Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Drug Metab Dispos ; 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38050055

ABSTRACT

The role of inflammatory cells and other components of the immune system in acetaminophen (APAP)-induced liver injury and repair has been extensively investigated. Although this has resulted in a wealth of information regarding the function and regulation of immune cells in the liver after injury, apparent contradictions have fueled controversy around the central question of whether the immune system is beneficial or detrimental after APAP overdose. Ultimately, this may not be a simple assignment of "good" or "bad." Clinical studies have clearly demonstrated an association between immune dysregulation and a poor outcome in patients with severe liver damage/liver failure induced by APAP overdose. To date, studies in mice have not uniformly replicated this connection. The apparent disconnect between clinical and experimental studies has perhaps stymied progress and further complicated investigation of the immune system in APAP-induced liver injury. Mouse models are often dismissed as not recapitulating the clinical scenario. Moreover, clinical investigation is most often focused on the most severe APAP overdose patients, those with liver failure. Notably, recent studies have made it apparent that the functional role of the immune system in the pathogenesis of APAP-induced liver injury is highly context dependent and greatly influenced by the experimental conditions. In this review, we highlight some of these recent findings, and suggest strategies seeking to resolve and build on existing disconnects in the literature. Significance Statement Acetaminophen overdose is the most frequent cause of acute liver failure in the United States. Studies indicate that dysregulated innate immunity contributes to the transition from acute liver injury to acute liver failure. In this review, we discuss the evidence for this and the potential underlying causes.

2.
Front Immunol ; 14: 1303921, 2023.
Article in English | MEDLINE | ID: mdl-38094302

ABSTRACT

Introduction: Systemic levels of the anti-inflammatory cytokine interleukin 10 (IL-10) are highest in acetaminophen (APAP)-induced acute liver failure (ALF) patients with the poorest prognosis. The mechanistic basis for this counterintuitive finding is not known, as induction of IL-10 is hypothesized to temper the pathological effects of immune cell activation. Aberrant production of IL-10 after severe liver injury could conceivably interfere with the beneficial, pro-reparative actions of immune cells, such as monocytes. Methods: To test this possibility, we determined whether IL-10 levels are dysregulated in mice with APAP-induced ALF and further evaluated whether aberrant production of IL-10 prevents monocyte recruitment and/or the resolution of necrotic lesions by these cells. Results: Our studies demonstrate that in mice challenged with 300 mg/kg acetaminophen (APAP), a hepatotoxic dose of APAP that fails to produce ALF (i.e., APAP-induced acute liver injury; AALI), Ly6Chi monocytes were recruited to the liver and infiltrated the necrotic lesions by 48 hours coincident with the clearance of dead cell debris. At 72 hours, IL-10 was upregulated, culminating in the resolution of hepatic inflammation. By contrast, in mice treated with 600 mg/kg APAP, a dose that produces clinical features of ALF (i.e., APAP-induced ALF; AALF), IL-10 levels were markedly elevated by 24 hours. Early induction of IL-10 was associated with a reduction in the hepatic numbers of Ly6Chi monocytes resulting in the persistence of dead cell debris. Inhibition of IL-10 in AALF mice, beginning at 24 hours after APAP treatment, increased the hepatic numbers of monocytes which coincided with a reduction in the necrotic area. Moreover, pharmacologic elevation of systemic IL-10 levels in AALI mice reduced hepatic myeloid cell numbers and increased the area of necrosis. Discussion: Collectively, these results indicate that during ALF, aberrant production of IL-10 disrupts the hepatic recruitment of monocytes, which prevents the clearance of dead cell debris. These are the first studies to document a mechanistic basis for the link between high IL-10 levels and poor outcome in patients with ALF.


Subject(s)
Acetaminophen , Liver Failure, Acute , Humans , Animals , Mice , Acetaminophen/adverse effects , Interleukin-10 , Monocytes , Necrosis/chemically induced
3.
Adv Pharmacol ; 91: 213-228, 2021.
Article in English | MEDLINE | ID: mdl-34099109

ABSTRACT

Liver fibrosis is a leading cause of death worldwide, accounting for approximately 2 million deaths annually. Despite its wide prevalence, there are currently no pharmacological therapies that directly reverse the fibrotic process in patients. Studies over the last decade have revealed that liver fibrosis is reversible in patients and in animal models. Further, studies aimed at elucidating the mechanism of fibrosis reversal have revealed that macrophages are central to this process. During resolution of fibrosis, proinflammatory macrophages shift phenotype to pro-resolution macrophages which produce matrix degrading enzymes and mediators that inactivate hepatic stellate cells, the cell type principally involved in matrix production during fibrosis development. Since fibrosis reversal begins when disease-causing macrophages transition to disease-reversing macrophages, studies have focused on identifying pharmacological agents that stimulate this process to occur. If successful, these "drugs" would constitute a first-in-class, macrophage-targeted therapeutic approach to reverse liver fibrosis. In the following review, we summarize the current approaches under investigation to modify macrophage phenotype for liver disease treatment. Further we discuss the potential of other approaches to identify novel macrophage-targeted drugs that modify the phenotype of these cells.


Subject(s)
Liver Cirrhosis , Macrophages , Animals , Fibrosis , Hepatic Stellate Cells/pathology , Humans , Liver/pathology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Macrophages/pathology , Phenotype
4.
Adv Pharmacol ; 91: xi-xii, 2021.
Article in English | MEDLINE | ID: mdl-34099114
5.
Res Pract Thromb Haemost ; 4(5): 906-917, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32685902

ABSTRACT

BACKGROUND: Blood coagulation protease activity is proposed to drive hepatic fibrosis through activation of protease-activated receptors (PARs). Whole-body PAR-1 deficiency reduces experimental hepatic fibrosis, and in vitro studies suggest a potential contribution by PAR-1 expressed by hepatic stellate cells. However, owing to a lack of specific tools, the cell-specific role of PAR-1 in experimental hepatic fibrosis has never been formally investigated. Using a novel mouse expressing a conditional PAR-1 allele, we tested the hypothesis that PAR-1 expressed by hepatic stellate cells contributes to hepatic fibrosis. METHODS: PAR-1flox/flox mice were crossed with mice expressing Cre recombinase controlled by the lecithin retinol acyltransferase (LRAT) promoter, which induces recombination in hepatic stellate cells. Male PAR-1flox/flox/LRATCre and PAR-1flox/flox mice were challenged twice weekly with carbon tetrachloride (CCl4, 1 mL/kg i.p.) for 6 weeks to induce liver fibrosis. RESULTS: PAR-1 mRNA levels were reduced (>95%) in hepatic stellate cells isolated from PAR-1flox/flox/LRATCre mice. Hepatic stellate cell activation was evident in CCl4-challenged PAR-1flox/flox mice, indicated by increased α-smooth muscle actin labeling and induction of several profibrogenic genes. CCl4-challenged PAR-1flox/flox mice displayed robust hepatic collagen deposition, indicated by picrosirius red staining and type I collagen immunolabeling. Notably, stellate cell activation and collagen deposition were significantly reduced (>30%) in PAR-1flox/flox/LRATCre mice. Importantly, the reduction in liver fibrosis was not a consequence of reduced acute CCl4 hepatotoxicity in PAR-1flox/flox/LRATCre mice. CONCLUSIONS: The results constitute the first direct experimental evidence that PAR-1 expressed by stellate cells directly promotes their profibrogenic phenotype and hepatic fibrosis in vivo.

6.
Life Sci Alliance ; 3(8)2020 08.
Article in English | MEDLINE | ID: mdl-32571802

ABSTRACT

Genome-wide association studies have implicated the TAM receptor tyrosine kinase (RTK) Mer in liver disease, yet our understanding of the role that Mer and its related RTKs Tyro3 and Axl play in liver homeostasis and the response to acute injury is limited. We find that Mer and Axl are most prominently expressed in hepatic Kupffer and endothelial cells and that as mice lacking these RTKs age, they develop profound liver disease characterized by apoptotic cell accumulation and immune activation. We further find that Mer is critical to the phagocytosis of apoptotic hepatocytes generated in settings of acute hepatic injury, and that Mer and Axl act in concert to inhibit cytokine production in these settings. In contrast, we find that Axl is uniquely important in mitigating liver damage during acetaminophen intoxication. Although Mer and Axl are protective in acute injury models, we find that Axl exacerbates fibrosis in a model of chronic injury. These divergent effects have important implications for the design and implementation of TAM-directed therapeutics that might target these RTKs in the liver.


Subject(s)
Liver/injuries , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , c-Mer Tyrosine Kinase/metabolism , Animals , Apoptosis/genetics , Endothelial Cells/metabolism , Female , Genome-Wide Association Study , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Phagocytosis/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction/genetics , c-Mer Tyrosine Kinase/genetics , Axl Receptor Tyrosine Kinase
7.
World J Gastroenterol ; 26(16): 1879-1887, 2020 Apr 28.
Article in English | MEDLINE | ID: mdl-32390699

ABSTRACT

The liver functions, in part, to prevent exposure of the body to potentially harmful substances ingested in the diet. While it is highly efficient at accomplishing this, it is frequently prone to liver injury due to the biotransformation of xenobiotics into toxic metabolites. To counter this injury, the liver has evolved a unique capacity to rapidly and efficiently repair itself. Successful resolution of acute liver injury relies on hepatic macrophage populations that orchestrate the reparative response. After injury, Kupffer cells, the resident macrophages of the liver, become activated and secrete proinflammatory cytokines. These cytokines recruit other immune cells, including monocyte-derived macrophages, to the liver where they contribute to the repair process. Monocyte-derived macrophages traffic into the necrotic foci where they rapidly phagocytose dead cell debris. Simultaneous with this process, these cells change phenotype from a proinflammatory macrophage to a pro-restorative macrophage that produce pro-mitogenic growth factors and anti-inflammatory cytokines. Ultimately this process triggers resolution of inflammation, and along with proliferation of other hepatic cells, restores the liver architecture and function. While the mechanisms regulating specific macrophage functions during repair remain to be elucidated, recent studies indicate a key role for the fibrinolytic system in coordinating macrophage function during repair. In this review, we will highlight the function and role of hepatic macrophages in repair after acute liver injury, and will discuss the role of the fibrinolytic enzyme, plasmin, in regulation of these various processes.


Subject(s)
Chemical and Drug Induced Liver Injury/immunology , Fibrinolysis/immunology , Kupffer Cells/immunology , Liver Regeneration/immunology , Macrophage Activation , Acetaminophen/poisoning , Animals , Cell Proliferation , Chemical and Drug Induced Liver Injury/pathology , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Hepatic Stellate Cells/immunology , Humans , Inflammation Mediators/metabolism , Kupffer Cells/metabolism , Liver/drug effects , Liver/immunology , Liver/pathology
8.
Cytokine ; 127: 154986, 2020 03.
Article in English | MEDLINE | ID: mdl-31951966

ABSTRACT

INTRODUCTION: Cxcl12, or stromal-derived factor-1, is a chemokine produced by several hepatic cell types, including hepatocytes, after liver injury and surgical resection. Studies have revealed that Cxcl12 is important for regeneration of the liver after surgical resection and for development of liver fibrosis during chronic liver injury. While the function of Cxcl12 in the liver is well established, the mechanism by which Cxcl12 is upregulated is not fully understood. Because regions of hypoxia develop in the liver following injury, we tested the hypothesis that hypoxia upregulates Cxcl12 in hepatocytes by a hypoxia-inducible factor (HIF)-dependent mechanism. METHODS: To test this hypothesis, primary mouse hepatocytes were isolated from the livers of HIF-1α-deficient mice or HIF-1ß-deficient mice and exposed to 1% oxygen. Cxcl12 expression was increased following exposure of primary mouse hepatocytes to 1% oxygen. Previously we have shown, that in addition to HIFs, transforming growth factor-ß is required for upregulation of a subset of genes in hypoxic hepatocytes. To examine the role of TGF-ß in regulation of Cxcl12 during hypoxia, hepatocytes were pretreated with the TGF-ß receptor I inhibitor, SB431542. RESULTS: Upregulation of Cxcl12 by hypoxia was partially prevented in hepatocytes from HIF-1α-deficient mice and completely prevented in hepatocytes from HIF-1ß-deficient hepatocytes. This suggests that under hypoxic conditions, both HIF-1α and HIF-2α regulate Cxcl12 in hepatocytes. Pretreatment of hepatocytes with SB431542 completely prevented upregulation Cxcl12 by hypoxia. Further, treatment of hepatocytes with recombinant TGF-ß1 upregulated Cxcl12 in hepatocytes cultured in room air. CONCLUSION: Collectively, these studies demonstrate that hypoxia upregulates Cxcl12 in primary mouse hepatocytes by a mechanism that involves HIFs and TGF-ß.


Subject(s)
Chemokine CXCL12/metabolism , Hepatocytes/metabolism , Hypoxia/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation/physiology , Animals , Cells, Cultured , Liver/metabolism , Liver Cirrhosis/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction/physiology , Transcriptional Activation/physiology
9.
Article in English | MEDLINE | ID: mdl-31555773

ABSTRACT

The liver contains two distinct populations of macrophages, monocyte-derived macrophages (MDMs), which primarily reside proximal to the Glisson's capsule and Kupffer cells, which reside within the sinusoids. Kupffer cells infiltrate the liver during embryogenesis and are replenished from local proliferation of mature Kupffer cells. By contrast MDMs arise from hematopoietic stem cells in the bone marrow and are replenishedfrom circulating monocytes. Studies have revealed that these two hepatic macrophage populations possess distinct transcriptomic profiles, suggesting that they may be functionally distinct. In the present study, we tested the hypothesis that MDMs and Kupffer cells are differentially sensitive to bacterial lipopolysaccharide (LPS). MDMs and Kupffer cells were purified to greater than 90% from the livers of mice by using magnetic beads labeled with Cx3cr1 antibody for MDMs and F4/80 antibody for Kupffer cells. Basal levels of tumor necrosis factor-α (TNF-α) mRNA were higher in MDMs when compared to Kupffer cells. After treatment with LPS, mRNA levels of TNF-α, Cxcll, and Cxcl2 were increased to a greater extent in MDMs when compared to Kupffer cells. To confirm these findings, Kupffer cells and MDMs were isolated from mice in which bone marrow transplantation was used to selectively tag cells arising from hematopoietic stem cells in adult mice. Similar to above, treatment of MDMs with LPS increased TNF-α, Cxcll, and Cxcl2 to a greater extent when compared to Kupffer cells. Collectively, these results indicate that MDMs exhibit a greater pro-inflammatory phenotype in the liver when exposed to LPS.

10.
Am J Pathol ; 189(10): 1986-2001, 2019 10.
Article in English | MEDLINE | ID: mdl-31381887

ABSTRACT

Kupffer cells and monocyte-derived macrophages are critical for liver repair after acetaminophen (APAP) overdose. These cells produce promitogenic cytokines and growth factors, and they phagocytose dead cell debris, a process that is critical for resolution of inflammation. The factors that regulate these dynamic functions of macrophages after APAP overdose, however, are not fully understood. We tested the hypothesis that the fibrinolytic enzyme, plasmin, is a key regulator of macrophage function after APAP-induced liver injury. In these studies, inhibition of plasmin in mice with tranexamic acid delayed up-regulation of proinflammatory cytokines after APAP overdose. In culture, plasmin directly, and in synergy with high-mobility group B1, stimulated Kupffer cells and bone marrow-derived macrophages to produce cytokines by a mechanism that required NF-κB. Inhibition of plasmin in vivo also prevented trafficking of monocyte-derived macrophages into necrotic lesions after APAP overdose. This prevented phagocytic removal of dead cells, prevented maturation of monocyte-derived macrophages into F4/80-expressing macrophages, and prevented termination of proinflammatory cytokine production. Our studies reveal further that phagocytosis is an important stimulus for cessation of proinflammatory cytokine production as treatment of proinflammatory, monocyte-derived macrophages, isolated from APAP-treated mice, with necrotic hepatocytes decreased expression of proinflammatory cytokines. Collectively, these studies demonstrate that plasmin is an important regulator of macrophage function after APAP overdose.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Chemical and Drug Induced Liver Injury/pathology , Fibrinolysin/metabolism , Kupffer Cells/pathology , Macrophages/pathology , Animals , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Drug Overdose , Inflammation Mediators/metabolism , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Necrosis
11.
Am J Physiol Gastrointest Liver Physiol ; 315(4): G569-G579, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29927321

ABSTRACT

Zrt/Irt-like protein 8 (ZIP8) (encoded by Slc39a8) is a multifunctional membrane transporter that influxes essential metal cations Zn2+, Mn2+, Fe2+, and nonmetal inorganic selenite (HSeO3-). Physiological roles of ZIP8 in different cell types and tissues remain to be elucidated. We aimed to investigate ZIP8 functions in liver. Two mouse models were used in this study: 1) 13- to 21-mo-old Slc39a8(+/neo) hypomorphs having diminished ZIP8 levels and 2) a liver-specific ZIP8 acute knockdown mouse (Ad-shZip8). Histology, immunohistochemistry, and Western blotting were used to investigate ZIP8-deficiency effects on hepatic injury, inflammatory changes, and oxidative stress. Selenium (Se) and zinc (Zn) were quantified in tissues by inductively coupled plasma-mass spectrophotometry. We found that ZIP8 is required to maintain normal liver function; moderate or acute decreases in ZIP8 activity resulted in hepatic pathology. Spontaneous liver neoplastic nodules appeared in ~50% of Slc39a8(+/neo) between 13 and 21 mo of age, exhibiting features of inflammation, fibrosis, and liver injury. In Ad-shZip8 mice, significant hepatomegaly was observed; histology showed ZIP8 deficiency was associated with hepatocyte injury, inflammation, and proliferation. Significant decreases in Se, but not Zn, were found in Ad-shZip8 liver. Consistent with this Se deficit, liver expression of selenoproteins glutathione peroxidases 1 and 2 was downregulated, along with decreases in antioxidant superoxide dismutases 1 and 2, consistent with increased oxidative stress. Thus, ZIP8 plays an important role in maintaining normal hepatic function, likely through regulating Se homeostasis and redox balance. Hepatic ZIP8 deficiency is associated with liver pathology, including oxidative stress, inflammation, proliferation, and hepatocellular injury. NEW & NOTEWORTHY Zrt/Irt-like protein 8 (ZIP8) is a multifunctional membrane transporter that facilitates biometal and mineral uptake. The role of ZIP8 in liver physiology has not been previously investigated. Liu et al. discovered unique ZIP8 functions, i.e., regulation of hepatic selenium content and association of ZIP8 deficiency in mouse liver with liver defects.


Subject(s)
Cation Transport Proteins/deficiency , Hepatocytes/metabolism , Homeostasis , Liver Neoplasms/metabolism , Selenium/metabolism , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line , Cells, Cultured , Glutathione Peroxidase/metabolism , Hepatocytes/pathology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress , Superoxide Dismutase/metabolism , Zinc/metabolism
12.
Pharmacol Res ; 104: 9-21, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26706784

ABSTRACT

For many years, bile acids were thought to only function as detergents which solubilize fats and facilitate the uptake of fat-soluble vitamins in the intestine. Many early observations; however, demonstrated that bile acids regulate more complex processes, such as bile acids synthesis and immune cell function through activation of signal transduction pathways. These studies were the first to suggest that receptors may exist for bile acids. Ultimately, seminal studies by many investigators led to the discovery of several bile acid-activated receptors including the farnesoid X receptor, the vitamin D receptor, the pregnane X receptor, TGR5, α5 ß1 integrin, and sphingosine-1-phosphate receptor 2. Several of these receptors are expressed outside of the gastrointestinal system, indicating that bile acids may have diverse functions throughout the body. Characterization of the functions of these receptors over the last two decades has identified many important roles for these receptors in regulation of bile acid synthesis, transport, and detoxification; regulation of glucose utilization; regulation of fatty acid synthesis and oxidation; regulation of immune cell function; regulation of energy expenditure; and regulation of neural processes such as gastric motility. Through these many functions, bile acids regulate many aspects of digestion ranging from uptake of essential vitamins to proper utilization of nutrients. Accordingly, within a short time period, bile acids moved beyond simple detergents and into the realm of complex signaling molecules. Because of the important processes that bile acids regulate through activation of receptors, drugs that target these receptors are under development for the treatment of several diseases, including cholestatic liver disease and metabolic syndrome. In this review, we will describe the various bile acid receptors, the signal transduction pathways activated by these receptors, and briefly discuss the physiological processes that these receptors regulate.


Subject(s)
Bile Acids and Salts/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Humans , Signal Transduction
13.
Cell Mol Gastroenterol Hepatol ; 1(6): 589-597, 2015 Nov.
Article in English | MEDLINE | ID: mdl-28210703

ABSTRACT

Liver fibrosis remains a significant clinical problem in the United States and throughout the world. Although important advances in the understanding of this disease have been made, no effective pharmacologic agents have been developed that directly prevent or reverse the fibrotic process. Many of the successes in liver fibrosis treatment have been targeted toward treating the cause of fibrosis, such as the development of new antivirals that eradicate hepatitis virus. For many patients, however, this is not feasible, so a liver transplant remains the only viable option. Thus, there is a critical need to identify new therapeutic targets that will slow or reverse the progression of fibrosis in such patients. Research over the last 16 years has identified hypoxia-inducible factors (HIFs) as key transcription factors that drive many aspects of liver fibrosis, making them potential targets of therapy. In this review, we discuss the latest work on HIFs and liver fibrosis, including the cell-specific functions of these transcription factors in the development of liver fibrosis.

14.
PLoS One ; 9(12): e115849, 2014.
Article in English | MEDLINE | ID: mdl-25536043

ABSTRACT

Chronic alcohol causes liver hypoxia and steatosis, which eventually develops into alcoholic liver disease (ALD). While it has been known that alcohol consumption activates hepatic hypoxia inducing factor-1α (HIF-1α), conflicting results regarding the role of HIF-1α in alcohol-induced liver injury and steatosis in mice have been reported. In the present study, we aimed to use hepatocyte-specific HIF-1ß knockout mice to eliminate the possible compensatory effects of the single knockout of the 1α subunit of HIF to study the role of HIFs in ALD. C57BL/6 wild type mice were treated with acute ethanol to mimic human binge drinking. Matched wild-type and hepatocyte specific HIF-1ß knockout mice were also subjected to a recently established Gao-binge alcohol model to mimic chronic plus binge conditions, which is quite common in human alcoholics. We found that acute alcohol treatment increased BNIP3 and BNIP3L/NIX expression in primary cultured hepatocytes and in mouse livers, suggesting that HIF may be activated in these models. We further found that hepatocyte-specific HIF-1ß knockout mice developed less steatosis and liver injury following the Gao-binge model or acute ethanol treatment compared with their matched wild type mice. Mechanistically, protection against Gao-binge treatment-induced steatosis and liver injury was likely associated with increased FoxO3a activation and subsequent induction of autophagy in hepatocyte-specific HIF-1ß knockout mice.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Autophagy/drug effects , Fatty Liver, Alcoholic/metabolism , Fatty Liver, Alcoholic/pathology , Liver/pathology , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Cells, Cultured , Ethanol , Fatty Liver, Alcoholic/genetics , Gene Expression Regulation , Hepatocytes/metabolism , Hepatocytes/pathology , Liver/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism
15.
J Hepatol ; 61(3): 617-25, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24815875

ABSTRACT

BACKGROUND & AIMS: Autophagy is an intracellular lysosomal degradation process that plays an important role in regulating normal physiological functions of the liver. The purpose of the present study was to investigate the mechanism(s) by which the loss of hepatic autophagy leads to liver inflammation, fibrosis and tumorigenesis. METHODS: Hepatocyte-specific Atg5 knockout mice were generated by crossing Atg5 Flox/Flox mice with albumin Cre mice. These mice were also crossed with Nrf2 knockout mice to generate Atg5 Flox/Flox, Albumin Cre(+)/Nrf2(-/-) double knockout mice. These mice were housed for various time points up to 15 months, and blood and liver tissues were harvested for biochemical and histological analysis. RESULTS: Hepatocyte-specific deletion of Atg5 resulted in increased apoptosis, inflammation and fibrosis in the liver. Increased apoptosis in hepatocyte-specific Atg5 knockout mice was likely due to accumulation of aberrant polyubiquitinated proteins (proteotoxicity) and disruption of the homeostasis of pro-and anti-apoptotic proteins. All of these pathological changes started as early as one month and persisted for 12-15 months. At 9-15 months of age, these mice also developed hepatocellular adenomas. Interestingly, deletion of Nrf2 in Atg5 liver-specific knockout mice markedly abolished these pathological changes, indicating a key role for this transcription factor in the mechanism of hepatic pathology. CONCLUSIONS: Our results provide genetic evidence that loss of autophagy in hepatocytes causes cell death resulting in liver inflammation, fibrosis and tumorigenesis. We also demonstrate that persistent activation of Nrf2 is critical for liver inflammation, fibrosis and eventual tumorigenesis that occur in mice with defects in hepatocyte autophagy.


Subject(s)
Autophagy/physiology , Carcinogenesis , Liver Cirrhosis/physiopathology , Liver/physiopathology , NF-E2-Related Factor 2/physiology , Animals , Apoptosis/physiology , Autophagy-Related Protein 5 , Disease Models, Animal , Female , Hepatitis/physiopathology , Hepatocytes/metabolism , Hepatocytes/pathology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/physiology , NF-E2-Related Factor 2/deficiency , NF-E2-Related Factor 2/genetics
16.
J Immunol ; 192(8): 3847-3857, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24639359

ABSTRACT

Hypoxia-inducible factor-1α (HIF-1α) is activated in hepatic stellate cells (HSCs) by hypoxia and regulates genes important for tissue repair. Whether HIF-1α is activated in HSCs after acute injury and contributes to liver regeneration, however, is not known. To investigate this, mice were generated with reduced levels of HIF-1α in HSCs by crossing HIF-1α floxed mice with mice that express Cre recombinase under control of the glial fibrillary acidic protein (GFAP) promoter (i.e., HIF-1α-GFAP Cre+ mice). These mice and control mice (i.e., HIF-1α-GFAP Cre- mice) were treated with a single dose of carbon tetrachloride, and liver injury and repair were assessed. After carbon tetrachloride, HIF-1α was activated in HSCs. Although liver injury was not different between the two strains of mice, during resolution of injury, clearance of necrotic cells was decreased in HIF-1α-GFAP Cre+ mice. In these mice, the persistence of necrotic cells stimulated a fibrotic response characterized by extensive collagen deposition. Hepatic accumulation of macrophages, which clear necrotic cells from the liver after carbon tetrachloride, was not affected by HIF-1α deletion in HSCs. Conversion of macrophages to M1-like, proinflammatory macrophages, which have increased phagocytic activity, however, was reduced in HIF-1α-GFAP Cre+ mice as indicated by a decrease in proinflammatory cytokines and a decrease in the percentage of Gr1(hi) macrophages. Collectively, these studies have identified a novel function for HSCs and HIF-1α in orchestrating the clearance of necrotic cells from the liver and demonstrated a key role for HSCs in modulating macrophage phenotype during acute liver injury.


Subject(s)
Hepatic Stellate Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Macrophages/immunology , Macrophages/metabolism , Phenotype , Animals , Carbon Tetrachloride/pharmacology , Cell Proliferation , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Gene Deletion , Hepatic Stellate Cells/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Macrophages/pathology , Mice , Mice, Transgenic , Necrosis , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
17.
Hepatology ; 60(3): 919-30, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24619556

ABSTRACT

UNLABELLED: E2F transcription factor 1 (E2F1) is an important regulator of metabolic diseases; however, its role in liver function remains elusive. This study unraveled a regulatory cascade involving E2F1, early growth response-1 (Egr-1), nuclear receptor small heterodimer partner (SHP, NR0B2), and EIA-like inhibitor of differentiation 1 (EID1) in cholestatic liver fibrosis. Liver E2F1 messenger RNA (mRNA) and protein expression was strongly up-regulated in human nonalcoholic steatohepatitis (NASH) and alcohol cirrhosis; the latter was inversely correlated with diminished SHP expression. E2F1 was also highly induced by 3,5-diethoxycarbonyl-1, 4-dihydrocollidine (DDC) feeding and bile-duct ligation (BDL) in mice. E2F1-/- mice exhibited reduced biliary fibrosis by DDC as determined by Masson Trichrome and Picro Sirius red staining, and decreased serum bile acid (BA), BA pool size, and fecal BA excretion. In addition, cholestatic liver fibrosis induced by BDL, as determined by immunohistochemistry analysis of a1 collagen expression, was increased in SHP-/- mice but attenuated in hepatocyte SHP-overexpressed transgenic (STG) mice. Egr-1 exhibited marked induction in livers of SHP-/- mice compared to the wild-type mice in both sham and BDL groups, and reduction in STG livers. Egr-1 promoter was activated by E2F1, and the activation was abrogated by expression of SHP and its co-repressor EID1 in hepatoma cells Huh7, Hepa1, and stellate cells LX2. Chromatin immunoprecipitation assays further confirmed the association of E2F1, SHP, and EID1 proteins with the Egr-1 promoter, and their direct protein interactions were determined by glutathione S-transferase pull-down assays. Interestingly, E2F1 activated Egr-1 expression in a biphasic fashion as described in both human and mouse hepatocytes. CONCLUSION: E2F1 is a fibrogenic gene and could serve as a potential new diagnostic marker for nonalcoholic and alcoholic liver fibrosis/cirrhosis.


Subject(s)
Cholestasis/metabolism , Cholestasis/pathology , E2F1 Transcription Factor/physiology , Early Growth Response Protein 1/physiology , Gene Regulatory Networks/physiology , Liver Cirrhosis/genetics , Nuclear Proteins/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Repressor Proteins/physiology , Animals , Cell Cycle Proteins , Cholestasis/genetics , E2F1 Transcription Factor/genetics , Gene Regulatory Networks/genetics , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nuclear Proteins/genetics , Promoter Regions, Genetic/genetics , Pyridines , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Repressor Proteins/genetics
18.
Toxicol Lett ; 224(2): 186-95, 2014 Jan 13.
Article in English | MEDLINE | ID: mdl-24188933

ABSTRACT

Osteopontin (OPN) is a chemotactic factor which can be cleaved to the pro-inflammatory form by matrix metalloproteinases (MMPs). To test the hypothesis that OPN can modulate inflammatory liver injury during cholestasis, wild-type (WT) C57BL/6 and OPN knockout (OPN-KO) mice underwent bile duct ligation (BDL). OPN-KO mice showed significant reduction in liver injury (plasma ALT and necrosis) and neutrophil recruitment compared with WT animals at 24h but not 72h after BDL. In WT mice, a 4-fold increase in hepatic MMP-3 mRNA and elevated MMP activities and cleaved OPN levels were observed in bile. WT mice subjected to BDL in the presence of the MMP inhibitor BB-94 showed reduced liver injury, less neutrophil extravasation and diminished levels of cleaved OPN in bile. Thus, during obstructive cholestasis, OPN released from biliary epithelial cells could be cleaved by MMPs in bile. When the biliary system leaks, cleaved OPN enters the parenchyma and attracts neutrophils. In the absence of OPN, other chemoattractants, e.g. chemokines, mediate a delayed inflammatory response and injury. Taken together, our data suggest that OPN is the pro-inflammatory mediator that initiates the early neutrophil-mediated injury phase during obstructive cholestasis in mice.


Subject(s)
Cholestasis/complications , Inflammation/etiology , Osteopontin/physiology , Alanine Transaminase/blood , Animals , Bile Acids and Salts/toxicity , Bile Ducts/surgery , Ligation , Male , Matrix Metalloproteinase 3/genetics , Mice , Mice, Inbred C57BL , Neutrophil Infiltration , Osteopontin/genetics , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Thiophenes/pharmacology
19.
Am J Pathol ; 183(5): 1498-1507, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24012680

ABSTRACT

During obstructive cholestasis, increased concentrations of bile acids activate ERK1/2 in hepatocytes, which up-regulates early growth response factor 1, a key regulator of proinflammatory cytokines, such as macrophage inflammatory protein 2 (MIP-2), which, in turn, exacerbates cholestatic liver injury. Recent studies have indicated that IL-17A contributes to hepatic inflammation during obstructive cholestasis, suggesting that bile acids and IL-17A may interact to regulate hepatic inflammatory responses. We treated mice with an IL-17A neutralizing antibody or control IgG and subjected them to bile duct ligation. Neutralization of IL-17A prevented up-regulation of proinflammatory cytokines, hepatic neutrophil accumulation, and liver injury, indicating an important role for IL-17A in neutrophilic inflammation during cholestasis. Treatment of primary mouse hepatocytes with taurocholic acid (TCA) increased the expression of MIP-2. Co-treatment with IL-17A synergistically enhanced up-regulation of MIP-2 by TCA. In contrast to MIP-2, IL-17A did not affect up-regulation of Egr-1 by TCA, indicating that IL-17A does not affect bile acid-induced activation of signaling pathways upstream of early growth response factor 1. In addition, bile acids increased expression of IL-23, a key regulator of IL-17A production in hepatocytes in vitro and in vivo. Collectively, these data identify bile acids as novel triggers of the IL-23/IL-17A axis and suggest that IL-17A promotes hepatic inflammation during cholestasis by synergistically enhancing bile acid-induced production of proinflammatory cytokines by hepatocytes.


Subject(s)
Cholestasis/metabolism , Cholestasis/pathology , Inflammation/metabolism , Inflammation/pathology , Interleukin-17/metabolism , Actins/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Bile Acids and Salts/administration & dosage , Bile Ducts/drug effects , Bile Ducts/pathology , Biomarkers/metabolism , Cell Count , Chemokine CXCL2/genetics , Chemokine CXCL2/metabolism , Cholestasis/complications , Collagen Type I/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Inflammation/complications , Interleukin-23/genetics , Interleukin-23/metabolism , Ligation , Liver/drug effects , Liver/injuries , Liver/pathology , Macrophages/drug effects , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Neutralization Tests , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Taurocholic Acid/pharmacology , Up-Regulation/drug effects
20.
Am J Pathol ; 181(4): 1287-95, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22841818

ABSTRACT

Thrombin generation is increased in patients with nonalcoholic fatty liver disease (NAFLD) and in mouse models of diet-induced obesity. Deficiency in the thrombin receptor protease activated receptor-1 reduces hepatic inflammation and steatosis in mice fed a Western diet. However, it is currently unclear whether thrombin inhibitors can modify the pathogenesis of established NAFLD. We tested the hypothesis that thrombin inhibition could reverse hepatic steatosis and inflammation in mice with established diet-induced NAFLD. Low-density lipoprotein receptor-deficient LDLr(-/-) mice were fed a control diet or a Western diet for 19 weeks. Mice were given the direct thrombin inhibitor argatroban ∼15 mg/kg/day or its vehicle via a miniosmotic pump for the final 4 weeks of the study. Argatroban administration significantly reduced hepatic proinflammatory cytokine expression and reduced macrophage and neutrophil accumulation in livers of mice fed a Western diet. Argatroban did not significantly impact hepatic steatosis, as indicated by histopathology, Oil Red O staining, and hepatic triglyceride levels. Argatroban reduced serum triglyceride and cholesterol levels in mice fed a Western diet. Argatroban reduced both α-smooth muscle actin expression and Type 1 collagen mRNA levels in livers of mice fed a Western diet, indicating reduced activation of hepatic stellate cells. This study indicates that therapeutic intervention with a thrombin inhibitor attenuates hepatic inflammation and several profibrogenic changes in mice fed a Western diet.


Subject(s)
Fatty Liver/complications , Fatty Liver/drug therapy , Inflammation/drug therapy , Pipecolic Acids/pharmacology , Pipecolic Acids/therapeutic use , Thrombin/antagonists & inhibitors , Animals , Arginine/analogs & derivatives , Chemokine CCL2/metabolism , Chemokine CXCL2/genetics , Chemokine CXCL2/metabolism , Collagen/metabolism , Diet , Fatty Liver/blood , Feeding Behavior/drug effects , Gene Expression Regulation/drug effects , Inflammation/blood , Inflammation/complications , Inflammation/pathology , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Lipids/blood , Liver/drug effects , Liver/pathology , Liver Cirrhosis/blood , Liver Cirrhosis/complications , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/metabolism , Pipecolic Acids/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, LDL/deficiency , Receptors, LDL/metabolism , Sulfonamides , Thrombin/metabolism , Weight Gain/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL