Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Traffic ; 25(1): e12920, 2024 01.
Article in English | MEDLINE | ID: mdl-37886910

ABSTRACT

Wilson disease (WD) is caused by mutations in the ATP7B gene that encodes a copper (Cu) transporting ATPase whose trafficking from the Golgi to endo-lysosomal compartments drives sequestration of excess Cu and its further excretion from hepatocytes into the bile. Loss of ATP7B function leads to toxic Cu overload in the liver and subsequently in the brain, causing fatal hepatic and neurological abnormalities. The limitations of existing WD therapies call for the development of new therapeutic approaches, which require an amenable animal model system for screening and validation of drugs and molecular targets. To achieve this objective, we generated a mutant Caenorhabditis elegans strain with a substitution of a conserved histidine (H828Q) in the ATP7B ortholog cua-1 corresponding to the most common ATP7B variant (H1069Q) that causes WD. cua-1 mutant animals exhibited very poor resistance to Cu compared to the wild-type strain. This manifested in a strong delay in larval development, a shorter lifespan, impaired motility, oxidative stress pathway activation, and mitochondrial damage. In addition, morphological analysis revealed several neuronal abnormalities in cua-1 mutant animals exposed to Cu. Further investigation suggested that mutant CUA-1 is retained and degraded in the endoplasmic reticulum, similarly to human ATP7B-H1069Q. As a consequence, the mutant protein does not allow animals to counteract Cu toxicity. Notably, pharmacological correctors of ATP7B-H1069Q reduced Cu toxicity in cua-1 mutants indicating that similar pathogenic molecular pathways might be activated by the H/Q substitution and, therefore, targeted for rescue of ATP7B/CUA-1 function. Taken together, our findings suggest that the newly generated cua-1 mutant strain represents an excellent model for Cu toxicity studies in WD.


Subject(s)
Hepatolenticular Degeneration , Animals , Humans , Hepatolenticular Degeneration/genetics , Hepatolenticular Degeneration/drug therapy , Hepatolenticular Degeneration/metabolism , Copper/toxicity , Copper/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , Hepatocytes/metabolism
2.
Nature ; 606(7915): 761-768, 2022 06.
Article in English | MEDLINE | ID: mdl-35551511

ABSTRACT

SARS-CoV-2, like other coronaviruses, builds a membrane-bound replication organelle to enable RNA replication1. The SARS-CoV-2 replication organelle is composed of double-membrane vesicles (DMVs) that are tethered to the endoplasmic reticulum (ER) by thin membrane connectors2, but the viral proteins and the host factors involved remain unknown. Here we identify the viral non-structural proteins (NSPs) that generate the SARS-CoV-2 replication organelle. NSP3 and NSP4 generate the DMVs, whereas NSP6, through oligomerization and an amphipathic helix, zippers ER membranes and establishes the connectors. The NSP6(ΔSGF) mutant, which arose independently in the Alpha, Beta, Gamma, Eta, Iota and Lambda variants of SARS-CoV-2, behaves as a gain-of-function mutant with a higher ER-zippering activity. We identified three main roles for NSP6: first, to act as a filter in communication between the replication organelle and the ER, by allowing lipid flow but restricting the access of ER luminal proteins to the DMVs; second, to position and organize DMV clusters; and third, to mediate contact with lipid droplets (LDs) through the LD-tethering complex DFCP1-RAB18. NSP6 thus acts as an organizer of DMV clusters and can provide a selective means of refurbishing them with LD-derived lipids. Notably, both properly formed NSP6 connectors and LDs are required for the replication of SARS-CoV-2. Our findings provide insight into the biological activity of NSP6 of SARS-CoV-2 and of other coronaviruses, and have the potential to fuel the search for broad antiviral agents.


Subject(s)
Coronavirus Nucleocapsid Proteins , SARS-CoV-2 , Viral Nonstructural Proteins , Virus Replication , COVID-19/virology , Carrier Proteins , Cell Line , Coronavirus Nucleocapsid Proteins/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Humans , Lipid Droplets , SARS-CoV-2/genetics , SARS-CoV-2/growth & development , Viral Nonstructural Proteins/metabolism , rab GTP-Binding Proteins
3.
Cells ; 11(2)2022 01 10.
Article in English | MEDLINE | ID: mdl-35053335

ABSTRACT

ATP7B is a hepato-specific Golgi-located ATPase, which plays a key role in the regulation of copper (Cu) homeostasis and signaling. In response to elevated Cu levels, ATP7B traffics from the Golgi to endo-lysosomal structures, where it sequesters excess copper and further promotes its excretion to the bile at the apical surface of hepatocytes. In addition to liver, high ATP7B expression has been reported in tumors with elevated resistance to platinum (Pt)-based chemotherapy. Chemoresistance to Pt drugs represents the current major obstacle for the treatment of large cohorts of cancer patients. Although the mechanisms underlying Pt-tolerance are still ambiguous, accumulating evidence suggests that lysosomal sequestration of Pt drugs by ion transporters (including ATP7B) might significantly contribute to drug resistance development. In this context, signaling mechanisms regulating the expression of transporters such as ATP7B are of great importance. Considering this notion, we investigated whether ATP7B expression in Pt-resistant cells might be driven by transcription factor EB (TFEB), a master regulator of lysosomal gene transcription. Using resistant ovarian cancer IGROV-CP20 cells, we found that TFEB directly binds to the predicted coordinated lysosomal expression and regulation (CLEAR) sites in the proximal promoter and first intron region of ATP7B upon Pt exposure. This binding accelerates transcription of luciferase reporters containing ATP7B CLEAR regions, while suppression of TFEB inhibits ATP7B expression and stimulates cisplatin toxicity in resistant cells. Thus, these data have uncovered a Pt-dependent transcriptional mechanism that contributes to cancer chemoresistance and might be further explored for therapeutic purposes.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Copper-Transporting ATPases/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/genetics , Platinum/pharmacology , Base Sequence , Cell Line, Tumor , Copper-Transporting ATPases/metabolism , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Platinum/toxicity , Transcription, Genetic/drug effects
5.
JPGN Rep ; 2(3): e112, 2021 Aug.
Article in English | MEDLINE | ID: mdl-37205945

ABSTRACT

Wilson disease (WD) is a rare autosomal recessive disorder of copper metabolism typically presenting after 3 years of age. We describe a girl presenting with neonatal cholestasis rapidly progressing to end-stage liver disease. She presented hepatosplenomegaly, neurological impairment, Coombs-negative hemolytic anemia, central hypothyroidism. A patient-parents whole exome sequencing identified a homozygous state for ATP7B mutations causing WD in the proband (p.Gln7fs/p.His1069Gln) and her mother (p.His1069Gln/p.His1069Gln), who was then confirmed to have cirrhotic WD. A causative role of copper toxicity due to ATP7B loss of function was indicated by the presence of extrahepatic features of WD, consistent tests of copper metabolism-including a 7-fold increase in liver copper-and similarity of patient's liver gene expression profile and ultrastructure with that of WD models. This exceptionally early presentation could result from the combination of the ATP7B impairment and the intrauterine copper overload due to maternal undiagnosed WD.

6.
J Neurosci ; 41(2): 215-233, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33208468

ABSTRACT

Rare genetic diseases preponderantly affect the nervous system causing neurodegeneration to neurodevelopmental disorders. This is the case for both Menkes and Wilson disease, arising from mutations in ATP7A and ATP7B, respectively. The ATP7A and ATP7B proteins localize to the Golgi and regulate copper homeostasis. We demonstrate genetic and biochemical interactions between ATP7 paralogs with the conserved oligomeric Golgi (COG) complex, a Golgi apparatus vesicular tether. Disruption of Drosophila copper homeostasis by ATP7 tissue-specific transgenic expression caused alterations in epidermis, aminergic, sensory, and motor neurons. Prominent among neuronal phenotypes was a decreased mitochondrial content at synapses, a phenotype that paralleled with alterations of synaptic morphology, transmission, and plasticity. These neuronal and synaptic phenotypes caused by transgenic expression of ATP7 were rescued by downregulation of COG complex subunits. We conclude that the integrity of Golgi-dependent copper homeostasis mechanisms, requiring ATP7 and COG, are necessary to maintain mitochondria functional integrity and localization to synapses.SIGNIFICANCE STATEMENT Menkes and Wilson disease affect copper homeostasis and characteristically afflict the nervous system. However, their molecular neuropathology mechanisms remain mostly unexplored. We demonstrate that copper homeostasis in neurons is maintained by two factors that localize to the Golgi apparatus, ATP7 and the conserved oligomeric Golgi (COG) complex. Disruption of these mechanisms affect mitochondrial function and localization to synapses as well as neurotransmission and synaptic plasticity. These findings suggest communication between the Golgi apparatus and mitochondria through homeostatically controlled cellular copper levels and copper-dependent enzymatic activities in both organelles.


Subject(s)
Copper/physiology , Golgi Apparatus/physiology , Homeostasis/physiology , Organelle Biogenesis , Synapses/physiology , Adenosine Triphosphatases/metabolism , Animals , Animals, Genetically Modified , Cell Line , Copper/toxicity , Copper-Transporting ATPases/genetics , Drosophila , Electric Stimulation , Extracellular Space/metabolism , Female , Humans , Male , RNA, Small Interfering , Synapses/ultrastructure
7.
Proc Natl Acad Sci U S A ; 117(51): 32453-32463, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33288711

ABSTRACT

Pathogenic mutations in the copper transporter ATP7B have been hypothesized to affect its protein interaction landscape contributing to loss of function and, thereby, to hepatic copper toxicosis in Wilson disease. Although targeting mutant interactomes was proposed as a therapeutic strategy, druggable interactors for rescue of ATP7B mutants remain elusive. Using proteomics, we found that the frequent H1069Q substitution promotes ATP7B interaction with HSP70, thus accelerating endoplasmic reticulum (ER) degradation of the mutant protein and consequent copper accumulation in hepatic cells. This prompted us to use an HSP70 inhibitor as bait in a bioinformatics search for structurally similar Food and Drug Administration-approved drugs. Among the hits, domperidone emerged as an effective corrector that recovered trafficking and function of ATP7B-H1069Q by impairing its exposure to the HSP70 proteostatic network. Our findings suggest that HSP70-mediated degradation can be safely targeted with domperidone to rescue ER-retained ATP7B mutants and, hence, to counter the onset of Wilson disease.


Subject(s)
Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , Domperidone/pharmacology , HSP70 Heat-Shock Proteins/metabolism , Hepatolenticular Degeneration/genetics , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Cells, Cultured , Copper/metabolism , Domperidone/chemistry , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Hep G2 Cells , Hepatocytes/metabolism , Hepatolenticular Degeneration/drug therapy , Hepatolenticular Degeneration/metabolism , Hepatolenticular Degeneration/pathology , Humans , Mutation, Missense , Nipecotic Acids/chemistry , Nipecotic Acids/pharmacology , Protein Transport/drug effects , Protein Transport/genetics , Proteomics/methods
9.
Cancers (Basel) ; 12(3)2020 Mar 06.
Article in English | MEDLINE | ID: mdl-32155756

ABSTRACT

Tumor resistance to chemotherapy represents an important challenge in modern oncology. Although platinum (Pt)-based drugs have demonstrated excellent therapeutic potential, their effectiveness in a wide range of tumors is limited by the development of resistance mechanisms. One of these mechanisms includes increased cisplatin sequestration/efflux by the copper-transporting ATPase, ATP7B. However, targeting ATP7B to reduce Pt tolerance in tumors could represent a serious risk because suppression of ATP7B might compromise copper homeostasis, as happens in Wilson disease. To circumvent ATP7B-mediated Pt tolerance we employed a high-throughput screen (HTS) of an FDA/EMA-approved drug library to detect safe therapeutic molecules that promote cisplatin toxicity in the IGROV-CP20 ovarian carcinoma cells, whose resistance significantly relies on ATP7B. Using a synthetic lethality approach, we identified and validated three hits (Tranilast, Telmisartan, and Amphotericin B) that reduced cisplatin resistance. All three drugs induced Pt-mediated DNA damage and inhibited either expression or trafficking of ATP7B in a tumor-specific manner. Global transcriptome analyses showed that Tranilast and Amphotericin B affect expression of genes operating in several pathways that confer tolerance to cisplatin. In the case of Tranilast, these comprised key Pt-transporting proteins, including ATOX1, whose suppression affected ability of ATP7B to traffic in response to cisplatin. In summary, our findings reveal Tranilast, Telmisartan, and Amphotericin B as effective drugs that selectively promote cisplatin toxicity in Pt-resistant ovarian cancer cells and underscore the efficiency of HTS strategy for identification of biosafe compounds, which might be rapidly repurposed to overcome resistance of tumors to Pt-based chemotherapy.

10.
Dent J (Basel) ; 7(4)2019 Nov 17.
Article in English | MEDLINE | ID: mdl-31744265

ABSTRACT

This controlled in vitro study compared the effects of varying the thickness of a TiO2 layer on cellular activity using commercially available miniscrew samples with identical surface features to derive information with direct clinical impact. Titanium grade V plates with four different thicknesses of TiO2 layer/color were used: absent/gray (Control group), 40-50 nm/pink (Pink group), 130 nm/gold (Gold group) and 140 nm/rosé (Rosé group). In vitro experiments used Saos-2 cells and included cell growth analysis, phospho-Histone H3 and procollagen I staining, cell viability analysis, and a cell migration assay at 12, 24, 40 and to 48 h. Few differences were seen among the groups, with no clear behavior of cellular activity according to the TiO2 thickness. The Control group showed a greater cell count. Phospho-Histone H3 staining was similar among the groups and procollagen I staining was greater in the Rosé group. Cell viability analysis showed a significant difference for live cell counts (greater in the Rosé group) and no difference for the dead cell counts. The cell migration assay showed a delay for the Rosé group up to 40 h, where full repopulation of cell-free areas was obtained at 48 h. The results suggest that the TiO2 layers of the commercial miniscrews have minimal biological effects, including cytotoxicity, with possibly negligible or minimal clinical implications.

11.
Cells ; 8(9)2019 09 13.
Article in English | MEDLINE | ID: mdl-31540259

ABSTRACT

Membrane trafficking pathways emanating from the Golgi regulate a wide range of cellular processes. One of these is the maintenance of copper (Cu) homeostasis operated by the Golgi-localized Cu-transporting ATPases ATP7A and ATP7B. At the Golgi, these proteins supply Cu to newly synthesized enzymes which use this metal as a cofactor to catalyze a number of vitally important biochemical reactions. However, in response to elevated Cu, the Golgi exports ATP7A/B to post-Golgi sites where they promote sequestration and efflux of excess Cu to limit its potential toxicity. Growing tumors actively consume Cu and employ ATP7A/B to regulate the availability of this metal for oncogenic enzymes such as LOX and LOX-like proteins, which confer higher invasiveness to malignant cells. Furthermore, ATP7A/B activity and trafficking allow tumor cells to detoxify platinum (Pt)-based drugs (like cisplatin), which are used for the chemotherapy of different solid tumors. Despite these noted activities of ATP7A/B that favor oncogenic processes, the mechanisms that regulate the expression and trafficking of Cu ATPases in malignant cells are far from being completely understood. This review summarizes current data on the role of ATP7A/B in the regulation of Cu and Pt metabolism in malignant cells and outlines questions and challenges that should be addressed to understand how ATP7A and ATP7B trafficking mechanisms might be targeted to counteract tumor development.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Carcinogenesis/metabolism , Copper-Transporting ATPases/physiology , Copper/metabolism , Platinum/metabolism , Animals , Biological Transport , Cisplatin/pharmacokinetics , Drug Resistance, Neoplasm , Golgi Apparatus , Humans , Mice
12.
FEBS Lett ; 593(17): 2341-2350, 2019 09.
Article in English | MEDLINE | ID: mdl-31408533

ABSTRACT

Recent studies highlight the continued growth in the identification of a variety of cellular functions that involve the Golgi apparatus. Apart from well-known membrane sorting/trafficking and glycosylation machineries, the Golgi harbors molecular platforms operating in intracellular signaling, cytoskeleton organization, and protein quality control mechanisms. One of new emerging Golgi functions consists in the regulation of copper homeostasis by coordinating the relocation and activity of copper transporters. Of these, the Cu-transporting ATPase ATP7B (known as Wilson disease protein) plays a key role in the maintenance of the Cu balance in the body via the supply of essential Cu to the systemic circulation and via elimination of excess Cu into the bile. These activities require tightly regulated shuttling of ATP7B between the Golgi and different post-Golgi compartments. Despite significant progress over recent years, a number of issues regarding ATP7B trafficking remain to be clarified. This review summarizes current views on ATP7B trafficking pathways from and to the Golgi and underscores the challenges that should be addressed to define the ATP7B trafficking routes and mechanisms in health and disease.


Subject(s)
Copper-Transporting ATPases/metabolism , Golgi Apparatus/metabolism , Animals , Humans , Protein Transport
13.
Nutrients ; 11(6)2019 Jun 17.
Article in English | MEDLINE | ID: mdl-31213024

ABSTRACT

In humans, copper is an important micronutrient because it is a cofactor of ubiquitous and brain-specific cuproenzymes, as well as a secondary messenger. Failure of the mechanisms supporting copper balance leads to the development of neurodegenerative, oncological, and other severe disorders, whose treatment requires a detailed understanding of copper metabolism. In the body, bioavailable copper exists in two stable oxidation states, Cu(I) and Cu(II), both of which are highly toxic. The toxicity of copper ions is usually overcome by coordinating them with a wide range of ligands. These include the active cuproenzyme centers, copper-binding protein motifs to ensure the safe delivery of copper to its physiological location, and participants in the Cu(I) ↔ Cu(II) redox cycle, in which cellular copper is stored. The use of modern experimental approaches has allowed the overall picture of copper turnover in the cells and the organism to be clarified. However, many aspects of this process remain poorly understood. Some of them can be found out using abiogenic silver ions (Ag(I)), which are isoelectronic to Cu(I). This review covers the physicochemical principles of the ability of Ag(I) to substitute for copper ions in transport proteins and cuproenzyme active sites, the effectiveness of using Ag(I) to study copper routes in the cells and the body, and the limitations associated with Ag(I) remaining stable in only one oxidation state. The use of Ag(I) to restrict copper transport to tumors and the consequences of large-scale use of silver nanoparticles for human health are also discussed.


Subject(s)
Copper/metabolism , Ions/metabolism , Silver/metabolism , Biological Transport , Humans , Metal Nanoparticles
14.
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
15.
Tissue Cell ; 57: 103-110, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30497685

ABSTRACT

Labeling of specific molecules for electron microscopy provides an extremely powerful tool to investigate subcellular distribution of proteins, lipids and sugars with resolution of a few nanometers. Moreover, this technology offers a unique opportunity to see how the pattern of these molecules changes in different physiological or pathological conditions. Pre-embedding labeling evolved as one of the easiest method for molecule detection in electron microscopy. It was named "pre-embedding" because the procedure involves a labeling step before the embedding of the specimen in the resin, its subsequent sectioning and analysis of sections under the electron microscope. Here we review different strategies and technical tips of the pre-embedding procedure, the potential of this method for detection and quantification of molecular components at the ultrastructural level, and the integration of the pre-embedding approach with rapidly developing light and electron microscopy technologies.


Subject(s)
Immunohistochemistry/methods , Microscopy, Electron/methods , Staining and Labeling/methods , Animals , Humans
16.
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
17.
Sci Rep ; 8(1): 6247, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29674751

ABSTRACT

H1069Q substitution represents the most frequent mutation of the copper transporter ATP7B causing Wilson disease in Caucasian population. ATP7B localizes to the Golgi complex in hepatocytes but moves in response to copper overload to the endo-lysosomal compartment to support copper excretion via bile canaliculi. In heterologous or hepatoma-derived cell lines, overexpressed ATP7B-H1069Q is strongly retained in the ER and fails to move to the post-Golgi sites, resulting in toxic copper accumulation. However, this pathogenic mechanism has never been tested in patients' hepatocytes, while animal models recapitulating this form of WD are still lacking. To reach this goal, we have reprogrammed skin fibroblasts of homozygous ATP7B-H1069Q patients into induced pluripotent stem cells and differentiated them into hepatocyte-like cells. Surprisingly, in HLCs we found one third of ATP7B-H1069Q localized in the Golgi complex and able to move to the endo-lysosomal compartment upon copper stimulation. However, despite normal mRNA levels, the expression of the mutant protein was only 20% compared to the control because of endoplasmic reticulum-associated degradation. These results pinpoint rapid degradation as the major cause for loss of ATP7B function in H1069Q patients, and thus as the primary target for designing therapeutic strategies to rescue ATP7B-H1069Q function.


Subject(s)
Copper-Transporting ATPases/genetics , Hepatocytes/pathology , Hepatolenticular Degeneration/genetics , Induced Pluripotent Stem Cells/cytology , Polymorphism, Single Nucleotide , Cell Line , Cellular Reprogramming , Copper/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum-Associated Degradation , Fibroblasts/cytology , Golgi Apparatus/metabolism , Hepatolenticular Degeneration/pathology , Humans , Mutant Proteins/metabolism , Skin/cytology
18.
J Cell Sci ; 130(24): 4155-4167, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29093023

ABSTRACT

Many secretory cells increase the synthesis and secretion of cargo proteins in response to specific stimuli. How cells couple increased cargo load with a coordinate rise in secretory capacity to ensure efficient transport is not well understood. We used thyroid cells stimulated with thyrotropin (TSH) to demonstrate a coordinate increase in the production of thyroid-specific cargo proteins and ER-Golgi transport factors, and a parallel expansion of the Golgi complex. TSH also increased expression of the CREB3L1 transcription factor, which alone caused amplified transport factor levels and Golgi enlargement. Furthermore, CREB3L1 potentiated the TSH-induced increase in Golgi volume. A dominant-negative CREB3L1 construct hampered the ability of TSH to induce Golgi expansion, implying that this transcription factor contributes to Golgi expansion. Our findings support a model in which CREB3L1 acts as a downstream effector of TSH to regulate the expression of cargo proteins, and simultaneously increases the synthesis of transport factors and the expansion of the Golgi to synchronize the rise in cargo load with the amplified capacity of the secretory pathway.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Golgi Apparatus/genetics , Nerve Tissue Proteins/genetics , Thyroid Gland/metabolism , Thyrotropin/genetics , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Gene Expression Regulation/genetics , Golgi Apparatus/metabolism , Humans , Nerve Tissue Proteins/metabolism , Secretory Pathway/genetics , Thyrotropin/metabolism
19.
J Biol Chem ; 292(40): 16605-16615, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28860195

ABSTRACT

SLC30A10 and SLC39A14 are manganese efflux and influx transporters, respectively. Loss-of-function mutations in genes encoding either transporter induce hereditary manganese toxicity. Patients have elevated manganese in the blood and brain and develop neurotoxicity. Liver manganese is increased in patients lacking SLC30A10 but not SLC39A14. These organ-specific changes in manganese were recently recapitulated in knockout mice. Surprisingly, Slc30a10 knockouts also had elevated thyroid manganese and developed hypothyroidism. To determine the mechanisms of manganese-induced hypothyroidism and understand how SLC30A10 and SLC39A14 cooperatively mediate manganese detoxification, here we produced Slc39a14 single and Slc30a10/Slc39a14 double knockout mice and compared their phenotypes with that of Slc30a10 single knockouts. Compared with wild-type controls, Slc39a14 single and Slc30a10/Slc39a14 double knockouts had higher manganese levels in the blood and brain but not in the liver. In contrast, Slc30a10 single knockouts had elevated manganese levels in the liver as well as in the blood and brain. Furthermore, SLC30A10 and SLC39A14 localized to the canalicular and basolateral domains of polarized hepatic cells, respectively. Thus, transport activities of both SLC39A14 and SLC30A10 are required for hepatic manganese excretion. Compared with Slc30a10 single knockouts, Slc39a14 single and Slc30a10/Slc39a14 double knockouts had lower thyroid manganese levels and normal thyroid function. Moreover, intrathyroid thyroxine levels of Slc30a10 single knockouts were lower than those of controls. Thus, the hypothyroidism phenotype of Slc30a10 single knockouts is induced by elevated thyroid manganese, which blocks thyroxine production. These findings provide new insights into the mechanisms of manganese detoxification and manganese-induced thyroid dysfunction.


Subject(s)
Cation Transport Proteins/deficiency , Hypothyroidism , Manganese/metabolism , Thyroxine/biosynthesis , Animals , Cation Transport Proteins/metabolism , Hypothyroidism/genetics , Hypothyroidism/metabolism , Liver/metabolism , Mice , Mice, Knockout
20.
Nat Commun ; 7: 12148, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27401954

ABSTRACT

Membrane fission is an essential cellular process by which continuous membranes split into separate parts. We have previously identified CtBP1-S/BARS (BARS) as a key component of a protein complex that is required for fission of several endomembranes, including basolateral post-Golgi transport carriers. Assembly of this complex occurs at the Golgi apparatus, where BARS binds to the phosphoinositide kinase PI4KIIIß through a 14-3-3γ dimer, as well as to ARF and the PKD and PAK kinases. We now report that, when incorporated into this complex, BARS binds to and activates a trans-Golgi lysophosphatidic acid (LPA) acyltransferase type δ (LPAATδ) that converts LPA into phosphatidic acid (PA); and that this reaction is essential for fission of the carriers. LPA and PA have unique biophysical properties, and their interconversion might facilitate the fission process either directly or indirectly (via recruitment of proteins that bind to PA, including BARS itself).


Subject(s)
Acyltransferases/metabolism , Alcohol Oxidoreductases/metabolism , Cytokinesis/physiology , DNA-Binding Proteins/metabolism , Golgi Apparatus/metabolism , Intracellular Membranes/metabolism , 14-3-3 Proteins/metabolism , HeLa Cells , Humans , Lysophospholipids/metabolism , Phosphatidic Acids/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Kinase C/metabolism , p21-Activated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...