseases. J Hepatol 2017;66(01):212–227 [DOI] [PubMed] [Google Scholar]
- 5.Kostallari E, Shah VH. Angiocrine signaling in the hepatic sinusoids in health and disease. Am J Physiol Gastrointest Liver Physiol 2016;311(02):G246–G251 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.McConnell MJ, Kostallari E, Ibrahim SH, Iwakiri Y. The evolving role of liver sinusoidal endothelial cells in liver health and disease. Hepatology 2023;78(02):649–669. Doi: 10.1097/HEP.0000000000000207 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Wisse E An electron microscopic study of the fenestrated endothelial lining of rat liver sinusoids. J Ultrastruct Res 1970;31(01):125–150 [DOI] [PubMed] [Google Scholar]
- 8.Halpern KB, Shenhav R, Massalha H, et al. Paired-cell sequencing enables spatial gene expression mapping of liver endothelial cells. Nat Biotechnol 2018;36(10):962–970 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Ramachandran P, Matchett KP, Dobie R, Wilson-Kanamori JR, Henderson NC. Single-cell technologies in hepatology: new insights into liver biology and disease pathogenesis. Nat Rev Gastroenterol Hepatol 2020;17(08):457–472 [DOI] [PubMed] [Google Scholar]
- 10.Inverso D, Shi J, Lee KH, et al. A spatial vascular transcriptomic, proteomic, and phosphoproteomic atlas unveils an angiocrine Tie-Wnt signaling axis in the liver. Dev Cell 2021;56(11):1677–1693.e10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Duan JL, Ruan B, Yan XC, et al. Endothelial Notch activation reshapes the angiocrine of sinusoidal endothelia to aggravate liver fibrosis and blunt regeneration in mice. Hepatology 2018;68(02):677–690 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Duan JL, Zhou ZY, Ruan B, et al. Notch-regulated c-Kit-positive liver sinusoidal endothelial cells contribute to liver zonation and regeneration. Cell Mol Gastroenterol Hepatol 2022;13(06):1741–1756 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Baumann K Endothelial cell diversity in the liver. Nat Rev Mol Cell Biol 2022;23(05):305. [DOI] [PubMed] [Google Scholar]
- 14.de Haan W, Øie C, Benkheil M, et al. Unraveling the transcriptional determinants of liver sinusoidal endothelial cell specialization. Am J Physiol Gastrointest Liver Physiol 2020;318(04):G803–G815 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Gómez-Salinero JM, Izzo F, Lin Y, et al. Specification of fetal liver endothelial progenitors to functional zonated adult sinusoids requires c-Maf induction. Cell Stem Cell 2022;29(04):593–609.e7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Koch PS, Lee KH, Goerdt S, Augustin HG. Angiodiversity and organotypic functions of sinusoidal endothelial cells. Angiogenesis 2021;24(02):289–310 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Zhu S, Rao X, Qian Y, et al. Liver endothelial Heg regulates vascular/biliary network patterning and metabolic zonation via Wnt signaling. Cell Mol Gastroenterol Hepatol 2022;13(06):1757–1783 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Koch PS, Sandorski K, Heil J, et al. Imbalanced activation of Wnt-/β-catenin-signaling in liver endothelium alters normal sinusoidal differentiation. Front Physiol 2021;12:722394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Leibing T, Géraud C, Augustin I, et al. Angiocrine Wnt signaling controls liver growth and metabolic maturation in mice. Hepatology 2018;68(02):707–722 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Manco R, Itzkovitz S. Liver zonation. J Hepatol 2021;74(02):466–468 [DOI] [PubMed] [Google Scholar]
- 21.Paris J, Henderson NC. Liver zonation, revisited. Hepatology 2022;76(04):1219–1230 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Ribatti D Liver angiocrine factors. Tissue Cell 2023;81:102027. [DOI] [PubMed] [Google Scholar]
- 23.MacParland SA, Liu JC, Ma XZ, et al. Single cell RNA sequencing of human liver reveals distinct intrahepatic macrophage populations. Nat Commun 2018;9(01):4383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Su T, Yang Y, Lai S, et al. Single-cell transcriptomics reveals zone-specific alterations of liver sinusoidal endothelial cells in cirrhosis. Cell Mol Gastroenterol Hepatol 2021;11(04):1139–1161 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Song X, Shen Y, Lao Y, et al. CXCL9 regulates acetaminophen-induced liver injury via CXCR3. Exp Ther Med 2019;18(06):4845–4851 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Bonnardel J, T’Jonck W, Gaublomme D, et al. Stellate cells, hepatocytes, and endothelial cells imprint the Kupffer cell identity on monocytes colonizing the liver macrophage niche. Immunity 2019;51(04):638–654.e9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Gainullina A, Mogilenko DA, Huang LH, et al. ; ImmGen Consortium. Network analysis of large-scale ImmGen and Tabula Muris datasets highlights metabolic diversity of tissue mononuclear phagocytes. Cell Rep 2023;42(02):112046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Guilliams M, Bonnardel J, Haest B, et al. Spatial proteogenomics reveals distinct and evolutionarily conserved hepatic macrophage niches. Cell 2022;185(02):379–396.e38 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Guilliams M, Scott CL. Does niche competition determine the origin of tissue-resident macrophages? Nat Rev Immunol 2017;17(07):451–460 [DOI] [PubMed] [Google Scholar]
- 30.Scott CL, Zheng F, De Baetselier P, et al. Bone marrow-derived monocytes give rise to self-renewing and fully differentiated Kupffer cells. Nat Commun 2016;7:10321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.van de Laar L, Saelens W, De Prijck S, et al. Yolk sac macrophages, fetal liver, and adult monocytes can colonize an empty niche and develop into functional tissue-resident macrophages. Immunity 2016;44(04):755–768 [DOI] [PubMed] [Google Scholar]
- 32.Guilliams M, Scott CL. Liver macrophages in health and disease. Immunity 2022;55(09):1515–1529 [DOI] [PubMed] [Google Scholar]
- 33.Andrews TS, Atif J, Liu JC, et al. Single-cell, single-nucleus, and spatial RNA sequencing of the human liver identifies cholangiocyte and mesenchymal heterogeneity. Hepatol Commun 2022;6(04):821–840 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Dobie R, Wilson-Kanamori JR, Henderson BEP, et al. Single-cell transcriptomics uncovers zonation of function in the mesenchyme during liver fibrosis. Cell Rep 2019;29(07):1832–1847.e8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Kostallari E, Wei B, Sicard D, et al. Stiffness is associated with hepatic stellate cell heterogeneity during liver fibrosis. Am J Physiol Gastrointest Liver Physiol 2022;322(02):G234–G246 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Ramachandran P, Dobie R, Wilson-Kanamori JR, et al. Resolving the fibrotic niche of human liver cirrhosis at single-cell level. Nature 2019;575(7783):512–518 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Bellanti F, Vendemiale G. The aging liver: redox biology and liver regeneration. Antioxid Redox Signal 2021;35(10):832–847 [DOI] [PubMed] [Google Scholar]
- 38.Ben-Moshe S, Veg T, Manco R, et al. The spatiotemporal program of zonal liver regeneration following acute injury. Cell Stem Cell 2022;29(06):973–989.e10 [DOI] [PubMed] [Google Scholar]
- 39.Chen Y, Ding BS. Comprehensive review of the vascular niche in regulating organ regeneration and fibrosis. Stem Cells Transl Med 2022;11(11):1135–1142 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Chen Y, Pu Q, Ma Y, et al. Aging reprograms the hematopoietic-vascular niche to impede regeneration and promote fibrosis. Cell Metab 2021;33(02):395–410.e4 [DOI] [PubMed] [Google Scholar]
- 41.Ding BS, Cao Z, Lis R, et al. Divergent angiocrine signals from vascular niche balance liver regeneration and fibrosis. Nature 2014;505(7481):97–102 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Ding BS, Nolan DJ, Butler JM, et al. Inductive angiocrine signals from sinusoidal endothelium are required for liver regeneration. Nature 2010;468(7321):310–315 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Elchaninov A, Vishnyakova P, Menyailo E, Sukhikh G, Fatkhudinov T. An eye on Kupffer cells: development, phenotype and the macrophage niche. Int J Mol Sci 2022;23(17):9868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Große-Segerath L, Lammert E. Role of vasodilation in liver regeneration and health. Biol Chem 2021;402(09):1009–1019 [DOI] [PubMed] [Google Scholar]
- 45.Hu S, Liu S, Bian Y, et al. Single-cell spatial transcriptomics reveals a dynamic control of metabolic zonation and liver regeneration by endothelial cell Wnt2 and Wnt9b. Cell Rep Med 2022;3(10):100754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Huebert RC, Shah VH. Sinusoidal endothelial cells direct traffic at the intersection of regeneration and fibrosis. Hepatology 2014;60(02):754–756 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kim A, Wu X, Allende DS, Nagy LE. Gene deconvolution reveals aberrant liver regeneration and immune cell infiltration in alcohol-associated hepatitis. Hepatology 2021;74(02):987–1002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Ma R, Martínez-Ramírez AS, Borders TL, Gao F, Sosa-Pineda B. Metabolic and non-metabolic liver zonation is established non-synchronously and requires sinusoidal Wnts. eLife 2020;9:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Pellicoro A, Ramachandran P, Iredale JP, Fallowfield JA. Liver fibrosis and repair: immune regulation of wound healing in a solid organ. Nat Rev Immunol 2014;14(03):181–194 [DOI] [PubMed] [Google Scholar]
- 50.Ramasamy SK, Kusumbe AP, Adams RH. Regulation of tissue morphogenesis by endothelial cell-derived signals. Trends Cell Biol 2015;25(03):148–157 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Shido K, Chavez D, Cao Z, Ko J, Rafii S, Ding BS. Platelets prime hematopoietic and vascular niche to drive angiocrine-mediated liver regeneration. Signal Transduct Target Ther 2017;2:16044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Takakura N. Discovery of a vascular endothelial stem cell (VESC) population required for vascular regeneration and tissue maintenance. Circ J 2018;83(01):12–17 [DOI] [PubMed] [Google Scholar]
- 53.Xu F, Hua C, Tautenhahn HM, Dirsch O, Dahmen U. The role of autophagy for the regeneration of the aging liver. Int J Mol Sci 2020;21(10):3606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Zhang XJ, Olsavszky V, Yin Y, et al. Angiocrine hepatocyte growth factor signaling controls physiological organ and body size and dynamic hepatocyte proliferation to prevent liver damage during regeneration. Am J Pathol 2020;190(02):358–371 [DOI] [PubMed] [Google Scholar]
- 55.He J, Deng C, Krall L, Shan Z. ScRNA-seq and ST-seq in liver research. Cell Regen (Lond) 2023;12(01):11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Schoen JM, Wang HH, Minuk GY, Lautt WW. Shear stress-induced nitric oxide release triggers the liver regeneration cascade. Nitric Oxide 2001;5(05):453–464 [DOI] [PubMed] [Google Scholar]
- 57.Shah V, Haddad FG, Garcia-Cardena G, et al. Liver sinusoidal endothelial cells are responsible for nitric oxide modulation of resistance in the hepatic sinusoids. J Clin Invest 1997;100(11):2923–2930 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Vázquez-Chantada M, Ariz U, Varela-Rey M, et al. Evidence for LKB1/AMP-activated protein kinase/endothelial nitric oxide synthase cascade regulated by hepatocyte growth factor, S-adenosylmethionine, and nitric oxide in hepatocyte proliferation. Hepatology 2009;49(02):608–617 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Wan Y, Li X, Slevin E, et al. Endothelial dysfunction in pathological processes of chronic liver disease during aging. FASEB J 2022;36(01):e22125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kron P, Linecker M, Limani P, et al. Hypoxia-driven Hif2a coordinates mouse liver regeneration by coupling parenchymal growth to vascular expansion. Hepatology 2016;64(06):2198–2209 [DOI] [PubMed] [Google Scholar]
- 61.Braun L, Mead JE, Panzica M, Mikumo R, Bell GI, Fausto N. Transforming growth factor beta mRNA increases during liver regeneration: a possible paracrine mechanism of growth regulation. Proc Natl Acad Sci U S A 1988;85(05):1539–1543 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Arab JP, Cabrera D, Sehrawat TS, et al. Hepatic stellate cell activation promotes alcohol-induced steatohepatitis through Igfbp3 and SerpinA12. J Hepatol 2020;73(01):149–160 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Drinane MC, Yaqoob U, Yu H, et al. Synectin promotes fibrogenesis by regulating PDGFR isoforms through distinct mechanisms. JCI Insight 2017;2(24):e92821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Gao J, Wei B, de Assuncao TM, et al. Hepatic stellate cell autophagy inhibits extracellular vesicle release to attenuate liver fibrosis. J Hepatol 2020;73(05):1144–1154 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Kostallari E, Hirsova P, Prasnicka A, et al. Hepatic stellate cell-derived platelet-derived growth factor receptor-alpha-enriched extracellular vesicles promote liver fibrosis in mice through SHP2. Hepatology 2018;68(01):333–348 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Maiers JL, Kostallari E, Mushref M, et al. The unfolded protein response mediates fibrogenesis and collagen I secretion through regulating TANGO1 in mice. Hepatology 2017;65(03):983–998 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Yaqoob U, Luo F, Greuter T, et al. GIPC-regulated IGFBP-3 promotes HSC migration in vitro and portal hypertension in vivo through a β1-integrin pathway. Cell Mol Gastroenterol Hepatol 2020;10(03):545–559 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Gao J, Wei B, Liu M, et al. Endothelial p300 promotes portal hypertension and hepatic fibrosis through C-C motif chemokine ligand 2-mediated angiocrine signaling. Hepatology 2021;73(06):2468–2483 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Hilscher MB, Sehrawat T, Arab JP, et al. Mechanical stretch increases expression of CXCL1 in liver sinusoidal endothelial cells to recruit neutrophils, generate sinusoidal microthombi, and promote portal hypertension. Gastroenterology 2019;157(01):193–209.e9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Gracia-Sancho J, Caparrós E, Fernández-Iglesias A, Francés R. Role of liver sinusoidal endothelial cells in liver diseases. Nat Rev Gastroenterol Hepatol 2021;18(06):411–431 [DOI] [PubMed] [Google Scholar]
- 71.Gracia-Sancho J, Marrone G, Fernández-Iglesias A. Hepatic microcirculation and mechanisms of portal hypertension. Nat Rev Gastroenterol Hepatol 2019;16(04):221–234 [DOI] [PubMed] [Google Scholar]
- 72.Liu S, Premont RT, Rockey DC. Endothelial nitric-oxide synthase (eNOS) is activated through G-protein-coupled receptor kinase-interacting protein 1 (GIT1) tyrosine phosphorylation and Src protein. J Biol Chem 2014;289(26):18163–18174 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Deleve LD, Wang X, Guo Y. Sinusoidal endothelial cells prevent rat stellate cell activation and promote reversion to quiescence. Hepatology 2008;48(03):920–930 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Kostallari E, Shah VH. Pericytes in the liver. Adv Exp Med Biol 2019;1122:153–167 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Tacke F, Puengel T, Loomba R, Friedman SL. An integrated view of anti-inflammatory and antifibrotic targets for the treatment of NASH. J Hepatol 2023:S0168-8278(23)00218-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ruan B, Duan JL, Xu H, et al. Capillarized liver sinusoidal endothelial cells undergo partial endothelial-mesenchymal transition to actively deposit sinusoidal ECM in liver fibrosis. Front Cell Dev Biol 2021;9:671081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Winkler M, Staniczek T, Kürschner SW, et al. Endothelial GATA4 controls liver fibrosis and regeneration by preventing a pathogenic switch in angiocrine signaling. J Hepatol 2021;74(02):380–393 [DOI] [PubMed] [Google Scholar]
- 78.Drzewiecki K, Choi J, Brancale J, et al. GIMAP5 maintains liver endothelial cell homeostasis and prevents portal hypertension. J Exp Med 2021;218(07):e20201745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Xu M, Xu HH, Lin Y, et al. LECT2, a ligand for Tie1, plays a crucial role in liver fibrogenesis. Cell 2019;178(06):1478–1492.e20 [DOI] [PubMed] [Google Scholar]
- 80.Bosch J, Groszmann RJ, 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 2015;62(1, Suppl):S121–S130 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Engelmann C, Clària J, Szabo G, Bosch J, Bernardi M. Pathophysiology of decompensated cirrhosis: portal hypertension, circulatory dysfunction, inflammation, metabolism and mitochondrial dysfunction. J Hepatol 2021;75(Suppl 1):S49–S66 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Gunarathne LS, Rajapaksha H, Shackel N, Angus PW, Herath CB. Cirrhotic portal hypertension: from pathophysiology to novel therapeutics. World J Gastroenterol 2020;26(40):6111–6140 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Iwakiri Y, Trebicka J. Portal hypertension in cirrhosis: pathophysiological mechanisms and therapy. JHEP Rep 2021;3(04):100316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Bhatia R, Matsushita K, Yamakuchi M, Morrell CN, Cao W, Lowenstein CJ. Ceramide triggers Weibel-Palade body exocytosis. Circ Res 2004;95(03):319–324 [DOI] [PubMed] [Google Scholar]
- 85.Riddell DR, Owen JS. Nitric oxide and platelet aggregation. Vitam Horm 1999;57:25–48 [DOI] [PubMed] [Google Scholar]
- 86.Greuter T, Yaqoob U, Gan C, 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 2022;77(03):723–734 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Younossi ZM. Non-alcoholic fatty liver disease - a global public health perspective. J Hepatol 2019;70(03):531–544 [DOI] [PubMed] [Google Scholar]
- 88.Kus E, Kaczara P, Czyzynska-Cichon I, et al. LSEC fenestrae are preserved despite pro-inflammatory phenotype of liver sinusoidal endothelial cells in mice on high fat diet. Front Physiol 2019;10:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Guo Q, Furuta K, Islam S, et al. Liver sinusoidal endothelial cell expressed vascular cell adhesion molecule 1 promotes liver fibrosis. Front Immunol 2022;13:983255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Kawashita E, Ozaki T, Ishihara K, Kashiwada C, Akiba S. Endothelial group IVA phospholipase A2 promotes hepatic fibrosis with sinusoidal capillarization in the early stage of non-alcoholic steatohepatitis in mice. Life Sci 2022;294:120355. [DOI] [PubMed] [Google Scholar]
- 91.Miyao M, Kotani H, Ishida T, et al. Pivotal role of liver sinusoidal endothelial cells in NAFLD/NASH progression. Lab Invest 2015;95(10):1130–1144 [DOI] [PubMed] [Google Scholar]
- 92.Verhaegh P, Wisse E, de Munck T, et al. Electron microscopic observations in perfusion-fixed human non-alcoholic fatty liver disease biopsies. Pathology 2021;53(02):220–228 [DOI] [PubMed] [Google Scholar]
- 93.Francque S, Laleman W, Verbeke L, et al. Increased intrahepatic resistance in severe steatosis: endothelial dysfunction, vasoconstrictor overproduction and altered microvascular architecture. Lab Invest 2012;92(10):1428–1439 [DOI] [PubMed] [Google Scholar]
- 94.Pasarín M, La Mura V, Gracia-Sancho J, et al. Sinusoidal endothelial dysfunction precedes inflammation and fibrosis in a model of NAFLD. PLoS One 2012;7(04):e32785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Wei A, Gu Z, Li J, et al. Clinical adverse effects of endothelin receptor antagonists: insights from the meta-analysis of 4894 patients from 24 randomized double-blind placebo-controlled clinical trials. J Am Heart Assoc 2016;5(11):e003896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Fang ZQ, Ruan B, Liu JJ, et al. Notch-triggered maladaptation of liver sinusoidal endothelium aggravates nonalcoholic steatohepatitis through endothelial nitric oxide synthase. Hepatology 2022;76(03):742–758 [DOI] [PubMed] [Google Scholar]
- 97.Furuta K, Guo Q, Pavelko KD, et al. Lipid-induced endothelial vascular cell adhesion molecule 1 promotes nonalcoholic steatohepatitis pathogenesis. J Clin Invest 2021;131(06):e143690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Miyachi Y, Tsuchiya K, Komiya C, et al. Roles for cell-cell adhesion and contact in obesity-induced hepatic myeloid cell accumulation and glucose intolerance. Cell Rep 2017;18(11):2766–2779 [DOI] [PubMed] [Google Scholar]
- 99.Weston CJ, Shepherd EL, Claridge LC, et al. Vascular adhesion protein-1 promotes liver inflammation and drives hepatic fibrosis. J Clin Invest 2015;125(02):501–520 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Ibrahim SH. Sinusoidal endotheliopathy in nonalcoholic steatohepatitis: therapeutic implications. Am J Physiol Gastrointest Liver Physiol 2021;321(01):G67–G74 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Martin-Armas M, Simon-Santamaria J, Pettersen I, Moens U, Smedsrød B, Sveinbjørnsson B. Toll-like receptor 9 (TLR9) is present in murine liver sinusoidal endothelial cells (LSECs) and mediates the effect of CpG-oligonucleotides. J Hepatol 2006;44(05):939–946 [DOI] [PubMed] [Google Scholar]
- 102.Wu J, Meng Z, Jiang M, et al. Toll-like receptor-induced innate immune responses in non-parenchymal liver cells are cell type-specific. Immunology 2010;129(03):363–374 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Pruenster M, Mudde L, Bombosi P, et al. The Duffy antigen receptor for chemokines transports chemokines and supports their promigratory activity. Nat Immunol 2009;10(01):101–108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Hammoutene A, Biquard L, Lasselin J, et al. A defect in endothelial autophagy occurs in patients with non-alcoholic steatohepatitis and promotes inflammation and fibrosis. J Hepatol 2020;72(03):528–538 [DOI] [PubMed] [Google Scholar]
- 105.Maeso-Díaz R, Gracia-Sancho J. Aging and chronic liver disease. Semin Liver Dis 2020;40(04):373–384 [DOI] [PubMed] [Google Scholar]
- 106.Morsiani C, Bacalini MG, Santoro A, et al. The peculiar aging of human liver: a geroscience perspective within transplant context. Ageing Res Rev 2019;51:24–34 [DOI] [PubMed] [Google Scholar]
- 107.Pinto C, Ninfole E, Gaggiano L, Benedetti A, Marzioni M, Maroni L. Aging and the biological response to liver injury. Semin Liver Dis 2020;40(03):225–232 [DOI] [PubMed] [Google Scholar]
- 108.Zhao Y, Yang Y, Li Q, Li J. Understanding the unique microenvironment in the aging liver. Front Med (Lausanne) 2022;9:842024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Maeso-Díaz R, Ortega-Ribera M, Lafoz E, et al. Aging influences hepatic microvascular biology and liver fibrosis in advanced chronic liver disease. Aging Dis 2019;10(04):684–698 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Goligorsky MS. Microvascular rarefaction: the decline and fall of blood vessels. Organogenesis 2010;6(01):1–10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Hilmer SN, Cogger VC, Fraser R, McLean AJ, Sullivan D, Le Couteur DG. Age-related changes in the hepatic sinusoidal endothelium impede lipoprotein transfer in the rat. Hepatology 2005;42(06):1349–1354 [DOI] [PubMed] [Google Scholar]
- 112.Grunewald M, Kumar S, Sharife H, et al. Counteracting age-related VEGF signaling insufficiency promotes healthy aging and extends life span. Science 2021;373(6554):eabc8479. [DOI] [PubMed] [Google Scholar]
- 113.Hunt NJ, Kang SWS, Lockwood GP, Le Couteur DG, Cogger VC. Hallmarks of aging in the liver. Comput Struct Biotechnol J 2019;17:1151–1161 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Rohn F, Kordes C, Buschmann T, et al. Impaired integrin α5/β1-mediated hepatocyte growth factor release by stellate cells of the aged liver. Aging Cell 2020;19(04):e13131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Warren A, Cogger VC, Fraser R, Deleve LD, McCuskey RS, Le Couteur DG. The effects of old age on hepatic stellate cells. Curr Gerontol Geriatr Res 2011:439835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.McCuskey RS, Bethea NW, Wong J, et al. Ethanol binging exacerbates sinusoidal endothelial and parenchymal injury elicited by acetaminophen. J Hepatol 2005;42(03):371–377 [DOI] [PubMed] [Google Scholar]
- 117.Yang Y, Sangwung P, Kondo R, et al. Alcohol-induced Hsp90 acetylation is a novel driver of liver sinusoidal endothelial dysfunction and alcohol-related liver disease. J Hepatol 2021;75 (02):377–386 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Halpern KB, Shenhav R, Matcovitch-Natan O, et al. Single-cell spatial reconstruction reveals global division of labour in the mammalian liver. Nature 2017;542(7641):352–356 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Novak RF, Woodcroft KJ. The alcohol-inducible form of cytochrome P450 (CYP 2E1): role in toxicology and regulation of expression. Arch Pharm Res 2000;23(04):267–282 [DOI] [PubMed] [Google Scholar]
- 120.Galdieri L, Zhang T, Rogerson D, Lleshi R, Vancura A. Protein acetylation and acetyl coenzyme a metabolism in budding yeast. Eukaryot Cell 2014;13(12):1472–1483 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Kou K, Sun X, Tian G, Zhi Y, Fan Z, Lv G. The mechanisms of systemic inflammatory and immunosuppressive acute-on-chronic liver failure and application prospect of single-cell sequencing. J Immunol Res 2022:5091275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Liu M, Cao S, He L, et al. Super enhancer regulation of cytokine-induced chemokine production in alcoholic hepatitis. Nat Commun 2021;12(01):4560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Neumann K, Erben U, Kruse N, et al. Chemokine transfer by liver sinusoidal endothelial cells contributes to the recruitment of CD4+ T cells into the murine liver. PLoS One 2015;10(06):e0123867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Chang B, Xu MJ, Zhou Z, et al. Short- or long-term high-fat diet feeding plus acute ethanol binge synergistically induce acute liver injury in mice: an important role for CXCL1. Hepatology 2015;62(04):1070–1085 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Pott S, Lieb JD. What are super-enhancers? Nat Genet 2015;47(01):8–12 [DOI] [PubMed] [Google Scholar]
- 126.Ito Y, Bethea NW, Abril ER, McCuskey RS. Early hepatic microvascular injury in response to acetaminophen toxicity. Microcirculation 2003;10(05):391–400 [DOI] [PubMed] [Google Scholar]
- 127.Badmann A, Langsch S, Keogh A, Brunner T, Kaufmann T, Corazza N. TRAIL enhances paracetamol-induced liver sinusoidal endothelial cell death in a Bim- and Bid-dependent manner. Cell Death Dis 2012;3(12):e447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Biagioli M, Marchiano S, di Giorgio C, et al. Combinatorial targeting of G-protein-coupled bile acid receptor 1 and cysteinyl leukotriene receptor 1 reveals a mechanistic role for bile acids and leukotrienes in drug-induced liver injury. Hepatology 2023;78(01):26–44. Doi: 10.1002/hep.32787 [DOI] [PubMed] [Google Scholar]
- 129.Holt MP, Yin H, Ju C. Exacerbation of acetaminophen-induced disturbances of liver sinusoidal endothelial cells in the absence of Kupffer cells in mice. Toxicol Lett 2010;194(1–2):34–41 [DOI] [PubMed] [Google Scholar]
- 130.You Q, Holt M, Yin H, Li G, Hu CJ, Ju C. Role of hepatic resident and infiltrating macrophages in liver repair after acute injury. Biochem Pharmacol 2013;86(06):836–843 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Teratani T, Tomita K, Suzuki T, et al. Free cholesterol accumulation in liver sinusoidal endothelial cells exacerbates acetaminophen hepatotoxicity via TLR9 signaling. J Hepatol 2017;67(04):780–790 [DOI] [PubMed] [Google Scholar]
- 132.Rampes S, Ma D. Hepatic ischemia-reperfusion injury in liver transplant setting: mechanisms and protective strategies. J Biomed Res 2019;33(04):221–234 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Hide D, Warren A, Fernández-Iglesias A, et al. Ischemia/Reperfusion injury in the aged liver: the importance of the sinusoidal endothelium in developing therapeutic strategies for the elderly. J Gerontol A Biol Sci Med Sci 2020;75(02):268–277 [DOI] [PubMed] [Google Scholar]
- 134.Miyashita T, Nakanuma S, Ahmed AK, et al. Ischemia reperfusion-facilitated sinusoidal endothelial cell injury in liver transplantation and the resulting impact of extravasated platelet aggregation. Eur Surg 2016;48:92–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Bautista AP, Spitzer JJ. Platelet activating factor stimulates and primes the liver, Kupffer cells and neutrophils to release superoxide anion. Free Radic Res Commun 1992;17(03):195–209 [DOI] [PubMed] [Google Scholar]
- 136.Jaeschke H Molecular mechanisms of hepatic ischemia-reperfusion injury and preconditioning. Am J Physiol Gastrointest Liver Physiol 2003;284(01):G15–G26 [DOI] [PubMed] [Google Scholar]
- 137.Upadhya GA, Topp SA, Hotchkiss RS, Anagli J, Strasberg SM. Effect of cold preservation on intracellular calcium concentration and calpain activity in rat sinusoidal endothelial cells. Hepatology 2003;37(02):313–323 [DOI] [PubMed] [Google Scholar]
- 138.Guixé-Muntet S, de Mesquita FC, Vila S, et al. Cross-talk between autophagy and KLF2 determines endothelial cell phenotype and microvascular function in acute liver injury. J Hepatol 2017;66(01):86–94 [DOI] [PubMed] [Google Scholar]
- 139.Mandili G, Alchera E, Merlin S, et al. Mouse hepatocytes and LSEC proteome reveal novel mechanisms of ischemia/reperfusion damage and protection by A2aR stimulation. J Hepatol 2015;62(03):573–580 [DOI] [PubMed] [Google Scholar]
- 140.Russo L, Gracia-Sancho J, García-Calderó H, et al. Addition of simvastatin to cold storage solution prevents endothelial dysfunction in explanted rat livers. Hepatology 2012;55(03):921–930 [DOI] [PubMed] [Google Scholar]
- 141.Xin J, Yang T, Wu X, et al. Spatial transcriptomics analysis of zone-dependent hepatic ischemia-reperfusion injury murine model. Commun Biol 2023;6(01):194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Furuta K, Tang X, Islam S, Tapia A, Chen ZB, Ibrahim SH. Endotheliopathy in the metabolic syndrome: mechanisms and clinical implications. Pharmacol Ther 2023;244:108372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Tripathi DM, Rohilla S, Kaur I, et al. Immunonano-lipocarrier-mediated liver sinusoidal endothelial cell-specific RUNX1 inhibition impedes immune cell infiltration and hepatic inflammation in murine model of NASH. Int J Mol Sci 2021;22 (16):8489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Zhang LF, Wang XH, Zhang CL, et al. Sequential nano-penetrators of capillarized liver sinusoids and extracellular matrix barriers for liver fibrosis therapy. ACS Nano 2022;16(09):14029–14042 [DOI] [PubMed] [Google Scholar]
- 145.Marrone G, Maeso-Díaz R, García-Cardena G, et al. KLF2 exerts antifibrotic and vasoprotective effects in cirrhotic rat livers: behind the molecular mechanisms of statins. Gut 2015;64(09):1434–1443 [DOI] [PubMed] [Google Scholar]
- 146.Marrone G, Russo L, Rosado E, et al. The transcription factor KLF2 mediates hepatic endothelial protection and paracrine endothelial-stellate cell deactivation induced by statins. J Hepatol 2013;58(01):98–103 [DOI] [PubMed] [Google Scholar]
- 147.Cui H, Divakaran A, Pandey AK, et al. Selective N-terminal BET bromodomain inhibitors by targeting non-conserved residues and structured water displacement. Angew Chem Int Ed Engl 2021;60(03):1220–1226 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Tateya S, Rizzo NO, Handa P, et al. Endothelial NO/cGMP/VASP signaling attenuates Kupffer cell activation and hepatic insulin resistance induced by high-fat feeding. Diabetes 2011;60(11):2792–2801 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Turaga RC, Satyanarayana G, Sharma M, et al. Targeting integrin αvβ3 by a rationally designed protein for chronic liver disease treatment. Commun Biol 2021;4(01):1087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Kalucka J, de Rooij LPMH, Goveia J, et al. Single-cell transcriptome atlas of murine endothelial cells. Cell 2020;180(04):764–779. e20 [DOI] [PubMed] [Google Scholar]
- 151.Aizarani N, Saviano A, Sagar, et al. A human liver cell atlas reveals heterogeneity and epithelial progenitors. Nature 2019;572(7768):199–204 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Pita-Juarez Y, Karagkouni D, Kalavros N, et al. A single-nucleus and spatial transcriptomic atlas of the COVID-19 liver reveals topological, functional, and regenerative organ disruption in patients. bioRxiv 2022. Doi: 10.1101/2022.10.27.514070 [DOI] [Google Scholar]
- 153.Holland CH, Ramirez Flores RO, Myllys M, et al. Transcriptomic cross-species analysis of chronic liver disease reveals consistent regulation between humans and mice. Hepatol Commun 2022;6 (01):161–177 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Bondareva O, Rodríguez-Aguilera JR, Oliveira F, et al. Single-cell profiling of vascular endothelial cells reveals progressive organ-specific vulnerabilities during obesity. Nat Metab 2022;4(11):1591–1610 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Betsholtz C. Toward a granular molecular-anatomic map of the blood vasculature - single-cell RNA sequencing makes the leap. Ups J Med Sci 2022;127:127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Tabula Muris C, Overall C, Logistical C, et al. ; Tabula Muris Consortium Overall coordination Logistical coordination Organ collection and processing Library preparation and sequencing Computational data analysis Cell type annotation Writing group Supplemental text writing group Principal investigators. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature 2018;562(7727):367–372 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Paik DT, Tian L, Williams IM, et al. Single-cell RNA sequencing unveils unique transcriptomic signatures of organ-specific endothelial cells. Circulation 2020;142(19):1848–1862 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Massalha H, Bahar Halpern K, Abu-Gazala S, et al. A single cell atlas of the human liver tumor microenvironment. Mol Syst Biol 2020;16(12):e9682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Cavalli M, Diamanti K, Pan G, et al. A multi-omics approach to liver diseases: integration of single nuclei transcriptomics with proteomics and HiCap bulk data in human liver. OMICS 2020;24(04):180–194 [DOI] [PMC free article] [PubMed] [Google Scholar]