Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Hepatol ; 63(1): 191-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25724365

ABSTRACT

BACKGROUND & AIMS: Alcoholic liver disease (ALD) is characterized by the development of fatty liver, alcoholic hepatitis, fibrosis and cirrhosis. However, the underlying mechanism(s) associated with progression remains elusive. Pro-inflammatory cytokines have been implicated in ALD progression due to pro-apoptotic effects on hepatocytes. Wnt/ß-catenin signaling recently has been shown to promote inflammation and apoptosis, suggesting that activation of this signaling pathway may modulate ALD progression. The current study was designed to test whether pharmacological activation of Wnt/ß-catenin signaling altered ALD development and progression in a rat model. METHODS: Adult male Long Evans rats were fed with isocaloric liquid diets containing 0% or 37% ethanol for 8 weeks, and also treated with Wnt agonist during the last 3 weeks of the feeding regimen. Liver and blood samples were subjected to histology, TUNEL assay, immunoblot analysis, real-time quantitative PCR, and alanine transaminase (ALT) assay. RESULTS: Wnt/ß-catenin signaling was negatively correlated with Foxo3A expression and reduced steatosis, cellular injury and apoptosis in ALD rats. Mutation experiments demonstrated that Foxo3A was critical for modulating these effects. Activation of Wnt/ß-catenin signaling suppressed Foxo3A-induced apoptosis through upregulation of serum/glucocorticoid regulated kinase 1 (SGK1). Moreover, pharmacological restoration of Wnt/ß-catenin signaling reduced ALD progression in vivo. CONCLUSIONS: Wnt/ß-catenin signaling plays a protective role in ALD progression via antagonizing Foxo3A-induced apoptosis, and activation of the Wnt/ß-catenin signaling cascade attenuates ALD progression.


Subject(s)
DNA/genetics , Liver Diseases, Alcoholic/genetics , Up-Regulation , Wnt Signaling Pathway/genetics , beta Catenin/genetics , Animals , Apoptosis , Cells, Cultured , Disease Models, Animal , Disease Progression , Immunoblotting , Immunohistochemistry , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Male , Rats , Rats, Long-Evans , Real-Time Polymerase Chain Reaction , beta Catenin/metabolism
2.
Alcohol Clin Exp Res ; 38(4): 897-910, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24460535

ABSTRACT

BACKGROUND: Chronic ethanol (EtOH) consumption impairs the ability of insulin to suppress hepatic glucose production in a strain-dependent manner, with hepatic insulin resistance being greater in Long-Evans (LE) than Sprague-Dawley (SD) rats. We assessed whether strain differences exist for whole-body and tissue glucose uptake under basal and insulin-stimulated conditions and whether they were associated with coordinate strain-dependent elevations in muscle cytokines. METHODS: Male rats (160 g) were provided the Lieber-DeCarli EtOH-containing (36% total energy) diet or pair-fed a control diet for 8 weeks. Rats were studied in the basal state or during a euglycemic hyperinsulinemic clamp, and whole-body glucose flux assessed using (3) H-glucose and in vivo tissue glucose uptake by (14) C-2-deoxyglucose. RESULTS: EtOH impaired whole-body insulin-mediated glucose uptake (IMGU) more in SD than LE rats. This difference was due to impaired IMGU by gastrocnemius and heart in EtOH-fed SD versus LE rats. However, decreased IMGU in adipose tissue (epididymal and perirenal) produced by EtOH was comparable between strains. EtOH-induced insulin resistance in muscle from SD rats was associated with reduced AKT and AS160 phosphorylation and plasma membrane-localized GLUT4 protein as well as enhanced phosphorylation of c-Jun N-terminal kinase (JNK) and IRS-1 (S307), changes which were absent in muscle from LE rats. EtOH increased tumor necrosis factor alpha (TNFα) mRNA in gastrocnemius and fat under basal conditions in both SD and LE rats; however, hyperinsulinemia decreased TNFα in skeletal muscle from LE, but not SD rats. Interleukin (IL)-6 mRNA in gastrocnemius was increased under basal conditions and increased further in response to insulin in SD rats, but no EtOH- or insulin-induced change was detected in muscle IL-6 of LE rats. CONCLUSIONS: These data indicate strain-dependent differences in EtOH-induced IMGU in skeletal and cardiac muscle, but not fat, associated with sustained increases in TNFα and IL-6 mRNA and JNK activation and decreased plasma membrane GLUT4 in response to insulin.


Subject(s)
Ethanol/administration & dosage , Glucose/metabolism , Muscle, Skeletal/metabolism , Myocardium/metabolism , Animals , Glucose Clamp Technique/methods , Insulin/pharmacology , Male , Muscle, Skeletal/drug effects , Random Allocation , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Species Specificity
3.
J Hepatol ; 58(4): 785-91, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23211317

ABSTRACT

BACKGROUND & AIMS: p53 and its transcriptional target miRNA34a have been implicated in the pathogenesis of fatty liver. We tested the efficacy of a p53 inhibitor, pifithrin-α p-nitro (PFT) in attenuating steatosis, associated oxidative stress and apoptosis in a murine model of non-alcoholic fatty liver disease (NAFLD). METHODS: C57BL/6 mice were fed a high-fat (HFD) or control diet for 8 weeks; PFT or DMSO (vehicle) was administered three times per week. Markers of oxidative stress and apoptosis as well as mediators of hepatic fatty acid metabolism were assessed by immunohistochemistry, Western blot, real-time PCR, and biochemical assays. RESULTS: PFT administration suppressed HFD-induced weight gain, ALT elevation, steatosis, oxidative stress, and apoptosis. PFT treatment blunted the HFD-induced upregulation of miRNA34a and increased SIRT1 expression. In the livers of HFD-fed, PFT-treated mice, activation of the SIRT1/PGC1α/PPARα axis increased the expression of malonyl-CoA decarboxylase (MLYCD), an enzyme responsible for malonyl-CoA (mCoA) degradation. Additionally, the SIRT1/LKB1/AMPK pathway (upstream activator of MLYCD) was promoted by PFT. Thus, induction of these two pathways by PFT diminished the hepatic mCoA content by enhancing MLYCD expression and function. Since mCoA inhibits carnitine palmitoyltransferase 1 (CPT1), the decrease of hepatic mCoA in the PFT-treated, HFD-fed mice increased CPT1 activity, favored fatty acid oxidation, and decreased steatosis. Additionally, we demonstrated that PFT abrogated steatosis and promoted MLYCD expression in palmitoleic acid-treated human HepaRG cells. CONCLUSIONS: The p53 inhibitor PFT diminished hepatic triglyceride accumulation and lipotoxicity in mice fed a HFD, by depleting mCoA and favoring the ß-oxidation of fatty acids.


Subject(s)
Benzothiazoles/pharmacology , Fatty Liver/prevention & control , Toluene/analogs & derivatives , Tumor Suppressor Protein p53/antagonists & inhibitors , Alanine Transaminase/metabolism , Animals , Apoptosis/drug effects , Cell Line , Diet, High-Fat/adverse effects , Disease Models, Animal , Fatty Acids/metabolism , Fatty Liver/metabolism , Fatty Liver/pathology , Humans , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Malonyl Coenzyme A/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Models, Biological , Non-alcoholic Fatty Liver Disease , Oxidative Stress/drug effects , Toluene/pharmacology , Triglycerides/metabolism , Tumor Suppressor Protein p53/metabolism , Weight Gain/drug effects
4.
Liver Int ; 32(5): 761-70, 2012 May.
Article in English | MEDLINE | ID: mdl-22292946

ABSTRACT

BACKGROUND: Previous studies demonstrated that the Long-Evans (LE) rats exhibited liver injury and lipid metabolic abnormalities after 8 weeks of ethanol feeding. AIMS: The goal of this study was to investigate if the LE rats develop more advanced hepatic abnormalities (e.g., fibrosis) after long-term feeding with an ethanol-containing Lieber-DeCarli diet. In addition, the contribution of early growth response-1 (EGR1) transcription factor to these pathological changes was assessed. METHODS: Long-Evans rats were fed an ethanol-containing or isocaloric control liquid diet for 18 months. Livers were processed for histological analyses, studies of fibrosis-related gene expression, cell fractionation and triglyceride measurement. Serum alanine aminotransferase (ALT) levels were assessed. DNA binding activities of p53 and the sterol regulatory element-binding protein-1c (SREBP1c) were analysed. The abundance of EGR1 and enzymes involved in fatty acid synthesis were determined. Chromatin immunoprecipitation was employed to study EGR1 binding to the SREBP1c promoter region. RESULTS: Ethanol feeding generated steatosis, chicken wire fibrosis and ALT elevations in the LE rats. Fibrosis was associated with the upregulation of EGR1 and its downstream target genes. EGR1 upregulation was associated with enhanced p53 activity and an increase in the cellular p66(shc) abundance. Steatosis was linked to the activation of SREBP1c. Importantly, EGR1 upregulation paralleled the expression and transcriptional activity of SREBP1c. Finally, EGR1 was shown to bind to the SREBP1c promoter region. CONCLUSIONS: Long-term ethanol feeding promoted steatosis and fibrosis in LE rats via EGR1 activation. The highly abundant EGR1 bound to the SREBP1c promoter and contributed to the steatosis observed in the LE rat model.


Subject(s)
Central Nervous System Depressants/toxicity , Early Growth Response Protein 1/genetics , Ethanol/toxicity , Fatty Liver/chemically induced , Liver Cirrhosis/chemically induced , Alanine Transaminase/blood , Animal Feed , Animals , Biomarkers/metabolism , Cell Fractionation , Central Nervous System Depressants/administration & dosage , Disease Models, Animal , Early Growth Response Protein 1/biosynthesis , Ethanol/administration & dosage , Fatty Liver/genetics , Fatty Liver/pathology , Gene Expression/drug effects , Gene Expression Profiling , Liver/chemistry , Liver/drug effects , Liver/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Male , Protein Binding , Rats , Rats, Long-Evans , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides/analysis , Up-Regulation
5.
J Gastroenterol Hepatol ; 27 Suppl 2: 33-41, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22320914

ABSTRACT

Chronic alcohol exposure inhibits insulin and insulin-like growth factor signaling in the liver and brain by impairing the signaling cascade at multiple levels. These alterations produced by alcohol cause severe hepatic and central nervous system insulin resistance as the cells fail to adequately transmit signals downstream through Erk/mitogen-activated protein kinase (MAPK), which is needed for DNA synthesis and liver regeneration, and phosphatidylinositol 3-kinase (PI3K), which promotes growth, survival, cell motility, glucose utilization, plasticity, and energy metabolism. The robust inhibition of insulin signaling in liver and brain is augmented by additional factors involving the activation of phosphatases such as phosphatase and tensin homologue (PTEN), which further impairs insulin signaling through PI3K/Akt. Thus, intact insulin signaling is important for neuronal survival. Chronic alcohol consumption produces steatohepatitis, which also promotes hepatic insulin resistance, oxidative stress and injury, with the attendant increased generation of "toxic lipids" such as ceramides that increase insulin resistance. The PI3K/Akt signaling cascade is altered by direct interaction with ceramides as well as through PTEN upregulation as a downstream target gene of enhanced p53 transcriptional activity. Cytotoxic ceramides transferred from the liver to the blood can enter the brain due to their lipid-soluble nature, and thereby exert neurodegenerative effects via a liver-brain axis. We postulate that the neurotoxic and neurodegenerative effects of liver-derived ceramides activate pro-inflammatory cytokines and increase lipid adducts and insulin resistance in the brain to impair cognitive and motor function. These observations are discussed in the context of insulin sensitizers as potential cytoprotective agents against liver and brain injury induced by alcohol.


Subject(s)
Alcohol-Induced Disorders, Nervous System/etiology , Alcoholism/complications , Brain/metabolism , Insulin Resistance , Liver Diseases, Alcoholic/etiology , Liver/metabolism , Alcohol-Induced Disorders, Nervous System/metabolism , Alcohol-Induced Disorders, Nervous System/pathology , Alcohol-Induced Disorders, Nervous System/physiopathology , Alcoholism/drug therapy , Alcoholism/metabolism , Alcoholism/pathology , Alcoholism/physiopathology , Animals , Brain/pathology , Brain/physiopathology , DNA Damage , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/metabolism , Humans , Insulin/metabolism , Liver/drug effects , Liver/pathology , Liver/physiopathology , Liver Diseases, Alcoholic/drug therapy , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Liver Diseases, Alcoholic/physiopathology , Liver Regeneration , PPAR gamma/agonists , PPAR gamma/metabolism , PTEN Phosphohydrolase/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction
6.
Nano Lett ; 11(7): 2678-83, 2011 Jul 13.
Article in English | MEDLINE | ID: mdl-21644516

ABSTRACT

We present the development of a new imaging technique for the early diagnosis of hepatocellular carcinoma that utilizes surface-modified gold nanoparticles in combination with X-ray imaging. Tissues labeled with these electron-dense particles show enhanced X-ray scattering over normal tissues, distinguishing cells containing gold nanoparticles from cells without gold in X-ray scatter images. Our results suggest that this novel approach could enable the in vivo detection of tumors as small as a few millimeters in size.


Subject(s)
Carcinoma, Hepatocellular/diagnosis , Gold/chemistry , Liver Neoplasms/diagnosis , Nanostructures/chemistry , Nanotechnology , Humans , Molecular Imaging , Particle Size , Scattering, Radiation , Surface Properties , X-Rays
7.
J Hepatol ; 54(1): 164-72, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20961644

ABSTRACT

BACKGROUND & AIMS: Chronic ethanol consumption in the Long-Evans (LE) rat has been associated with hepatic p53 activation, and inhibition of the insulin/PI3K/AKT signal transduction cascade due to increased expression of PTEN. We hypothesize that p53 activation and altered insulin signaling may influence the susceptibility of rats to ethanol-induced liver damage. Furthermore, p53 not only activates programmed cell death pathways and suppresses hepatocellular survival signals, but also promotes gluconeogenesis to increase systemic insulin resistance due to a novel metabolic function. METHODS: Fischer (F), Sprague-Dawley (SD) and LE rats were fed ethanol-containing or control liquid diet for 8 weeks. Histopathological and biochemical changes were assessed. RESULTS: Here, we demonstrate that chronic ethanol feeding in rats promotes p53 activation, hepatic steatosis, oxidative stress, PUMA, and PTEN expression, which contribute to hepatocellular death and diminished insulin signaling in the liver. Such changes are pronounced in the LE, less prominent in SD, and virtually absent in the F rat strain. More importantly, there is activation of Tp53-induced glycolysis and apoptosis regulator (TIGAR) in the ethanol-fed LE rat. This event generates low hepatic fructose-2,6-bisphosphate (Fru-2,6-P2) levels, reduced lactate/pyruvate ratio and may contribute to increased basal glucose turnover and high residual hepatic glucose production during euglycemic hyperinsulinemic clamp. CONCLUSIONS: p53 activation correlates with the susceptibility to ethanol-induced liver damage in different rat strains. p53 not only orchestrates apoptosis and suppresses cell survival, but by activating TIGAR and decreasing hepatic Fru-2,6-P2) levels it promotes insulin resistance and therefore, contributes to the metabolic abnormalities associated with hepatic steatosis.


Subject(s)
Apoptosis/physiology , Insulin Resistance/physiology , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Tumor Suppressor Protein p53/metabolism , Alanine Transaminase/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Caspase 3/metabolism , DNA Damage , DNA, Mitochondrial/metabolism , Disease Models, Animal , Fatty Liver, Alcoholic/metabolism , Fatty Liver, Alcoholic/pathology , Fructosediphosphates/metabolism , Gluconeogenesis , Male , Oxidative Stress , PTEN Phosphohydrolase/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Inbred F344 , Rats, Long-Evans , Rats, Sprague-Dawley , Signal Transduction , Species Specificity , Sterol Regulatory Element Binding Protein 1/metabolism
8.
Methods Mol Biol ; 559: 205-17, 2009.
Article in English | MEDLINE | ID: mdl-19609759

ABSTRACT

There is an increasing evidence that uncoupling protein-2 (UCP2), a recently identified molecular sensor and suppressor of mitochondrial reactive oxygen species (ROS), plays an important role in -regulating apoptosis in different cell systems. A great technical difficulty that many groups have encountered is the reliable detection of endogenously or exogenously expressed UCP2 protein. The goal of this -chapter is to introduce the reader to techniques that we have successfully used over the years to detect UCP2 protein in various mouse and human specimens. These techniques include mitochondrial isolation and submitochondrial fractionation followed by Western blotting and UCP2 immunohistochemistry. We find that sample preparation is a key to success and it allows one to produce relevant and important data using commercially available antibodies.


Subject(s)
Apoptosis , Cell Fractionation/methods , Ion Channels/analysis , Ion Channels/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/analysis , Mitochondrial Proteins/metabolism , Alkalies , Animals , Blotting, Western/methods , Digitonin , Humans , Immunohistochemistry/methods , Mice , Oxidative Stress , Uncoupling Protein 2
9.
Br J Clin Pharmacol ; 66(3): 366-74, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18492126

ABSTRACT

AIMS: Human serum paraoxonase-1 (PON1) protects lipoproteins against oxidation by hydrolysing lipid peroxides in oxidized low-density lipoprotein, therefore it may protect against atherosclerosis. One of the two common PON1 gene polymorphisms within the PON1 gene is the Q192R, whose prevalence can be estimated by phenotype distribution analysis. The goal of this study was to clarify the role of PON1 phenotypes on the effect of three different statins on paraoxonase activity and lipid parameters. METHODS: One hundred and sixty-four patients with type IIb hypercholesterolaemia were involved in the study. We examined the effect of 10 mg day(-1) atorvastatin, 10/20 mg day(-1) simvastatin and 80 mg day(-1) extended-release fluvastatin treatment on lipid levels and paraoxonase activity in patients with different PON1 phenotypes. The phenotype distribution of PON1 was determined by the dual substrate method. RESULTS: Three months of statin treatment significantly increased paraoxonase activity in every statin-treated group. In patients with AB+BB phenotype, statin treatment was significantly more effective on paraoxonase activity than in the AA group. Statin treatment more effectively decreased triglyceride levels in the AB+BB group compared with the AA group in the whole study population and in the simvastatin-treated group. Atorvastatin treatment was significantly more effective on apolipoprotein B levels in patients with AB+BB phenotype than in the AA phenotype group. CONCLUSIONS: The PON1 phenotype may be a novel predictive factor for the effectiveness of statin treatment on PON1 activity and serum lipid levels; however, different types of statins may exert different effects on these parameters.


Subject(s)
Aryldialkylphosphatase/therapeutic use , Fatty Acids, Monounsaturated/therapeutic use , Heptanoic Acids/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypercholesterolemia/drug therapy , Indoles/therapeutic use , Pyrroles/therapeutic use , Simvastatin/therapeutic use , Aryldialkylphosphatase/genetics , Atorvastatin , Cholesterol, LDL/genetics , Female , Fluvastatin , Humans , Hypercholesterolemia/genetics , Lipids/blood , Male , Middle Aged , Phenotype , Polymorphism, Genetic
10.
Phys Med Biol ; 53(23): 6911-23, 2008 Dec 07.
Article in English | MEDLINE | ID: mdl-19001700

ABSTRACT

We report a new preparative method for providing contrast through reduction in electron density that is uniquely suited for propagation-based differential x-ray phase contrast imaging. The method, which results in an air or fluid filled vasculature, makes possible visualization of the smallest microvessels, roughly down to 15 microm, in an excised murine liver, while preserving the tissue for subsequent histological workup. We show the utility of spatial frequency filtering for increasing the visibility of minute features characteristic of phase contrast imaging, and the capability of tomographic reconstruction to reveal microvessel structure and three-dimensional visualization of the sample. The effect of water evaporation from livers during x-ray imaging on the visibility of blood vessels is delineated. The deformed vascular tree in a cancerous murine liver is imaged.


Subject(s)
Angiography/methods , Contrast Media , Microvessels/diagnostic imaging , Air , Animals , Female , Formaldehyde , Image Processing, Computer-Assisted , Imaging, Three-Dimensional/methods , Liver/anatomy & histology , Liver/blood supply , Liver/pathology , Liver Neoplasms/blood supply , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Microvessels/anatomy & histology , Microvessels/pathology , Tomography, X-Ray Computed/methods
11.
Sci Rep ; 5: 15673, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26511147

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common malignant tumors worldwide and is almost uniformly fatal. Current methods of detection include ultrasound examination and imaging by CT scan or MRI; however, these techniques are problematic in terms of sensitivity and specificity, and the detection of early tumors (<1 cm diameter) has proven elusive. Better, more specific, and more sensitive detection methods are therefore urgently needed. Here we discuss the application of a newly developed x-ray imaging technique called Spatial Frequency Heterodyne Imaging (SFHI) for the early detection of HCC. SFHI uses x-rays scattered by an object to form an image and is more sensitive than conventional absorption-based x-radiography. We show that tissues labeled in vivo with gold nanoparticle contrast agents can be detected using SFHI. We also demonstrate that directed targeting and SFHI of HCC tumors in a mouse model is possible through the use of HCC-specific antibodies. The enhanced sensitivity of SFHI relative to currently available techniques enables the x-ray imaging of tumors that are just a few millimeters in diameter and substantially reduces the amount of nanoparticle contrast agent required for intravenous injection relative to absorption-based x-ray imaging.


Subject(s)
Carcinoma, Hepatocellular/diagnostic imaging , Contrast Media , Liver Neoplasms, Experimental/diagnostic imaging , Nanoparticles/chemistry , X-Ray Diffraction/methods , Animals , Cell Line, Tumor , Contrast Media/chemistry , Contrast Media/pharmacology , Humans , Mice , NIH 3T3 Cells , Radiography
12.
Phys Med Biol ; 60(2): 769-84, 2015 Jan 21.
Article in English | MEDLINE | ID: mdl-25559398

ABSTRACT

Innovations that improve sensitivity and reduce cost are of paramount importance in diagnostic imaging. The novel x-ray imaging modality called spatial frequency heterodyne imaging (SFHI) is based on a linear arrangement of x-ray source, tissue, and x-ray detector, much like that of a conventional x-ray imaging apparatus. However, SFHI rests on a complete paradigm reversal compared to conventional x-ray absorption-based radiology: while scattered x-rays are carefully rejected in absorption-based x-ray radiology to enhance the image contrast, SFHI forms images exclusively from x-rays scattered by the tissue. In this study we use numerical processing to produce x-ray scatter images of hepatocellular carcinoma labeled with a nanoparticle contrast agent. We subsequently compare the sensitivity of SFHI in this application to that of both conventional x-ray imaging and magnetic resonance imaging (MRI). Although SFHI is still in the early stages of its development, our results show that the sensitivity of SFHI is an order of magnitude greater than that of absorption-based x-ray imaging and approximately equal to that of MRI. As x-ray imaging modalities typically have lower installation and service costs compared to MRI, SFHI could become a cost effective alternative to MRI, particularly in areas of the world with inadequate availability of MRI facilities.


Subject(s)
Carcinoma, Hepatocellular/diagnosis , Contrast Media/pharmacokinetics , Diagnostic Imaging/methods , Liver Neoplasms/diagnosis , Metal Nanoparticles/chemistry , Spectrometry, X-Ray Emission/methods , Gold/chemistry , Humans , Image Processing, Computer-Assisted , In Vitro Techniques , Magnetic Resonance Imaging/methods , Tissue Distribution , Tumor Cells, Cultured , X-Rays
13.
Metabolism ; 63(5): 607-17, 2014 May.
Article in English | MEDLINE | ID: mdl-24629562

ABSTRACT

Obesity has been recognized as a key component of the metabolic syndrome, a cluster of risk factors associated with diabetes and cardiovascular morbidity. In addition, obesity has been linked to higher frequency of cancers in a variety of tissues including the liver. Liver cancer most often occurs as hepatocellular carcinoma (HCC) complicating cirrhosis due to chronic viral infection or toxic injury and remains the third leading cause of cancer death in the world. However, HCC is increasingly diagnosed among individuals with obesity and related disorders. As these metabolic conditions have become globally prevalent, they coexist with well-established risk factors of HCC and create a unique challenge for the liver as a chronically diseased organ. Obesity-associated HCC has recently been attributed to molecular mechanisms such as chronic inflammation due to adipose tissue remodeling and pro-inflammatory adipokine secretion, ectopic lipid accumulation and lipotoxicity, altered gut microbiota, and disrupted senescence in stellate cells, as well as insulin resistance leading to increased levels of insulin and insulin-like growth factors. These mechanisms synergize with those occurring in chronic liver disease resulting from other etiologies and accelerate the development of HCC before or after the onset of cirrhosis. Increasingly common interactions between oncogenic pathways linked to obesity and chronic liver disease may explain why HCC is one of the few malignancies with rising incidence in developed countries. Better understanding of this complex process will improve our strategies of cancer prevention, prediction, and surveillance.


Subject(s)
Carcinoma, Hepatocellular/etiology , Liver Neoplasms/etiology , Obesity/complications , Adipokines/adverse effects , Adipokines/metabolism , Adipose Tissue/physiology , Animals , Carcinoma, Hepatocellular/epidemiology , Cell Transformation, Neoplastic/genetics , Humans , Inflammation Mediators/adverse effects , Inflammation Mediators/metabolism , Insulin/physiology , Liver Neoplasms/epidemiology , Obesity/epidemiology , Somatomedins/physiology
15.
Transl Res ; 153(4): 190-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19304278

ABSTRACT

Human serum paraoxonase-1 (PON1) protects lipoproteins against oxidation by hydrolyzing lipid peroxides in oxidized low-density lipoprotein (LDL); therefore, it may protect against atherosclerosis. Changes in the ratio of high-density lipoprotein (HDL) subfractions may alter the stability and the antioxidant capacity of PON1. The aim of the study was to examine the effect of atorvastatin treatment on the distribution of HDL subfractions, LDL size, cholesteryl ester transfer protein (CETP), lecithin-cholesterol acyltransferase (LCAT), and PON1 activity. In all, 33 patients with type IIa and IIb hypercholesterolemia were involved in the study. LDL sizes and HDL subfractions were determined by gradient gel electrophoresis. CETP, LCAT, and PON1 activities were measured spectrophotometrically. Three months of treatment with atorvastatin 20 mg daily significantly increased the HDL3 (+8.13%) and decreased the HDL2a and HDL2b subfractions (-1.57% and -6.55%, respectively). The mean LDL size was significantly increased (+3.29%). The level of oxidized LDL was significantly decreased (-46.0%). The PON1 activity was augmented by the atorvastatin treatment (+5.0%). The CETP activity positively correlated with the HDL2b level and negatively correlated with the HDL3 and HDL2a levels. Atorvastatin alters the HDL subfractions, which may improve its antiatherogenic effect via enhancement of the PON1 activity.


Subject(s)
Aryldialkylphosphatase/blood , Heptanoic Acids/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lipoproteins, HDL/blood , Pyrroles/pharmacology , Adult , Aged , Atorvastatin , Cholesterol Ester Transfer Proteins/blood , Humans , Middle Aged , Phosphatidylcholine-Sterol O-Acyltransferase/blood
16.
Cancer Res ; 68(8): 2813-9, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18413749

ABSTRACT

Cancer cells acquire drug resistance as a result of selection pressure dictated by unfavorable microenvironments. This survival process is facilitated through efficient control of oxidative stress originating from mitochondria that typically initiates programmed cell death. We show this critical adaptive response in cancer cells to be linked to uncoupling protein-2 (UCP2), a mitochondrial suppressor of reactive oxygen species (ROS). UCP2 is present in drug-resistant lines of various cancer cells and in human colon cancer. Overexpression of UCP2 in HCT116 human colon cancer cells inhibits ROS accumulation and apoptosis after exposure to chemotherapeutic agents. Tumor xenografts of UCP2-overexpressing HCT116 cells retain growth in nude mice receiving chemotherapy. Augmented cancer cell survival is accompanied by altered NH(2)-terminal phosphorylation of the pivotal tumor suppressor p53 and induction of the glycolytic phenotype (Warburg effect). These findings link UCP2 with molecular mechanisms of chemoresistance. Targeting UCP2 may be considered a novel treatment strategy for cancer.


Subject(s)
Drug Resistance, Neoplasm , Ion Channels/genetics , Mitochondrial Proteins/genetics , Animals , Antineoplastic Agents/toxicity , Apoptosis/genetics , Cell Division/genetics , Cell Line, Tumor , Colonic Neoplasms , Gene Expression Regulation, Neoplastic , Glycolysis , Humans , Ion Channels/drug effects , Ion Channels/metabolism , Mice , Mice, Nude , Mitochondria/drug effects , Mitochondrial Proteins/drug effects , Mitochondrial Proteins/metabolism , Polymerase Chain Reaction , Reactive Oxygen Species/antagonists & inhibitors , Transplantation, Heterologous , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays , Uncoupling Protein 2
17.
Am J Physiol Gastrointest Liver Physiol ; 294(4): G1017-24, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18292186

ABSTRACT

Uncoupling protein-2 (UCP2) regulates insulin secretion by controlling ATP levels in beta-cells. Although UCP2 deficiency improves glycemic control in mice, increased expression of UCP2 interferes with glucose-stimulated insulin secretion. These observations link UCP2 to beta-cell dysfunction in type 2 diabetes with a perplexing evolutionary role. We found higher residual serum insulin levels and blunted lipid metabolic responses in fasted ucp2(-/-) mice, supporting the concept that UCP2 evolved to suppress insulin effects and to accommodate the fuel switch to fatty acids during starvation. In the absence of UCP2, fasting initially promotes peripheral lipolysis and hepatic fat accumulation at less than expected rates but culminates in protracted steatosis, indicating diminished hepatic utilization and clearance of fatty acids. We conclude that UCP2-mediated control of insulin secretion is a physiologically relevant mechanism of the metabolic response to fasting.


Subject(s)
Fasting/metabolism , Fatty Acids, Nonesterified/metabolism , Fatty Liver/metabolism , Ion Channels/metabolism , Lipid Mobilization , Liver/metabolism , Mitochondrial Proteins/metabolism , Adaptation, Physiological , Animals , Blood Glucose/metabolism , Fatty Liver/genetics , Fatty Liver/physiopathology , Gene Expression Regulation, Enzymologic , Insulin/blood , Ion Channels/deficiency , Ion Channels/genetics , Lipid Mobilization/genetics , Liver/enzymology , Liver/physiopathology , Male , Mice , Mice, Knockout , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Oxidation-Reduction , Time Factors , Uncoupling Protein 2
18.
Hepatology ; 44(3): 592-601, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16941708

ABSTRACT

Fatty liver is vulnerable to conditions that challenge hepatocellular energy homeostasis. Lipid-laden hepatocytes highly express uncoupling protein-2 (UCP2), a mitochondrial carrier that competes with adenosine triphosphate (ATP) synthesis by mediating proton leak. However, evidence for a link between UCP2 expression and susceptibility of liver to acute injury is lacking. We asked whether absence of UCP2 protects ob/ob mice from Fas-mediated acute liver damage. UCP2-deficient ob/ob mice (ob/ob:ucp2-/-) and UCP2-competent littermates (ob/ob:ucp2+/+) received a single dose of agonistic anti-Fas antibody (Jo2). Low-dose Jo2 (0.15 mg/kg intraperitoneally) caused less serum alanine aminotransferase (ALT) elevation and lower apoptosis rates in ob/ob:ucp2-/- mice. High-dose Jo2 (0.40 mg/kg intraperitoneally) proved uniformly fatal; however, ob/ob:ucp2-/- mice survived longer with less depletion of liver ATP stores, indicating that fatty hepatocytes may benefit from lack of UCP2 during Jo2 challenge. Although UCP2 reportedly controls mitochondrial oxidant production, its absence had no apparent effect on fatty liver tissue malondialdehyde levels augmented by Jo2. This finding prompted us to determine UCP2 expression in Kupffer cells, a major source of intrahepatic oxidative stress. UCP2 expression was found diminished in Kupffer cells of untreated ob/ob:ucp2+/+ mice, conceivably contributing to increased oxidative stress in fatty liver and limiting the impact of UCP2 ablation. In conclusion, whereas UCP2 abundance in fatty hepatocytes exacerbates Fas-mediated injury by compromising ATP stores, downregulation of UCP2 in Kupffer cells may account for persistent oxidative stress in fatty liver. Our data support a cell-specific approach when considering the therapeutic effects of mitochondrial uncoupling in fatty liver disease.


Subject(s)
DNA/genetics , Fatty Liver/metabolism , Gene Expression , Membrane Transport Proteins/genetics , Mitochondrial Proteins/genetics , Acute Disease , Animals , Apoptosis , Disease Models, Animal , Fatty Liver/genetics , Fatty Liver/pathology , Female , Hepatocytes/metabolism , Hepatocytes/pathology , Ion Channels , Membrane Transport Proteins/metabolism , Mice , Mitochondrial Proteins/metabolism , Oxidative Stress , Polymerase Chain Reaction , Uncoupling Protein 2 , fas Receptor/toxicity
19.
Carcinogenesis ; 27(5): 956-61, 2006 May.
Article in English | MEDLINE | ID: mdl-16401637

ABSTRACT

Oxidative stress has a complex effect on cancer development. To further study this process, we induced colon tumors with azoxymethane (AOM) in mice deficient for uncoupling protein-2 (UCP2). UCP2 has recently emerged as a negative regulator of mitochondrial oxidant production. When overexpressed, UCP2 protects cells from oxidative stress, while its absence may cause abundance of reactive oxygen species, release of pro-inflammatory cytokines and persistent activation of nuclear factor kappaB (NF-kappaB), a pleiotropic transcription factor with an increasingly recognized role in cancer. Here we show that Ucp2-/- mice develop more aberrant crypt foci and colon tumors than Ucp2+/+ littermates when examined 24 weeks after the completion of treatment with AOM (10 mg/kg i.p. weekly for a total of 6 weeks, n = 8-12). This effect is primarily seen in the proximal colon of Ucp2-/- mice (P < 0.05), in association with changes indicative of increased oxidative stress (increased staining for malondialdehyde and inducible nitric oxide synthase), enhanced NF-kappaB activation (increased levels of phosphorylated IkappaB and increased nuclear presence of p65) and a disrupted balance between intestinal epithelial cell proliferation (greater 5-bromo-2'-deoxy-uridine incorporation rates and increased phosphorylation of ERK1/2 and AKT) and apoptosis (decreased number of terminal deoxynucleotidyltransferase-mediated nick-end-labeling (TUNEL)-positive cells and increased expression of Bcl-2). In conclusion, our findings provide the first in vivo evidence for a link between UCP2 and tumorigenesis and indicate the need for additional studies to assess the role of mitochondrial uncoupling in cancer development.


Subject(s)
Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Membrane Transport Proteins/genetics , Mitochondrial Proteins/genetics , NF-kappa B/genetics , Oxidative Stress , Animals , Disease Models, Animal , Epigenesis, Genetic , In Situ Nick-End Labeling , Ion Channels , Male , Membrane Transport Proteins/physiology , Mice , Mice, Transgenic , Mitochondrial Proteins/physiology , NF-kappa B/physiology , Reactive Oxygen Species , Time Factors , Uncoupling Protein 2
20.
Hepatology ; 39(2): 386-92, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14767991

ABSTRACT

The control of liver regeneration remains elusive. Because reactive oxygen species (ROS) are able to mediate cell growth arrest and activate proteins that inhibit the cell cycle, ROS production may have a negative impact on liver regeneration. We examined how liver regeneration is affected by uncoupling protein-2 (UCP2), an inner mitochondrial membrane carrier that senses and negatively regulates superoxide production. Liver regeneration was monitored up to 5 days and was found to be significantly delayed in UCP2(-/-) mice after partial hepatectomy. Apoptosis rates in UCP2(+/+) and UCP2(- /-) liver remnants were similar, while parameters of cell proliferation indicated a diminished response in UCP2(- /-) mice with corresponding changes in the expression of key cell cycle regulatory proteins and prolonged activation of stress-responsive protein kinase p38. Levels of malondialdehyde, a marker of ROS generation and oxidant stress, were elevated in UCP2(- /-) livers at every examined time point. Liver remnants of UCP2(+ /+) mice 48 hours post-hepatectomy showed a fourfold increase in the expression of UCP2 protein primarily detected in hepatocytes. In conclusion, our results suggest that absent or insufficient UCP2 function in the regenerating liver results in increased ROS production and negatively modulates the control of cell cycle.


Subject(s)
Liver Regeneration/physiology , Liver/physiology , Membrane Transport Proteins/genetics , Mitochondrial Proteins/genetics , Oxidative Stress/physiology , Animals , Apoptosis/physiology , Cell Cycle Proteins/metabolism , Cell Division/physiology , Female , Hepatectomy , Ion Channels , Membrane Transport Proteins/metabolism , Mice , Mice, Transgenic , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Reactive Oxygen Species/metabolism , Uncoupling Protein 2
SELECTION OF CITATIONS
SEARCH DETAIL