Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 12(5): 441-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21423173

ABSTRACT

Subcompartments of the plasma membrane are believed to be critical for lymphocyte responses, but few genetic tools are available to test their function. Here we describe a previously unknown X-linked B cell-deficiency syndrome in mice caused by mutations in Atp11c, which encodes a member of the P4 ATPase family thought to serve as 'flippases' that concentrate aminophospholipids in the cytoplasmic leaflet of cell membranes. Defective ATP11C resulted in a lower rate of phosphatidylserine translocation in pro-B cells and much lower pre-B cell and B cell numbers despite expression of pre-rearranged immunoglobulin transgenes or enforced expression of the prosurvival protein Bcl-2 to prevent apoptosis and abolished pre-B cell population expansion in response to a transgene encoding interleukin 7. The only other abnormalities we noted were anemia, hyperbilirubinemia and hepatocellular carcinoma. Our results identify an intimate connection between phospholipid transport and B lymphocyte function.


Subject(s)
Adenosine Triphosphatases/immunology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Endocytosis/immunology , Phosphoserine/immunology , Adenosine Triphosphatases/genetics , Animals , B-Lymphocytes/enzymology , Base Sequence , Female , Flow Cytometry , Genes, bcl-2/immunology , Interleukin-7/genetics , Interleukin-7/immunology , Liver/cytology , Liver/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Mutagenesis/immunology , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
2.
J Hepatol ; 73(1): 140-148, 2020 07.
Article in English | MEDLINE | ID: mdl-32302728

ABSTRACT

BACKGROUND & AIMS: Obesity and type 2 diabetes increase hepatocellular carcinoma (HCC) incidence in humans and accelerate diethylnitrosamine (DEN)-induced hepatocarcinogenesis in mice. We investigated whether exercise reduces HCC development in obese/diabetic Alms1 mutant (foz/foz) mice and studied protective mechanisms. METHODS: We measured HCC development in DEN-injected male foz/foz and wild-type (WT) littermates housed with or without an exercise wheel from week 4 until 12 or 24 weeks, and in foz/foz mice pair-fed to WT littermates. We also studied HCC development in DEN-injected Jnk1-/-.foz/foz mice generated by cross breeding, as well as their genetic controls. Dysplastic hepatocytes were identified by glutathione-S-transferase pi form (GST-pi) immunohistochemistry, liver nodules were counted, and HCC was analysed by histopathology. RESULTS: Exercising foz/foz mice maintained similar weight as WT mice up to 10 weeks, but then gained weight and were obese by 24 weeks; a similar body weight profile was obtained by pair-feeding foz/foz mice to WT. At 12 weeks, livers of exercising foz/foz mice exhibited fewer GST-pi positive hepatocytes than sedentary counterparts; by 24 weeks, fewer exercising foz/foz mice developed HCC (15% vs. 64%, p <0.05). Conversely, pair-feeding foz/foz mice failed to reduce HCC incidence. In these insulin-resistant foz/foz mice, exercise failed to activate hepatic AMPK or Akt/mTORC1. Instead, it improved insulin sensitivity, ameliorated steatosis and liver injury, activated p53 to increase p27 expression, and prevented JNK activation. This was associated with suppression of hepatocellular proliferation. DEN-injected Jnk1-/-.foz/foz mice failed to develop liver tumours or HCC at 24 weeks. CONCLUSIONS: Direct effects of exercise dampen proliferation of dysplastic hepatocytes to reduce 3-month dysplastic foci and 6-month incidence of DEN-induced HCC in obese, insulin-resistant mice. The effects of exercise that potentially slow hepatocarcinogenesis include p53-mediated induction of p27 and prevention of JNK activation. LAY SUMMARY: Fatty liver disease commonly occurs alongside obesity and diabetes, contributing to rapidly increasing rates of liver cancer throughout the world. Herein, we show that exercise reduces the incidence and progression of hepatocellular carcinoma in mouse models. The effect of exercise on cancer risk was shown to be independent of changes in weight. Exercise could be a protective mechanism against liver cancer in at-risk individuals.


Subject(s)
Carcinogenesis , Carcinoma, Hepatocellular , Diabetes Mellitus, Type 2 , Liver Neoplasms , Motor Activity/physiology , Obesity , Animals , Body Weight/physiology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Immunohistochemistry , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , MAP Kinase Signaling System/physiology , Mice , Mice, Obese , Mitogen-Activated Protein Kinases/metabolism , Obesity/metabolism , Obesity/physiopathology , Physical Conditioning, Animal , Proliferating Cell Nuclear Antigen/metabolism , Protective Factors , Risk Factors , Tumor Suppressor Protein p53/metabolism
3.
Hepatology ; 69(5): 2241-2257, 2019 05.
Article in English | MEDLINE | ID: mdl-30372785

ABSTRACT

Nonalcoholic steatohepatitis (NASH) arises from a variable interplay between environmental factors and genetic determinants that cannot be completely replicated in animals. Notwithstanding, preclinical models are needed to understand NASH pathophysiology and test mechanism-based therapies. Among several mouse models of NASH, some exhibit the key pathophysiologic as well as histopathologic criteria for human NASH, whereas others may be useful to address specific questions. Models based on overnutrition with adipose restriction/inflammation and metabolic complications, particularly insulin resistance, may be most useful to investigate critical etiopathogenic factors. In-depth pathologic description is required for all models. Some models demonstrate hepatocyte ballooning, which can be confused with microvesicular steatosis, whereas demonstration of an inflammatory infiltrate and pattern of liver fibrosis compatible with human NASH is desirable in models used for pharmacologic testing. When mice with specific genetic strains or mutations that cause overeating consume a diet enriched with fat, modest amounts of cholesterol, and/or simple sugars ("Western diet"), they readily develop obesity with liver disease similar to human NASH, including significant fibrosis. Purely dietary models, such as high-fat/high-cholesterol, Western diet, and choline-deficient, amino acid-defined, are similarly promising. We share concern about using models without weight gain, adipose pathology, or insulin resistance/hyperinsulinemia and with inadequate documentation of liver pathology. NASH-related fibrosis is a key endpoint in trials of possible therapies. When studied for this purpose, NASH models should be reproducible and show steatohepatitis (ideally with ballooning) and at least focal bridging fibrosis, while metabolic factors/disordered lipid partitioning should contribute to etiopathogenesis. Because murine models are increasingly used to explore pharmacologic therapies for NASH, we propose a minimum set of requirements that investigators, drug companies, and journals should consider to optimize their translational value.


Subject(s)
Disease Models, Animal , Non-alcoholic Fatty Liver Disease , Animals , Disease Progression , Liver Cirrhosis , Mice
4.
J Gastroenterol Hepatol ; 35(6): 953-959, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31867782

ABSTRACT

Liver stiffness measurement (LSM) by FibroScan-determined transient elastography is a noninvasive approach to estimate liver fibrosis severity. In non-alcoholic fatty liver disease (NAFLD), advanced liver fibrosis is excluded by normal liver stiffness, but a wide range of cutoffs have been used to predict advanced liver fibrosis or cirrhosis. This may be partly because steatosis (measured by controlled attenuation parameter [CAP]) contributes to liver stiffness and also because LSM fluctuates in NAFLD. In a recent pivotal study, one-third of patients with liver stiffness > 12.0 kPa showed reversal after 4-6 months; these cases did not have advanced liver fibrosis on biopsy. We performed serial FibroScans 6-36 months apart in 73 NAFLD patients, 38 with LSM > 10 kPa at entry. Those who lost ≥ 1 kg of weight (n = 31) significantly reduced liver stiffness (3.6 ± 6.1 vs 0.53 ± 4.1 kPa, P < 0.05) and CAP score (39 ± 63 dB/m of loss vs 24 ± 65 dB/m of gain, P < 0.05) compared with those who did not (n = 29). Patients who reported increased physical activity (n = 25) also reduced liver stiffness (3.6 ± 6 vs 0.35 ± 6 kPa) and CAP (20 ± 71 dB/m of loss vs 32 ± 71 dB/m of gain). Overall, those with improved LSM were significantly more likely to have lost weight and/or improved physical activity. These effects of lifestyle adjustments partly explain why a single measurement of 12.0 kPa is not a reliable cutoff for advanced liver fibrosis in NAFLD. In addition to repeating the study after 6-12 months, documentation of response to lifestyle advice and weight reduction should be determined before assuming any cutoff indicates advanced liver fibrosis. Despite this reservation about diagnostic accuracy, we consider that measurement of liver stiffness and CAP score serve to motivate patients to enact lifestyle modifications that can improve NAFLD severity.


Subject(s)
Elasticity Imaging Techniques/methods , Elasticity , Liver Cirrhosis/diagnostic imaging , Liver Cirrhosis/physiopathology , Liver/diagnostic imaging , Liver/physiopathology , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/diagnostic imaging , Exercise , Humans , Life Style , Liver Cirrhosis/etiology , Motivation , Patient Education as Topic , Patients/psychology , Severity of Illness Index
5.
Intern Med J ; 50(8): 951-959, 2020 08.
Article in English | MEDLINE | ID: mdl-31821680

ABSTRACT

BACKGROUND: Inequalities in access to renal services and acute care for rural and remote populations in Australia have been described but not quantified. AIM: To describe: the coverage of renal disease management services in rural and remote Australia; and the characteristics of patients who had an aeromedical retrieval for renal disease by Australia's Royal Flying Doctor Service (RFDS). METHODS: Data from the RFDS, the Australian Bureau of Statistics, and Health Direct were used to estimate provision of renal disease management services by geographic area. RFDS patient diagnostic data were prospectively collected from 2014 to 2018. RESULTS: Many rural and remote areas have limited access to regular renal disease management services. Most RFDS retrievals for renal disease are from regions without such services. The RFDS conducted 1636 aeromedical retrievals for renal disease, which represented 1.6% of all retrievals. Among retrieved patients, there was a higher proportion of men than women (54.6% vs 45.4%, P < 0.01), while indigenous patients (n = 546, 33.4%) were significantly younger than non-indigenous patients (40.9 vs 58.5, P < 0.01). There were significant differences in underlying diagnoses triggering retrievals between genders, with males being more likely than females to be transferred with acute renal failure, calculus of the kidney and ureter, renal colic, obstructive uropathy, and kidney failure (all P < 0.01). Conversely, females were more likely to have chronic kidney disease, disorders of the urinary system, acute nephritic syndrome, tubulo-interstitial nephritis, and nephrotic syndrome (all P < 0.01). CONCLUSION: Aeromedical retrievals for acute care were from rural areas without regular access to renal disease prevention or management services.


Subject(s)
Air Ambulances , Rural Health Services , Australia/epidemiology , Disease Management , Female , Health Services Accessibility , Humans , Kidney , Male , Rural Population
6.
Exp Cell Res ; 356(1): 48-56, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28408319

ABSTRACT

Dysplastic hepatocytes (DH) represent altered hepatocytes with potential for malignant transformation. To date, most research on pathways to hepatocarcinogenesis has focused on use of "hepatoma" cell lines derived from hepatocellular carcinoma (HCC). We describe a novel technique for deriving/culturing DH and demonstrate their utility for functional studies in vitro, compared to primary hepatocytes (PH) and HCC. PH and DH were prepared by portal vein collagenase perfusion from C57BL/6J mice. DH were subsequently subjected to FACS. HCC from diethylnitrosamine (DEN)-injected mice were mechanically isolated. Cell cycle analyses were performed by flow cytometry and PCNA immunohistochemistry. To establish utility of DH, we studied pathways of p53 turnover, apoptosis and cell proliferation using pfithrin-α (PFT) and nutlin-3. Like PH, DH were minimally proliferative compared to HCC. Only 30±0.03% of DH were in G2/M phase versus 51±0.01% of HCC; this difference corroborated with PCNA-immunostaining of dysplastic nodules from DEN-injected mice. In DH and HCC, nutlin-3 suppressed p53 mRNA, induced p53 and mdm2 activation but paradoxically resulted in increased anti-apoptotic and proliferative activity. Primary murine DH display distinctive biological characteristics compared with PH and HCC. As an intermediate cell type to HCC, they offer a new pathobiologically relevant primary cell culture system with which to interrogate the molecular changes in hepatocarcinogenesis.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/pathology , Hepatocytes/pathology , Liver Neoplasms, Experimental/pathology , Liver/pathology , Animals , Apoptosis/genetics , Cell Cycle/physiology , Cell Proliferation/genetics , Cells, Cultured , Diethylnitrosamine , Enzyme Activation , Imidazoles/pharmacology , Liver/cytology , Male , Mice , Mice, Inbred C57BL , Piperazines/pharmacology , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
J Hepatol ; 66(5): 1037-1046, 2017 05.
Article in English | MEDLINE | ID: mdl-28167322

ABSTRACT

BACKGROUND & AIMS: NOD-like receptor protein 3 (NLRP3) inflammasome activation occurs in Non-alcoholic fatty liver disease (NAFLD). We used the first small molecule NLRP3 inhibitor, MCC950, to test whether inflammasome blockade alters inflammatory recruitment and liver fibrosis in two murine models of steatohepatitis. METHODS: We fed foz/foz and wild-type mice an atherogenic diet for 16weeks, gavaged MCC950 or vehicle until 24weeks, then determined NAFLD phenotype. In mice fed an methionine/choline deficient (MCD) diet, we gavaged MCC950 or vehicle for 6weeks and determined the effects on liver fibrosis. RESULTS: In vehicle-treated foz/foz mice, hepatic expression of NLRP3, pro-IL-1ß, active caspase-1 and IL-1ß increased at 24weeks, in association with cholesterol crystal formation and NASH pathology; plasma IL-1ß, IL-6, MCP-1, ALT/AST all increased. MCC950 treatment normalized hepatic caspase 1 and IL-1ß expression, plasma IL-1ß, MCP-1 and IL-6, lowered ALT/AST, and reduced the severity of liver inflammation including designation as NASH pathology, and liver fibrosis. In vitro, cholesterol crystals activated Kupffer cells and macrophages to release IL-1ß; MCC950 abolished this, and the associated neutrophil migration. MCD diet-fed mice developed fibrotic steatohepatitis; MCC950 suppressed the increase in hepatic caspase 1 and IL-1ß, lowered numbers of macrophages and neutrophils in the liver, and improved liver fibrosis. CONCLUSION: MCC950, an NLRP3 selective inhibitor, improved NAFLD pathology and fibrosis in obese diabetic mice. This is potentially attributable to the blockade of cholesterol crystal-mediated NLRP3 activation in myeloid cells. MCC950 reduced liver fibrosis in MCD-fed mice. Targeting NLRP3 is a logical direction in pharmacotherapy of NASH. LAY SUMMARY: Fatty liver disease caused by being overweight with diabetes and a high risk of heart attack, termed non-alcoholic steatohepatitis (NASH), is the most common serious liver disease with no current treatment. There could be several causes of inflammation in NASH, but activation of a protein scaffold within cells termed the inflammasome (NLRP3) has been suggested to play a role. Here we show that cholesterol crystals could be one pathway to activate the inflammasome in NASH. We used a drug called MCC950, which has already been shown to block NLRP3 activation, in an attempt to reduce liver injury in NASH. This drug partly reversed liver inflammation, particularly in obese diabetic mice that most closely resembles the human context of NASH. In addition, such dampening of liver inflammation in NASH achieved with MCC950 partly reversed liver scarring, the process that links NASH to the development of cirrhosis.


Subject(s)
Hepatitis/prevention & control , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Liver Cirrhosis, Experimental/prevention & control , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Non-alcoholic Fatty Liver Disease/drug therapy , Sulfones/therapeutic use , Animals , Disease Models, Animal , Female , Furans , Indenes , Interleukin-1beta/blood , Mice , NF-kappa B/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Non-alcoholic Fatty Liver Disease/complications , Reactive Oxygen Species/metabolism , Sulfonamides
8.
Clin Sci (Lond) ; 131(16): 2145-2159, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28687713

ABSTRACT

Background and aims: TLR9 deletion protects against steatohepatitis due to choline-amino acid depletion and high-fat diet. We measured TLR9 in human non-alcoholic steatohepatitis (NASH) livers, and tested whether TLR9 mediates inflammatory recruitment in three murine models of non-alcoholic fatty liver disease (NAFLD). Methods: We assayed TLR mRNA in liver biopsies from bariatric surgery patients. Wild-type (Wt), appetite-dysregulated Alms1 mutant (foz/foz), Tlr9-/-, and Tlr9-/-foz/foz C57BL6/J mice and bone marrow (BM) chimeras were fed 0.2% cholesterol, high-fat, high sucrose (atherogenic[Ath]) diet or chow, and NAFLD activity score (NAS)/NASH pathology, macrophage/neutrophil infiltration, cytokines/chemokines, and cell death markers measured in livers. Results: Hepatic TLR9 and TLR4 mRNA were increased in human NASH but not simple steatosis, and in Ath-fed foz/foz mice with metabolic syndrome-related NASH. Ath-fed Tlr9-/- mice showed simple steatosis and less Th1 cytokines than Wt. Tlr9-/-foz/foz mice were obese and diabetic, but necroinflammatory changes were less severe than Tlr9+/+.foz/foz mice. TLR9-expressing myeloid cells were critical for Th1 cytokine production in BM chimeras. BM macrophages from Tlr9-/- mice showed M2 polarization, were resistant to M1 activation by necrotic hepatocytes/other pro-inflammatory triggers, and provoked less neutrophil chemotaxis than Wt Livers from Ath-fed Tlr9-/- mice appeared to exhibit more markers of necroptosis [receptor interacting protein kinase (RIP)-1, RIP-3, and mixed lineage kinase domain-like protein (MLKL)] than Wt, and ∼25% showed portal foci of mononuclear cells unrelated to NASH pathology. CONCLUSION: Our novel clinical data and studies in overnutrition models, including those with diabetes and metabolic syndrome, clarify TLR9 as a pro-inflammatory trigger in NASH. This response is mediated via M1-macrophages and neutrophil chemotaxis.


Subject(s)
Inflammation Mediators/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Toll-Like Receptor 9/biosynthesis , Up-Regulation/physiology , Adiponectin/deficiency , Adult , Animals , Bariatric Surgery , Biopsy , Cells, Cultured , Cytokines/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Gene Deletion , Hepatocytes/metabolism , Hepatomegaly/prevention & control , Humans , Liver/metabolism , Liver/pathology , Macrophages/metabolism , Metabolic Syndrome/metabolism , Metabolism, Inborn Errors/prevention & control , Mice, Knockout , Neutrophils/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Obesity/prevention & control , RNA, Messenger/genetics , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/genetics
9.
J Gastroenterol Hepatol ; 31(6): 1210-9, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26574916

ABSTRACT

BACKGROUND AND AIM: While gender differences in hepatocellular carcinoma (HCC) are profound, the mechanism is unclear. Using castration and hormone replacement strategies, we tested whether these gender differences are attributable to testosterone or estradiol/progesterone effects on cell cycle regulators and p53. METHODS: We studied dysplastic liver and HCCs in intact and castrated diethylnitrosamine-injected C57BL/6J male and female mice, with or without hormonal replacement. Effects of sex steroids on proliferation and survival of primary hepatocytes and primary HCC cells were also characterized. RESULTS: Diethylnitrosamine-injected female mice displayed fewer dysplastic foci and slower onset of HCC than male mice, with smaller/more differentiated tumors and fewer metastases. Castration of diethylnitrosamine-injected male mice reduced cyclin E kinase and augmented hepatocyte apoptosis compared with intact male mice; estradiol/progesterone enhanced these effects. In intact female mice, cyclin E kinase activity was less than in males; testosterone administered to ovariectomized female mice upregulated cyclin E, increased cyclin E kinase, and accelerated hepatocarcinogenesis. In vitro, testosterone increased expression of cell cycle regulators (cyclin D1, cyclin E, and cyclin-dependent kinase 2) and reduced p53 and p21, which enhanced hepatocyte viability. In contrast, estradiol both suppressed hepatocyte cell cycle markers, upregulated p53 and reduced viability of hepatocytes and HCC cells. CONCLUSIONS: Testosterone is the positive regulator of hepatocyte cell cycle via cyclin E, while estradiol plays a negative role by effects of p53 and p21. Together, both sex hormones determine the male predominance of gender differences in hepatocarcinogenesis.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Cell Transformation, Neoplastic/chemically induced , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , Hormone Replacement Therapy/adverse effects , Liver Neoplasms, Experimental/enzymology , Testosterone/pharmacology , Testosterone/toxicity , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/pathology , Castration , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Diethylnitrosamine , Estradiol/toxicity , Estrogen Replacement Therapy/adverse effects , Female , Hepatocytes/drug effects , Hepatocytes/enzymology , Hepatocytes/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/pathology , Male , Mice, Inbred C57BL , Primary Cell Culture , Sex Factors , Time Factors , Tumor Suppressor Protein p53/metabolism
10.
J Lipid Res ; 56(2): 277-85, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25520429

ABSTRACT

Cholesterol crystals form within hepatocyte lipid droplets in human and experimental nonalcoholic steatohepatitis (NASH) and are the focus of crown-like structures (CLSs) of activated Kupffer cells (KCs). Obese, diabetic Alms1 mutant (foz/foz) mice were a fed high-fat (23%) diet containing 0.2% cholesterol for 16 weeks and then assigned to four intervention groups for 8 weeks: a) vehicle control, b) ezetimibe (5 mg/kg/day), c) atorvastatin (20 mg/kg/day), or d) ezetimibe and atorvastatin. Livers of vehicle-treated mice developed fibrosing NASH with abundant cholesterol crystallization within lipid droplets calculated to extend over 3.3% (SD, 2.2%) of liver surface area. Hepatocyte lipid droplets with prominent cholesterol crystallization were surrounded by TNFα-positive (activated) KCs forming CLSs (≥ 3 per high-power field). KCs that formed CLSs stained positive for NLRP3, implicating activation of the NLRP3 inflammasome in response to cholesterol crystals. In contrast, foz/foz mice treated with ezetimibe and atorvastatin showed near-complete resolution of cholesterol crystals [0.01% (SD, 0.02%) of surface area] and CLSs (0 per high-power field), with amelioration of fibrotic NASH. Ezetimibe or atorvastatin alone had intermediate effects on cholesterol crystallization, CLSs, and NASH. These findings are consistent with a causative link between exposure of hepatocytes and KCs to cholesterol crystals and with the development of NASH possibly mediated by NLRP3 activation.


Subject(s)
Anticholesteremic Agents/therapeutic use , Cholesterol/metabolism , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Animals , Anticholesteremic Agents/pharmacology , Atorvastatin , Azetidines/pharmacology , Azetidines/therapeutic use , Ezetimibe , Female , Heptanoic Acids/pharmacology , Heptanoic Acids/therapeutic use , Inflammasomes/drug effects , Inflammasomes/metabolism , Liver/drug effects , Liver/metabolism , Mice , Mice, Mutant Strains , Non-alcoholic Fatty Liver Disease/drug therapy , Pyrroles/pharmacology , Pyrroles/therapeutic use
11.
J Biol Chem ; 289(28): 19531-7, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24898253

ABSTRACT

Transmembrane lipid transporters are believed to establish and maintain phospholipid asymmetry in biological membranes; however, little is known about the in vivo function of the specific transporters involved. Here, we report that developing erythrocytes from mice lacking the putative phosphatidylserine flippase ATP11C showed a lower rate of PS translocation in vitro compared with erythrocytes from wild-type littermates. Furthermore, the mutant mice had an elevated percentage of phosphatidylserine-exposing mature erythrocytes in the periphery. Although erythrocyte development in ATP11C-deficient mice was normal, the mature erythrocytes had an abnormal shape (stomatocytosis), and the life span of mature erythrocytes was shortened relative to that in control littermates, resulting in anemia in the mutant mice. Thus, our findings uncover an essential role for ATP11C in erythrocyte morphology and survival and provide a new candidate for the rare inherited blood disorder stomatocytosis with uncompensated anemia.


Subject(s)
Adenosine Triphosphatases/metabolism , Erythrocyte Membrane/enzymology , Phospholipids/metabolism , Acid-Base Imbalance/genetics , Acid-Base Imbalance/metabolism , Acid-Base Imbalance/pathology , Adenosine Triphosphatases/genetics , Anemia, Hemolytic, Congenital/genetics , Anemia, Hemolytic, Congenital/metabolism , Anemia, Hemolytic, Congenital/pathology , Animals , Biological Transport, Active , Cell Survival/physiology , Erythrocyte Membrane/genetics , Erythrocytes, Abnormal/metabolism , Erythrocytes, Abnormal/pathology , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/metabolism , Metabolism, Inborn Errors/pathology , Mice , Mice, Mutant Strains , Phospholipids/genetics
12.
Liver Int ; 35(9): 2174-86, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25786512

ABSTRACT

BACKGROUND & AIMS: Steatosis accentuates the severity of hepatic ischaemia-reperfusion injury (IRI); 'statins' (HMG-CoA reductase inhibitors) protect the heart and brain against post-ischaemic injury. We tested whether short-term administration of atorvastatin protects fatty livers in obese mice against IRI. METHODS: Mice with dietary or genetic simple steatosis (SS) or non-alcoholic steatohepatitis (NASH) were subjected to 60 min partial hepatic ischaemia/24 h reperfusion. Atorvastatin was injected intravenously (5 mg/kg) 1 h before IRI. Liver injury, Toll-like receptor-4 (TLR4), cytokines/chemokines, iNOS/eNOS expression, eNOS activity and thromboxane B2 (TXB2) production were determined. RESULTS: Ischaemia-reperfusion injury was exaggerated by two- to five-fold in SS and NASH compared with lean liver. Atorvastatin pretreatment conferred 70-90% hepatic protection in all animals. Atorvastatin increased post-ischaemic eNOS mRNA/protein and strikingly enhanced eNOS activity (by phospho-eNOS). It also attenuated microparticle (MP) production, NF-κB activation, significantly dampened post-ischaemic thromboxane B2 production, induction of TNF-α, IL-6, MIP-1a, MCP-1, GM-CSF and vascular cell adhesion molecule-1 (VCAM), with a resultant reduction on macrophage and polymorphonuclear neutrophil recruitment. Up-regulation of HMGB1 and TLR4 after IRI was marked in fatty livers; 1 h pretreatment with atorvastatin reduced HMGB1 and TLR4 expression in all livers. CONCLUSIONS: Acute (1 h) atorvastatin administration is highly hepatoprotective against IRI in NASH, fatty and lean livers. Key mechanisms include suppression of inflammation by prevention of NF-κB activation, microvascular protection via eNOS activation and suppression of TXB2 and MP release. Short-term intravenous statin treatment is a readily available and effective preventive agent against hepatic IRI, irrespective of obesity and fatty liver disease, and merits clinical trials in at-risk patients.


Subject(s)
Atorvastatin/administration & dosage , Chemokines/blood , Cytokines/blood , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Non-alcoholic Fatty Liver Disease/drug therapy , Reperfusion Injury/drug therapy , Animals , HMGB1 Protein , Liver/pathology , Male , Mice , Mice, Obese , NF-kappa B/metabolism , Nitric Oxide Synthase Type III/metabolism , Thromboxane B2/metabolism , Toll-Like Receptor 4/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
13.
Liver Int ; 34(7): 1084-93, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24107103

ABSTRACT

BACKGROUND & AIMS: Obese Alms1 mutant (foz/foz) NOD.B10 mice develop diabetes and fibrotic NASH when fed high-fat(HF) diet. To establish whether diabetes or obesity is more closely associated with NASH fibrosis, we compared diabetic foz/foz C57BL6/J with non-diabetic foz/foz BALB/c mice. We also determined hepatic cytokines, growth factors and related profibrotic pathways. METHODS: Male and female foz/foz BALB/c and C57BL6/J mice were fed HF or chow for 24 weeks before determining metabolic indices, liver injury, cytokines, growth factors, pathology/fibrosis and matrix deposition pathways. RESULTS: All foz/foz mice were obese. Hepatomegaly, hyperinsulinemia, hyperglycaemia and hypoadiponectinaemia occurred only in foz/foz C57BL6/J mice, whereas foz/foz BALB/c formed more adipose. Serum ALT, steatosis, ballooning, liver inflammation and NAFLD activity score were worse in C57BL6/J mice. In HF-fed mice, fibrosis was severe in foz/foz C57BL6/J, appreciable in WT C57BL6/J, but absent in foz/foz BALB/c mice. Hepatic mRNA expression of TNF-α, IL-12, IL-4, IL-10 was increased (but not IFN-γ, IL-1ß, IL-17A), and IL-4:IFN-γ ratio (indicating Th-2 predominance) was higher in HF-fed foz/foz C57BL6/J than BALB/c mice. In livers of HF-fed foz/foz C57BL6/J mice, TGF-ß was unaltered but PDGFα and CTGF were increased in association with enhanced α-SMA, CD147and MMP activity. CONCLUSIONS: In mice with equivalent genetic/dietary obesity, NASH development is linked to strain differences in hyperinsulinaemia and hyperglycaemia inversely related to lipid partitioning between adipose and liver. Diabetes-mediated CTGF-regulation of MMPs as well as cytokines/growth factors (Th-2 cytokine predominant, PDGFα, not TGF-ß) mobilized in the resultant hepatic necroinflammatory change may contribute to strain differences in NASH fibrosis.


Subject(s)
DNA-Binding Proteins/genetics , Diabetes Mellitus, Type 2/etiology , Diet, High-Fat/adverse effects , Liver Cirrhosis/etiology , Non-alcoholic Fatty Liver Disease/etiology , Analysis of Variance , Animals , Cell Cycle Proteins , Cytokines/metabolism , Diabetes Mellitus, Type 2/metabolism , Female , Fluorescent Antibody Technique , Intercellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Liver/pathology , Liver Cirrhosis/pathology , Male , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Non-alcoholic Fatty Liver Disease/pathology , Species Specificity
15.
J Gastroenterol Hepatol ; 29(3): 435-41, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24199670

ABSTRACT

Coffee is one of the most popular beverages in the world. Several studies consistently show that coffee drinkers with chronic liver disease have a reduced risk of cirrhosis and a lower incidence of hepatocellular carcinoma regardless of primary etiology. With the increasing prevalence of non-alcoholic fatty liver disease (NAFLD) worldwide, there is renewed interest in the effect of coffee intake on NAFLD severity and positive clinical outcomes. This review gives an overview of growing epidemiological and clinical evidence which indicate that coffee consumption reduces severity of NAFLD. These studies vary in methodology, and potential confounding factors have not always been completely excluded. However, it does appear that coffee, and particular components other than caffeine, reduce NAFLD prevalence and inflammation of non-alcoholic steatohepatitis. Several possible mechanisms underlying coffee's hepatoprotective effects in NAFLD include antioxidative, anti-inflammatory, and antifibrotic effects, while a chemopreventive effect against hepatocarcinogenesis seems likely. The so-far limited data supporting such effects will be discussed, and the need for further study is highlighted.


Subject(s)
Coffee , Fatty Liver/prevention & control , Anti-Inflammatory Agents , Antioxidants , Carcinoma, Hepatocellular/prevention & control , Chemoprevention , Humans , Liver Cirrhosis/prevention & control , Liver Neoplasms/prevention & control , Non-alcoholic Fatty Liver Disease , Prevalence , Prognosis , Severity of Illness Index
16.
J Gastroenterol Hepatol ; 29(6): 1258-64, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24955454

ABSTRACT

BACKGROUND AND AIM: Pegylated-interferon-α/ribavirin (PEG-IFN/RBV) treatment can cure hepatitis C virus (HCV) infection but has frequent neuropsychiatric side-effects. Patients with pre-existing psychiatric illness may not be offered therapy. We established prevalence of self-reported psychiatric comorbidity among HCV-infected patients in a hospital-liver clinic, and determined the impact of such diagnoses on uptake and tolerance to PEG-IFN/RBV. METHODS: All HCV cases referred for assessment in Australian Capital Territory/surrounding regions April 2004-March 2012 were entered into a clinical database. We conducted univariate and multivariate analyses of variables correlating with uptake of antiviral therapy and frequency of treatment-related side-effects. RESULTS: Of 773 referred patients, 235 (30%) described pre-existing psychiatric illness. Among these, 26% received antiviral therapy, compared with 30% of 538 without psychiatric comorbidity. History of depression (usually validated by liaison psychiatry) was associated with higher incidence of treatment-related neuropsychiatric side-effects (odds ratio 2.79 [1.35-5.70], P < 0.05) but did not affect treatment outcome. Twenty-seven patients reported schizophrenia: three (11%) received antiviral therapy, compared with 30% admitting depression and 20% with bipolar affective disorder (all assessed by psychiatrist). In most schizophrenia cases, the reason for not offering antiviral treatment was psychological illness, yet none of five treated (these three plus two others in a psychiatric rehabilitation facility) experienced worsening psychiatric symptoms. CONCLUSIONS: A history of depression is common with hepatitis C but does not affect initiation of antiviral treatment, despite substantially increased risk of psychiatric side-effects. In contrast, pre-existing schizophrenia appears to influence treatment decisions, despite little evidence that PEG-IFN/RBV exacerbates the psychiatric condition, and well-supervised antiviral therapy can have good outcomes.


Subject(s)
Antiviral Agents/adverse effects , Hepatitis C/complications , Hepatitis C/drug therapy , Interferon-alpha/adverse effects , Mental Disorders/chemically induced , Mental Disorders/complications , Ribavirin/adverse effects , Adult , Depression/chemically induced , Depression/complications , Disease Progression , Female , Humans , Male , Middle Aged , Multivariate Analysis , Schizophrenia/chemically induced , Schizophrenia/complications
18.
Sci Adv ; 10(4): eadh3409, 2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38277448

ABSTRACT

The innate immune response contributes to the development or attenuation of acute and chronic diseases, including cancer. Microbial DNA and mislocalized DNA from damaged host cells can activate different host responses that shape disease outcomes. Here, we show that mice and humans lacking a single allele of the DNA repair protein Ku70 had increased susceptibility to the development of intestinal cancer. Mechanistically, Ku70 translocates from the nucleus into the cytoplasm where it binds to cytosolic DNA and interacts with the GTPase Ras and the kinase Raf, forming a tripartite protein complex and docking at Rab5+Rab7+ early-late endosomes. This Ku70-Ras-Raf signalosome activates the MEK-ERK pathways, leading to impaired activation of cell cycle proteins Cdc25A and CDK1, reducing cell proliferation and tumorigenesis. We also identified the domains of Ku70, Ras, and Raf involved in activating the Ku70 signaling pathway. Therapeutics targeting components of the Ku70 signalosome could improve the treatment outcomes in cancer.


Subject(s)
Neoplasms , Signal Transduction , Animals , Humans , Mice , Cell Proliferation , DNA , MAP Kinase Signaling System , Neoplasms/genetics
19.
J Hepatol ; 59(1): 144-52, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23500152

ABSTRACT

BACKGROUND & AIMS: We have recently showed that hyperinsulinemia promotes hepatic free cholesterol (FC) accumulation in obese, insulin-resistant Alms1 mutant (foz/foz) mice with NASH. Here we tested whether cholesterol-lowering drugs reduce stress-activated c-Jun N-terminal kinase (JNK) activation, hepatocyte injury/apoptosis, inflammation, and fibrosis in this metabolic syndrome NASH model. METHODS: Female foz/foz and WT mice were fed HF (0.2% cholesterol) 16 weeks, before adding ezetimibe (5 mg/kg), atorvastatin (20 mg/kg), or both to diet, another 8 weeks. Hepatic lipidomic analysis, ALT, liver histology, Sirius Red morphometry, hepatic mRNA and protein expression and immunohistochemistry (IHC) for apoptosis (M30), macrophages (F4/80), and polymorphs (myeloperoxidase) were determined. RESULTS: In mice with NASH, ezetimibe/atorvastatin combination normalized hepatic FC but did not alter saturated free fatty acids (FFA) and had minimal effects on other lipids; ezetimibe and atorvastatin had similar but less profound effects. Pharmacological lowering of FC abolished JNK activation, improved serum ALT, apoptosis, liver inflammation/NAFLD activity score, designation as "NASH", macrophage chemotactic protein-1 expression, reduced macrophage and polymorph populations, and liver fibrosis. CONCLUSIONS: Cholesterol lowering with ezetimibe/atorvastatin combination reverses hepatic FC but not saturated FFA accumulation. This dampens JNK activation, ALT release, hepatocyte apoptosis, and inflammatory recruitment, with reversal of steatohepatitis pathology and liver fibrosis. Ezetimibe/statin combination is a potent, mechanism-based treatment that could reverse NASH and liver fibrosis.


Subject(s)
Anticholesteremic Agents/therapeutic use , Diabetes Complications/drug therapy , Fatty Liver/drug therapy , Metabolic Syndrome/drug therapy , Adiponectin/blood , Animals , Anticholesteremic Agents/administration & dosage , Atorvastatin , Azetidines/administration & dosage , Azetidines/therapeutic use , Cholesterol/metabolism , Diabetes Complications/metabolism , Diabetes Complications/pathology , Drug Therapy, Combination , Ezetimibe , Fatty Liver/complications , Fatty Liver/metabolism , Female , Heptanoic Acids/administration & dosage , Heptanoic Acids/therapeutic use , Insulin/blood , Liver/drug effects , Liver/metabolism , Liver/pathology , MAP Kinase Signaling System/drug effects , Metabolic Syndrome/complications , Metabolic Syndrome/metabolism , Mice , Mice, Mutant Strains , Mice, Obese , Non-alcoholic Fatty Liver Disease , Pyrroles/administration & dosage , Pyrroles/therapeutic use
20.
J Gastroenterol Hepatol ; 28(9): 1545-54, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23574010

ABSTRACT

BACKGROUND AND AIM: By array-comparative genomic hybridization, we demonstrated cyclin E as one of seven genes associated with hepatocellular carcinoma (HCC) development in Ku70 DNA repair-deficient mice. We therefore explored the hypothesis that during hepatocarcinogenesis, cyclin E kinase can overcome the inhibitory effects of p53 and establish whether abnormal miRNA(mi-R)-34, a co-regulator of cyclin E and p53, can account for their interactions as "drivers" of HCC. METHODS: Dysplastic hepatocytes (DNs) and HCCs were generated from diethylnitrosamine (DEN)-injected C57BL/6 male mice at 3-12 months. RESULTS: Cyclin E/cdk2 was barely expressed in normal liver, but was readily detected in dysplastic hepatocytes, localizing to glutathione-S transferase pi-form positive cells dissected by laser-dissection. Cyclin E kinase activity preceded cyclin D1, proliferating cell nuclear antigen expression in DNs and HCCs despite maximal p53 and p21 expression. We confirmed that cyclin E, rather than cyclin D1, is the proliferative driver in hepatocarcinogenesis by immunoprecipitation experiments demonstrating preferential binding of p21 to cyclin D1, allowing cyclin E-mediated "escape" from G1/S checkpoint. We then showed cyclin E was responsible for regulating wild-type p53 by knockdown experiments in primary HCC cells; cyclin E-knockdown increased p53 and p21, diminished anti-apoptotic Bcl-XL and reduced cell viability. Conversely, blocking p53 augmented cyclin E, Bcl-XL expression and increased proliferation. Physiological interactions between cyclin E/p53/p21 were confirmed in primary hepatocytes. miR-34a,c were upregulated in dysplastic murine, human liver and HCCs compared with normal liver, and appeared to be linked to cyclin E/p53. CONCLUSION: Upregulation of functionally active cyclin E via miR34 with loss of p53 function is associated with cell-cycle checkpoint failure increasing proliferative drive that favors hepatocarcinogenesis.


Subject(s)
Cell Transformation, Neoplastic/pathology , Cyclin E/physiology , G1 Phase Cell Cycle Checkpoints/physiology , Hepatocytes/pathology , Liver Neoplasms, Experimental/pathology , Animals , Apoptosis , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cyclin E/biosynthesis , Cyclin-Dependent Kinase 2 , Diethylnitrosamine , Gene Knockdown Techniques , Hepatocytes/metabolism , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/physiology
SELECTION OF CITATIONS
SEARCH DETAIL