Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 31
1.
Traffic ; 25(1): e12920, 2024 01.
Article En | MEDLINE | ID: mdl-37886910

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.


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 En | MEDLINE | ID: mdl-35551511

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.


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
4.
Int J Mol Sci ; 22(11)2021 May 23.
Article En | MEDLINE | ID: mdl-34071094

Three main approaches are used to combat severe viral respiratory infections. The first is preemptive vaccination that blocks infection. Weakened or dead viral particles, as well as genetic constructs carrying viral proteins or information about them, are used as an antigen. However, the viral genome is very evolutionary labile and changes continuously. Second, chemical agents are used during infection and inhibit the function of a number of viral proteins. However, these drugs lose their effectiveness because the virus can rapidly acquire resistance to them. The third is the search for points in the host metabolism the effect on which would suppress the replication of the virus but would not have a significant effect on the metabolism of the host. Here, we consider the possibility of using the copper metabolic system as a target to reduce the severity of influenza infection. This is facilitated by the fact that, in mammals, copper status can be rapidly reduced by silver nanoparticles and restored after their cancellation.


Copper/metabolism , Influenza A virus/physiology , Influenza, Human/metabolism , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Ceruloplasmin/physiology , Copper Transport Proteins/metabolism , Copper-Transporting ATPases/physiology , Drug Resistance, Viral , Host-Pathogen Interactions , Humans , Influenza Vaccines , Influenza, Human/drug therapy , Influenza, Human/prevention & control , Influenza, Human/virology , Mammals/metabolism , Metal Nanoparticles/therapeutic use , Molecular Chaperones/metabolism , PrPC Proteins/physiology , RNA, Viral/physiology , Silver/therapeutic use , Superoxide Dismutase-1/physiology , Viral Proteins/physiology , Virus Replication
6.
Nanotechnol Sci Appl ; 13: 137-157, 2020.
Article En | MEDLINE | ID: mdl-33408467

PURPOSE: The ability of silver nanoparticles (AgNPs) of different sizes to influence copper metabolism in mice is assessed. MATERIALS AND METHODS: AgNPs with diameters of 10, 20, and 75 nm were fabricated through a chemical reduction of silver nitrate and characterized by UV/Vis spectrometry, transmission and scanning electronic microscopy, and laser diffractometry. To test their bioactivity, Escherichia coli cells, cultured A549 cells, and C57Bl/6 mice were used. The antibacterial activity of AgNPs was determined by inhibition of colony-forming ability, and cytotoxicity was tested using the MTT test (viability, %). Ceruloplasmin (Cp, the major mammalian extracellular copper-containing protein) concentration and enzymatic activity were measured using gel-assay analyses and WB, respectively. In vitro binding of AgNPs with serum proteins was monitored with UV/Vis spectroscopy. Metal concentrations were measured using atomic absorption spectrometry. RESULTS: The smallest AgNPs displayed the largest dose- and time-dependent antibacterial activity. All nanoparticles inhibited the metabolic activity of A549 cells in accordance with dose and time, but no correlation between cytotoxicity and nanoparticle size was found. Nanosilver was not uniformly distributed through the body of mice intraperitoneally treated with low AgNP concentrations. It was predominantly accumulated in liver. There, nanosilver was included in ceruloplasmin, and Ag-ceruloplasmin with low oxidase activity level was formed. Larger nanoparticles more effectively interfered with the copper metabolism of mice. Large AgNPs quickly induced a drop of blood serum oxidase activity to practically zero, but after cancellation of AgNP treatment, the activity was rapidly restored. A major fraction of the nanosilver was excreted in the bile with Cp. Nanosilver was bound by alpha-2-macroglobulin in vitro and in vivo, but silver did not substitute for the copper atoms of Cp in vitro. CONCLUSION: The data showed that even at low concentrations, AgNPs influence murine copper metabolism in size-dependent manner. This property negatively correlated with the antibacterial activity of AgNPs.

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

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.

8.
FEBS Lett ; 593(17): 2341-2350, 2019 09.
Article En | MEDLINE | ID: mdl-31408533

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.


Copper-Transporting ATPases/metabolism , Golgi Apparatus/metabolism , Animals , Humans , Protein Transport
9.
Nutrients ; 11(6)2019 Jun 17.
Article En | MEDLINE | ID: mdl-31213024

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.


Copper/metabolism , Ions/metabolism , Silver/metabolism , Biological Transport , Humans , Metal Nanoparticles
10.
Gastroenterology ; 156(4): 1173-1189.e5, 2019 03.
Article En | MEDLINE | ID: mdl-30452922

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.


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
11.
Tissue Cell ; 57: 103-110, 2019 Apr.
Article En | MEDLINE | ID: mdl-30497685

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.


Immunohistochemistry/methods , Microscopy, Electron/methods , Staining and Labeling/methods , Animals , Humans
12.
Cell Mol Gastroenterol Hepatol ; 7(3): 571-596, 2019.
Article En | MEDLINE | ID: mdl-30586623

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.


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
13.
Int J Mol Sci ; 21(1)2019 Dec 26.
Article En | MEDLINE | ID: mdl-31888107

Nephropathic cystinosis is a rare lysosomal storage disorder caused by mutations in CTNS gene leading to Fanconi syndrome. Independent studies reported defective clearance of damaged mitochondria and mitochondrial fragmentation in cystinosis. Proteins involved in the mitochondrial dynamics and the mitochondrial ultrastructure were analyzed in CTNS-/- cells treated with cysteamine, the only drug currently used in the therapy for cystinosis but ineffective to treat Fanconi syndrome. CTNS-/- cells showed an overexpression of parkin associated with deregulation of ubiquitination of mitofusin 2 and fission 1 proteins, an altered proteolytic processing of optic atrophy 1 (OPA1), and a decreased OPA1 oligomerization. According to molecular findings, the analysis of electron microscopy images showed a decrease of mitochondrial cristae number and an increase of cristae lumen and cristae junction width. Cysteamine treatment restored the fission 1 ubiquitination, the mitochondrial size, number and lumen of cristae, but had no effect on cristae junction width, making CTNS-/- tubular cells more susceptible to apoptotic stimuli.


Amino Acid Transport Systems, Neutral/genetics , Cysteamine/pharmacology , Cystinosis/genetics , Mitochondria/metabolism , Cells, Cultured , Cystinosis/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , GTP Phosphohydrolases/metabolism , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Membrane Proteins/metabolism , Mitochondria/drug effects , Mitochondrial Dynamics/drug effects , Mitochondrial Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
14.
Sci Rep ; 8(1): 6247, 2018 04 19.
Article En | MEDLINE | ID: mdl-29674751

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.


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
15.
J Biol Chem ; 292(40): 16605-16615, 2017 10 06.
Article En | MEDLINE | ID: mdl-28860195

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.


Cation Transport Proteins/deficiency , Hypothyroidism , Manganese/metabolism , Thyroxine/biosynthesis , Animals , Cation Transport Proteins/metabolism , Hypothyroidism/genetics , Hypothyroidism/metabolism , Liver/metabolism , Mice , Mice, Knockout
16.
EMBO Mol Med ; 9(5): 605-621, 2017 05.
Article En | MEDLINE | ID: mdl-28283651

TFE3 and TFEB are members of the MiT family of HLH-leucine zipper transcription factors. Recent studies demonstrated that they bind overlapping sets of promoters and are post-transcriptionally regulated through a similar mechanism. However, while Tcfeb knockout (KO) mice die during early embryonic development, no apparent phenotype was reported in Tfe3 KO mice. Thus raising the need to characterize the physiological role of TFE3 and elucidate its relationship with TFEB TFE3 deficiency resulted in altered mitochondrial morphology and function both in vitro and in vivo due to compromised mitochondrial dynamics. In addition, Tfe3 KO mice showed significant abnormalities in energy balance and alterations in systemic glucose and lipid metabolism, resulting in enhanced diet-induced obesity and diabetes. Conversely, viral-mediated TFE3 overexpression improved the metabolic abnormalities induced by high-fat diet (HFD). Both TFEB overexpression in Tfe3 KO mice and TFE3 overexpression in Tcfeb liver-specific KO mice (Tcfeb LiKO) rescued HFD-induced obesity, indicating that TFEB can compensate for TFE3 deficiency and vice versa Analysis of Tcfeb LiKO/Tfe3 double KO mice demonstrated that depletion of both TFE3 and TFEB results in additive effects with an exacerbation of the hepatic phenotype. These data indicate that TFE3 and TFEB play a cooperative, rather than redundant, role in the control of the adaptive response of whole-body metabolism to environmental cues such as diet and physical exercise.


Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Energy Metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Diet, High-Fat/adverse effects , Glucose/metabolism , Lipid Metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Dynamics , Physical Conditioning, Animal , Up-Regulation
17.
Cell Metab ; 25(1): 182-196, 2017 01 10.
Article En | MEDLINE | ID: mdl-28011087

The transcription factor EB (TFEB) is an essential component of lysosomal biogenesis and autophagy for the adaptive response to food deprivation. To address the physiological function of TFEB in skeletal muscle, we have used muscle-specific gain- and loss-of-function approaches. Here, we show that TFEB controls metabolic flexibility in muscle during exercise and that this action is independent of peroxisome proliferator-activated receptor-γ coactivator1α (PGC1α). Indeed, TFEB translocates into the myonuclei during physical activity and regulates glucose uptake and glycogen content by controlling expression of glucose transporters, glycolytic enzymes, and pathways related to glucose homeostasis. In addition, TFEB induces the expression of genes involved in mitochondrial biogenesis, fatty acid oxidation, and oxidative phosphorylation. This coordinated action optimizes mitochondrial substrate utilization, thus enhancing ATP production and exercise capacity. These findings identify TFEB as a critical mediator of the beneficial effects of exercise on metabolism.


Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Metabolism , Physical Conditioning, Animal , Adenylate Kinase/metabolism , Animals , Autophagy/genetics , Cell Nucleus/metabolism , Energy Metabolism/genetics , Genes, Mitochondrial , Genome , Glucose/metabolism , Homeostasis/genetics , Insulin/metabolism , Metabolism/genetics , Mice, Knockout , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Organelle Biogenesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Protein Transport , Signal Transduction/genetics
18.
Metallomics ; 8(9): 853-62, 2016 09 01.
Article En | MEDLINE | ID: mdl-27339113

The lysosomal system operates as a focal point where a number of important physiological processes such as endocytosis, autophagy and nutrient sensing converge. One of the key functions of lysosomes consists of regulating the metabolism/homeostasis of metals. Metal-containing components are carried to the lysosome through incoming membrane flows, while numerous transporters allow metal ions to move across the lysosome membrane. These properties enable lysosomes to direct metal fluxes to the sites where metal ions are either used by cellular components or sequestered. Copper belongs to a group of metals that are essential for the activity of vitally important enzymes, although it is toxic when in excess. Thus, copper uptake, supply and intracellular compartmentalization have to be tightly regulated. An increasing number of publications have indicated that these processes involve lysosomes. Here we review studies that reveal the expanding role of the lysosomal system as a hub for the control of Cu homeostasis and for the regulation of key Cu-dependent processes in health and disease.


Copper-Transporting ATPases/metabolism , Copper/metabolism , Homeostasis/physiology , Lysosomes/metabolism , Animals , Humans
19.
Hepatology ; 63(6): 1842-59, 2016 06.
Article En | MEDLINE | ID: mdl-26660341

UNLABELLED: Wilson disease (WD) is an autosomal recessive disorder that is caused by the toxic accumulation of copper (Cu) in the liver. The ATP7B gene, which is mutated in WD, encodes a multitransmembrane domain adenosine triphosphatase that traffics from the trans-Golgi network to the canalicular area of hepatocytes, where it facilitates excretion of excess Cu into the bile. Several ATP7B mutations, including H1069Q and R778L that are two of the most frequent variants, result in protein products, which, although still functional, remain in the endoplasmic reticulum. Thus, they fail to reach Cu excretion sites, resulting in the toxic buildup of Cu in the liver of WD patients. Therefore, correcting the location of these mutants by leading them to the appropriate functional sites in the cell should restore Cu excretion and would be beneficial to help large cohorts of WD patients. However, molecular targets for correction of endoplasmic reticulum-retained ATP7B mutants remain elusive. Here, we show that expression of the most frequent ATP7B mutant, H1069Q, activates p38 and c-Jun N-terminal kinase signaling pathways, which favor the rapid degradation of the mutant. Suppression of these pathways with RNA interference or specific chemical inhibitors results in the substantial rescue of ATP7B(H1069Q) (as well as that of several other WD-causing mutants) from the endoplasmic reticulum to the trans-Golgi network compartment, in recovery of its Cu-dependent trafficking, and in reduction of intracellular Cu levels. CONCLUSION: Our findings indicate p38 and c-Jun N-terminal kinase as intriguing targets for correction of WD-causing mutants and, hence, as potential candidates, which could be evaluated for the development of novel therapeutic strategies to combat WD. (Hepatology 2016;63:1842-1859).


Adenosine Triphosphatases/genetics , Cation Transport Proteins/genetics , Hepatolenticular Degeneration/genetics , MAP Kinase Signaling System , Copper/metabolism , Copper-Transporting ATPases , HeLa Cells , Hep G2 Cells , Hepatolenticular Degeneration/metabolism , Humans , Liver/metabolism , Mutation , Secretory Pathway
20.
Hum Mol Genet ; 24(23): 6811-25, 2015 Dec 01.
Article En | MEDLINE | ID: mdl-26420842

Stargardt disease (STGD1) due to mutations in the large ABCA4 gene is the most common inherited macular degeneration in humans. We have shown that dual adeno-associated viral (AAV) vectors effectively transfer ABCA4 to the retina of Abca4-/- mice. However, they express both lower levels of transgene compared with a single AAV and truncated proteins. To increase productive dual AAV concatemerization, which would overcome these limitations, we have explored the use of either various regions of homology or heterologous inverted terminal repeats (ITR). In addition, we tested the ability of various degradation signals to decrease the expression of truncated proteins. We found the highest levels of transgene expression using regions of homology based on either alkaline phosphatase or the F1 phage (AK). The use of heterologous ITR does not decrease the levels of truncated proteins relative to full-length ABCA4 and impairs AAV vector production. Conversely, the inclusion of the CL1 degradation signal results in the selective degradation of truncated proteins from the 5'-half without affecting full-length protein production. Therefore, we developed dual AAV hybrid ABCA4 vectors including homologous ITR2, the photoreceptor-specific G protein-coupled receptor kinase 1 promoter, the AK region of homology and the CL1 degradation signal. We show that upon subretinal administration these vectors are both safe in pigs and effective in Abca4-/- mice. Our data support the use of improved dual AAV vectors for gene therapy of STGD1.


ATP-Binding Cassette Transporters/genetics , Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors , Macular Degeneration/congenital , Administration, Ophthalmic , Animals , Disease Models, Animal , Female , Genetic Vectors/administration & dosage , HEK293 Cells , Humans , Macular Degeneration/genetics , Macular Degeneration/therapy , Mice , Retina/metabolism , Stargardt Disease , Swine , Terminal Repeat Sequences , Transgenes
...