Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Gastroenterology ; 165(1): 187-200.e7, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36966941

ABSTRACT

BACKGROUND & AIMS: Excess copper causes hepatocyte death in hereditary Wilson's disease (WD). Current WD treatments by copper-binding chelators may gradually reduce copper overload; they fail, however, to bring hepatic copper close to normal physiological levels. Consequently, lifelong daily dose regimens are required to hinder disease progression. This may result in severe issues due to nonadherence or unwanted adverse drug reactions and also due to drug switching and ultimate treatment failures. This study comparatively tested bacteria-derived copper binding agents-methanobactins (MBs)-for efficient liver copper depletion in WD rats as well as their safety and effect duration. METHODS: Copper chelators were tested in vitro and in vivo in WD rats. Metabolic cage housing allowed the accurate assessment of animal copper balances and long-term experiments related to the determination of minimal treatment phases. RESULTS: We found that copper-binding ARBM101 (previously known as MB-SB2) depletes WD rat liver copper dose dependently via fecal excretion down to normal physiological levels within 8 days, superseding the need for continuous treatment. Consequently, we developed a new treatment consisting of repetitive cycles, each of ∼1 week of ARBM101 applications, followed by months of in-between treatment pauses to ensure a healthy long-term survival in WD rats. CONCLUSIONS: ARBM101 safely and efficiently depletes excess liver copper from WD rats, thus allowing for short treatment periods as well as prolonged in-between rest periods.


Subject(s)
Hepatolenticular Degeneration , Rats , Animals , Hepatolenticular Degeneration/drug therapy , Hepatolenticular Degeneration/metabolism , Copper , Hepatobiliary Elimination , Liver/metabolism , Chelating Agents/pharmacology , Chelating Agents/therapeutic use
2.
ISME J ; 16(1): 211-220, 2022 01.
Article in English | MEDLINE | ID: mdl-34290379

ABSTRACT

Aerobic methanotrophy is strongly controlled by copper, and methanotrophs are known to use different mechanisms for copper uptake. Some methanotrophs secrete a modified polypeptide-methanobactin-while others utilize a surface-bound protein (MopE) and a secreted form of it (MopE*) for copper collection. As different methanotrophs have different means of sequestering copper, competition for copper significantly impacts methanotrophic activity. Herein, we show that Methylomicrobium album BG8, Methylocystis sp. strain Rockwell, and Methylococcus capsulatus Bath, all lacking genes for methanobactin biosynthesis, are not limited for copper by multiple forms of methanobactin. Interestingly, Mm. album BG8 and Methylocystis sp. strain Rockwell were found to have genes similar to mbnT that encodes for a TonB-dependent transporter required for methanobactin uptake. Data indicate that these methanotrophs "steal" methanobactin and such "theft" enhances the ability of these strains to degrade methylmercury, a potent neurotoxin. Further, when mbnT was deleted in Mm. album BG8, methylmercury degradation in the presence of methanobactin was indistinguishable from when MB was not added. Mc. capsulatus Bath lacks anything similar to mbnT and was unable to degrade methylmercury either in the presence or absence of methanobactin. Rather, Mc. capsulatus Bath appears to rely on MopE/MopE* for copper collection. Finally, not only does Mm. album BG8 steal methanobactin, it synthesizes a novel chalkophore, suggesting that some methanotrophs utilize both competition and cheating strategies for copper collection. Through a better understanding of these strategies, methanotrophic communities may be more effectively manipulated to reduce methane emissions and also enhance mercury detoxification in situ.


Subject(s)
Methylmercury Compounds , Methylosinus trichosporium , Copper/metabolism , Imidazoles/metabolism , Methylmercury Compounds/metabolism , Methylosinus trichosporium/genetics , Methylosinus trichosporium/metabolism , Oligopeptides/metabolism
3.
Life Sci Alliance ; 5(3)2022 03.
Article in English | MEDLINE | ID: mdl-34857647

ABSTRACT

In Wilson disease, excessive copper accumulates in patients' livers and may, upon serum leakage, severely affect the brain according to current viewpoints. Present remedies aim at avoiding copper toxicity by chelation, for example, by D-penicillamine (DPA) or bis-choline tetrathiomolybdate (ALXN1840), the latter with a very high copper affinity. Hence, ALXN1840 may potentially avoid neurological deterioration that frequently occurs upon DPA treatment. As the etiology of such worsening is unclear, we reasoned that copper loosely bound to albumin, that is, mimicking a potential liver copper leakage into blood, may damage cells that constitute the blood-brain barrier, which was found to be the case in an in vitro model using primary porcine brain capillary endothelial cells. Such blood-brain barrier damage was avoided by ALXN1840, plausibly due to firm protein embedding of the chelator bound copper, but not by DPA. Mitochondrial protection was observed, a prerequisite for blood-brain barrier integrity. Thus, high-affinity copper chelators may minimize such deterioration in the treatment of neurologic Wilson disease.


Subject(s)
Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Copper/metabolism , Molybdenum/pharmacology , Penicillamine/pharmacology , Animals , Biological Transport , Biomarkers , Blood-Brain Barrier/diagnostic imaging , Brain/diagnostic imaging , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Survival , Chelating Agents/pharmacology , Copper/adverse effects , Copper/chemistry , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Mice, Transgenic , Mitochondria/metabolism , Mitochondria/ultrastructure , Models, Molecular , Positron-Emission Tomography , Protein Binding , Rats , Serum Albumin/chemistry , Serum Albumin/metabolism , Structure-Activity Relationship
4.
Gastroenterology ; 156(4): 1173-1189.e5, 2019 03.
Article in English | MEDLINE | ID: mdl-30452922

ABSTRACT

BACKGROUND & AIMS: Wilson disease (WD) is an inherited disorder of copper metabolism that leads to copper accumulation and toxicity in the liver and brain. It is caused by mutations in the adenosine triphosphatase copper transporting ß gene (ATP7B), which encodes a protein that transports copper from hepatocytes into the bile. We studied ATP7B-deficient cells and animals to identify strategies to decrease copper toxicity in patients with WD. METHODS: We used RNA-seq to compare gene expression patterns between wild-type and ATP7B-knockout HepG2 cells exposed to copper. We collected blood and liver tissues from Atp7b-/- and Atp7b+/- (control) rats (LPP) and mice; some mice were given 5 daily injections of an autophagy inhibitor (spautin-1) or vehicle. We obtained liver biopsies from 2 patients with WD in Italy and liver tissues from patients without WD (control). Liver tissues were analyzed by immunohistochemistry, immunofluorescence, cell viability, apoptosis assays, and electron and confocal microscopy. Proteins were knocked down in cell lines using small interfering RNAs. Levels of copper were measured in cell lysates, blood samples, liver homogenates, and subcellular fractions by spectroscopy. RESULTS: After exposure to copper, ATP7B-knockout cells had significant increases in the expression of 103 genes that regulate autophagy (including MAP1LC3A, known as LC3) compared with wild-type cells. Electron and confocal microscopy visualized more autophagic structures in the cytoplasm of ATP7B-knockout cells than wild-type cells after copper exposure. Hepatocytes in liver tissues from patients with WD and from Atp7b-/- mice and rats (but not controls) had multiple autophagosomes. In ATP7B-knockout cells, mammalian target of rapamycin (mTOR) had decreased activity and was dissociated from lysosomes; this resulted in translocation of the mTOR substrate transcription factor EB to the nucleus and activation of autophagy-related genes. In wild-type HepG2 cells (but not ATP7B-knockout cells), exposure to copper and amino acids induced recruitment of mTOR to lysosomes. Pharmacologic inhibitors of autophagy or knockdown of autophagy proteins ATG7 and ATG13 induced and accelerated the death of ATP7B-knockout HepG2 cells compared with wild-type cells. Autophagy protected ATP7B-knockout cells from copper-induced death. CONCLUSION: ATP7B-deficient hepatocytes, such as in those in patients with WD, activate autophagy in response to copper overload to prevent copper-induced apoptosis. Agents designed to activate this autophagic pathway might decrease copper toxicity in patients with WD.


Subject(s)
Apoptosis , Autophagy/genetics , Copper-Transporting ATPases/genetics , Hepatocytes/physiology , Hepatolenticular Degeneration/physiopathology , Liver/physiopathology , Animals , Autophagosomes/ultrastructure , Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Benzylamines/pharmacology , Cell Survival , Copper/toxicity , Copper-Transporting ATPases/metabolism , Female , Hep G2 Cells , Hepatocytes/ultrastructure , Humans , Male , Mice , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron , Mitochondria/ultrastructure , Protein Transport , Quinazolines/pharmacology , Rats , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
5.
Cell Mol Gastroenterol Hepatol ; 7(3): 571-596, 2019.
Article in English | MEDLINE | ID: mdl-30586623

ABSTRACT

BACKGROUND & AIMS: In Wilson disease, ATP7B mutations impair copper excretion into bile. Hepatic copper accumulation may induce mild to moderate chronic liver damage or even acute liver failure. Etiologic factors for this heterogeneous phenotype remain enigmatic. Liver steatosis is a frequent finding in Wilson disease patients, suggesting that impaired copper homeostasis is linked with liver steatosis. Hepatic mitochondrial function is affected negatively both by copper overload and steatosis. Therefore, we addressed the question of whether a steatosis-promoting high-calorie diet aggravates liver damage in Wilson disease via amplified mitochondrial damage. METHODS: Control Atp7b+/- and Wilson disease Atp7b-/- rats were fed either a high-calorie diet (HCD) or a normal diet. Copper chelation using the high-affinity peptide methanobactin was used in HCD-fed Atp7b-/- rats to test for therapeutic reversal of mitochondrial copper damage. RESULTS: In comparison with a normal diet, HCD feeding of Atp7b-/- rats resulted in a markedly earlier onset of clinically apparent hepatic injury. Strongly increased mitochondrial copper accumulation was observed in HCD-fed Atp7b-/- rats, correlating with severe liver injury. Mitochondria presented with massive structural damage, increased H2O2 emergence, and dysfunctional adenosine triphosphate production. Hepatocellular injury presumably was augmented as a result of oxidative stress. Reduction of mitochondrial copper by methanobactin significantly reduced mitochondrial impairment and ameliorated liver damage. CONCLUSIONS: A high-calorie diet severely aggravates hepatic mitochondrial and hepatocellular damage in Wilson disease rats, causing an earlier onset of the disease and enhanced disease progression.


Subject(s)
Diet , Hepatolenticular Degeneration/pathology , Liver/pathology , Mitochondria/pathology , Animals , Bile Acids and Salts/biosynthesis , Copper/blood , Copper-Transporting ATPases/metabolism , Disease Progression , Fatty Liver/pathology , Female , Hepatocytes/pathology , Hepatocytes/ultrastructure , Hepatolenticular Degeneration/blood , Inflammation/pathology , Lipids/biosynthesis , Liver/metabolism , Liver/ultrastructure , Male , Mitochondria/metabolism , Mitochondria/ultrastructure , Peptides/pharmacology , Proteome/metabolism , Rats
6.
J Trace Elem Med Biol ; 49: 119-127, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29895360

ABSTRACT

Wilson disease (WD) is a rare genetic disorder of the copper metabolism leading to systemic copper accumulation, predominantly in the liver. The therapeutic approach in WD patients is the generation of a negative copper balance and the maintenance of copper homeostasis, currently by the use of copper chelators such as D-penicillamine (D-PA). However, in circumstances of delayed diagnosis, poor treatment compliance, or treatment failure, mortality is almost certain without hepatic transplantation. Moreover, even after years of D-PA treatment, high liver copper levels are present in WD patients. We have recently suggested the use of the bacterial peptide Methanobactin (MB), which has an outstanding binding affinity for copper, as potentially efficient and patient-friendly remedy against copper damage in WD. Here we substantiate these findings considerably, by demonstrating a significant removal of copper from liver samples of WD rats upon short, one week only, MB treatments. Using laser ablation-inductively coupled plasma-mass spectrometry with a spatial resolution down to 4 µm, we demonstrate that only small copper hotspots remain in MB treated animal livers. We further demonstrate in WD rat liver, seven weeks after the stopped MB treatment, a lower liver copper concentration as compared to untreated control animals. Thus, MB highly efficiently depletes liver copper overload with a sustained therapeutic effect.


Subject(s)
Copper/metabolism , Hepatolenticular Degeneration/drug therapy , Hepatolenticular Degeneration/metabolism , Imidazoles/therapeutic use , Liver/drug effects , Liver/metabolism , Mass Spectrometry/methods , Oligopeptides/therapeutic use , Animals , Mice, Knockout , Rats
7.
Toxicol In Vitro ; 51: 11-22, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29715505

ABSTRACT

Wilson disease (WD) is characterized by a disrupted copper homeostasis resulting in dramatically increased copper levels, mainly in liver and brain. While copper damage to mitochondria is an established feature in WD livers, much less is known about such detrimental copper effects in other organs. We therefore assessed the mitochondrial sensitivity to copper in a tissue specific manner, namely of isolated rat liver, kidney, heart, and brain mitochondria. Brain mitochondria presented with exceptional copper sensitivity, as evidenced by a comparatively early membrane potential loss, profound structural changes already at low copper dose, and a dose-dependent reduced capacity to produce ATP. This sensitivity was likely due to a copper-dependent attack on free protein thiols and due to a decreased copper reactive defense system, as further evidenced in neuroblastoma SHSY5Y cells. In contrast, an increased production of reactive oxygen species was found to be a late-stage event, only occurring in destroyed mitochondria. We therefore propose mitochondrial protein thiols as major targets of mitochondrial copper toxicity.


Subject(s)
Copper/toxicity , Mitochondria/drug effects , Adenosine Triphosphate/metabolism , Animals , Brain , Cell Line, Tumor , Glutathione/metabolism , Heart , Humans , Kidney , Liver , Microscopy, Electron , Mitochondria/metabolism , Mitochondria/ultrastructure , Rats , Sulfhydryl Compounds/metabolism
8.
J Clin Invest ; 126(7): 2721-35, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27322060

ABSTRACT

In Wilson disease (WD), functional loss of ATPase copper-transporting ß (ATP7B) impairs biliary copper excretion, leading to excessive copper accumulation in the liver and fulminant hepatitis. Current US Food and Drug Administration- and European Medicines Agency-approved pharmacological treatments usually fail to restore copper homeostasis in patients with WD who have progressed to acute liver failure, leaving liver transplantation as the only viable treatment option. Here, we investigated the therapeutic utility of methanobactin (MB), a peptide produced by Methylosinus trichosporium OB3b, which has an exceptionally high affinity for copper. We demonstrated that ATP7B-deficient rats recapitulate WD-associated phenotypes, including hepatic copper accumulation, liver damage, and mitochondrial impairment. Short-term treatment of these rats with MB efficiently reversed mitochondrial impairment and liver damage in the acute stages of liver copper accumulation compared with that seen in untreated ATP7B-deficient rats. This beneficial effect was associated with depletion of copper from hepatocyte mitochondria. Moreover, MB treatment prevented hepatocyte death, subsequent liver failure, and death in the rodent model. These results suggest that MB has potential as a therapeutic agent for the treatment of acute WD.


Subject(s)
Hepatolenticular Degeneration/drug therapy , Imidazoles/pharmacology , Liver Failure, Acute/drug therapy , Oligopeptides/pharmacology , Adenosine Triphosphatases/metabolism , Animals , Bile/chemistry , Cation Transport Proteins/metabolism , Chelating Agents/chemistry , Copper/chemistry , Copper-Transporting ATPases , Disease Models, Animal , Hepatocytes/metabolism , Humans , Liver/drug effects , Mitochondria/drug effects , Phenotype , Rats
9.
Methods Mol Biol ; 1295: 75-86, 2015.
Article in English | MEDLINE | ID: mdl-25820715

ABSTRACT

Mitochondria are key organelles for cellular energy production and cell death decisions. Consequently, a plethora of conditions which are toxic to cells are known to directly attack these organelles. However, mitochondria originating from different tissues differ in their sensitivity to toxic insults. Thus, in order to predict the potential organ-specific toxicity of a given drug or pathological condition at the mitochondrial level, test settings are needed that directly compare the responses and vulnerabilities of mitochondria from different organs. As a prerequisite for such test strategies, we provide here a robust, prompt, and easy-to-follow step-by-step protocol to simultaneously isolate functional and intact mitochondria from rat liver, kidney, heart, and brain. This isolation procedure ensures mitochondrial preparations of comparable purity and reproducible quantities which can be subsequently analyzed for organ-specific mitochondrial toxicity.


Subject(s)
Brain , Cell Fractionation/methods , Kidney , Mitochondria, Liver , Mitochondria, Muscle , Mitochondria , Animals , Brain/metabolism , Kidney/metabolism , Mitochondria/metabolism , Mitochondria, Liver/metabolism , Mitochondria, Muscle/metabolism , Rats
10.
Ann N Y Acad Sci ; 1315: 6-15, 2014 May.
Article in English | MEDLINE | ID: mdl-24517326

ABSTRACT

In Wilson's disease (WD) and related animal models, liver mitochondria are confronted with an increasing copper burden. Physiologically, the mitochondrial matrix may act as a dynamic copper buffer that efficiently distributes the metal to its copper-dependent enzymes. Mitochondria are the first responders in the event of an imbalanced copper homeostasis, as typical changes of their structure are among the earliest observable pathological features in WD. These changes are due to accumulating copper in the mitochondrial membranes and can be reversed by copper-chelating therapies. At the early stage, copper-dependent oxidative stress does not seem to occur. On the contrary, however, when copper is massively deposited in mitochondria, severe structural and respiratory impairments are observed upon disease progression. This provokes reactive oxygen species and consequently causes the mitochondrial membranes to disintegrate, which triggers hepatocyte death. Thus, in WD mitochondria are prime targets for copper, and the excessive copper burden causes their destruction, subsequently provoking tissue failure and death.


Subject(s)
Copper/metabolism , Hepatolenticular Degeneration/metabolism , Mitochondria, Liver/metabolism , Adenosine Triphosphatases/deficiency , Adenosine Triphosphatases/genetics , Animals , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Chelating Agents/therapeutic use , Copper/toxicity , Copper-Transporting ATPases , Disease Models, Animal , Hepatolenticular Degeneration/drug therapy , Hepatolenticular Degeneration/pathology , Homeostasis , Humans , Mice , Mice, Knockout , Mitochondria, Liver/drug effects , Mitochondria, Liver/pathology , Rats , Rats, Inbred LEC , Reactive Oxygen Species/metabolism
11.
Toxicol Sci ; 136(2): 344-58, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24068676

ABSTRACT

The gaseous olefin ethylene (ET) is metabolized in mammals to the carcinogenic epoxide ethylene oxide (EO). Although ET is the largest volume organic chemical worldwide, the EO burden in ET-exposed humans is still uncertain, and only limited data are available on the EO burden in ET-exposed rodents. Therefore, EO was quantified in blood of mice, rats, or 4 volunteers that were exposed once to constant atmospheric ET concentrations of between 1 and 10 000 ppm (rodents) or 5 and 50 ppm (humans). Both the compounds were determined by gas chromatography. At ET concentrations of between 1 and 10 000 ppm, areas under the concentration-time curves of EO in blood (µmol × h/l) ranged from 0.039 to 3.62 in mice and from 0.086 to 11.6 in rats. At ET concentrations ≤ 30 ppm, EO concentrations in blood were 8.7-fold higher in rats and 3.9-fold higher in mice than that in the volunteer with the highest EO burdens. Based on measured EO concentrations, levels of EO adducts to hemoglobin and lymphocyte DNA were calculated for diverse ET concentrations and compared with published adduct levels. For given ET exposure concentrations, there were good agreements between calculated and measured levels of adducts to hemoglobin in rats and humans and to DNA in rats and mice. Reported hemoglobin adduct levels in mice were higher than calculated ones. Furthermore, information is given on species-specific background adduct levels. In summary, the study provides most relevant data for an improved assessment of the human health risk from exposure to ET.


Subject(s)
Ethylene Oxide/blood , Ethylenes/toxicity , Adult , Animals , Ethylenes/pharmacokinetics , Gas Chromatography-Mass Spectrometry , Half-Life , Humans , Male , Mice , Middle Aged , Rats , Rats, Inbred F344
12.
Anal Biochem ; 443(1): 66-74, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-23969012

ABSTRACT

Mitochondrial dysfunctions decisively contribute to the progression of human diseases, implying that functional tests of isolated mitochondria may furnish conclusive information for diagnosis and therapy. Classical mitochondrial isolation methods, however, lack precisely adjustable settings for cell rupture, which is the most critical step in this procedure, and this complicates subsequent analyses. Here, we present an efficient method to isolate functionally active, intact mitochondria from cultured or primary cells and minute tissue samples in a rapid, highly reproducible manner.


Subject(s)
Hepatocytes/ultrastructure , Mitochondria, Liver/ultrastructure , Neurons/ultrastructure , Animals , Automation, Laboratory , Biomarkers/metabolism , Cell Fractionation , Cell Line, Tumor , Hepatocytes/metabolism , Humans , Mice , Mice, Inbred C57BL , Mitochondria, Liver/metabolism , Neurons/metabolism , Primary Cell Culture , Rats , Rats, Inbred BUF , Rats, Inbred WKY
13.
Biochim Biophys Acta ; 1828(9): 2121-33, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23685124

ABSTRACT

The cell-toxic bile salt glycochenodeoxycholic acid (GCDCA) and taurochenodeoxycholic acid (TCDCA) are responsible for hepatocyte demise in cholestatic liver diseases, while tauroursodeoxycholic acid (TUDCA) is regarded hepatoprotective. We demonstrate the direct mitochondrio-toxicity of bile salts which deplete the mitochondrial membrane potential and induce the mitochondrial permeability transition (MPT). The bile salt mediated mechanistic mode of destruction significantly differs from that of calcium, the prototype MPT inducer. Cell-toxic bile salts initially bind to the mitochondrial outer membrane. Subsequently, the structure of the inner boundary membrane disintegrates. And it is only thereafter that the MPT is induced. This progressive destruction occurs in a dose- and time-dependent way. We demonstrate that GCDCA and TCDCA, but not TUDCA, preferentially permeabilize liposomes containing the mitochondrial membrane protein ANT, a process resembling the MPT induction in whole mitochondria. This suggests that ANT is one decisive target for toxic bile salts. To our knowledge this is the first report unraveling the consecutive steps leading to mitochondrial destruction by cell-toxic bile salts.


Subject(s)
Glycochenodeoxycholic Acid/toxicity , Mitochondria, Liver/drug effects , Mitochondrial ADP, ATP Translocases/agonists , Taurochenodeoxycholic Acid/pharmacology , Animals , Cell Membrane Permeability/drug effects , Dose-Response Relationship, Drug , Liposomes/chemistry , Liver/chemistry , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/chemistry , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Mitochondrial ADP, ATP Translocases/isolation & purification , Mitochondrial Membrane Transport Proteins/agonists , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/chemistry , Mitochondrial Membranes/drug effects , Mitochondrial Permeability Transition Pore , Myocardium/chemistry , Rats , Taurochenodeoxycholic Acid/toxicity , Voltage-Dependent Anion Channels/chemistry , Voltage-Dependent Anion Channels/isolation & purification
14.
Toxicology ; 309: 107-16, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23639626

ABSTRACT

Previous quantitative proteomic studies on the actions of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in 5L rat hepatoma cells, a cell model frequently used for investigating the mechanisms of TCDD toxicity, had indicated that dioxin exposure reduced the abundance of numerous proteins which are regulated at the level of protein synthesis initiation. In the present study, we have analysed the mechanism mediating this inhibition. TCDD treatment of the cells largely prevented the activation of eukaryotic translation initiation factor 4E-binding protein 1, a regulator of translation initiation and substrate of the mammalian target of rapamycin (mTOR). By "working upwards" from mTOR, we observed that TCDD inhibited endogenous and IGF-I-induced AKT and ERK activation by interfering with tyrosine phosphorylation of insulin receptor substrate 1. This inhibition was mediated by a TCDD-induced secreted factor which was identified as insulin-like growth factor binding protein 4 (IGFBP-4). The induction of IGFBP-4 protein was dependent on a functional aryl hydrocarbon receptor and was preceded by a rapid increase in the level of IGFBP-4 mRNA indicating that IGFBP-4 is a previously unknown transcriptional target of TCDD in 5L cells. IGFBP-4 was not induced by TCDD in the parental cell line of 5L cells, Fao, and in various closely related rat hepatoma cell lines as well as in other unrelated cell types. Analysis of 5L cell chromosomes by multicolour spectral karyotyping (SKY) revealed that the cells carry several hitherto uncharacterised chromosomal translocations. The observations suggest that in 5L cells the Igfbp-4 gene may have got under the control of a promoter containing dioxin responsive element(s) leading to the induction of IGFBP-4 by TCDD. These findings emphasise a particular caution when interpreting and extrapolating results on the action mechanisms of TCDD obtained in studies using 5L cells as a model system.


Subject(s)
Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor Binding Protein 4/biosynthesis , Liver Neoplasms, Experimental/metabolism , Polychlorinated Dibenzodioxins/toxicity , Animals , Cell Line, Tumor , Dioxins/toxicity , Gene Expression Regulation, Neoplastic/drug effects , Insulin-Like Growth Factor Binding Protein 4/genetics , Liver Neoplasms, Experimental/genetics , Rats
15.
J Clin Invest ; 121(4): 1508-18, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21364284

ABSTRACT

Wilson disease (WD) is a rare hereditary condition that is caused by a genetic defect in the copper-transporting ATPase ATP7B that results in hepatic copper accumulation and lethal liver failure. The present study focuses on the structural mitochondrial alterations that precede clinical symptoms in the livers of rats lacking Atp7b, an animal model for WD. Liver mitochondria from these Atp7b­/­ rats contained enlarged cristae and widened intermembrane spaces, which coincided with a massive mitochondrial accumulation of copper. These changes, however, preceded detectable deficits in oxidative phosphorylation and biochemical signs of oxidative damage, suggesting that the ultrastructural modifications were not the result of oxidative stress imposed by copper- dependent Fenton chemistry. In a cell-free system containing a reducing dithiol agent, isolated mitochondria exposed to copper underwent modifications that were closely related to those observed in vivo. In this cell-free system, copper induced thiol modifications of three abundant mitochondrial membrane proteins, and this correlated with reversible intramitochondrial membrane crosslinking, which was also observed in liver mitochondria from Atp7b­/­ rats. In vivo, copper-chelating agents reversed mitochondrial accumulation of copper, as well as signs of intra-mitochondrial membrane crosslinking, thereby preserving the functional and structural integrity of mitochondria. Together, these findings suggest that the mitochondrion constitutes a pivotal target of copper in WD.


Subject(s)
Hepatolenticular Degeneration/pathology , Mitochondria, Liver/pathology , Adenosine Triphosphatases/deficiency , Adenosine Triphosphatases/genetics , Animals , Cation Transport Proteins , Cell-Free System , Chelating Agents/pharmacology , Copper/metabolism , Copper-Transporting ATPases , Cross-Linking Reagents , Disease Models, Animal , Gene Knockout Techniques , Hepatolenticular Degeneration/genetics , Hepatolenticular Degeneration/metabolism , Humans , In Vitro Techniques , Microscopy, Electron, Transmission , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Mitochondrial Proteins/metabolism , Rats , Sulfhydryl Compounds/metabolism
16.
J Trace Elem Med Biol ; 25(1): 36-41, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21242075

ABSTRACT

Copper is an essential redox-active metal ion which in excess becomes toxic due to the formation of reactive oxygen species. In Wilson disease the elevated copper level in liver leads to chronic oxidative stress and subsequent hepatitis. This study was designed to evaluate the copper chelating efficiency of the bacterial methanobactin (MB) in a rat model for Wilson disease. Methanobactin is a small peptide produced by the methanotrophic bacterium Methylosinus trichosporium OB3b and has an extremely high affinity for copper. Methanobactin treatment of the rats was started at high liver copper and serum aspartate aminotransferase (AST) levels. Two dosing schedules with either 6 or 13 intraperitoneal doses of 200mg methanobactin per kg body weight were applied. Methanobactin treatment led to a return of serum AST values to basal levels and a normalization of liver histopathology. Concomitantly, copper levels declined to 45% and 24% of untreated animals after 6 and 13 doses, respectively. Intravenous application of methanobactin led to a prompt release of copper from liver into bile and the copper was shown to be associated with methanobactin. In vitro experiments with liver cytosol high in copper metallothionein demonstrated that methanobactin removes copper from metallothionein confirming the potent copper chelating activity of methanobactin.


Subject(s)
Chelating Agents/therapeutic use , Disease Models, Animal , Hepatolenticular Degeneration/drug therapy , Imidazoles/therapeutic use , Oligopeptides/therapeutic use , Animals , Chromatography, High Pressure Liquid , Rats , Spectrometry, Mass, Electrospray Ionization , Spectrophotometry, Ultraviolet
17.
Biochemistry ; 48(5): 891-7, 2009 Feb 10.
Article in English | MEDLINE | ID: mdl-19146437

ABSTRACT

Wilson's disease is a human genetic disorder which results in copper accumulation in liver and brain. Treatments such as copper chelation therapy or dietary supplementation with zinc can ameliorate the effects of the disease, but if left untreated, it results in hepatitis, neurological complications, and death. Tetrathiomolybdate (TTM) is a promising new treatment for Wilson's disease which has been demonstrated both in an animal model and in clinical trials. X-ray absorption spectroscopy suggests that TTM acts as a novel copper chelator, forming a complex with accumulated copper in liver. We have used X-ray absorption spectroscopy and X-ray fluorescence imaging to trace the molecular form and distribution of the complex in liver and kidney of an animal model of human Wilson's disease. Our work allows new insights into metabolism of the metal complex in the diseased state.


Subject(s)
Copper/chemistry , Hepatolenticular Degeneration/drug therapy , Molybdenum/administration & dosage , Molybdenum/chemistry , Adenosine Triphosphatases/deficiency , Adenosine Triphosphatases/genetics , Animals , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Chelating Agents/administration & dosage , Chelating Agents/chemistry , Chelating Agents/metabolism , Copper/metabolism , Copper-Transporting ATPases , Disease Models, Animal , Hepatolenticular Degeneration/diagnosis , Hepatolenticular Degeneration/enzymology , Kidney/metabolism , Kidney/pathology , Liver/metabolism , Liver/pathology , Molybdenum/metabolism , Prospective Studies , Rats , Rats, Inbred LEC , Rats, Mutant Strains
18.
Anal Chem ; 80(13): 5051-8, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18510346

ABSTRACT

A pathological increase of the permeability of the mitochondrial membranes may culminate in the irreversible rupture of the mitochondrial outer membrane. Such a permeability transition is lethal because it results in the release of death-inducing molecules from mitochondria and/or metabolic failure. Current methods to assess this outer membrane damage are mostly indirect or scarcely representative of the overall mitochondrial population. Here we present an analytical and preparative approach using free flow electrophoresis to directly distinguish rat liver mitochondria that have undergone the permeability transition from unaffected organelles or from organelles that are damaged to a minor degree. Mitochondrial populations, which considerably differ in outer membrane integrity or cytochrome c content, were separated by this means. We further show that the relative abundance of each population depends on the dose of the permeability transition inducer and the duration of the treatment time. Finally, we have employed this approach to investigate the impairment of mitochondria that were isolated from livers subjected to ischemia/reperfusion damage.


Subject(s)
Electrophoresis/methods , Mitochondria, Liver/chemistry , Mitochondria, Liver/physiology , Mitochondrial Membranes/chemistry , Mitochondrial Membranes/physiology , Animals , Cell Membrane Permeability/physiology , Electrophoresis/instrumentation , Male , Membrane Potential, Mitochondrial/physiology , Mice , Rats , Rats, Sprague-Dawley
19.
Methods Mol Biol ; 424: 333-48, 2008.
Article in English | MEDLINE | ID: mdl-18369873

ABSTRACT

This protocol describes the purification of mitochondria from rat liver with the aid of zone electrophoresis in a free flow device (ZE-FFE). Starting from liver homogenate, cell debris and nuclei are removed by low speed centrifugation. A crude mitochondrial fraction is obtained by medium speed centrifugation and is further purified by washing followed by a Nycodenz gradient centrifugation. Lysosomes and microsomes are located at the upper parts of the gradient, whereas mitochondria are found in the medium part of the gradient. A subsequent purification step with ZE-FFE efficiently removes remaining lysosomes and microsomes and, importantly, damaged mitochondrial structures. The resulting purified mitochondria can be concentrated by centrifugation and used for further experiments. Finally, possible modifications of this protocol with respect to the isolation of pure lysosomes are discussed.


Subject(s)
Electrophoresis/instrumentation , Electrophoresis/methods , Mitochondria, Liver , Animals , Centrifugation, Density Gradient/methods , Mitochondria, Liver/chemistry , Mitochondria, Liver/ultrastructure , Rats
20.
Mol Cell Proteomics ; 7(2): 394-410, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17998243

ABSTRACT

As part of a comprehensive survey of the impact of the environmental pollutant and hepatocarcinogen 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on the proteome of hepatic cells, we have performed a high resolution two-dimensional gel electrophoresis study on the rat hepatoma cell line 5L. 78 protein species corresponding to 73 different proteins were identified as up- or down-regulated following exposure of the cells to 1 nm TCDD for 8 h. There was an overlap of only nine proteins with those detected as altered by TCDD in our recent study using the non-gel-based isotope-coded protein label method (Sarioglu, H., Brandner, S., Jacobsen, C., Meindl, T., Schmidt, A., Kellermann, J., Lottspeich, F., and Andrae, U. (2006) Quantitative analysis of 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced proteome alterations in 5L rat hepatoma cells using isotope-coded protein labels. Proteomics 6, 2407-2421) indicating a strong complementarity of the two approaches. For the majority of the altered proteins, an effect of TCDD on their abundance or posttranslational modifications had not been known before. Several observations suggest that a sizable fraction of the proteins with altered abundance was induced as an adaptive response to TCDD-induced oxidative stress that was demonstrated using the fluorescent probe dihydrorhodamine 123. A prominent group of these proteins comprised various enzymes for which there is evidence that their expression is regulated via the Keap1/Nrf2/antioxidant response element pathway. Other proteins included several involved in the maintenance of mitochondrial energy production and the regulation of the mitochondrial apoptotic pathway. A particularly intriguing finding was the up-regulation of the mitochondrial outer membrane pore protein, voltage-dependent anion channel-selective protein 2 (VDAC2), which was dependent on the presence of a functional aryl hydrocarbon receptor. The regulatability of VDAC2 protein abundance has not been described previously. In view of the recently discovered central role of VDAC2 as an inhibitor of the activation of the proapoptotic protein BAK and the mitochondrial apoptotic pathway, the present data point to a hitherto unrecognized mechanism by which TCDD may affect cellular homeostasis and survival.


Subject(s)
Apoptosis/drug effects , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Mitochondrial Proteins/analysis , Polychlorinated Dibenzodioxins/toxicity , Proteome/analysis , Voltage-Dependent Anion Channel 2/metabolism , Animals , Cell Line, Tumor , Electrophoresis, Gel, Two-Dimensional , Mitochondrial Proteins/chemistry , Neoplasm Proteins/analysis , Neoplasm Proteins/chemistry , Oxidative Stress/drug effects , Proteome/chemistry , Rats , Reactive Oxygen Species/metabolism , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...