Skip to main content
American Journal of Physiology - Cell Physiology logoLink to American Journal of Physiology - Cell Physiology
. 2020 May 6;318(6):C1200–C1213. doi: 10.1152/ajpcell.00062.2020

New aspects of hepatic endothelial cells in physiology and nonalcoholic fatty liver disease

Xinghui Sun 1,2,3,, Edward N Harris 1,2,4,
PMCID: PMC7311747  PMID: 32374676

Abstract

The liver is the central metabolic hub for carbohydrate, lipid, and protein metabolism. It is composed of four major types of cells, including hepatocytes, endothelial cells (ECs), Kupffer cells, and stellate cells. Hepatic ECs are highly heterogeneous in both mice and humans, representing the second largest population of cells in liver. The majority of them line hepatic sinusoids known as liver sinusoidal ECs (LSECs). The structure and biology of LSECs and their roles in physiology and liver disease were reviewed recently. Here, we do not give a comprehensive review of LSEC structure, function, or pathophysiology. Instead, we focus on the recent progress in LSEC research and other hepatic ECs in physiology and nonalcoholic fatty liver disease and other hepatic fibrosis-related conditions. We discuss several current areas of interest, including capillarization, scavenger function, autophagy, cellular senescence, paracrine effects, and mechanotransduction. In addition, we summarize the strengths and weaknesses of evidence for the potential role of endothelial-to-mesenchymal transition in liver fibrosis.

Keywords: autophagy, cellular senescence, fibrosis, hepatic endothelium, LSECs, NAFLD, scavenger function

INTRODUCTION

The liver is the central metabolic hub for carbohydrate, lipid, and protein metabolism (160). It can uptake, store, synthesize, metabolize, and export glucose for distal tissues. It can absorb dietary lipids, convert carbohydrates into lipids, synthesize and secrete triglycerides, absorb and synthesize cholesterol, and produce ketone bodies from fatty acids. In addition, it can also produce glucose and ketone bodies using the carbon skeleton of specific amino acids while removing the excess nitrogen through the urea cycle. These metabolic processes are tightly controlled by hormones such as insulin and glucagon. Impaired liver function is involved in the pathogenesis of different diseases, including type 2 diabetes, cardiovascular disease, and cancer (16, 142).

The liver is composed of four major types of cells, including hepatocytes, endothelial cells (ECs), Kupffer cells (KCs), and stellate cells (87). Immunocytochemical staining and quantification revealed that hepatocytes and ECs constitute ∼52% and 22% of all labeled cells, respectively (6). In healthy liver, these cells cooperate to maintain normal liver homeostasis and function. Under pathological conditions, they undergo phenotypic changes at molecular and cellular levels. ECs represent a major subpopulation of nonparenchymal cells, and a majority of them are defined as noncontinuous ECs that line the hepatic sinusoids and are known as liver sinusoidal ECs (LSECs). Liver sinusoidal endothelium is structurally and functionally unique in multiple ways (34, 88, 130, 152): 1) discontinuous and the presence of open pores (see more details in the section of fenestrae and capillarization); 2) the lack of a basement membrane and tight junctions; 3) very high endocytotic capacity; 4) frequently exposed to the high levels of macromolecules, toxins, and waste products; and 5) potent immunological functions. The phenotypic changes of LSECs in nonalcoholic fatty liver disease (NAFLD) and their consequences on NAFLD progression and complications were elegantly reviewed recently (56). In addition to LSECs, there are other types of hepatic ECs such as periportal and pericentral ECs in mice (170) or hepatic artery, central vein, and lymphatic ECs in humans (136) clustered from single-cell RNA sequencing data. In this review, different aspects of hepatic ECs are discussed mainly in NAFLD as well as in other fibrotic liver-related conditions where it is appropriate (Table 1).

Table 1.

The key points of phenotypes and functions of hepatic ECs in pathophysiology

Phenotype and Function Key Points Reference No.
Fenestrae and capillarization Fenestrae are very dynamic with a mean life span of about 18 minutes. The diameter and number of fenestrae vary from species to species and in response to different stimuli. Aging, a major risk factor for NAFLD, is associated with the loss of fenestrae in mice and humans. Fenestrae porosity did not differ in isolated LSECs between lean and obese mice. (13, 75, 94, 107, 110, 176)
Scavenger function Clearance of endogenous and exogenous material that contributes to inflammation is just beginning to be understood in a physiological context. (58, 80, 109, 131, 151, 152)
Autophagy Autophagy is impaired in NASH patients. The deficiency in endothelial autophagy induces liver injury and inflammation and promotes liver fibrosis in mouse NAFLD. (55, 59, 141)
EndMT A small percentage of hepatic ECs undergo mesenchymal transition in mice and humans under disease states. More studies are needed to reveal the functional significance of EndMT in liver fibrosis. (44, 86, 138)
Cellular senescence The knowledge about cellular senescence of hepatic endothelium is very limited in liver diseases; cGAS-STING signaling likely plays a key role; hepatic cellular senescence promotes aging or NAFLD-related liver steatosis. (105, 119, 126, 175)
Mechanotransduction Mechanotransduction in hepatic endothelium induces hepatocyte proliferation and liver growth in mice during development. Pathological mechanical stretching of hepatic endothelium induces the recruitment of neutrophils, which in turn induces microvascular thrombosis and fibrosis. (69, 104)

cGAS-STING, cyclic GMP-AMP (cGAMP) synthase-stimulator of interferon genes; ECs, endothelial cells; EndMT, endothelial-to-mesenchymal transition; LSECs, liver sinusoidal endothelial cells; NAFLD, nonalcoholic fatty liver disease; NASH, nonalcoholic steatohepatitis.

NAFLD

NAFLD is a spectrum of liver diseases in which excess fat builds up in livers without secondary causes of hepatic fat accumulation, such as significant alcohol consumption (18). Scientists have proposed that the name of NAFLD should be changed to obesity-associated fatty liver disease to emphasize the major driver of NAFLD, obesity (150). The early stage of NAFLD is known as steatosis (simple fatty liver) which has been chemically defined as excessive intrahepatic triglyceride content more than 5% of liver volume or liver weight (46). At this stage of NAFLD, there is no evidence of hepatocellular injury in the form of hepatocytes ballooning and no evidence of fibrosis (18). The progressive subtype of NAFLD is nonalcoholic steatohepatitis (NASH). It is characterized by steatosis (≥5%), hepatocyte injury (e.g., ballooning), and lobular inflammation with or without any fibrosis (18). The most advanced stage of NAFLD is NASH cirrhosis, in which liver is scarred and permanently damaged. It is defined as the presence of cirrhosis with current or previous histological evidence of steatosis or steatohepatitis (18). NASH can progress to cirrhosis in about 22% NASH patients with advanced fibrosis (143). While lifestyle modifications can control risk factors, there are no FDA-approved pharmacological therapies available to treat NAFLD (18).

FENESTRAE AND CAPILLARIZATION

Capillarization, also known as defenestration, refers to the loss of fenestrae. Fenestrae are unique morphological features of LSECs in that they are defined as holes that go through the cytoplasm (Fig. 1) and are organized in clusters termed sieve plates (167). They are transmembrane pores with a diameter of 50–150 nm in mouse LSECs. The lifespan, formation, and disappearance of LSEC fenestrae were recently monitored in mouse LSEC culture using atomic force microscopy (176). The major findings are that 1) 75% of the newly formed fenestrae remain open for ≤20 min, whereas the mean fenestrae lifespan is ∼18 min, 2) fenestrae are not static but instead continue to change their positions (≤5 µm) and diameter (≤200%) during their lifespan, 3) some of the fenestrae can close for a while and open again, 4) three distinct ways (the process of formation of new fenestrae becomes slower, the coalescence of neighboring fenestrae lead to the formation of gaps, and fenestrae number may drop because of the formation of defenestration centers) can result in the defenestration of LSECs, and 5) cytochalasin B, which promotes actin depolymerization (106), can induce fenestrae in initially defenestrated cells (176). In addition to atomic force microscopy, fenestrae can be visualized by several other microscopy techniques (113, 114). Mönkemöller et al. (114) resolved structural details of fenestrae in rat LSECs using direct stochastic optical reconstruction microscopy. They demonstrated that this technique can identify individual fenestrae with a diameter of <50 nm. In a more recent study, Mönkemöller et al. (113) combined two superresolution microscopy techniques, direct stochastic optical reconstruction microscopy and three-dimensional structured illumination microscopy, to reveal the new insights of fenestrae. The combination enables the authors to observe the subdiffraction-sized LSEC fenestrae with great detail down to 20 nm. They also found that microtubules appear to surround and separate sieve plates, whereas actin filaments surround each fenestrae within a sieve plate (113). With advances in microscopy techniques, new physiological facets of fenestrae will be revealed in the future.

Fig. 1.

Fig. 1.

Scanning electron microscope image shows mouse liver sinusoids. Liver sinusoidal endothelial cells (LSEC) line hepatic sinusoids. The circle indicates a group of fenestrae that form a sieve plate in the LSEC. Two arrows indicate the space of Disse that is the space between sinusoidal endothelium and microvilli of hepatocytes. This image was obtained under protocol #607 and approved by the Institutional Animal Care and Use Committee on Nov. 23, 2010.

The diameter and number of fenestrae vary from species to species and in response to different stimuli (13). Although aging is not a disease, it is a major risk factor for liver disease (74, 85). Aging is associated with markedly diminished fenestrae porosity, increased hepatic vascular resistance, and elevated portal pressure (107). Isolated LSECs from old mice and rats have reduced fenestrae porosity (75, 96). Aging in humans is also associated with the loss of fenestrae in the sinusoidal endothelium (110). Although aging correlates with the loss of fenestrae, age-matched animals fed a high-fat diet (HFD) do not appear to affect the numbers of fenestrae. In a mouse model of diet-induced obesity, LSEC fenestrae are functionally preserved in either the early (2–8 wk) or late (15–20 wk) stages of NAFLD progression, despite LSECs displaying a proinflammatory phenotype (94). Overall, LSEC fenestrae porosity did not differ between lean and obese mice. However, the mean diameter of fenestrae was larger in the obese mice fed a HFD (60 kcal% fat) to induce NAFLD. LSEC morphology was visualized using scanning electron microscopy and atomic force microscopy in primary cells within 24 h from the isolation. It remains unknown whether fenestrae would change during cell isolation that may affect the overall results.

There are several signaling pathways involved in capillarization. Delta-like 4, a ligand of the Notch signaling pathway, was upregulated in the LSECs of human and carbon tetrachloride (CCl4)-induced murine fibrotic liver (21). It mediates LSEC capillarization and the vicious cycle between fibrosis and pathological sinusoidal remodeling (21). Bone morphogenetic protein 9 (BMP9) is a circulating factor produced by hepatic stellate cells. Loss of BMP9 led to the expression of several terminal differentiation markers such as LYVE1 and Stab2 and triggered hepatic perisinusoidal fibrosis (38). In addition, LSECs from BMP9 knockout mice had a reduced number of fenestrae, indicating that they are capillarized (38). These data demonstrated that BMP9 protects against hepatic fibrosis by influencing LSEC fenestration.

The strategies to induce the reformation of fenestrae have been explored by several groups. Metformin treatment caused an increase in fenestrae porosity in LSECs isolated from both young and old mice without altering fenestrae diameter (75). The use of 0.1% metformin diet for 12 mo reduced the age-related loss of fenestrae porosity and frequency by 50% in mice (75). Agents that target nitric oxide, actin, or lipid rafts promote changes in fenestrae in murine LSECs (76). Interestingly, LSECs from young and old mice respond differently to these agents. Bosentan, an endothelin receptor agonist used in the treatment for pulmonary hypertension, and 2,5-dimethoxy-4-iodoamphetamine, a hallucinogen that activates neurotrophin receptors, can increase fenestration porosity and/or frequency in old mice only (76). LSECs lose their fenestrae in cell culture in a time-dependent manner once they are isolated from rodents (12). Interestingly, actin depolymerization by cytochalasin D can induce fenestrae reformation in dedifferentiated LSECs devoid of fenestrate (39). These studies demonstrated that age-related defenestration can be reversed pharmacologically.

LSECs SCAVENGER FUNCTION AND THE ROLE OF LSECs IN INFLAMMATORY RESPONSE

Fenestrated LSECs allow large macromolecules into the Space of Disse; however, LSECs also have a high surface area and are covered with various scavenger receptors to recognize danger-associated molecular patterns along with a host of indigenous and foreign molecules. LSECs are among of the most endocytic cells of the mammalian system (146) and are unique in that their endocytic vesicles retain their clathrin coat longer than in any other cells. Endocytic vesicles are conveyed along a network of microtubules in which cargo is delivered to a robust lysosomal network to degrade cargo (47). The scavenger receptors at the cellular surface include SR-A (MSR1, SR-A1), SR-B (SR-B1, SCARB1), SR-E1 (LOX1), SR-H (stabilin-1, stabilin-2/HARE), FcγRII/CD32b, CD206/mannose receptor, and C-type lectin receptors (L-SIGN, LSECTin). Some of these receptors, such as SR-H, are constitutive and do not change significantly in their expression, even in chronic diseased states (8). However, some diseased states induce higher expression of these receptor and some of their splice variants. These would include SR-B, which increases in expression during steatohepatitis (134, 137), and SR-E under similar conditions in which there is hypercholesterolemia and hyperlipidemia (19, 22). It should be noted that LSECs take up molecules and small particles amenable for pinocytosis (<100 nm), and larger particles are primarily taken up by KCs (149, 151) in a coordinated fashion. Ligands for the known scavenger receptors expressed by LSECs are extensive and beyond the scope of this review. For suitable references on these ligands, the reader is invited to obtain this information from Refs. 34, 58, 80, 109, 131, and 152.

During NAFLD or lipotoxic conditions, many cells of the liver increase their expression of tumor necrosis factor receptors (31, 171); however, a coordinated effort between the nonparenchymal cells of the liver and immune cells promotes tolerance for foreign antigens (158). Fas receptor, which is normally absent on LSECs, is expressed during lipotoxic conditions to bind with activated immune cells expressing Fas ligand (37, 168). LSECs have a higher tolerance than hepatocytes and stellate cells to resist undergoing apoptosis under these conditions and instead start expressing ICAM-1 and VCAM-1 (7, 17) in addition to the constitutive, vascular adhesion protein-1 (VAP-1) (95). These adhesion molecules are thought to recruit KCs to clean up cellular debris from apoptotic hepatocytes and other cells. Without this coordinated effort between KCs and LSECs, the viability of LSECs decreases (70, 77). Overall, LSECs promote tolerance and immune dampening through their expression of programmed death ligand-1 (PD-1L). The interaction of programmed cell death protein 1 (PD-1) expression on KCs and CD8+ cells promotes immune tolerance, as the blood supply for the liver is coming from the gut circulation via the portal vein and contains a host of immune stimulatory molecules such as lipopolysaccharides and food antigens (82). PD-1L/PD-1 interactions between LSECs and TH1/TH17 cells, respectively, promote suppression of cytokine release from these cells. However, immunotolerance does have its limits, and a breach beyond this undefined threshold stimulates activation of the SHP-1 and SHP-2 phosphatases through the FAS and PD-1 pathways to promote activation of immune cells and cytokine release (120, 148). More detailed and in-depth information on LSEC activation in NAFLD was reviewed by Hammoutene and Rautou (56) and neutrophil recruitment in the liver by Liew and Kubes (102) and Zindel and Kubes (181).

Quiescence in the liver is promoted by the production of nitric oxide and hepatocyte growth factor (HGF) secreted by LSECs and VEGF secreted by hepatocytes (33, 93, 159). Cross-talk between liver cell types typically dampens inflammatory responses, but it may also stimulate repair and cleanup operations by immune cells. For example, lipopolysaccharide-stimulated KCs secrete prostaglandin E2, which is a suppressor of T cell receptor signaling. In this setting, it helps protect and increase the survival of LSECs against the harmful effects of ischemia-reperfusion (I/R) (2, 172). Naturally, during an acute injury such as I/R, which leads to increased reactive oxygen species and inflammation (79) and reduced nitric oxide (122), the endocannabinoid system is involved and upregulated in both parenchymal and nonparenchymal cells. The cannabinoid CB2 receptor acts to dampen the damage of the inflammatory response. Activation of CB2 through a synthetic agonist, JWH133, decreased levels of alanine aminotransferase and aspartate aminotransferase, hematopoietic markers of liver damage. Similarly, activation of CB2 counteracted TNFα and lowered ICAM-1 and VCAM-1 expression in LSECs, which lowered the levels of neutrophil adhesion and possibly invasion within the parenchyma (7). On the other end, an antagonist of CB1 with rimonabant (SR141716) was hepatoprotective by decreasing inflammation through TNFα in a hepatic steatosis rat model (51). Although CB1 is found predominantly in the brain, it still has effects in liver that may be induced during specific injuries. For example, CB1 is involved with liver regeneration and may cause scarring or fibrosis if inflammation is not controlled (115, 132). Generally, agonists of CB2 and antagonists of CB1 are beneficial for liver health. Effective therapeutic treatment of chronic (steatosis) or acute (I/R) liver disease may be managed by stimulating the endocannabinoid system, although such efforts are still controversial due to side effects in other organ systems and unintended behavioral consequences (5, 68, 108, 157).

AUTOPHAGY

Autophagy (the elimination/recycling of organelles) is critical for the proper functionality of the liver in physiological conditions, and defects in autophagy cause liver disease (59). The important role of autophagy in hepatic adaptation to stress was reviewed recently (59). In this section, we will focus on autophagy in hepatic ECs (Fig. 2A).

Fig. 2.

Fig. 2.

Hepatic endothelial cells (ECs) undergo phenotypic changes under disease condition. A: liver sinusoidal endothelial cells (LSEC) have impaired autophagy in nonalcoholic fatty liver disease (NAFLD). The loss of fenestrae is also known as capillarization or defenestration. B: hepatic ECs undergo cellular senescence in different liver diseases. The role of the cCAG-STING axis in the cellular senescence of hepatic ECs has not been examined, while it plays an important role in the cellular senescence of other cell types. It is very likely that cyclic GMP-AMP (cGAMP) synthase (cGAS)-stimulator of interferon genes (STING) axis is activated in hepatic ECs upon DNA damage or mitochondrial stress in different liver diseases. C: hepatic ECs undergo endothelial to mesenchymal transition (EndMT) under disease conditions. It is interesting to compare mitochondrial respiration and substrate preference for ATP production before and after EndMT. AC: hepatic ECs can affect the function of hepatocytes and hepatic stellate cells through paracrine effects.

Endothelial autophagy plays a protective role in both acute and chronic liver injury. In patients with nonalcoholic steatohepatitis (a severe form of NAFLD), LSECs contain fewer and smaller autophagic vacuoles than in patients with no or mild histological abnormalities or with simple steatosis (55). In vitro mechanistic studies revealed that the deficiency in autophagy in hepatic endothelium induces endothelial inflammation, endothelial-to-mesenchymal transition (EndMT), and EC apoptosis. These in vitro experiments were performed in the immortalized murine liver EC line (transformed LSECs). Evidence from primary LSECs will strengthen the results (55). In a mouse model of NAFLD, the deficiency in endothelial autophagy induces liver injury and inflammation and promotes liver fibrosis (55). In rats with mild acute liver injury induced by CCl4, endothelial autophagy was upregulated (141). To examine the function of endothelial autophagy during acute liver injury, EC-specific Atg7 knockout mice were treated with CCl4. Their LSECs exhibited a reduction in the number of fenestrae, increased oxidative stress, decreased nitric oxide bioavailability, and aggravated liver fibrosis. In an acute liver injury induced by cold ischemia-reperfusion, endothelial autophagy improved cell viability and ameliorated hepatic damage and microvascular function (54). These data demonstrate that endothelial autophagy protects liver from injury during the early stages of liver diseases.

The autophagy of mitochondria, also known as mitophagy, has not been examined in hepatic endothelium. However, mitochondrial energetics was recently examined in LSECs isolated from lean and obese mice (94). Basal glycolysis and maximal glycolysis capacity were increased in LSECs from obese mice after 2 wk of HFD, while mitochondrial respiration was impaired in LSECs from mice after 4 wk of HFD. Mitochondrial oxidative phosphorylation was impaired, as evidenced by decreased ATP production, increased maximal mitochondrial respiration, and increased proton leak. Interestingly, mitochondrial bioenergetics were not changed at any other examined time points, including 8, 15, and 20 wk of HFD. The relative contribution of mitochondrial respiration and glycolysis to ATP production was not compared in this study, although it is known that LSECs contain few mitochondria (11) and are not highly glycolytic like other ECs (30, 140, 154). The experiments were conducted in LSECs within 24 h from the isolation using CD146 immunomagnetic sorting. However, the in vitro culture condition may not reflect in vivo status. Improving our understanding of mitophagy in hepatic ECs may provide new insights for targets against liver disease.

ENDOTHELIAL TO MESENCHYMAL TRANSITION

Liver fibrosis is a pathology caused by chronic infection or metabolic stress that is observed in different liver diseases such as alcoholic liver disease, NAFLD, and viral hepatitis. It may lead to liver cirrhosis or hepatocellular carcinoma that eventually causes the death of patients. Liver stellate cells are traditionally considered as the major cellular source of myofibroblasts that drive liver fibrosis during liver injury (67, 133). In the CCl4-induced liver injury model, more than 87% of myofibroblasts originate from hepatic stellate cells (78). In a separate study, it was demonstrated that hepatic stellate cells give rise to 82–96% of myofibroblasts in bile duct ligation-induced, CCl4-induced, or methionine- and choline-deficient diet-induced liver fibrosis (111). Recently, it has become appreciated that ECs also contribute to liver fibrosis through a process known as EndMT (Fig. 2C). It describes a process in which an EC undergoes transcriptomic reprograming that leads to phenotypic changes toward a mesenchymal cell (90, 129). It is a very complex biological process in which ECs lose EC phenotype and functions and acquire mesenchymal cell morphology and functions. At molecular levels, ECs lose the expression of EC markers such as platelet-EC adhesion molecule, von Willebrand factor, and vascular-endothelial cadherin, while they gain the ability to produce mesenchymal proteins, including α-smooth muscle actin (α-SMA), N-cadherin, vimentin, fibroblast-specific protein-1 (also known as S100A4 protein), fibroblast activating protein, and fibrillar collagens type I and type III. Many transcription factors, noncoding RNAs, and different signaling pathways participate in EndMT. It plays an important role in organ development and remodeling as well as the pathogenesis of human diseases involving fibrosis.

There are different methods that can be used to detect EndMT in vivo, such as simple immunostaining and genetic lineage tracing with inherent limitations (90). Immunostaining cannot identify cells that have substantially reduced or lost EC marker expression, while genetic lineage tracing such as Tie2-lineage cells can label a very small population of nonendothelial hematopoietic cells. These methods revealed that EndMT is present in mice and humans during liver injury. A very small subpopulation of liver ECs from cirrhotic patients and mice with liver fibrosis undergo EndMT (86, 138). Liver ECs from cirrhotic patients with alcohol-induced liver disease and hepatitis C virus are α-SMA positive, indicating an EndMT phenotype. In patients with idiopathic portal hypertension, double immunofluorescence staining revealed the co-localization of CD34 and S100A4 or CD34 and collagen (COL1A1) in the portal venous endothelium, indicating the presence of EndMT (86). In a mouse model of liver fibrosis induced by intraperitoneal injection of CCl4, α-SMA expression was detected in CD31+ cells in parenchymatous areas (138). In the same study, genetic lineage-tracing revealed that about 3.8% of the tdTomato positive cells express mesenchymal markers in the livers of Tie2-tdTomato mice (138). This demonstrated the direct evidence of in vivo EndMT in mouse fibrotic livers. However, the percentage of myofibroblasts originating from hepatic ECs remains unknown, which very likely is not high because hepatic stellate cells account for 87% or 82–96% of myofibroblasts in different mouse models of fibrosis (78, 111). In other mouse models of fibrotic diseases, where EndMT is a key contributor to fibrosis, the percentage of EndMT-derived cells varies among the fibroblast population (90). In advanced atherosclerosis, EndMT-derived cells comprise almost half of the fibroblast population (45). In cardiac overload and fibrosis, EndMT-derived cells (Tie1 was used as endothelial marker) contribute to about 27–33% of cardiac fibroblasts (177).

Despite the small percentage of hepatic ECs that undergo mesenchymal transition, hepatic ECs could play important roles in liver fibrosis. For example, the endothelial transcription factor ETS-related gene (ERG) promotes the SMAD1 pathway, while it represses SMAD2/3 activity in vitro and in vivo (44), loss of endothelial ERG results in EndMT and spontaneous liver fibrosis in mouse models, ERG expression was reduced in CD31-positive cells from human fibrotic liver specimens, and importantly, loss of endothelial ERG correlates with EndMT and liver fibrosis in patients (44), indicating that ERG likely inhibits EndMT and liver fibrosis in end-stage human liver disease. Second, chromatin remodeler controls EndMT in liver fibrosis, brahma related gene 1 (Brg1) is a chromatin remodeling protein, and endothelial Brg1 deficiency attenuates bile duct ligation-induced EndMT and liver fibrosis in mice by reducing NADPH oxidase 4 expression and reactive oxygen species production (101). Third, the transcriptional modulator megakaryocytic leukemia 1 (MKL1) activates EndMT by activating TWIST1 expression in ECs, a master regulator of EndMT (100); EC-specific deletion of MKL1 suppresses EndMT and attenuates liver fibrosis in mice (100). In the last two studies (100, 101), primary LSECs isolated from Brg1 or MKL1 EC knockout mice display higher expression of EC markers and lower expression of mesenchymal markers at mRNA levels in liver fibrosis induced by bile duct ligation, and there was no other in vivo evidence examined for the role of Brg1 or MKL1 in EndMT. In all three studies (44, 100, 101), the potent effects of different genes on liver fibrosis could be attributed to EndMT-independent mechanisms or the paracrine effects of these EndMT-derived cells. Further studies are required to tease it out. In summary, there is evidence supporting that hepatic ECs undergo EndMT during fibrosis in human liver disease and animal studies. However, more studies are needed to demonstrate the significant functional effects of EndMT on liver fibrosis development. Improving our understanding of EndMT in liver fibrosis could provide new therapeutic targets for liver disease.

CELLULAR SENESCENCE

Cellular senescence is a stable form of cell cycle arrest characterized by different features such as DNA damage response, mitochondrial dysfunction, an increase in the expression of cyclin-dependent kinase inhibitors like p16 and p21, an increase in senescence-associated secretory phenotype (SASP), and an increase in senescence-associated β-galactosidase activity (SA-β-Gal) (4, 53, 63, 65). Cells become senescent in response to different stresses such as persistent DNA damage and oxidative stress (50, 63, 139). This is known as premature senescence, which is different from replicative senescence caused by telomere shortening (92). Although cell senescence can exert beneficial effects in human health and disease (36, 48, 116, 144, 155), it plays a causative role in the pathogenesis of obesity and associated vascular complications (14, 23, 84, 112, 117, 119, 123, 124, 164). Senescent cells are found in obesity (14, 112, 119, 164), diabetes (14, 123), and atherosclerosis (23). How to safely counter the detrimental effects of cellular senescence is an emerging research area of great interest (15, 24, 81, 121, 124, 162). For example, elimination of senescent cells improved glucose tolerance, enhanced insulin sensitivity, reduced macrophage accumulation in adipose tissue, lowered circulating inflammatory mediators, and promoted adipogenesis in obese mice (124). In the first clinical trial of senolytics, the combination of Dasatinib and Quercetin improved physical function in patients with idiopathic pulmonary fibrosis (81) and reduced adipose tissue senescent cell burden within 11 days in patients with diabetic kidney disease (66).

The role of senescent cells in liver diseases was recently summarized in two review papers with minimal information about the cellular senescence of hepatic endothelium (73, 126). Hepatocyte senescence was observed in acute live injury induced by acetaminophen or CCl4 (10) and in nonalcoholic fatty liver disease (119) in mice. The accumulation of senescent hepatocytes promotes hepatic fat accumulation and steatosis, because mitochondria lose the ability to metabolize fatty acids efficiently (119). Hepatocyte senescence was also observed in patients with NAFLD (3). Importantly, hepatocyte senescence correlated closely with fibrosis stage, diabetes mellitus, and clinical outcome; an adverse liver-related outcome was strongly associated with higher hepatocyte p21 expression (3). Whereas the cellular senescence of hepatocytes aggravates fatty liver, senescent hepatic stellate cells appear to limit liver fibrosis. Krizhanovsky et al. (91) reported that in response to liver injury, senescent hepatic stellate cells accumulate in fibrotic livers to limit the extent of fibrosis. They found that if hepatic stellate cells were deficient in key senescent genes in the p53 or retinoblastoma protein pathways, these cells continued to proliferate and secrete excessive extracellular matrix components. The senescence of activated hepatic stellate cells also facilitated the resolution of fibrosis upon withdrawal of CCl4. In this study, the authors also demonstrated that these senescent hepatic stellate cells stimulate immune surveillance, leading to the clearance of these senescent cells by natural killer cells. These findings suggest that senescent hepatic stellate cells are beneficial for the injured liver when the production of senescent cells does not outpace their clearance.

As discussed above and reviewed by others, the important roles of cellular senescence of hepatocytes and hepatic stellate cells have been revealed by many studies in liver diseases. In contrast, we barely know anything about the role of cellular senescence of hepatic endothelium or LSECs, although cellular senescence has been broadly studied in ECs. Aging-related senescent cells are observed in many organs, including liver (163). γ‐H2AX (phosphorylated H2AX at Ser139) is a molecular marker of DNA damage and used to identify senescent cells. In liver, the frequency of γ‐H2AX foci‐positive cells increases significantly with age, and the vast majority of foci‐positive cells in liver are hepatocytes, as judged by morphological criteria (163). Senescent cells induce senescence in bystander cells in vitro (118) and in vivo (27). We speculate that a subpopulation of hepatic ECs or LSECs is senescent with aging. The LSECs-related phenotypic changes during aging were reviewed previously (74). These changes are reduction in the number and size of fenestrations, thickening of the endothelium, deposition of basal lamina and collagen, altered expression of antigens such as von Willebrand factor, CD31 and collagen, and increased perisinusoidal staining with Masson’s Trichrome and Sirius Red. These changes are observed in rodents, nonhuman primates, and humans (25, 26, 74, 107, 110). In addition, the endocytotic activity is impaired with aging. In vitro culture of rodent LSECs causes a rapid loss of the EC phenotype and an onset of spontaneous cellular senescence several days after isolation, which is blocked by the deletion of p19ARF (89). Whether p19ARF is involved in the cellular senescence of hepatic endothelium remains unknown in liver diseases.

What signaling pathways promote senescence in hepatic ECs? The role of p53‐p21 and p16‐retinoblastoma protein pathways in cellular senescence has been well established (20, 126). Below, we briefly review the cytoplasmic DNA-sensing pathway in cellular senescence (Fig. 2B). Cyclic GMP-AMP (cGAMP) synthase (cGAS) is a cytosolic DNA sensor that is activated by binding to double-stranded DNA (156). Activated cGAS catalyzes the synthesis of cGAMP by using ATP and GTP as substrates. cGAMP acts as a second messenger to bind and activate the adaptor protein stimulator of interferon genes (STING). This triggers a signaling cascade that leads to the production of type I interferons. Both DNA damage and mitochondrial damage can activate cGAS-STING signaling (99). Nuclear DNA damage causes the accumulation of cytoplasmic DNA, notably in the form of micronuclei. The rupture of micronuclei exposes genomic DNA to cytosolic cGAS. This activates cGAS-STING signaling and type I interferon response. Indeed, DNA damage activates cell-autonomous induction of IFN-β (174). DNA damage-induced IFN-β promotes cell growth arrest and senescence, which is attenuated by anti-IFN-β antibody or knockdown of IFN-β receptor. Mitochondrial stress leads to the release of mitochondrial DNA into the cytosol (166) and activates DNA sensor cGAS that potentiates type I interferon responses and elevates interferon-stimulated gene expression (166). This gene expression profile confers broad viral resistance, indicating that mitochondria play important roles in innate immunity.

The role of cGAS in mediating cellular senescence has been demonstrated in vitro and in vivo (43, 52, 173). In human and mice fibroblasts, cGAS deletion abrogates cell senescence phenotypes induced by replication, DNA damage, or ionizing radiation (173). Increased SA-β-Gal activity and SASP are two important features of senescence, among others, and both are reduced in cGAS-deficient cells (173). Studies from two more groups also demonstrate that different stressors such as oxidative stress and activation of oncogenes can trigger cellular senescence, depending upon cGAS-STING signaling to drive the production of SASP components not only in cells but also in mice (43, 52). Interestingly, cGAS can be recruited to chromosomes during mitosis (57, 173). The functional consequences of exposing cGAS to chromosomes upon mitotic nuclear envelope breakdown were examined recently (180). In normal mitosis, nucleosomes suppress cGAS, so cells divide without cGAS activation. When cells are arrested in mitosis, cGAS-dependent IRF3 activation triggers intrinsic apoptosis by inducing mitochondrial outer membrane permeabilization.

STING-deficient mice carry a missense mutation (I199N) in the STING protein rendering the protein inactive. These HFD-fed STING-deficient mice display higher insulin sensitivity, less hepatic steatosis, and fibrosis (175). A similar study showed that STING-deficient mice developed less severe hepatic steatosis, inflammation, and/or fibrosis in mice with NAFLD or NASH than control mice (105). Bone marrow transplantation revealed that myeloid cell-specific STING disruption leads to similar phenotypes, while myeloid reconstitution of STING has an opposite effect. Mechanistically, STING disruption in macrophages reduces macrophage pro-inflammatory activation, which exerts paracrine effects on hepatocytes by decreasing hepatocyte fat accumulation, pro-inflammatory responses, and the expression of genes involved in fatty acid synthesis. Moreover, cell-autonomous and cell-nonautonomous STING signaling promote the activation of hepatic stellate cells. These data demonstrate that STING plays an important role in regulating diet-induced hepatic steatosis, inflammation, and liver fibrosis. STING expression is increased in livers of human patients with NAFLD and in livers of mice with HFD-induced steatosis (105). Interestingly, it was found that hepatocytes in livers of adult mice did not express STING protein (175). In contrast, STING was expressed in KCs and hepatic ECs (175). Thus, the role of endothelial cGAS-STING signaling axis in hepatic senescence, steatosis, and fibrosis warrants further investigation (Fig. 2B).

PARACRINE EFFECTS OF LSEC

Perturbed EC communication with other cell types plays a key role in the development of diseases such as obesity (98, 127, 128). In liver, ECs can impair insulin action in hepatocytes via tyrosine nitration of insulin receptors, leading to impaired ability of insulin to suppress glucose production (161). Here, we will briefly discuss how LSECs exert paracrine effects on the function of hepatocytes and hepatic stellate cells.

LSECs protect hepatocytes and promote hepatocyte proliferation in partially hepatectomized mice through the release of angiocrine factors Wnt2 and hepatocyte growth factor (42). In a separate study, LSEC-derived angiopoietin-2 displays a dynamic expression pattern after partial hepatectomy (72). In the early inductive phase of liver regeneration, the reduction of angiopoietin-2 enables hepatocyte proliferation through the release of an angiocrine proliferative brake. In the later angiogenic phase of liver regeneration, the expression of angiopoietin-2 was restored in LSEC, which enables regenerative angiogenesis by controlling LSEC vascular endothelial growth factor receptor 2 expression and signaling (72).

Healthy LSECs prevent hepatic stellate cells activation through nitric oxide (35). Moreover, healthy LSECs promote the reversion of activated hepatic stellate cells to quiescence (169), although the paracrine mediator has not been identified. Divergent angiocrine signals from LSECs stimulate regeneration after immediate injury and provoke fibrosis after chronic insult (41). After chronic injury, LSECs promote liver fibrosis by activating hepatic stellate cells. This profibrotic pathway is driven by fibroblast growth factor receptor 1 and C-X-C motif chemokine receptor 4 angiocrine pathway that counteracts the pro-regenerative response (41). EC-derived exosomes regulate hepatic stellate cell function in a paracrine manner (165). Exosomes are cell-derived extracellular vesicles that, in some cases, promote intercellular or interorgan communication. EC-derived exosomes contain the sphingosine kinase 1 protein (165). After being delivered to hepatic stellate cells, the sphingosine kinase 1 protein increased the levels of sphingosine-1-phosphate and Akt phosphorylation, resulting in HSC activation and migration in vitro. In a murine model of liver fibrosis, sphingosine kinase 1 was upregulated in exosomes derived from CCl4-treated mice compared with the control group, which may activate HSC and contribute to liver fibrosis in vivo (165).

MECHANOTRANSDUCTION

The effects of mechanical force on the functional properties of extrahepatic vascular endothelium have been well demonstrated, as previously reviewed (29, 60, 179), while the role of hepatic endothelium in mechanical sensing is less well understood (83, 153). LSECs are subjected to two major forces (135): 1) cyclic stretch resulted from mechanical distortion and tension of the vessel wall and 2) fluid shear stress caused by blood flow. Studies have demonstrated LSECs as important sensors that link mechanical forces to LSEC function and liver homeostasis. Lorenz et al. (104) found that vascular perfusion induces the proliferation of hepatocytes and the growth of liver in mice during development (Fig. 3A). Mechanistically, vascular perfusion leads to the mechanotransduction in hepatic endothelium, which in turn induces production of HGF through the β1-integrin-VEGFR3 signaling axis. HGF is one of the key angiocrine signals required for liver growth and survival. Mechanical stretching also induces HGF expression by activating the β1-integrin-VEGFR3 signaling axis in hepatic ECs in vitro, which is necessary and sufficient to promote hepatocyte proliferation. The authors also found that mechanical forces in the blood vascular system are associated with liver size in metabolically healthy individuals. These data demonstrated that mechanical forces generated by vascular perfusion in liver vessels play key roles in promoting embryonic liver development (Fig. 3A). Whether it also contributes to regeneration in adult liver remains unexamined. Recently, the impact of pathologic levels of mechanical stretch on LSEC signaling was examined in portal hypertension (Fig. 3B) (69), a common sequelae of chronic liver disease. Mechanical stretch of LSECs induces Notch-dependent upregulation and secretion of the neutrophil chemotactic chemokine CXCL1, which in turn recruit neutrophils in vitro and in vivo. These neutrophils interact with platelets in liver sinusoids, leading to the formation of prothrombotic neutrophil extracellular traps that contribute to microvascular thrombosis, fibrosis, and portal hypertension in different mouse models of liver disease (Fig. 3B). These results revealed a novel pathway of endothelial mechanocrine signaling upon pathological mechanical stretching within hepatic sinusoids that participates in the liver pathophysiology.

Fig. 3.

Fig. 3.

Mechanotransduction in liver sinusoidal endothelial cells (LSECs). A: mechanotransduction in LSECs induces hepatocyte proliferation and liver growth in mice during development. B: pathological mechanical stretching of LSECs induces the recruitment of neutrophils, which in turn induces microvascular thrombosis and fibrosis and promotes portal hypertension. CXCL1, C-X-C motif chemokine ligand 1; HGF, hepatocyte growth factor; NETs, neutrophil extracellular traps; VEGFR3, vascular endothelial growth factor receptor 3.

CHALLENGES AND FUTURE DIRECTIONS

The progression of chronic liver disease involves a complex interplay among the hepatic ECs, KCs, stellate cells, and hepatocytes. In rodent models, hepatic ECs are known to regulate fibrogenesis and cirrhosis. In human livers, certain subpopulations of ECs express pro-fibrogenic genes and inhabit the fibrotic niche in cirrhotic livers (136). This indicates that hepatic ECs are important regulators of human liver fibrosis and cirrhosis. However, our understanding of hepatic ECs in liver disease is very limited. Below, we discuss some challenges and future directions about the heterogeneity of hepatic ECs, cellular senescence, mechanotransduction, and the role of noncoding RNAs.

Hepatic ECs are highly heterogeneous in both mice and humans, representing the second largest population of cells in liver revealed by single-cell RNA sequencing (136, 170). In mice, liver ECs can be divided into four sub-clusters representing periportal and pericentral ECs and two clusters of LSECs that line the surface of liver sinusoids (170). In humans, clustering of liver ECs identified seven subpopulations, including LSEC, hepatic artery, central vein, lymphatic, two clusters of disease-specific ECs, and one unannotated cluster (136). The heterogeneity of liver ECs revealed by the studies raise two challenges for future studies. First, identification of common molecular signatures that can be used to cluster mouse and human liver ECs is important for improving our understanding about liver EC function in chronic liver disease. Second, generation of LSEC-specific Cre mouse line is important for loss-of-function studies in mice. The common approach to knock out a floxed gene of interest in hepatic endothelium is to breed floxed mice with VE-cadherin promoter-driven Cre mouse line (55, 141). However, this strategy cannot differentiate the heterogeneous population of hepatic ECs.

It is increasingly recognized that cell senescence is heterogeneous and cell specific with pleiotropic function (6164, 97). There are no specific markers of cellular senescence, and a combination of several markers are often required to identify senescent cells (53). SA-β-Gal activity and expression of p16 are the most commonly used senescence markers (32, 40, 103, 147). However, p16 is not expressed by all senescent cells, while it is also expressed in nonsenescent cells and SA-β-Gal is neither required nor a determinant of the senescent phenotype (53). It is particularly challenging to identify, quantify, and characterize senescent cells in human tissues by SA-β-Gal staining because it is not suitable for fixed tissues or by p16 staining because of the lack of reliable antibodies. Recently, a quantitative approach has been established to identify and characterize senescent cells in aging and disease using ImageStreamX (9, 49). This new approach combines the quantitative power of flow cytometry with high content image analysis. It detects SA-β-gal activities using the bright-field channel and additional senescence-related markers such as p16 and Ki67 (cell proliferation markers) via multiple fluorescence channels (49). It overcomes an important limitation of standard methods by detecting several senescence markers at a time. This also helps identify the specific cell origin of senescent cells (49). However, it requires the specific instrument that is the ImageStreamX imaging flow cytometer.

The mechanobiology in LSECs is an exciting research topic that can potentially provide novel therapeutic targets for chronic liver disease (83, 153). There are several knowledge gaps in this research field. First, it is important to improve our understanding about how to specifically target mechano-sensing pathways in LSECs. The reasons are: 1) several mechano-signaling pathways, including Notch signaling, PIEZO channels, and YAP1 all mediate signaling transduction upon mechanical force in not only LSECs but also in ECs of other vascular beds (83, 153) and 2) although mechano-sensing pathways are involved in the pathogenesis of chronic liver disease, they are also important in maintaining tissue homeostasis and organ regeneration. Second, we need to study the synergistic effects of mechanotransduction and other biochemical signaling on LSEC function, which could lead to the development of combined strategies and agents that can improve the clinical outcomes of chronic liver disease (83). Third, the knowledge about mechanical force in mitochondrial function is limited in hepatic ECs. It has been proposed that mechanical forces regulate mitochondrial calcium signaling in the endothelium (1). Further studies are required to examine the role of mechanical forces in mitochondrial structure and function in hepatic ECs as well as their role in cell metabolism.

Not only protein-coding genes but also noncoding genes control many aspects of physiology and disease. Long noncoding RNAs (lncRNAs) are an important class of RNAs lacking protein coding capacity, and they are typically longer than 200 nucleotides in length. There are 19,175 potentially functional lncRNAs in the human genome, and the vast majority of them are functionally uncharacterized (71). Recently, several lncRNAs have emerged as crucial regulators of obesity and obesity-related pathologies (28, 145, 178). However, the role of lncRNAs in hepatic ECs is completely unknown. By integrating the lncRNA research into the existing body of biological knowledge, we will improve our understanding about how hepatic ECs respond to stress by changing gene expression and signal transduction in liver diseases. This may lead to the development of more effective therapies for liver disease and other chronic metabolic diseases such as obesity, diabetes, and cardiovascular disease.

GRANTS

This work was supported by the National Institutes of Health Funded COBRE Grant 1P20GM104320 (X.S. through the Nebraska Center for the Prevention of Obesity-related Diseases through Dietary Molecules), the American Heart Association SDG#15SDG25400012 (to X.S.), and the National Institutes of Health National Heart, Lung, and Blood Institute Grant R01 HL130864 to E.N.H.

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

X.S. and E.N.H. prepared figures; drafted manuscript; edited and revised manuscript; and approved final version of manuscript.

REFERENCES

  • 1.Alevriadou BR, Shanmughapriya S, Patel A, Stathopulos PB, Madesh M. Mitochondrial Ca2+ transport in the endothelium: regulation by ions, redox signalling and mechanical forces. J R Soc Interface 14: 20170672, 2017. doi: 10.1098/rsif.2017.0672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Arai M, Peng XX, Currin RT, Thurman RG, Lemasters JJ. Protection of sinusoidal endothelial cells against storage/reperfusion injury by prostaglandin E2 derived from Kupffer cells. Transplantation 68: 440–445, 1999. doi: 10.1097/00007890-199908150-00017. [DOI] [PubMed] [Google Scholar]
  • 3.Aravinthan A, Scarpini C, Tachtatzis P, Verma S, Penrhyn-Lowe S, Harvey R, Davies SE, Allison M, Coleman N, Alexander G. Hepatocyte senescence predicts progression in non-alcohol-related fatty liver disease. J Hepatol 58: 549–556, 2013. doi: 10.1016/j.jhep.2012.10.031. [DOI] [PubMed] [Google Scholar]
  • 4.Baker DJ, Petersen RC. Cellular senescence in brain aging and neurodegenerative diseases: evidence and perspectives. J Clin Invest 128: 1208–1216, 2018. doi: 10.1172/JCI95145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Baldassarre M, Giannone FA, Napoli L, Tovoli A, Ricci CS, Tufoni M, Caraceni P. The endocannabinoid system in advanced liver cirrhosis: pathophysiological implication and future perspectives. Liver Int 33: 1298–1308, 2013. doi: 10.1111/liv.12263. [DOI] [PubMed] [Google Scholar]
  • 6.Baratta JL, Ngo A, Lopez B, Kasabwalla N, Longmuir KJ, Robertson RT. Cellular organization of normal mouse liver: a histological, quantitative immunocytochemical, and fine structural analysis. Histochem Cell Biol 131: 713–726, 2009. doi: 10.1007/s00418-009-0577-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Bátkai S, Osei-Hyiaman D, Pan H, El-Assal O, Rajesh M, Mukhopadhyay P, Hong F, Harvey-White J, Jafri A, Haskó G, Huffman JW, Gao B, Kunos G, Pacher P. Cannabinoid-2 receptor mediates protection against hepatic ischemia/reperfusion injury. FASEB J 21: 1788–1800, 2007. doi: 10.1096/fj.06-7451com. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Beck JR, Cabral F, Rasineni K, Casey CA, Harris EN, Stains CI. A panel of protein kinase chemosensors distinguishes different types of fatty liver disease. Biochemistry 58: 3911–3917, 2019. doi: 10.1021/acs.biochem.9b00547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Biran A, Zada L, Abou Karam P, Vadai E, Roitman L, Ovadya Y, Porat Z, Krizhanovsky V. Quantitative identification of senescent cells in aging and disease. Aging Cell 16: 661–671, 2017. doi: 10.1111/acel.12592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Bird TG, Müller M, Boulter L, Vincent DF, Ridgway RA, Lopez-Guadamillas E, Lu WY, Jamieson T, Govaere O, Campbell AD, Ferreira-Gonzalez S, Cole AM, Hay T, Simpson KJ, Clark W, Hedley A, Clarke M, Gentaz P, Nixon C, Bryce S, Kiourtis C, Sprangers J, Nibbs RJB, Van Rooijen N, Bartholin L, McGreal SR, Apte U, Barry ST, Iredale JP, Clarke AR, Serrano M, Roskams TA, Sansom OJ, Forbes SJ. TGFβ inhibition restores a regenerative response in acute liver injury by suppressing paracrine senescence. Sci Transl Med 10: eaan1230, 2018. doi: 10.1126/scitranslmed.aan1230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Blouin A, Bolender RP, Weibel ER. Distribution of organelles and membranes between hepatocytes and nonhepatocytes in the rat liver parenchyma. A stereological study. J Cell Biol 72: 441–455, 1977. doi: 10.1083/jcb.72.2.441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Braet F, De Zanger R, Crabbé E, Wisse E. New observations on cytoskeleton and fenestrae in isolated rat liver sinusoidal endothelial cells. J Gastroenterol Hepatol 10, Suppl 1: S3–S7, 1995. doi: 10.1111/j.1440-1746.1995.tb01792.x. [DOI] [PubMed] [Google Scholar]
  • 13.Braet F, Wisse E. Structural and functional aspects of liver sinusoidal endothelial cell fenestrae: a review. Comp Hepatol 1: 1, 2002. doi: 10.1186/1476-5926-1-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Brodsky SV, Gealekman O, Chen J, Zhang F, Togashi N, Crabtree M, Gross SS, Nasjletti A, Goligorsky MS. Prevention and reversal of premature endothelial cell senescence and vasculopathy in obesity-induced diabetes by ebselen. Circ Res 94: 377–384, 2004. doi: 10.1161/01.RES.0000111802.09964.EF. [DOI] [PubMed] [Google Scholar]
  • 15.Bussian TJ, Aziz A, Meyer CF, Swenson BL, van Deursen JM, Baker DJ. Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature 562: 578–582, 2018. doi: 10.1038/s41586-018-0543-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Byrne CD, Targher G. NAFLD: a multisystem disease. J Hepatol 62, Suppl: S47–S64, 2015. doi: 10.1016/j.jhep.2014.12.012. [DOI] [PubMed] [Google Scholar]
  • 17.Cardier JE, Schulte T, Kammer H, Kwak J, Cardier M. Fas (CD95, APO-1) antigen expression and function in murine liver endothelial cells: implications for the regulation of apoptosis in liver endothelial cells. FASEB J 13: 1950–1960, 1999. doi: 10.1096/fasebj.13.14.1950. [DOI] [PubMed] [Google Scholar]
  • 18.Chalasani N, Younossi Z, Lavine JE, Charlton M, Cusi K, Rinella M, Harrison SA, Brunt EM, Sanyal AJ. The diagnosis and management of nonalcoholic fatty liver disease: practice guidance from the American Association for the Study of Liver Diseases. Hepatology 67: 328–357, 2018. doi: 10.1002/hep.29367. [DOI] [PubMed] [Google Scholar]
  • 19.Chen H, Li D, Sawamura T, Inoue K, Mehta JL. Upregulation of LOX-1 expression in aorta of hypercholesterolemic rabbits: modulation by losartan. Biochem Biophys Res Commun 276: 1100–1104, 2000. doi: 10.1006/bbrc.2000.3532. [DOI] [PubMed] [Google Scholar]
  • 20.Chen J, Huang X, Halicka D, Brodsky S, Avram A, Eskander J, Bloomgarden NA, Darzynkiewicz Z, Goligorsky MS. Contribution of p16INK4a and p21CIP1 pathways to induction of premature senescence of human endothelial cells: permissive role of p53. Am J Physiol Heart Circ Physiol 290: H1575–H1586, 2006. doi: 10.1152/ajpheart.00364.2005. [DOI] [PubMed] [Google Scholar]
  • 21.Chen L, Gu T, Li B, Li F, Ma Z, Zhang Q, Cai X, 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. doi: 10.1016/j.bbamcr.2019.06.011. [DOI] [PubMed] [Google Scholar]
  • 22.Chen M, Kakutani M, Minami M, Kataoka H, Kume N, Narumiya S, Kita T, Masaki T, Sawamura T. Increased expression of lectin-like oxidized low density lipoprotein receptor-1 in initial atherosclerotic lesions of Watanabe heritable hyperlipidemic rabbits. Arterioscler Thromb Vasc Biol 20: 1107–1115, 2000. doi: 10.1161/01.ATV.20.4.1107. [DOI] [PubMed] [Google Scholar]
  • 23.Childs BG, Baker DJ, Wijshake T, Conover CA, Campisi J, van Deursen JM. Senescent intimal foam cells are deleterious at all stages of atherosclerosis. Science 354: 472–477, 2016. doi: 10.1126/science.aaf6659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Childs BG, Gluscevic M, Baker DJ, Laberge RM, Marquess D, Dananberg J, van Deursen JM. Senescent cells: an emerging target for diseases of ageing. Nat Rev Drug Discov 16: 718–735, 2017. doi: 10.1038/nrd.2017.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Cogger VC, Svistounov D, Warren A, Zykova S, Melvin RG, Solon-Biet SM, O’Reilly JN, McMahon AC, Ballard JW, De Cabo R, Le Couteur DG, Lebel M. Liver aging and pseudocapillarization in a Werner syndrome mouse model. J Gerontol A Biol Sci Med Sci 69: 1076–1086, 2014. doi: 10.1093/gerona/glt169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Cogger VC, Warren A, Fraser R, Ngu M, McLean AJ, Le Couteur DG. Hepatic sinusoidal pseudocapillarization with aging in the non-human primate. Exp Gerontol 38: 1101–1107, 2003. doi: 10.1016/j.exger.2003.07.002. [DOI] [PubMed] [Google Scholar]
  • 27.da Silva PFL, Ogrodnik M, Kucheryavenko O, Glibert J, Miwa S, Cameron K, Ishaq A, Saretzki G, Nagaraja-Grellscheid S, Nelson G, von Zglinicki T. The bystander effect contributes to the accumulation of senescent cells in vivo. Aging Cell 18: e12848, 2019. doi: 10.1111/acel.12848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Dallner OS, Marinis JM, Lu YH, Birsoy K, Werner E, Fayzikhodjaeva G, Dill BD, Molina H, Moscati A, Kutalik Z, Marques-Vidal P, Kilpeläinen TO, Grarup N, Linneberg A, Zhang Y, Vaughan R, Loos RJF, Lazar MA, Friedman JM. Dysregulation of a long noncoding RNA reduces leptin leading to a leptin-responsive form of obesity. Nat Med 25: 507–516, 2019. doi: 10.1038/s41591-019-0370-1. [DOI] [PubMed] [Google Scholar]
  • 29.Davies PF. Hemodynamic shear stress and the endothelium in cardiovascular pathophysiology. Nat Clin Pract Cardiovasc Med 6: 16–26, 2009. doi: 10.1038/ncpcardio1397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.De Bock K, Georgiadou M, Schoors S, Kuchnio A, Wong BW, Cantelmo AR, Quaegebeur A, Ghesquière B, Cauwenberghs S, Eelen G, Phng LK, Betz I, Tembuyser B, Brepoels K, Welti J, Geudens I, Segura I, Cruys B, Bifari F, Decimo I, Blanco R, Wyns S, Vangindertael J, Rocha S, Collins RT, Munck S, Daelemans D, Imamura H, Devlieger R, Rider M, Van Veldhoven PP, Schuit F, Bartrons R, Hofkens J, Fraisl P, Telang S, Deberardinis RJ, Schoonjans L, Vinckier S, Chesney J, Gerhardt H, Dewerchin M, Carmeliet P. Role of PFKFB3-driven glycolysis in vessel sprouting. Cell 154: 651–663, 2013. doi: 10.1016/j.cell.2013.06.037. [DOI] [PubMed] [Google Scholar]
  • 31.Deaciuc IV, D’Souza NB, Spitzer JJ. Tumor necrosis factor-alpha cell-surface receptors of liver parenchymal and nonparenchymal cells during acute and chronic alcohol administration to rats. Alcohol Clin Exp Res 19: 332–338, 1995. doi: 10.1111/j.1530-0277.1995.tb01511.x. [DOI] [PubMed] [Google Scholar]
  • 32.Debacq-Chainiaux F, Erusalimsky JD, Campisi J, Toussaint O. Protocols to detect senescence-associated beta-galactosidase (SA-betagal) activity, a biomarker of senescent cells in culture and in vivo. Nat Protoc 4: 1798–1806, 2009. doi: 10.1038/nprot.2009.191. [DOI] [PubMed] [Google Scholar]
  • 33.DeLeve LD. Liver sinusoidal endothelial cells in hepatic fibrosis. Hepatology 61: 1740–1746, 2015. doi: 10.1002/hep.27376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.DeLeve LD, Maretti-Mira AC. Liver Sinusoidal Endothelial Cell: An Update. Semin Liver Dis 37: 377–387, 2017. doi: 10.1055/s-0037-1617455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.DeLeve LD, Wang X, Guo Y. Sinusoidal endothelial cells prevent rat stellate cell activation and promote reversion to quiescence. Hepatology 48: 920–930, 2008. doi: 10.1002/hep.22351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Demaria M, Ohtani N, Youssef SA, Rodier F, Toussaint W, Mitchell JR, Laberge RM, Vijg J, Van Steeg H, Dollé ME, Hoeijmakers JH, de Bruin A, Hara E, Campisi J. An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA. Dev Cell 31: 722–733, 2014. doi: 10.1016/j.devcel.2014.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Depraetere V, Golstein P. Fas and other cell death signaling pathways. Semin Immunol 9: 93–107, 1997. doi: 10.1006/smim.1997.0062. [DOI] [PubMed] [Google Scholar]
  • 38.Desroches-Castan A, Tillet E, Ricard N, Ouarné M, Mallet C, Belmudes L, Couté Y, Boillot O, Scoazec JY, Bailly S, 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. doi: 10.1002/hep.30655. [DOI] [PubMed] [Google Scholar]
  • 39.Di Martino J, Mascalchi P, Legros P, Lacomme S, Gontier E, Bioulac-Sage P, Balabaud C, Moreau V, Saltel F. Actin depolymerization in dedifferentiated liver sinusoidal endothelial cells promotes fenestrae re-formation. Hepatol Commun 3: 213–219, 2019. doi: 10.1002/hep4.1301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley C, Medrano EE, Linskens M, Rubelj I, Pereira-Smith O. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci USA 92: 9363–9367, 1995. doi: 10.1073/pnas.92.20.9363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ding BS, Cao Z, Lis R, Nolan DJ, Guo P, Simons M, Penfold ME, Shido K, Rabbany SY, Rafii S. Divergent angiocrine signals from vascular niche balance liver regeneration and fibrosis. Nature 505: 97–102, 2014. doi: 10.1038/nature12681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Ding BS, Nolan DJ, Butler JM, James D, Babazadeh AO, Rosenwaks Z, Mittal V, Kobayashi H, Shido K, Lyden D, Sato TN, Rabbany SY, Rafii S. Inductive angiocrine signals from sinusoidal endothelium are required for liver regeneration. Nature 468: 310–315, 2010. doi: 10.1038/nature09493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Dou Z, Ghosh K, Vizioli MG, Zhu J, Sen P, Wangensteen KJ, Simithy J, Lan Y, Lin Y, Zhou Z, Capell BC, Xu C, Xu M, Kieckhaefer JE, Jiang T, Shoshkes-Carmel M, Tanim KMAA, Barber GN, Seykora JT, Millar SE, Kaestner KH, Garcia BA, Adams PD, Berger SL. Cytoplasmic chromatin triggers inflammation in senescence and cancer. Nature 550: 402–406, 2017. doi: 10.1038/nature24050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Dufton NP, Peghaire CR, Osuna-Almagro L, Raimondi C, Kalna V, Chauhan A, Webb G, Yang Y, Birdsey GM, Lalor P, Mason JC, Adams DH, Randi AM. Dynamic regulation of canonical TGFβ signalling by endothelial transcription factor ERG protects from liver fibrogenesis. Nat Commun 8: 895, 2017. [Erratum in Nat Commun 11: 1301, 2020]. doi: 10.1038/s41467-017-01169-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Evrard SM, Lecce L, Michelis KC, Nomura-Kitabayashi A, Pandey G, Purushothaman KR, d’Escamard V, Li JR, Hadri L, Fujitani K, Moreno PR, Benard L, Rimmele P, Cohain A, Mecham B, Randolph GJ, Nabel EG, Hajjar R, Fuster V, Boehm M, Kovacic JC. Endothelial to mesenchymal transition is common in atherosclerotic lesions and is associated with plaque instability. Nat Commun 7: 11853, 2016. [Erratum in Nat Commun 8: 14710, 2017]. doi: 10.1038/ncomms11853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Fabbrini E, Sullivan S, Klein S. Obesity and nonalcoholic fatty liver disease: biochemical, metabolic, and clinical implications. Hepatology 51: 679–689, 2010. doi: 10.1002/hep.23280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Falkowska-Hansen B, Falkowski M, Metharom P, Krunic D, Goerdt S. Clathrin-coated vesicles form a unique net-like structure in liver sinusoidal endothelial cells by assembling along undisrupted microtubules. Exp Cell Res 313: 1745–1757, 2007. doi: 10.1016/j.yexcr.2007.02.026. [DOI] [PubMed] [Google Scholar]
  • 48.Feng T, Meng J, Kou S, Jiang Z, Huang X, Lu Z, Zhao H, Lau LF, Zhou B, Zhang H. CCN1-induced cellular senescence promotes heart regeneration. Circulation 139: 2495–2498, 2019. doi: 10.1161/CIRCULATIONAHA.119.039530. [DOI] [PubMed] [Google Scholar]
  • 49.Gal H, Porat Z, Krizhanovsky V. A multiparametric assay to evaluate senescent cells. Methods Mol Biol 1896: 107–117, 2019. doi: 10.1007/978-1-4939-8931-7_11. [DOI] [PubMed] [Google Scholar]
  • 50.Gambino V, De Michele G, Venezia O, Migliaccio P, Dall’Olio V, Bernard L, Minardi SP, Della Fazia MA, Bartoli D, Servillo G, Alcalay M, Luzi L, Giorgio M, Scrable H, Pelicci PG, Migliaccio E. Oxidative stress activates a specific p53 transcriptional response that regulates cellular senescence and aging. Aging Cell 12: 435–445, 2013. [Erratum in Aging Cell 18: e12962, 2019]. doi: 10.1111/acel.12060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Gary-Bobo M, Elachouri G, Gallas JF, Janiak P, Marini P, Ravinet-Trillou C, Chabbert M, Cruccioli N, Pfersdorff C, Roque C, Arnone M, Croci T, Soubrié P, Oury-Donat F, Maffrand JP, Scatton B, Lacheretz F, Le Fur G, Herbert JM, Bensaid M. Rimonabant reduces obesity-associated hepatic steatosis and features of metabolic syndrome in obese Zucker fa/fa rats. Hepatology 46: 122–129, 2007. doi: 10.1002/hep.21641. [DOI] [PubMed] [Google Scholar]
  • 52.Glück S, Guey B, Gulen MF, Wolter K, Kang TW, Schmacke NA, Bridgeman A, Rehwinkel J, Zender L, Ablasser A. Innate immune sensing of cytosolic chromatin fragments through cGAS promotes senescence. Nat Cell Biol 19: 1061–1070, 2017. doi: 10.1038/ncb3586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O, Bishop C, Campisi J, Collado M, Evangelou K, Ferbeyre G, Gil J, Hara E, Krizhanovsky V, Jurk D, Maier AB, Narita M, Niedernhofer L, Passos JF, Robbins PD, Schmitt CA, Sedivy J, Vougas K, von Zglinicki T, Zhou D, Serrano M, Demaria M. Cellular senescence: defining a path forward. Cell 179: 813–827, 2019. doi: 10.1016/j.cell.2019.10.005. [DOI] [PubMed] [Google Scholar]
  • 54.Guixé-Muntet S, de Mesquita FC, Vila S, Hernández-Gea V, Peralta C, García-Pagán JC, Bosch J, Gracia-Sancho J. Cross-talk between autophagy and KLF2 determines endothelial cell phenotype and microvascular function in acute liver injury. J Hepatol 66: 86–94, 2017. doi: 10.1016/j.jhep.2016.07.051. [DOI] [PubMed] [Google Scholar]
  • 55.Hammoutene A, Biquard L, Lasselin J, Kheloufi M, Tanguy M, Vion AC, Merian J, Colnot N, Loyer X, Tedgui A, Codogno P, Lotersztajn S, Paradis V, Boulanger CM, Rautou PE. A defect in endothelial autophagy occurs in patients with non-alcoholic steatohepatitis and promotes inflammation and fibrosis. J Hepatol 72: 528–538, 2020. doi: 10.1016/j.jhep.2019.10.028. [DOI] [PubMed] [Google Scholar]
  • 56.Hammoutene A, Rautou PE. Role of liver sinusoidal endothelial cells in non-alcoholic fatty liver disease. J Hepatol 70: 1278–1291, 2019. doi: 10.1016/j.jhep.2019.02.012. [DOI] [PubMed] [Google Scholar]
  • 57.Harding SM, Benci JL, Irianto J, Discher DE, Minn AJ, Greenberg RA. Mitotic progression following DNA damage enables pattern recognition within micronuclei. Nature 548: 466–470, 2017. doi: 10.1038/nature23470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Harris EN, Cabral F. Ligand binding and signaling of HARE/stabilin-2. Biomolecules 9: 273, 2019. doi: 10.3390/biom9070273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Hazari Y, Bravo-San Pedro JM, Hetz C, Galluzzi L, Kroemer G. Autophagy in hepatic adaptation to stress. J Hepatol 72: 183–196, 2020. doi: 10.1016/j.jhep.2019.08.026. [DOI] [PubMed] [Google Scholar]
  • 60.He M, Martin M, Marin T, Chen Z, Gongol B. Endothelial mechanobiology. APL Bioeng 4: 010904, 2020. doi: 10.1063/1.5129563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.He S, Sharpless NE. Senescence in health and disease. Cell 169: 1000–1011, 2017. doi: 10.1016/j.cell.2017.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Hernandez-Segura A, de Jong TV, Melov S, Guryev V, Campisi J, Demaria M. Unmasking transcriptional heterogeneity in senescent cells. Curr Biol 27: 2652–2660.e4, 2017. doi: 10.1016/j.cub.2017.07.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Hernandez-Segura A, Nehme J, Demaria M. Hallmarks of Cellular Senescence. Trends Cell Biol 28: 436–453, 2018. doi: 10.1016/j.tcb.2018.02.001. [DOI] [PubMed] [Google Scholar]
  • 64.Hernandez-Segura A, Rubingh R, Demaria M. Identification of stable senescence-associated reference genes. Aging Cell 18: e12911, 2019. doi: 10.1111/acel.12911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Herranz N, Gil J. Mechanisms and functions of cellular senescence. J Clin Invest 128: 1238–1246, 2018. doi: 10.1172/JCI95148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Hickson LJ, Langhi Prata LGP, Bobart SA, Evans TK, Giorgadze N, Hashmi SK, Herrmann SM, Jensen MD, Jia Q, Jordan KL, Kellogg TA, Khosla S, Koerber DM, Lagnado AB, Lawson DK, LeBrasseur NK, Lerman LO, McDonald KM, McKenzie TJ, Passos JF, Pignolo RJ, Pirtskhalava T, Saadiq IM, Schaefer KK, Textor SC, Victorelli SG, Volkman TL, Xue A, Wentworth MA, Wissler Gerdes EO, Zhu Y, Tchkonia T, Kirkland JL. Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease. EBioMedicine 47: 446–456, 2019. [Erratum in EBio Medicine 52: 102595, 2020]. doi: 10.1016/j.ebiom.2019.08.069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Higashi T, Friedman SL, Hoshida Y. Hepatic stellate cells as key target in liver fibrosis. Adv Drug Deliv Rev 121: 27–42, 2017. doi: 10.1016/j.addr.2017.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Hillard CJ. Circulating endocannabinoids: from whence do they come and where are they going? Neuropsychopharmacology 43: 155–172, 2018. doi: 10.1038/npp.2017.130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Hilscher MB, Sehrawat T, Arab JP, Zeng Z, Gao J, Liu M, Kostallari E, Gao Y, Simonetto DA, Yaqoob U, Cao S, Revzin A, Beyder A, Wang RA, Kamath PS, Kubes P, Shah VH. 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. doi: 10.1053/j.gastro.2019.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.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 194: 34–41, 2010. doi: 10.1016/j.toxlet.2010.01.020. [DOI] [PubMed] [Google Scholar]
  • 71.Hon CC, Ramilowski JA, Harshbarger J, Bertin N, Rackham OJ, Gough J, Denisenko E, Schmeier S, Poulsen TM, Severin J, Lizio M, Kawaji H, Kasukawa T, Itoh M, Burroughs AM, Noma S, Djebali S, Alam T, Medvedeva YA, Testa AC, Lipovich L, Yip CW, Abugessaisa I, Mendez M, Hasegawa A, Tang D, Lassmann T, Heutink P, Babina M, Wells CA, Kojima S, Nakamura Y, Suzuki H, Daub CO, de Hoon MJ, Arner E, Hayashizaki Y, Carninci P, Forrest AR. An atlas of human long non-coding RNAs with accurate 5′ ends. Nature 543: 199–204, 2017. doi: 10.1038/nature21374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Hu J, Srivastava K, Wieland M, Runge A, Mogler C, Besemfelder E, Terhardt D, Vogel MJ, Cao L, Korn C, Bartels S, Thomas M, Augustin HG. Endothelial cell-derived angiopoietin-2 controls liver regeneration as a spatiotemporal rheostat. Science 343: 416–419, 2014. doi: 10.1126/science.1244880. [DOI] [PubMed] [Google Scholar]
  • 73.Huda N, Liu G, Hong H, Yan S, Khambu B, Yin XM. Hepatic senescence, the good and the bad. World J Gastroenterol 25: 5069–5081, 2019. doi: 10.3748/wjg.v25.i34.5069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Hunt NJ, Kang SWS, Lockwood GP, Le Couteur DG, Cogger VC. hallmarks of aging in the liver. Comput Struct Biotechnol J 17: 1151–1161, 2019. doi: 10.1016/j.csbj.2019.07.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Hunt NJ, Lockwood GP, Kang SWS, Pulpitel T, Clark X, Mao H, McCourt PAG, Cooney GJ, Wali JA, Le Couteur FH, Le Couteur DG, Cogger VC. The effects of metformin on age-related changes in the liver sinusoidal endothelial cell. J Gerontol A Biol Sci Med Sci 75: 278–285, 2020. doi: 10.1093/gerona/glz153. [DOI] [PubMed] [Google Scholar]
  • 76.Hunt NJ, Lockwood GP, Warren A, Mao H, McCourt PAG, Le Couteur DG, Cogger VC. Manipulating fenestrations in young and old liver sinusoidal endothelial cells. Am J Physiol Gastrointest Liver Physiol 316: G144–G154, 2019. doi: 10.1152/ajpgi.00179.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Hutchins NA, Wang F, Wang Y, Chung CS, Ayala A. Kupffer cells potentiate liver sinusoidal endothelial cell injury in sepsis by ligating programmed cell death ligand-1. J Leukoc Biol 94: 963–970, 2013. doi: 10.1189/jlb.0113051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Iwaisako K, Jiang C, Zhang M, Cong M, Moore-Morris TJ, Park TJ, Liu X, Xu J, Wang P, Paik YH, Meng F, Asagiri M, Murray LA, Hofmann AF, Iida T, Glass CK, Brenner DA, Kisseleva T. Origin of myofibroblasts in the fibrotic liver in mice. Proc Natl Acad Sci USA 111: E3297–E3305, 2014. doi: 10.1073/pnas.1400062111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Jaeschke H. Mechanisms of liver injury. II. Mechanisms of neutrophil-induced liver cell injury during hepatic ischemia-reperfusion and other acute inflammatory conditions. Am J Physiol Gastrointest Liver Physiol 290: G1083–G1088, 2006. doi: 10.1152/ajpgi.00568.2005. [DOI] [PubMed] [Google Scholar]
  • 80.Józefowski S. [The role of the class A scavenger receptors, SR-A and MARCO, in the immune system. Part 1. The structure of receptors, their ligand binding repertoires and ability to initiate intracellular signaling]. Postepy Hig Med Dosw (Online) 66: 104–119, 2012. doi: 10.5604/17322693.984079. [DOI] [PubMed] [Google Scholar]
  • 81.Justice JN, Nambiar AM, Tchkonia T, LeBrasseur NK, Pascual R, Hashmi SK, Prata L, Masternak MM, Kritchevsky SB, Musi N, Kirkland JL. Senolytics in idiopathic pulmonary fibrosis: Results from a first-in-human, open-label, pilot study. EBioMedicine 40: 554–563, 2019. doi: 10.1016/j.ebiom.2018.12.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Kaczmarek J, Homsi Y, van Üüm J, Metzger C, Knolle PA, Kolanus W, Lang T, Diehl L. Liver sinusoidal endothelial cell-mediated CD8 T cell priming depends on co-inhibitory signal integration over time. PLoS One 9: e99574, 2014. doi: 10.1371/journal.pone.0099574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Kang N. Mechanotransduction in liver diseases. Semin Liver Dis 40: 84–90, 2020. doi: 10.1055/s-0039-3399502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Katsuumi G, Shimizu I, Yoshida Y, Minamino T. Vascular senescence in cardiovascular and metabolic diseases. Front Cardiovasc Med 5: 18, 2018. doi: 10.3389/fcvm.2018.00018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kim IH, Kisseleva T, Brenner DA. Aging and liver disease. Curr Opin Gastroenterol 31: 184–191, 2015. doi: 10.1097/MOG.0000000000000176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Kitao A, Sato Y, Sawada-Kitamura S, Harada K, Sasaki M, Morikawa H, Shiomi S, Honda M, Matsui O, Nakanuma Y. Endothelial to mesenchymal transition via transforming growth factor-beta1/Smad activation is associated with portal venous stenosis in idiopathic portal hypertension. Am J Pathol 175: 616–626, 2009. doi: 10.2353/ajpath.2009.081061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Kmieć Z. Cooperation of liver cells in health and disease. Adv Anat Embryol Cell Biol 161: III–XIII, 2001. doi: 10.1007/978-3-642-56553-3. [DOI] [PubMed] [Google Scholar]
  • 88.Knolle PA, Wohlleber D. Immunological functions of liver sinusoidal endothelial cells. Cell Mol Immunol 13: 347–353, 2016. doi: 10.1038/cmi.2016.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Koudelkova P, Weber G, Mikulits W. Liver sinusoidal endothelial cells escape senescence by loss of p19ARF. PLoS One 10: e0142134, 2015. doi: 10.1371/journal.pone.0142134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Kovacic JC, Dimmeler S, Harvey RP, Finkel T, Aikawa E, Krenning G, Baker AH. Endothelial to mesenchymal transition in cardiovascular disease: JACC state-of-the-art review. J Am Coll Cardiol 73: 190–209, 2019. doi: 10.1016/j.jacc.2018.09.089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Krizhanovsky V, Yon M, Dickins RA, Hearn S, Simon J, Miething C, Yee H, Zender L, Lowe SW. Senescence of activated stellate cells limits liver fibrosis. Cell 134: 657–667, 2008. doi: 10.1016/j.cell.2008.06.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Kuilman T, Michaloglou C, Mooi WJ, Peeper DS. The essence of senescence. Genes Dev 24: 2463–2479, 2010. doi: 10.1101/gad.1971610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Kus E, Jasiński K, Skórka T, Czyzynska-Cichon I, Chlopicki S. Short-term treatment with hepatoselective NO donor V-PYRRO/NO improves blood flow in hepatic microcirculation in liver steatosis in mice. Pharmacol Rep 70: 463–469, 2018. doi: 10.1016/j.pharep.2017.11.019. [DOI] [PubMed] [Google Scholar]
  • 94.Kus E, Kaczara P, Czyzynska-Cichon I, Szafranska K, Zapotoczny B, Kij A, Sowinska A, Kotlinowski J, Mateuszuk L, Czarnowska E, Szymonski M, Chlopicki S. LSEC fenestrae are preserved despite pro-inflammatory phenotype of liver sinusoidal endothelial cells in mice on high fat diet. Front Physiol 10: 6, 2019. doi: 10.3389/fphys.2019.00006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Lalor PF, Sun PJ, Weston CJ, Martin-Santos A, Wakelam MJ, Adams DH. Activation of vascular adhesion protein-1 on liver endothelium results in an NF-kappaB-dependent increase in lymphocyte adhesion. Hepatology 45: 465–474, 2007. doi: 10.1002/hep.21497. [DOI] [PubMed] [Google Scholar]
  • 96.Le Couteur DG, Cogger VC, Markus AM, Harvey PJ, Yin ZL, Ansselin AD, McLean AJ. Pseudocapillarization and associated energy limitation in the aged rat liver. Hepatology 33: 537–543, 2001. doi: 10.1053/jhep.2001.22754. [DOI] [PubMed] [Google Scholar]
  • 97.Lecot P, Alimirah F, Desprez PY, Campisi J, Wiley C. Context-dependent effects of cellular senescence in cancer development. Br J Cancer 114: 1180–1184, 2016. doi: 10.1038/bjc.2016.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Li M, Qian M, Xu J. Vascular endothelial regulation of obesity-associated insulin resistance. Front Cardiovasc Med 4: 51, 2017. doi: 10.3389/fcvm.2017.00051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Li T, Chen ZJ. The cGAS-cGAMP-STING pathway connects DNA damage to inflammation, senescence, and cancer. J Exp Med 215: 1287–1299, 2018. doi: 10.1084/jem.20180139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Li Z, Chen B, Dong W, Kong M, Fan Z, Yu L, Wu D, Lu J, Xu Y. MKL1 promotes endothelial-to-mesenchymal transition and liver fibrosis by activating TWIST1 transcription. Cell Death Dis 10: 899, 2019. doi: 10.1038/s41419-019-2101-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Li Z, Chen B, Dong W, Kong M, Shao Y, Fan Z, Yu L, Wu D, Lu J, Guo J, Xu Y. The chromatin remodeler Brg1 integrates ROS production and endothelial-mesenchymal transition to promote liver fibrosis in mice. Front Cell Dev Biol 7: 245, 2019. doi: 10.3389/fcell.2019.00245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Liew PX, Kubes P. The Neutrophil’s Role During Health and Disease. Physiol Rev 99: 1223–1248, 2019. doi: 10.1152/physrev.00012.2018. [DOI] [PubMed] [Google Scholar]
  • 103.Liu Y, Sanoff HK, Cho H, Burd CE, Torrice C, Ibrahim JG, Thomas NE, Sharpless NE. Expression of p16(INK4a) in peripheral blood T-cells is a biomarker of human aging. Aging Cell 8: 439–448, 2009. doi: 10.1111/j.1474-9726.2009.00489.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Lorenz L, Axnick J, Buschmann T, Henning C, Urner S, Fang S, Nurmi H, Eichhorst N, Holtmeier R, Bódis K, Hwang JH, Müssig K, Eberhard D, Stypmann J, Kuss O, Roden M, Alitalo K, Häussinger D, Lammert E. Mechanosensing by β1 integrin induces angiocrine signals for liver growth and survival. Nature 562: 128–132, 2018. doi: 10.1038/s41586-018-0522-3. [DOI] [PubMed] [Google Scholar]
  • 105.Luo X, Li H, Ma L, Zhou J, Guo X, Woo SL, Pei Y, Knight LR, Deveau M, Chen Y, Qian X, Xiao X, Li Q, Chen X, Huo Y, McDaniel K, Francis H, Glaser S, Meng F, Alpini G, Wu C. Expression of STING Is increased in liver tissues from patients with NAFLD and promotes macrophage-mediated hepatic inflammation and fibrosis in mice. Gastroenterology 155: 1971–1984.e4, 2018. doi: 10.1053/j.gastro.2018.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.MacLean-Fletcher S, Pollard TD. Mechanism of action of cytochalasin B on actin. Cell 20: 329–341, 1980. doi: 10.1016/0092-8674(80)90619-4. [DOI] [PubMed] [Google Scholar]
  • 107.Maeso-Díaz R, Ortega-Ribera M, Fernández-Iglesias A, Hide D, Muñoz L, Hessheimer AJ, Vila S, Francés R, Fondevila C, Albillos A, Peralta C, Bosch J, Tacke F, Cogger VC, Gracia-Sancho J. Effects of aging on liver microcirculatory function and sinusoidal phenotype. Aging Cell 17: e12829, 2018. doi: 10.1111/acel.12829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Mallat A, Teixeira-Clerc F, Deveaux V, Manin S, Lotersztajn S. The endocannabinoid system as a key mediator during liver diseases: new insights and therapeutic openings. Br J Pharmacol 163: 1432–1440, 2011. doi: 10.1111/j.1476-5381.2011.01397.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Martinez-Pomares L. The mannose receptor. J Leukoc Biol 92: 1177–1186, 2012. doi: 10.1189/jlb.0512231. [DOI] [PubMed] [Google Scholar]
  • 110.McLean AJ, Cogger VC, Chong GC, Warren A, Markus AM, Dahlstrom JE, Le Couteur DG. Age-related pseudocapillarization of the human liver. J Pathol 200: 112–117, 2003. doi: 10.1002/path.1328. [DOI] [PubMed] [Google Scholar]
  • 111.Mederacke I, Hsu CC, Troeger JS, Huebener P, Mu X, Dapito DH, Pradere JP, Schwabe RF. Fate tracing reveals hepatic stellate cells as dominant contributors to liver fibrosis independent of its aetiology. Nat Commun 4: 2823, 2013. doi: 10.1038/ncomms3823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Minamino T, Orimo M, Shimizu I, Kunieda T, Yokoyama M, Ito T, Nojima A, Nabetani A, Oike Y, Matsubara H, Ishikawa F, Komuro I. A crucial role for adipose tissue p53 in the regulation of insulin resistance. Nat Med 15: 1082–1087, 2009. doi: 10.1038/nm.2014. [DOI] [PubMed] [Google Scholar]
  • 113.Mönkemöller V, Øie C, Hübner W, Huser T, 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: 16279, 2015. doi: 10.1038/srep16279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Mönkemöller V, Schüttpelz M, McCourt P, Sørensen K, Smedsrød B, Huser T. Imaging fenestrations in liver sinusoidal endothelial cells by optical localization microscopy. Phys Chem Chem Phys 16: 12576–12581, 2014. doi: 10.1039/C4CP01574F. [DOI] [PubMed] [Google Scholar]
  • 115.Mukhopadhyay B, Cinar R, Yin S, Liu J, Tam J, Godlewski G, Harvey-White J, Mordi I, Cravatt BF, Lotersztajn S, Gao B, Yuan Q, Schuebel K, Goldman D, Kunos G. Hyperactivation of anandamide synthesis and regulation of cell-cycle progression via cannabinoid type 1 (CB1) receptors in the regenerating liver. Proc Natl Acad Sci USA 108: 6323–6328, 2011. doi: 10.1073/pnas.1017689108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Muñoz-Espín D, Cañamero M, Maraver A, Gómez-López G, Contreras J, Murillo-Cuesta S, Rodríguez-Baeza A, Varela-Nieto I, Ruberte J, Collado M, Serrano M. Programmed cell senescence during mammalian embryonic development. Cell 155: 1104–1118, 2013. doi: 10.1016/j.cell.2013.10.019. [DOI] [PubMed] [Google Scholar]
  • 117.Muñoz-Espín D, Serrano M. Cellular senescence: from physiology to pathology. Nat Rev Mol Cell Biol 15: 482–496, 2014. doi: 10.1038/nrm3823. [DOI] [PubMed] [Google Scholar]
  • 118.Nelson G, Wordsworth J, Wang C, Jurk D, Lawless C, Martin-Ruiz C, von Zglinicki T. A senescent cell bystander effect: senescence-induced senescence. Aging Cell 11: 345–349, 2012. doi: 10.1111/j.1474-9726.2012.00795.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Ogrodnik M, Miwa S, Tchkonia T, Tiniakos D, Wilson CL, Lahat A, Day CP, Burt A, Palmer A, Anstee QM, Grellscheid SN, Hoeijmakers JHJ, Barnhoorn S, Mann DA, Bird TG, Vermeij WP, Kirkland JL, Passos JF, von Zglinicki T, Jurk D. Cellular senescence drives age-dependent hepatic steatosis. Nat Commun 8: 15691, 2017. doi: 10.1038/ncomms15691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Okazaki T, Maeda A, Nishimura H, Kurosaki T, Honjo T. PD-1 immunoreceptor inhibits B cell receptor-mediated signaling by recruiting src homology 2-domain-containing tyrosine phosphatase 2 to phosphotyrosine. Proc Natl Acad Sci USA 98: 13866–13871, 2001. doi: 10.1073/pnas.231486598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Ovadya Y, Krizhanovsky V. Strategies targeting cellular senescence. J Clin Invest 128: 1247–1254, 2018. doi: 10.1172/JCI95149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Pacher P, Beckman JS, Liaudet L. Nitric oxide and peroxynitrite in health and disease. Physiol Rev 87: 315–424, 2007. doi: 10.1152/physrev.00029.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Palmer AK, Tchkonia T, LeBrasseur NK, Chini EN, Xu M, Kirkland JL. Cellular Senescence in Type 2 Diabetes: A Therapeutic Opportunity. Diabetes 64: 2289–2298, 2015. doi: 10.2337/db14-1820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Palmer AK, Xu M, Zhu Y, Pirtskhalava T, Weivoda MM, Hachfeld CM, Prata LG, van Dijk TH, Verkade E, Casaclang-Verzosa G, Johnson KO, Cubro H, Doornebal EJ, Ogrodnik M, Jurk D, Jensen MD, Chini EN, Miller JD, Matveyenko A, Stout MB, Schafer MJ, White TA, Hickson LJ, Demaria M, Garovic V, Grande J, Arriaga EA, Kuipers F, von Zglinicki T, LeBrasseur NK, Campisi J, Tchkonia T, Kirkland JL. Targeting senescent cells alleviates obesity-induced metabolic dysfunction. Aging Cell 18: e12950, 2019. doi: 10.1111/acel.12950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Papatheodoridi AM, Chrysavgis L, Koutsilieris M, Chatzigeorgiou A. The role of senescence in the development of nonalcoholic fatty liver disease and progression to nonalcoholic steatohepatitis. Hepatology 71: 363–374, 2020. doi: 10.1002/hep.30834. [DOI] [PubMed] [Google Scholar]
  • 127.Pellegrinelli V, Rouault C, Veyrie N, Clément K, Lacasa D. Endothelial cells from visceral adipose tissue disrupt adipocyte functions in a three-dimensional setting: partial rescue by angiopoietin-1. Diabetes 63: 535–549, 2014. doi: 10.2337/db13-0537. [DOI] [PubMed] [Google Scholar]
  • 128.Pi X, Xie L, Patterson C. Emerging roles of vascular endothelium in metabolic homeostasis. Circ Res 123: 477–494, 2018. doi: 10.1161/CIRCRESAHA.118.313237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Piera-Velazquez S, Jimenez SA. Endothelial to mesenchymal transition: role in physiology and in the pathogenesis of human diseases. Physiol Rev 99: 1281–1324, 2019. doi: 10.1152/physrev.00021.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Poisson J, Lemoinne S, Boulanger C, Durand F, Moreau R, Valla D, Rautou PE. Liver sinusoidal endothelial cells: Physiology and role in liver diseases. J Hepatol 66: 212–227, 2017. doi: 10.1016/j.jhep.2016.07.009. [DOI] [PubMed] [Google Scholar]
  • 131.Pombinho R, Sousa S, Cabanes D. Scavenger receptors: promiscuous players during microbial pathogenesis. Crit Rev Microbiol 44: 685–700, 2018. doi: 10.1080/1040841X.2018.1493716. [DOI] [PubMed] [Google Scholar]
  • 132.Pritchard MT, McCracken JM. Identifying novel targets for treatment of liver fibrosis: What can we learn from injured tissues which heal without a scar? Curr Drug Targets 16: 1332–1346, 2015. doi: 10.2174/1389450116666150825111439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Puche JE, Saiman Y, Friedman SL. Hepatic stellate cells and liver fibrosis. Compr Physiol 3: 1473–1492, 2013. doi: 10.1002/cphy.c120035. [DOI] [PubMed] [Google Scholar]
  • 134.Qiu Y, Liu S, Chen HT, Yu CH, Teng XD, Yao HT, Xu GQ. Upregulation of caveolin-1 and SR-B1 in mice with non-alcoholic fatty liver disease. Hepatobiliary Pancreat Dis Int 12: 630–636, 2013. doi: 10.1016/S1499-3872(13)60099-5. [DOI] [PubMed] [Google Scholar]
  • 135.Rabbany SY, Rafii S. Blood flow forces liver growth. Nature 562: 42–43, 2018. doi: 10.1038/d41586-018-06741-2. [DOI] [PubMed] [Google Scholar]
  • 136.Ramachandran P, Dobie R, Wilson-Kanamori JR, Dora EF, Henderson BEP, Luu NT, Portman JR, Matchett KP, Brice M, Marwick JA, Taylor RS, Efremova M, Vento-Tormo R, Carragher NO, Kendall TJ, Fallowfield JA, Harrison EM, Mole DJ, Wigmore SJ, Newsome PN, Weston CJ, Iredale JP, Tacke F, Pollard JW, Ponting CP, Marioni JC, Teichmann SA, Henderson NC. Resolving the fibrotic niche of human liver cirrhosis at single-cell level. Nature 575: 512–518, 2019. doi: 10.1038/s41586-019-1631-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Rein-Fischboeck L, Krautbauer S, Eisinger K, Pohl R, Meier EM, Weiss TS, Buechler C. Hepatic scavenger receptor BI is associated with type 2 diabetes but unrelated to human and murine non-alcoholic fatty liver disease. Biochem Biophys Res Commun 467: 377–382, 2015. doi: 10.1016/j.bbrc.2015.09.149. [DOI] [PubMed] [Google Scholar]
  • 138.Ribera J, Pauta M, Melgar-Lesmes P, Córdoba B, Bosch A, Calvo M, Rodrigo-Torres D, Sancho-Bru P, Mira A, Jiménez W, Morales-Ruiz M. A small population of liver endothelial cells undergoes endothelial-to-mesenchymal transition in response to chronic liver injury. Am J Physiol Gastrointest Liver Physiol 313: G492–G504, 2017. doi: 10.1152/ajpgi.00428.2016. [DOI] [PubMed] [Google Scholar]
  • 139.Rodier F, Coppé JP, Patil CK, Hoeijmakers WA, Muñoz DP, Raza SR, Freund A, Campeau E, Davalos AR, Campisi J. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat Cell Biol 11: 973–979, 2009. [Erratum in Nat Cell Biol 11: 1272, 2009.] doi: 10.1038/ncb1909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Rohlenova K, Veys K, Miranda-Santos I, De Bock K, Carmeliet P. Endothelial Cell Metabolism in Health and Disease. Trends Cell Biol 28: 224–236, 2018. doi: 10.1016/j.tcb.2017.10.010. [DOI] [PubMed] [Google Scholar]
  • 141.Ruart M, Chavarria L, Campreciós G, Suárez-Herrera N, Montironi C, Guixé-Muntet S, Bosch J, Friedman SL, Garcia-Pagán JC, 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. doi: 10.1016/j.jhep.2018.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Sanna C, Rosso C, Marietti M, Bugianesi E. Non-alcoholic fatty liver disease and extra-hepatic cancers. Int J Mol Sci 17: 717, 2016. doi: 10.3390/ijms17050717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Sanyal AJ, Harrison SA, Ratziu V, Abdelmalek MF, Diehl AM, Caldwell S, Shiffman ML, Aguilar Schall R, Jia C, McColgan B, Djedjos CS, McHutchison JG, Subramanian GM, Myers RP, Younossi Z, Muir AJ, Afdhal NH, Bosch J, Goodman Z. The natural history of advanced fibrosis due to nonalcoholic steatohepatitis: data from the Simtuzumab Trials. Hepatology 70: 1913–1927, 2019. doi: 10.1002/hep.30664. [DOI] [PubMed] [Google Scholar]
  • 144.Sarig R, Rimmer R, Bassat E, Zhang L, Umansky KB, Lendengolts D, Perlmoter G, Yaniv K, Tzahor E. Transient p53-mediated regenerative senescence in the injured heart. Circulation 139: 2491–2494, 2019. doi: 10.1161/CIRCULATIONAHA.119.040125. [DOI] [PubMed] [Google Scholar]
  • 145.Schmidt E, Dhaouadi I, Gaziano I, Oliverio M, Klemm P, Awazawa M, Mitterer G, Fernandez-Rebollo E, Pradas-Juni M, Wagner W, Hammerschmidt P, Loureiro R, Kiefer C, Hansmeier NR, Khani S, Bergami M, Heine M, Ntini E, Frommolt P, Zentis P, Ørom UA, Heeren J, Blüher M, Bilban M, Kornfeld JW. LincRNA H19 protects from dietary obesity by constraining expression of monoallelic genes in brown fat. Nat Commun 9: 3622, 2018. doi: 10.1038/s41467-018-05933-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Schurich A, Böttcher JP, Burgdorf S, Penzler P, Hegenbarth S, Kern M, Dolf A, Endl E, Schultze J, Wiertz E, Stabenow D, Kurts C, Knolle P. Distinct kinetics and dynamics of cross-presentation in liver sinusoidal endothelial cells compared to dendritic cells. Hepatology 50: 909–919, 2009. doi: 10.1002/hep.23075. [DOI] [PubMed] [Google Scholar]
  • 147.Sharpless NE, Sherr CJ. Forging a signature of in vivo senescence. Nat Rev Cancer 15: 397–408, 2015. [Erratum in Nat Rev Cancer 15: 509, 2015]. doi: 10.1038/nrc3960. [DOI] [PubMed] [Google Scholar]
  • 148.Sheppard KA, Fitz LJ, Lee JM, Benander C, George JA, Wooters J, Qiu Y, Jussif JM, Carter LL, Wood CR, Chaudhary D. PD-1 inhibits T-cell receptor induced phosphorylation of the ZAP70/CD3zeta signalosome and downstream signaling to PKCtheta. FEBS Lett 574: 37–41, 2004. doi: 10.1016/j.febslet.2004.07.083. [DOI] [PubMed] [Google Scholar]
  • 149.Shiratori Y, Tananka M, Kawase T, Shiina S, Komatsu Y, Omata M. Quantification of sinusoidal cell function in vivo. Semin Liver Dis 13: 39–49, 1993. doi: 10.1055/s-2007-1007336. [DOI] [PubMed] [Google Scholar]
  • 150.Softic S, Kahn CR. Fatty liver disease: is it nonalcoholic fatty liver disease or obesity-associated fatty liver disease? Eur J Gastroenterol Hepatol 31: 143, 2019. doi: 10.1097/MEG.0000000000001279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Sørensen KK, McCourt P, Berg T, Crossley C, Le Couteur D, Wake K, Smedsrød B. The scavenger endothelial cell: a new player in homeostasis and immunity. Am J Physiol Regul Integr Comp Physiol 303: R1217–R1230, 2012. doi: 10.1152/ajpregu.00686.2011. [DOI] [PubMed] [Google Scholar]
  • 152.Sørensen KK, Simon-Santamaria J, McCuskey RS, Smedsrød B. Liver sinusoidal endothelial cells. Compr Physiol 5: 1751–1774, 2015. doi: 10.1002/cphy.c140078. [DOI] [PubMed] [Google Scholar]
  • 153.Soydemir S, Comella O, Abdelmottaleb D, Pritchett J. Does mechanocrine signaling by liver sinusoidal endothelial cells offer new opportunities for the development of anti-fibrotics? Front Med (Lausanne) 6: 312, 2020. doi: 10.3389/fmed.2019.00312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Spolarics Z, Lang CH, Bagby GJ, Spitzer JJ. Glutamine and fatty acid oxidation are the main sources of energy for Kupffer and endothelial cells. Am J Physiol 261: G185–G190, 1991. doi: 10.1152/ajpgi.1991.261.2.G185. [DOI] [PubMed] [Google Scholar]
  • 155.Storer M, Mas A, Robert-Moreno A, Pecoraro M, Ortells MC, Di Giacomo V, Yosef R, Pilpel N, Krizhanovsky V, Sharpe J, Keyes WM. Senescence is a developmental mechanism that contributes to embryonic growth and patterning. Cell 155: 1119–1130, 2013. doi: 10.1016/j.cell.2013.10.041. [DOI] [PubMed] [Google Scholar]
  • 156.Sun L, Wu J, Du F, Chen X, Chen ZJ. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 339: 786–791, 2013. doi: 10.1126/science.1232458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Tam J, Liu J, Mukhopadhyay B, Cinar R, Godlewski G, Kunos G. Endocannabinoids in liver disease. Hepatology 53: 346–355, 2011. doi: 10.1002/hep.24077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Tiegs G, Lohse AW. Immune tolerance: what is unique about the liver. J Autoimmun 34: 1–6, 2010. doi: 10.1016/j.jaut.2009.08.008. [DOI] [PubMed] [Google Scholar]
  • 159.Timchenko NA. Aging and liver regeneration. Trends Endocrinol Metab 20: 171–176, 2009. doi: 10.1016/j.tem.2009.01.005. [DOI] [PubMed] [Google Scholar]
  • 160.Trefts E, Gannon M, Wasserman DH. The liver. Curr Biol 27: R1147–R1151, 2017. doi: 10.1016/j.cub.2017.09.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Tsuchiya K, Accili D. Liver sinusoidal endothelial cells link hyperinsulinemia to hepatic insulin resistance. Diabetes 62: 1478–1489, 2013. doi: 10.2337/db12-1296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.van Deursen JM. Senolytic therapies for healthy longevity. Science 364: 636–637, 2019. doi: 10.1126/science.aaw1299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Wang C, Jurk D, Maddick M, Nelson G, Martin-Ruiz C, von Zglinicki T. DNA damage response and cellular senescence in tissues of aging mice. Aging Cell 8: 311–323, 2009. doi: 10.1111/j.1474-9726.2009.00481.x. [DOI] [PubMed] [Google Scholar]
  • 164.Wang CY, Kim HH, Hiroi Y, Sawada N, Salomone S, Benjamin LE, Walsh K, Moskowitz MA, Liao JK. Obesity increases vascular senescence and susceptibility to ischemic injury through chronic activation of Akt and mTOR. Sci Signal 2: ra11, 2009. doi: 10.1126/scisignal.2000143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Wang R, Ding Q, Yaqoob U, de Assuncao TM, Verma VK, Hirsova P, Cao S, Mukhopadhyay D, Huebert RC, Shah VH. Exosome adherence and internalization by hepatic stellate cells triggers sphingosine 1-phosphate-dependent migration. J Biol Chem 290: 30684–30696, 2015. doi: 10.1074/jbc.M115.671735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.West AP, Khoury-Hanold W, Staron M, Tal MC, Pineda CM, Lang SM, Bestwick M, Duguay BA, Raimundo N, MacDuff DA, Kaech SM, Smiley JR, Means RE, Iwasaki A, Shadel GS. Mitochondrial DNA stress primes the antiviral innate immune response. Nature 520: 553–557, 2015. doi: 10.1038/nature14156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Wisse E, De Zanger RB, Charels K, Van Der Smissen P, McCuskey RS. The liver sieve: considerations concerning the structure and function of endothelial fenestrae, the sinusoidal wall and the space of Disse. Hepatology 5: 683–692, 1985. doi: 10.1002/hep.1840050427. [DOI] [PubMed] [Google Scholar]
  • 168.Wu LQ, Zhang WJ, Niu JX, Ye LY, Yang ZH, Grau GE, Lou JN. Phenotypic and functional differences between human liver cancer endothelial cells and liver sinusoidal endothelial cells. J Vasc Res 45: 78–86, 2008. doi: 10.1159/000109079. [DOI] [PubMed] [Google Scholar]
  • 169.Xie G, Wang X, Wang L, Wang L, Atkinson RD, Kanel GC, Gaarde WA, 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. doi: 10.1053/j.gastro.2011.12.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Xiong X, Kuang H, Ansari S, Liu T, Gong J, Wang S, Zhao XY, Ji Y, Li C, Guo L, Zhou L, Chen Z, Leon-Mimila P, Chung MT, Kurabayashi K, Opp J, Campos-Pérez F, Villamil-Ramírez H, Canizales-Quinteros S, Lyons R, Lumeng CN, Zhou B, Qi L, Huertas-Vazquez A, Lusis AJ, Xu XZS, Li S, Yu Y, Li JZ, Lin JD. Landscape of intercellular crosstalk in healthy and NASH liver revealed by single-cell secretome gene analysis. Mol Cell 75: 644–660.e5, 2019. doi: 10.1016/j.molcel.2019.07.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Yamashita M, Taniyama M, Tamai M. Cellular localization of tumor necrosis factor-alpha mRNA and interleukin-6 mRNA in the rat liver after hemorrhagic shock. Surg Today 32: 701–706, 2002. doi: 10.1007/s005950200130. [DOI] [PubMed] [Google Scholar]
  • 172.Yang H, Majno P, Morel P, Toso C, Triponez F, Oberholzer J, Mentha G, Lou J. Prostaglandin E(1) protects human liver sinusoidal endothelial cell from apoptosis induced by hypoxia reoxygenation. Microvasc Res 64: 94–103, 2002. doi: 10.1006/mvre.2002.2404. [DOI] [PubMed] [Google Scholar]
  • 173.Yang H, Wang H, Ren J, Chen Q, Chen ZJ. cGAS is essential for cellular senescence. Proc Natl Acad Sci USA 114: E4612–E4620, 2017. doi: 10.1073/pnas.1705499114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Yu Q, Katlinskaya YV, Carbone CJ, Zhao B, Katlinski KV, Zheng H, Guha M, Li N, Chen Q, Yang T, Lengner CJ, Greenberg RA, Johnson FB, Fuchs SY. DNA-damage-induced type I interferon promotes senescence and inhibits stem cell function. Cell Reports 11: 785–797, 2015. doi: 10.1016/j.celrep.2015.03.069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Yu Y, Liu Y, An W, Song J, Zhang Y, Zhao X. STING-mediated inflammation in Kupffer cells contributes to progression of nonalcoholic steatohepatitis. J Clin Invest 129: 546–555, 2019. doi: 10.1172/JCI121842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Zapotoczny B, Szafranska K, Kus E, Braet F, Wisse E, Chlopicki S, Szymonski M. Tracking fenestrae dynamics in live murine liver sinusoidal endothelial cells. Hepatology 69: 876–888, 2019. doi: 10.1002/hep.30232. [DOI] [PubMed] [Google Scholar]
  • 177.Zeisberg EM, Tarnavski O, Zeisberg M, Dorfman AL, McMullen JR, Gustafsson E, Chandraker A, Yuan X, Pu WT, Roberts AB, Neilson EG, Sayegh MH, Izumo S, Kalluri R. Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat Med 13: 952–961, 2007. doi: 10.1038/nm1613. [DOI] [PubMed] [Google Scholar]
  • 178.Zhao XY, Xiong X, Liu T, Mi L, Peng X, Rui C, Guo L, Li S, Li X, Lin JD. Long noncoding RNA licensing of obesity-linked hepatic lipogenesis and NAFLD pathogenesis. Nat Commun 9: 2986, 2018. doi: 10.1038/s41467-018-05383-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Zhou J, Li YS, Chien S. Shear stress-initiated signaling and its regulation of endothelial function. Arterioscler Thromb Vasc Biol 34: 2191–2198, 2014. doi: 10.1161/ATVBAHA.114.303422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Zierhut C, Yamaguchi N, Paredes M, Luo JD, Carroll T, Funabiki H. The cytoplasmic DNA sensor cGAS promotes mitotic cell death. Cell 178: 302–315.e23, 2019. doi: 10.1016/j.cell.2019.05.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Zindel J, Kubes P. DAMPs, PAMPs, and LAMPs in immunity and sterile inflammation. Annu Rev Pathol 15: 493–518, 2020. doi: 10.1146/annurev-pathmechdis-012419-032847. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Cell Physiology are provided here courtesy of American Physiological Society

RESOURCES