Topic Highlights Open Access
Copyright ©2009 The WJG Press and Baishideng. All rights reserved.
World J Gastroenterol. Mar 14, 2009; 15(10): 1194-1200
Published online Mar 14, 2009. doi: 10.3748/wjg.15.1194
Impact of asialoglycoprotein receptor deficiency on the development of liver injury
Serene ML Lee, Departments of Internal Medicine and Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68105, United States
Carol A Casey, Benita L McVicker, Liver Study Unit, Department of Veterans Affairs Medical Center, and Departments of Internal Medicine and Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68105, United States
Author contributions: All authors contributed to the writing of this manuscript.
Correspondence to: Carol A Casey, PhD, Veterans Affairs Medical Center, Research Service (151), 4101 Woolworth Avenue, Omaha, NE 68105, United States. ccasey@unmc.edu
Telephone: +1-402-9953737
Fax: +1-402-4490604
Received: December 17, 2008
Revised: January 6, 2009
Accepted: January 13, 2009
Published online: March 14, 2009

Abstract

The asialoglycoprotein (ASGP) receptor is a well-characterized hepatic receptor that is recycled via the common cellular process of receptor-mediated endocytosis (RME). The RME process plays an integral part in the proper trafficking and routing of receptors and ligands in the healthy cell. Thus, the mis-sorting or altered transport of proteins during RME is thought to play a role in several diseases associated with hepatocyte and liver dysfunction. Previously, we examined in detail alterations that occur in hepatocellular RME and associated receptor functions as a result of one particular liver injury, alcoholic liver disease (ALD). The studies revealed profound ethanol-mediated impairments to the ASGP receptor and the RME process, indicating the importance of this receptor and the maintenance of proper endocytic events in normal tissue. To further clarify these observations, studies were performed utilizing knockout mice (lacking a functional ASGP receptor) to which were administered several liver toxicants. In addition to alcohol, we examined the effects following administration of anti-Fas (CD95) antibody, carbon tetrachloride (CCl4) and lipopolysaccharide (LPS)/galactosamine. The results of these studies demonstrated that the knockout mice sustained enhanced liver injury in response to all of the treatments, as shown by increased indices of liver damage, such as enhancement of serum enzyme levels, histopathological scores, as well as hepatocellular death. Overall, the work completed to date suggests a possible link between hepatic receptors and liver injury. In particular, adequate function and content of the ASGP receptor may provide protection against various toxin-mediated liver diseases.

Key Words: Asialoglycoprotein receptor, Asialoglycoprotein receptor deficient mice, Receptor-mediated endocytosis, Alcohol, Carbon tetrachloride, Anti-Fas, Lipopolysaccharide/galactosamine, Toxicant-induced liver injury



THE ASIALOGLYCOPROTEIN RECEPTOR AND ITS POTENTIAL ROLE IN LIVER INJURY

The asialoglycoprotein (ASGP) receptor, also termed the hepatic binding protein or the Ashwell receptor, was discovered nearly four decades ago by Ashwell and Morell, and was described as a hepatocellular surface carbohydrate that binds glycoproteins lacking terminal sialic acid residues (asialoglycoproteins)[12]. Subsequently, many studies have contributed to the detailed characterization of the ASGP receptor, describing its functional role in the binding, internalization and transport of a wide range of glycoproteins, which have exposed galactose or N-acetylgalactosamine residues, via the process of receptor-mediated endocytosis (RME)[36]. However, translating altered ASGP receptor function and its altered clearance of serum glycoproteins to disease states remains a topic of current research efforts. This ongoing interest is fueled by the knowledge that the ASGP receptor can bind a variety of important plasma proteins that include transport proteins (i.e. transferrin)[7], enzymes such as alkaline phosphatase[8], immunoglobulins including IgA[9], apoptotic hepatocytes[1011], fibronectin[12] and platelets[13]. Additionally, the expression of the ASGP receptor has been clinically correlated to the level of hepatic function that is lost during liver diseases related to cancer, viral hepatitis, and cirrhosis[1415]. Overall, the quest to identify and understand the physiological role(s) of the ASGP receptor, and the consequences that may result from alterations in the function and/or expression of this abundant hepatocellular binding protein, continues.

In search of the physiological roles of the ASGP receptor, our lab initially concentrated on characterizing the role of the ASGP receptor and RME events during a serious and common form of liver injury, alcoholic liver disease (ALD). Alcoholism, and resultant ALD, are indeed significant biomedical problems. Specifically, recent data has noted that chronic liver disease and cirrhosis was the 12th leading cause of death in the United States in the year 2005, and that out of those deaths, approximately 47% of them were due to ALD[16]. Therefore, defining potential contributing mechanisms (such as altered protein trafficking and impaired hepatic receptor functions) may aid in the elucidation of potential therapeutic treatments for ALD. In that effort, our laboratory has extensively studied the RME process and parameters of the ASGP receptor following the administration of ethanol to rodents.

The ASGP receptor consists of major and minor subunits, which in the rat were identified as rat hepatic lectin (RHL) 1 and RHL 2/3, that have respective molecular weights of 42, 49 and 54 kDa[17]. The selective binding and uptake of terminal galactosyl bearing proteins requires the formation of hetero-oligomers between these major and minor forms, and that binding activity was calcium and pH dependent[2518]. Also, the subcellular distribution of the receptor revealed that approximately one-third of the total ASGP receptor pool was associated with the plasma membrane located on the basolateral surface of the hepatocyte[19]. Additionally, it was shown that the total ASGP receptor population consisted of two functionally distinct receptor populations, designated State 1 and State 2, which were involved in the endocytosis and intracellular processing of ligands by different pathways[2022].

Utilizing these known properties, we studied the effects of ethanol on the ASGP receptor itself, as well as endocytic processes, using isolated hepatocytes, whole liver sections, and perfused livers obtained from rats voluntarily fed an ethanol containing diet over a time course of administration. In summary, differential effects were observed over the time course of treatment in the ability of ethanol and resultant metabolites to affect the ASGP receptor and RME events. Specifically, after early periods of ethanol feeding (1-2 wk), we found that the observed decrease in ligand binding capacity of the ASGP receptor could be attributed to inactivation and redistribution of the receptor[23]. However, after more chronic ethanol administration (5-8 wk), the functional alterations of the receptor were found to be reflective of reductions in the content, synthesis, and mRNA expression of the receptor[23]. Also, it was determined that ethanol treatment caused equal inactivation of both State 1 and State 2 receptors, suggesting that ethanol may be unique compared to other agents (e.g. monensin, vanadate, and chloroquine) that are known to inflict post-translational modifications, such as acylation, selectively to just the State 2 population[24]. In other studies, it was revealed that the ASGP receptor was hyperphosphorylated over the time course of treatment, which could contribute to the aberrant activity of the receptor by disrupting the phosphorylation/dephosphorylation state associated with normal recycling of the receptor[25]. We were also able to demonstrate that the ASGP receptor is involved in the recognition and uptake of apoptotic cells and that this process was significantly altered in hepatocytes obtained from ethanol fed rats[11]. Overall, the results from these studies revealed that ethanol administration impairs multiple aspects of RME by the hepatic ASGP receptor, such that binding, internalization and degradation of ligands internalized by the receptor were found to be significantly altered. Additionally, it was shown that these defects are associated with alterations in the ASGP receptor’s physiologically relevant role of clearing apoptotic cells. Taken together, our findings have important implications for the pathogenesis of alcoholic liver injury and potentially for other forms of liver diseases in which RME is profoundly affected. In more recent studies, a mouse model lacking the ASGP receptor was used to gain a better understanding of the associations that may exist between alterations in receptor function and the generation of pathological liver injury.

THE ASGP RECEPTOR-DEFICIENT MOUSE MODEL

The ASGP receptor in mice is a hetero-oligomeric receptor composed of 2 subunits that are both required for its function. These subunits have been named murine hepatic lectin (MHL), with the major subunit called MHL-1 and the minor subunit called MHL-2[26]. ASGP receptor-deficient (RD) mice have a complete lack of the MHL-2 protein and were generated by homologous recombination with a gene replacement vector in embryonic stem cells[27]. MHL-2 appears to be required for the post-translational stability of MHL-1, as these mice have substantially reduced protein content of MHL-1, even though MHL-1 mRNA expression remains the same[27]. Although the MHL-1 protein is still detected in low levels in the RD mice, these levels are unable to induce a measurable clearance of 125I-labeled asialo-orosomucoid[27]. Despite lacking functional ASGP receptors, these knockout mice remain viable and fertile, and appear to have a normal lifespan. In addition, these mice do not display any obvious phenotypic abnormalities[2728].

As previously mentioned, we have found that chronic alcohol administration markedly decreased mRNA expression and content of the ASGP receptor in rats prior to the appearance of pathology such as fibrosis[2329]. Thus, it was felt that the RD mice might provide a powerful tool to examine the role of the ASGP receptor and help delineate pathways by which liver injury occurs in general, as well as during alcoholic liver injury. Currently, we are utilizing the knockout mouse model to examine the link between ASGP receptor function and liver injury, in the context of various models of toxic liver injury such as alcohol, anti-Fas, carbon tetrachloride (CCl4) and lipopolysaccharide (LPS)/galactosamine. In this report, we present a brief overview of our findings to date.

MODELS OF LIVER INJURY AND THEIR EFFECTS ON ASGP RECEPTOR-DEFICIENT MICE
Alcohol

Alcohol-induced liver injury has previously been found to be related to several events, including ethanol metabolism (via alcohol dehydrogenase[3033]), generation of reactive oxygen species (via cytochrome isoforms such as CYP2E1[3436]), interaction of other liver products (such as cytokines[3738]) and the induction of apoptosis through the Fas death receptor system[39]. In the search for cellular signaling and mechanisms resulting as a consequence of these events, studies were performed that examined the effects of ethanol on hepatocellular protein trafficking, particularly the process of RME utilizing the hepatic ASGP receptor.

As mentioned previously, we examined the effects of ethanol administration using a rat model exclusively; the rats showed decreased ligand binding, internalization and degradation of several ligands including asialo-orosomucoid, which are processed by RME[4234043]. In order to assess the effect of ethanol administration on RME by the ASGP receptor using a mouse model, we obtained wild-type (WT) mice possessing abundant ASGP receptor activity and ASGP receptor-deficient (RD) mice lacking MHL-2 from the Jackson Laboratories (Bar Harbor, ME). The mice were fed a Lieber de-Carli liquid diet (with or without 5% by volume ethanol) for ten days[44]. When hepatocytes from these mice were incubated with 125I-ASOR (a representative ligand for the ASGP receptor), WT mice showed ethanol-induced alterations that were consistent with our observations for rats, with an approximately 50% decrease in ligand binding, internalization and degradation in isolated hepatocytes[42344]. However, binding, internalization and degradation of the ligand by RD hepatocytes was negligible, regardless of diet[44]. In addition, the presence of apoptotic bodies was found to be approximately three-fold higher in the livers of RD mice compared to WT mice, irrespective of diet[44]. As a result of this work, it is hypothesized that a potential consequence of altered ASGP receptor function is impaired clearance of ethanol-generated apoptotic cells, resulting in the observed accumulation of apoptotic bodies. Furthermore, other work has shown that these bodies have the potential to promote a variety of responses within the liver, such as the activation of Kupffer cells and the subsequent release of proinflammatory and profibrogenic substances, leading to the enhanced susceptibility to hepatocellular damage that is observed following ethanol administration[1145].

Anti-Fas

Anti-Fas is an antibody that specifically recognizes and works as an agonist of the Fas antigen[46]. Fas is a member of the TNF receptor superfamily and is a key mediator of apoptosis[47]. This receptor is found in hepatocytes, cholangiocytes, sinusoidal endothelial cells, stellate cells and Kupffer cells[48]. Ligation of Fas results in the recruitment of adaptor proteins, such as Fas-associated death domain (FADD) and procaspase 8, to form the death-inducing signaling complex (DISC)[47]. Caspase 8 can then either directly or indirectly cleave procaspase 3 to mediate apoptosis[47]. A variety of studies have shown that the injection of anti-Fas into mice causes widespread apoptosis and ultimately results in focal hemorrhage and hepatocyte necrosis, making Fas injection a model for fulminant hepatic failure[464950].

From studies related to Fas-mediated cell death in our laboratory, we have shown that the metabolism of ethanol in WIF-B cells (hepatoma hybrid cells) was involved in enhanced Fas protein localization to the membrane, leading to increased activity of the upstream initiator caspases (caspase 2 and caspase 8) and the subsequent downstream activation of caspase 3[39]. As an extension to these studies, aimed to characterize the role of Fas-mediated death in injured hepatocytes, anti-Fas (0.1 or 0.2 &mgr;g/g body weight) was injected intraperitoneally into WT and RD mice, which were monitored for up to 48 h[51]. Receptor-deficient mice showed an enhancement of liver injury with higher aspartate transaminase (AST) and alanine transaminase (ALT) activities in the serum compared to the enzyme levels detected in WT mice[51]. Similarly, pathology showed that the RD mice had increased steatosis, inflammation and necrosis compared to the WT mice[51]. As expected, caspase 3 activities were found to be increased 5- to 6-fold in WT mice at 2 h and 16 h after anti-Fas injection, with caspase activities returning to baseline levels by 24 h[51]. However, the activity of caspase 3 remained elevated in the RD livers at all times following treatment and was significantly enhanced over the WT livers at 24 h and 48 h post anti-Fas injection[51]. Overall, the livers of the RD mice were found to be more susceptible than the livers of the WT mice to anti-Fas injection; showing greater apoptosis and increased ECM deposition of collagen and fibronectin[51].

Carbon tetrachloride

Another agent used to study liver injury is carbon tetrachloride (CCl4), which can cause liver damage through a number of mechanisms. Carbon tetrachloride is metabolized through the action of the mixed function cytochrome P450 system of the endoplasmic reticulum to form the trichloromethyl free radical (CCl3•), which can subsequently be converted to the trichloromethyl peroxy radical (CCl3OO•) in the presence of oxygen[5253]. These free radicals are highly reactive and can bind covalently to cellular macromolecules forming nucleic acid, protein and lipid adducts. When these radicals attack the polyunsaturated fatty acids of the cellular membranes, the fatty acid free radicals generated initiate autocatalytic lipid peroxidation, ultimately resulting in the loss of membrane integrity[5253]. Carbon tetrachloride can also induce cellular hypomethylation, leading to inhibition of protein synthesis (possibly through ribosomal RNA hypomethylation) and defects in lipid and lipoprotein metabolism[53]. Finally, CCl4 also affects hepatocellular calcium homeostasis, either by disrupting membrane integrity or by opening certain membrane calcium channels. High levels of Ca2+ in the cell can then activate Ca2+-responsive enzymes such as proteases, endonucleases and phospholipases and lead to cell death via apoptosis and necrosis[5253]. The consequences of CCl4 toxicity include centrilobular steatosis, inflammation, apoptosis and necrosis[5254].

In our studies, WT and RD mice were injected with CCl4 (1 mL/kg body weight) and monitored up to a week after injection[55]. Carbon tetrachloride injection caused greater liver injury in the RD mice, as evidenced by the RD mice having increased AST and ALT activities in the serum, compared to the WT mice 48 h post CCl4 injection[55]. Histologically, centrilobular liver damage was observed in WT mice by 48 h after injection[55]. At this time point, RD mice had more severe damage, showing a greater number of neutrophilic inflammatory infiltrates[55]. In order to elucidate the mechanisms by which this damage is caused, malondia dehyde (MDA), deposition of α-smooth muscle actin (α-SMA) and the percentage of TUNEL-positive hepatocytes was measured[55]. Levels of MDA in the WT mice were not significantly increased throughout the course of the experiment[55]. In contrast, RD mice showed increased contents of MDA as early as 24 h post CCl4 injection and these levels were maintained up to 48 h[55]. α-SMA was increased significantly in both the WT and RD mice, with the RD mice having a more prolonged increase (between 48 h to 72 h) than the WT mice (only at 72 h)[55]. In addition, α-SMA content was approximately 2-fold of that in the WT liver at 48 h, 96 h and 7 d following injection[55]. Finally, RD mice had significantly more TUNEL-positive hepatocytes than the WT mice at 48 h post injection (2.4-fold more)[55]. This suggests that the absence of functional ASGP receptor resulted in increased lipid peroxidation, perturbations in ECM turnover and increased apoptosis[55].

LPS/galactosamine

Lipopolysaccharide (endotoxin) and galactosamine can be used either alone or in combination with each other to cause liver injury in mice. The metabolism of galactosamine leads to hepatotoxicity by depleting uridine nucleotides and UDP-hexoses and concurrently increasing UDP-hexosamines, primarily in hepatocytes[5657]. The depletion of uridine nucleotides (as mentioned above) results in an inhibition of RNA and protein synthesis[58]. It has also been suggested that metabolism of galactosamine results in an impaired biosynthesis of macromolecular cell constituents[56]. This impairment leads to plasma membrane injury, which results in an influx of calcium ions and a commitment to cell death[58]. In rats, galactosamine leads to an inflammatory infiltrate of polymorphonuclear leukocytes and lymphocytes and foci of hepatocellular necrosis, resembling the effects of human viral hepatitis[5759].

Mice and rats are relatively resistant to the lethal effects of LPS[60]. Thus, LPS is injected in concert with galactosamine for a model of fulminant hepatitis[61]. It is thought that galactosamine-induced suppression of RNA synthesis leads to an increased tumor necrosis factor (TNF) production by macrophages, resulting in an increased susceptibility to LPS[6062]. Tumor necrosis factor was proposed as the agent responsible for the lethality, because lethality was retained by substituting LPS with TNF and was inhibited by anti-TNF antibody[6364]. Thus, in LPS/galactosamine injection, apoptosis induced by TNF occurs initially and is followed subsequently by necrosis[65].

In our studies, a sub-lethal dose of LPS (50 &mgr;g/kg body weight) combined with galactosamine (350 mg/kg body weight) was injected into WT and RD mice via the intraperitoneal route and the mice were monitored up to 4.5 h[6667]. After LPS/galactosamine injection, WT mice maintained normal liver lobular architecture[66]. However, RD mice showed considerable liver injury with areas of portal inflammation, hepatocellular necrosis, increased inflammatory cell infiltration and hemorrhage[66]. These histological observations were further corroborated by the RD mice having increased serum AST and ALT activities at 4.5 h after LPS/galactosamine injection, which were not observed in the WT mice[66]. Also, RD mice showed increased apoptosis, having significantly enhanced caspase 3 activities and TUNEL-positive cells at 4.5 h post injection, whilst there were no changes measured in WT mice[66]. Additionally, the serum content of the pro-inflammatory cytokine interleukin 6 (IL-6) was increased in RD mice compared to WT mice at 3 and 4.5 h after LPS/galactosamine injection[67]. Overall, the results demonstrate that the RD mice are more susceptible to the development fulminant liver injury as a result of sub-lethal treatment with LPS/galactosamine[6667]. Given that the induction of apoptosis is a consequence of LPS/galactosamine treatment, the enhanced susceptibility to liver damage observed in the RD mice may be related to the inability of hepatocytes to phagocytose and clear the dying apoptotic cells via the ASGP receptor.

THE LINK BETWEEN FUNCTIONAL ASGP RECEPTOR AND LIVER INJURY

After the administration of the four toxicants mentioned above (alcohol, anti-Fas, CCl4 and LPS/galactosamine), RD mice consistently sustained greater liver injury than WT mice, as evidenced by increased indices of liver damage (serum AST and ALT activities) and worse pathology (light microscopy). These four agents of liver injury cause damage through different biochemical pathways or signaling cascades. Therefore, it appears that proper functioning of the ASGP receptor may provide universal protection against liver injury from these toxicants and possibly others. Although it is not known exactly how an adequately functioning receptor can protect the hepatocyte, the use of the knockout mice treated with these four toxicants highlight some of the possible mechanisms.

It appears that with all four toxicants, caspase 3 or TUNEL-positive cells are increased. Apoptosis is a highly regulated mode of cell death that helps to maintain tissue homeostasis in a healthy organ[68]. However, it appears that when apoptotic death factors are inappropriately expressed due to the introduction of pathological stimuli, such as the four toxicants, apoptosis becomes one of the common pathways by which liver injury is caused. Increased accumulation of apoptotic cells has been shown to occur during alcohol-induced liver injury in a variety of species, including humans, and is thought to play an important role in the progression of liver injury[6975]. During the process of forming apoptotic cells, glycoconjugates on the cell surface losing their sialic acid masks the increase[1076]. Since the ASGP receptor binds to desialylated proteins, the receptor recognizes and binds the altered glycans on apoptotic bodies, resulting in phagocytosis and efficient removal of the dying cells[11]. Thus, we speculate that the ASGP receptor exerts protection by removing apoptotic bodies in a timely fashion.

Another way that the ASGP receptor might be protective is through its role in regulating turnover of ECM. Anti-Fas and CCl4 treatments lead to increased deposition of collagen, fibronectin or α-SMA in the RD mice in comparison to the WT mice. The ASGP receptor has a direct link to cellular fibronectin clearance because cellular fibronectin displays terminal galactose residues, making it a ligand of the ASGP receptor[12]. Cellular fibronectin is one of the first ECM proteins that accumulates during fibrosis[7779] and thus impairments of ASGP receptor function could lead to increased ECM deposition and hence lead to fibrosis and cirrhosis.

Two additional ways that liver injury could be mediated is through increased lipid peroxidation (MDA content) or by increased contents of pro-inflammatory cytokines, such as IL-6. At present, it is not known how ASGP receptor function is related to perturbations in levels of these two substances. In addition, there may be dysregulation of other asialoglycoproteins not examined at this point. The total absence of ASGP receptor function does not result in a measurable increase in the steady state concentrations of galactose-terminating glycoproteins in the plasma of knockout mice[28]. Thus, the ASGP receptor is unlikely to be involved in the normal turnover of serum glycoproteins. However, in toxicant-induced injuries, acute surges of asialoglycoproteins may overwhelm the alternative galactose recognition systems[28]. Thus, the ASGP receptor might function to prevent an acute increase in potentially harmful asialoglycoproteins. Altogether, the ASGP receptor knockout mouse model provides an excellent tool to elucidate the relationship between ASGP receptor function and liver injury.

CONCLUSION

The ASGP receptor is an abundant hepatic receptor that recognizes desialylated ligands. After binding to its ligand, the receptor internalizes and facilitates transport of specific ligands by the process of receptor-mediated endocytosis. Previously, studies have shown that ASGP receptor function is impaired in disease states such as alcoholic liver disease. This gave us the impetus to examine if proper ASGP receptor function offers protection against liver injury or if defects in function occurred as a result of liver damage. To examine this, we utilized a knockout mouse model, which lacked functional ASGP receptor in comparison to wild-type animals in the context of various toxic challenges (alcohol, anti-Fas, CCl4 and LPS/galactosamine). After all four challenges, the receptor-deficient mice consistently showed more liver injury than the wild-type animals (Table 1), proving that ASGP receptor function is protective. Thus, these studies highlight receptor-mediated endocytosis as a novel mechanism that may be involved in the induction of toxin-induced liver injury. At present, the precise nature of the specific ligands involved or the pathways that lead to further injury have not been determined. However, in the studies reviewed here, it is likely that impaired clearance of apoptotic bodies, perturbations in extracellular matrix deposition, oxidative stress, and cytokine dysregulation may play roles in the progression of disease. In the future, further clarification of the pathways by which liver injury occurs (including altered ASGP receptor-mediated endocytosis) will provide new therapeutic leads.

Table 1 Changes in ALT activity, TUNEL positive cells, AST activity, caspase 3 activity, collagen content, IL-6 content, MDA content and α-SMA content in RD mice compared to WT mice.
ChallengeRD mice compared to WT mice
AlcoholLiver TUNEL-positive hepatocytes
Anti-FasLiver TUNEL-positive hepatocytes
Serum ALT
Serum AST
Liver caspase 3 activity
Collagen deposition in extracellular matrix
Fibronectin deposition in the extracellular matrix
CCl4Liver TUNEL-positive hepatocytes
Serum ALT
Serum AST
Liver MDA content
Liver α-SMA content
LPS/galactosamineLiver TUNEL-positive hepatocytes
Serum ALT
Serum AST
Liver caspase 3 activity
Serum IL-6 content
Footnotes

Supported by The National Institute on Alcohol Abuse and Alcoholism and by the Department of Veterans Affairs

Peer reviewer: Dr. Robin Hughes, Institute of Liver Studies, King’s College London School of Medicine, Bessemer Road, London, SE5 9PJ, United Kingdom

S- Editor Li LF L- Editor Stewart GJ E- Editor Lin YP

References
1.  Ashwell G, Morell AG. The role of surface carbohydrates in the hepatic recognition and transport of circulating glycoproteins. Adv Enzymol Relat Areas Mol Biol. 1974;41:99-128.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Ashwell G, Kawasaki T. A protein from mammalian liver that specifically binds galactose-terminated glycoproteins. Methods Enzymol. 1978;50:287-288.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Ashwell G, Steer CJ. Hepatic recognition and catabolism of serum glycoproteins. JAMA. 1981;246:2358-2364.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Casey CA, Kragskow SL, Sorrell MF, Tuma DJ. Chronic ethanol administration impairs the binding and endocytosis of asialo-orosomucoid in isolated hepatocytes. J Biol Chem. 1987;262:2704-2710.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Stockert RJ. The asialoglycoprotein receptor: relationships between structure, function, and expression. Physiol Rev. 1995;75:591-609.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Weigel PH. Endocytosis and function of the hepatic asialoglycoprotein receptor. Subcell Biochem. 1993;19:125-161.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Debanne MT, Evans WH, Flint N, Regoeczi E. Receptor-rich intracellular membrane vesicles transporting asialotransferrin and insulin in liver. Nature. 1982;298:398-400.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Meijer DK, Scholtens HB, Hardonk MJ. The role of the liver in clearance of glycoproteins from the general circulation, with special reference to intestinal alkaline phosphatase. Pharm Weekbl Sci. 1982;4:57-70.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Tomana M, Phillips JO, Kulhavy R, Mestecky J. Carbohydrate-mediated clearance of secretory IgA from the circulation. Mol Immunol. 1985;22:887-892.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Dini L, Autuori F, Lentini A, Oliverio S, Piacentini M. The clearance of apoptotic cells in the liver is mediated by the asialoglycoprotein receptor. FEBS Lett. 1992;296:174-178.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  McVicker BL, Tuma DJ, Kubik JA, Hindemith AM, Baldwin CR, Casey CA. The effect of ethanol on asialoglycoprotein receptor-mediated phagocytosis of apoptotic cells by rat hepatocytes. Hepatology. 2002;36:1478-1487.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Rotundo RF, Rebres RA, Mckeown-Longo PJ, Blumenstock FA, Saba TM. Circulating cellular fibronectin may be a natural ligand for the hepatic asialoglycoprotein receptor: possible pathway for fibronectin deposition and turnover in the rat liver. Hepatology. 1998;28:475-485.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Grewal PK, Uchiyama S, Ditto D, Varki N, Le DT, Nizet V, Marth JD. The Ashwell receptor mitigates the lethal coagulopathy of sepsis. Nat Med. 2008;14:648-655.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Sawamura T, Nakada H, Hazama H, Shiozaki Y, Sameshima Y, Tashiro Y. Hyperasialoglycoproteinemia in patients with chronic liver diseases and/or liver cell carcinoma. Asialoglycoprotein receptor in cirrhosis and liver cell carcinoma. Gastroenterology. 1984;87:1217-1221.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Virgolini I, Muller C, Klepetko W, Angelberger P, Bergmann H, O’Grady J, Sinzinger H. Decreased hepatic function in patients with hepatoma or liver metastasis monitored by a hepatocyte specific galactosylated radioligand. Br J Cancer. 1990;61:937-941.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Kung HC, Hoyert DL, Xu J, Murphy SL. Deaths: final data for 2005. Natl Vital Stat Rep. 2008;56:1-120.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Drickamer K, Mamon JF, Binns G, Leung JO. Primary structure of the rat liver asialoglycoprotein receptor. Structural evidence for multiple polypeptide species. J Biol Chem. 1984;259:770-778.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Halberg DF, Wager RE, Farrell DC, Hildreth J 4th, Quesenberry MS, Loeb JA, Holland EC, Drickamer K. Major and minor forms of the rat liver asialoglycoprotein receptor are independent galactose-binding proteins. Primary structure and glycosylation heterogeneity of minor receptor forms. J Biol Chem. 1987;262:9828-9838.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Nakada H, Matsuura S, Sawamura T, Tashiro Y. Topology of asialoglycoprotein receptor in rat liver subcellular fractions: a ferritin immunoelectron microscopic study. Cell Struct Funct. 1984;9:391-406.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Stockert RJ, Potvin B, Tao L, Stanley P, Wolkoff AW. Human hepatoma cell mutant defective in cell surface protein trafficking. J Biol Chem. 1995;270:16107-16113.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Weigel PH, Medh JD, Oka JA. A novel cycle involving fatty acyl-coenzyme A regulates asialoglycoprotein receptor activity in permeable hepatocytes. Mol Biol Cell. 1994;5:227-235.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Weigel PH, Oka JA. Regulation of asialoglycoprotein receptor activity by a novel inactivation/reactivation cycle. Receptor reactivation in permeable rat hepatocytes is mediated by fatty acyl coenzyme A. J Biol Chem. 1993;268:27186-27190.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Tworek BL, Tuma DJ, Casey CA. Decreased binding of asialoglycoproteins to hepatocytes from ethanol-fed rats. Consequence of both impaired synthesis and inactivation of the asialoglycoprotein receptor. J Biol Chem. 1996;271:2531-2538.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Tworek BL, Oka JA, Casey CA, Weigel PH. Ethanol feeding causes inactivation of both state 1 and state 2 rat hepatic asialoglycoprotein receptors. Alcohol Clin Exp Res. 1997;21:1429-1434.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  McVicker BL, Tuma DJ, Casey CA. Hyperphosphorylation of the asialoglycoprotein receptor in isolated rat hepatocytes following ethanol administration. Biochem Pharmacol. 2000;60:343-351.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Hong W, Le AV, Doyle D. Identification and characterization of a murine receptor for galactose-terminated glycoproteins. Hepatology. 1988;8:553-558.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Ishibashi S, Hammer RE, Herz J. Asialoglycoprotein receptor deficiency in mice lacking the minor receptor subunit. J Biol Chem. 1994;269:27803-27806.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Braun JR, Willnow TE, Ishibashi S, Ashwell G, Herz J. The major subunit of the asialoglycoprotein receptor is expressed on the hepatocellular surface in mice lacking the minor receptor subunit. J Biol Chem. 1996;271:21160-21166.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Casey CA, Sorrell MF, Tuma DJ. Effect of ethanol on asialoglycoprotein receptor function. Targeted Diagn Ther. 1991;4:189-213.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Mailliard ME, Sorrell MF, Volentine GD, Tuma DJ. Impaired plasma membrane glycoprotein assembly in the liver following acute ethanol administration. Biochem Biophys Res Commun. 1984;123:951-958.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Sorrell MF, Tuma DJ. Hypothesis: alcoholic liver injury and the covalent binding of acetaldehyde. Alcohol Clin Exp Res. 1985;9:306-309.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Tuma DJ, Sorrell MF. Effects of ethanol on protein trafficking in the liver. Semin Liver Dis. 1988;8:69-80.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Tuma DJ, Casey CA, Sorrell MF. Effects of ethanol on hepatic protein trafficking: impairment of receptor-mediated endocytosis. Alcohol Alcohol. 1990;25:117-125.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Wu D, Cederbaum AI. Ethanol cytotoxicity to a transfected HepG2 cell line expressing human cytochrome P4502E1. J Biol Chem. 1996;271:23914-23919.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Castillo T, Koop DR, Kamimura S, Triadafilopoulos G, Tsukamoto H. Role of cytochrome P-450 2E1 in ethanol-, carbon tetrachloride- and iron-dependent microsomal lipid peroxidation. Hepatology. 1992;16:992-996.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Chen Q, Galleano M, Cederbaum AI. Cytotoxicity and apoptosis produced by arachidonic acid in Hep G2 cells overexpressing human cytochrome P4502E1. J Biol Chem. 1997;272:14532-14541.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Hoek JB, Pastorino JG. Ethanol, oxidative stress, and cytokine-induced liver cell injury. Alcohol. 2002;27:63-68.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  McClain CJ, Barve S, Deaciuc I, Kugelmas M, Hill D. Cytokines in alcoholic liver disease. Semin Liver Dis. 1999;19:205-219.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  McVicker BL, Tuma DJ, Kubik JL, Tuma PL, Casey CA. Ethanol-induced apoptosis in polarized hepatic cells possibly through regulation of the Fas pathway. Alcohol Clin Exp Res. 2006;30:1906-1915.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Casey CA, Kragskow SL, Sorrell MF, Tuma DJ. Ethanol-induced impairments in receptor-mediated endocytosis of asialoorosomucoid in isolated rat hepatocytes: time course of impairments and recovery after ethanol withdrawal. Alcohol Clin Exp Res. 1989;13:258-263.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Casey CA, Volentine GD, Jankovich CJ, Kragskow SL, Tuma DJ. Effect of chronic ethanol administration on the uptake and degradation of asialoglycoproteins by the perfused rat liver. Biochem Pharmacol. 1990;40:1117-1123.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Casey CA, Tuma DJ. Receptors and endocytosis. Molecular and Cell Biology of the Liver. Ann Arbor: CRC Press 1993; 117-141.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Casey CA, McVicker BL, Donohue TM Jr, McFarland MA, Wiegert RL, Nanji AA. Liver asialoglycoprotein receptor levels correlate with severity of alcoholic liver damage in rats. J Appl Physiol. 2004;96:76-80.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Dalton SR, Wiegert RL, Baldwin CR, Kassel KM, Casey CA. Impaired receptor-mediated endocytosis by the asialoglycoprotein receptor in ethanol-fed mice: implications for studying the role of this receptor in alcoholic apoptosis. Biochem Pharmacol. 2003;65:535-543.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  McVicker BL, Tuma DJ, Kharbanda KK, Kubik JL, Casey CA. Effect of chronic ethanol administration on the in vitro production of proinflammatory cytokines by rat Kupffer cells in the presence of apoptotic cells. Alcohol Clin Exp Res. 2007;31:122-129.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Ogasawara J, Watanabe-Fukunaga R, Adachi M, Matsuzawa A, Kasugai T, Kitamura Y, Itoh N, Suda T, Nagata S. Lethal effect of the anti-Fas antibody in mice. Nature. 1993;364:806-809.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Schutze S, Tchikov V, Schneider-Brachert W. Regulation of TNFR1 and CD95 signalling by receptor compartmentalization. Nat Rev Mol Cell Biol. 2008;9:655-662.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Malhi H, Gores GJ. Cellular and molecular mechanisms of liver injury. Gastroenterology. 2008;134:1641-1654.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Kakinuma C, Takagaki K, Yatomi T, Nakamura N, Nagata S, Uemura A, Shibutani Y. Acute toxicity of an anti-Fas antibody in mice. Toxicol Pathol. 1999;27:412-420.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Nishimura Y, Hirabayashi Y, Matsuzaki Y, Musette P, Ishii A, Nakauchi H, Inoue T, Yonehara S. In vivo analysis of Fas antigen-mediated apoptosis: effects of agonistic anti-mouse Fas mAb on thymus, spleen and liver. Int Immunol. 1997;9:307-316.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Baldwin CR, Radio , SJ , Kassel KM, Dalton , SR , McVicker BL, Kubik JL, Wiegert RL, Casey CA. Increased susceptibility to anti-fas antibody-induced liver injury in an asialoglycoprotein receptor deficient mouse. Hepatology. 2002;36:324A.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Manibusan MK, Odin M, Eastmond DA. Postulated carbon tetrachloride mode of action: a review. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev. 2007;25:185-209.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Weber LW, Boll M, Stampfl A. Hepatotoxicity and mechanism of action of haloalkanes: carbon tetrachloride as a toxicological model. Crit Rev Toxicol. 2003;33:105-136.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Shi J, Aisaki K, Ikawa Y, Wake K. Evidence of hepatocyte apoptosis in rat liver after the administration of carbon tetrachloride. Am J Pathol. 1998;153:515-525.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  McVicker BL, Nanji AA, Lee SM, Kharbanda KK, Casey CA. Asialoglycoprotein receptor deficiency increases carbon tetrachloride-induced liver damage in a mouse model. Hepatology. 2008;48:468A.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Decker K, Keppler D. Galactosamine hepatitis: key role of the nucleotide deficiency period in the pathogenesis of cell injury and cell death. Rev Physiol Biochem Pharmacol. 1974;48:77-106.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Farber JL, Gill G, Konishi Y. Prevention of galactosamine-induced liver cell necrosis by uridine. Am J Pathol. 1973;72:53-62.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  El-Mofty SK, Scrutton MC, Serroni A, Nicolini C, Farber JL. Early, reversible plasma membrane injury in galactosamine-induced liver cell death. Am J Pathol. 1975;79:579-596.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Keppler D, Lesch R, Reutter W, Decker K. Experimental hepatitis induced by D-galactosamine. Exp Mol Pathol. 1968;9:279-290.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Galanos C, Freudenberg MA, Reutter W. Galactosamine-induced sensitization to the lethal effects of endotoxin. Proc Natl Acad Sci USA. 1979;76:5939-5943.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Endo Y, Shibazaki M, Yamaguchi K, Kai K, Sugawara S, Takada H, Kikuchi H, Kumagai K. Enhancement by galactosamine of lipopolysaccharide(LPS)-induced tumour necrosis factor production and lethality: its suppression by LPS pretreatment. Br J Pharmacol. 1999;128:5-12.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Freudenberg MA, Galanos C. Induction of tolerance to lipopolysaccharide (LPS)-D-galactosamine lethality by pretreatment with LPS is mediated by macrophages. Infect Immun. 1988;56:1352-1357.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Freudenberg MA, Galanos C. Tumor necrosis factor alpha mediates lethal activity of killed gram-negative and gram-positive bacteria in D-galactosamine-treated mice. Infect Immun. 1991;59:2110-2115.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Tiegs G, Wolter M, Wendel A. Tumor necrosis factor is a terminal mediator in galactosamine/endotoxin-induced hepatitis in mice. Biochem Pharmacol. 1989;38:627-631.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Leist M, Gantner F, Bohlinger I, Tiegs G, Germann PG, Wendel A. Tumor necrosis factor-induced hepatocyte apoptosis precedes liver failure in experimental murine shock models. Am J Pathol. 1995;146:1220-1234.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Casey CA, Mulcahy EC, Wiegert RL, Wagner ZC, McVicker BL, Kharbanda KK. Mice lacking functional asialoglycoprotein receptor show increased susceptibility to D-galactosamine/lipopolysaccharide-induced liver injury. Hepatology. 2006;44:387A.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Lee SM, Casey CA, McVicker BL, Kharbanda KK. Increased production of interleukin-6 in asialoglycoprotein receptor deficient mice after D-galactosamine/lipopolysaccharide-induced liver injury. Alcohol Clin Exp Res. 2008;32:37A.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Hockenbery D. Defining apoptosis. Am J Pathol. 1995;146:16-19.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Benedetti A, Brunelli E, Risicato R, Cilluffo T, Jezequel AM, Orlandi F. Subcellular changes and apoptosis induced by ethanol in rat liver. J Hepatol. 1988;6:137-143.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Deaciuc IV, Fortunato F, D’Souza NB, Hill DB, Schmidt J, Lee EY, McClain CJ. Modulation of caspase-3 activity and Fas ligand mRNA expression in rat liver cells in vivo by alcohol and lipopolysaccharide. Alcohol Clin Exp Res. 1999;23:349-356.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Goldin RD, Hunt NC, Clark J, Wickramasinghe SN. Apoptotic bodies in a murine model of alcoholic liver disease: reversibility of ethanol-induced changes. J Pathol. 1993;171:73-76.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Natori S, Rust C, Stadheim LM, Srinivasan A, Burgart LJ, Gores GJ. Hepatocyte apoptosis is a pathologic feature of human alcoholic hepatitis. J Hepatol. 2001;34:248-253.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Yacoub LK, Fogt F, Griniuviene B, Nanji AA. Apoptosis and bcl-2 protein expression in experimental alcoholic liver disease in the rat. Alcohol Clin Exp Res. 1995;19:854-859.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Zhao M, Laissue JA, Zimmermann A. TUNEL-positive hepatocytes in alcoholic liver disease. A retrospective biopsy study using DNA nick end-labelling. Virchows Arch. 1997;431:337-344.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Ziol M, Tepper M, Lohez M, Arcangeli G, Ganne N, Christidis C, Trinchet JC, Beaugrand M, Guillet JG, Guettier C. Clinical and biological relevance of hepatocyte apoptosis in alcoholic hepatitis. J Hepatol. 2001;34:254-260.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Savill J, Dransfield I, Hogg N, Haslett C. Vitronectin receptor-mediated phagocytosis of cells undergoing apoptosis. Nature. 1990;343:170-173.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Owens MR, Cimino CD. Synthesis of fibronectin by the isolated perfused rat liver. Blood. 1982;59:1305-1309.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Ruoslahti E, Engvall E, Hayman EG. Fibronectin: current concepts of its structure and functions. Coll Relat Res. 1981;1:95-128.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Ballardini G, Faccani A, Fallani M, Berti S, Vasi V, Castaldini C, Biagini G, Garbisa S, Bianchi FB. Sequential behaviour of extracellular matrix glycoproteins in an experimental model of hepatic fibrosis. Virchows Arch B Cell Pathol Incl Mol Pathol. 1985;49:317-324.  [PubMed]  [DOI]  [Cited in This Article: ]