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










Publication year range
1.
Autophagy ; 18(4): 711-725, 2022 04.
Article in English | MEDLINE | ID: mdl-34554889

ABSTRACT

Getting to know Erwin Knecht is not so simple. This view was summarized in a comment from Dr. Marta Martinez Vicente, who obtained her PhD degree working in a lab that shared space with Erwin's group: "Erwin Knecht is a complex character, who awakens contradictory feelings. To define him I would say that he is a mixture of intelligence and madness, he's witty, very funny but also grumpy and cranky, all mixed and all simultaneously. Without a doubt he is a person who will not leave anyone indifferent, his original personality marked all who crossed his path, doctoral students, collaborators, people who attended his talks, etc … I remember the weekly lab meetings with him; we, the students, had a lot of respect for him (not to mention fear), but his comments were always smart, helpful and constructive, he has always been prone to helping everyone. In the lab, he was extremely demanding, but got people under his supervision to do their best. And despite his usual moodiness, he managed to make everyone love him and have a special affection for him. Above all, I remember his screams throughout the laboratory that could be heard from all over the building, calling out to his laboratory technician: 'Asunción!' They were like an old couple, arguing all day but they couldn't be without each other, it was like watching a sitcom every day." If you are intrigued, please read on.


Subject(s)
Autophagy , Intelligence , Humans , Male
2.
Biochim Biophys Acta Mol Cell Res ; 1867(2): 118613, 2020 02.
Article in English | MEDLINE | ID: mdl-31758957

ABSTRACT

Lafora progressive myoclonus epilepsy is a fatal rare neurodegenerative disorder characterized by the accumulation of insoluble abnormal glycogen deposits in the brain and peripheral tissues. Mutations in at least two genes are responsible for the disease: EPM2A, encoding the glucan phosphatase laforin, and EPM2B, encoding the RING-type E3-ubiquitin ligase malin. Both laforin and malin form a functional complex in which laforin recruits the substrates to be ubiquitinated by malin. We and others have described that, in cellular and animal models of this disease, there is an autophagy impairment which leads to the accumulation of dysfunctional mitochondria. In addition, we established that the autophagic defect occurred at the initial steps of autophagosome formation. In this work, we present evidence that in cellular models of the disease there is a decrease in the amount of phosphatidylinositol-3P. This is probably due to defective regulation of the autophagic PI3KC3 complex, in the absence of a functional laforin/malin complex. In fact, we demonstrate that the laforin/malin complex interacts physically and co-localizes intracellularly with core components of the PI3KC3 complex (Beclin1, Vps34 and Vps15), and that this interaction is specific and results in the polyubiquitination of these proteins. In addition, the laforin/malin complex is also able to polyubiquitinate ATG14L and UVRAG. Finally, we show that overexpression of the laforin/malin complex increases PI3KC3 activity. All these results suggest a new role of the laforin/malin complex in the activation of autophagy via regulation of the PI3KC3 complex and explain the defect in autophagy described in Lafora disease.


Subject(s)
Lafora Disease/pathology , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Autophagy , Autophagy-Related Proteins/metabolism , Beclin-1/chemistry , Beclin-1/metabolism , Cells, Cultured , Humans , Lafora Disease/metabolism , Microscopy, Fluorescence , Protein Binding , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Transcription Factors/chemistry , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitination
3.
Mol Neurobiol ; 57(3): 1607-1621, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31808062

ABSTRACT

Lafora disease (LD) is a rare, fatal form of progressive myoclonus epilepsy. The molecular basis of this devastating disease is still poorly understood, and no treatment is available yet, which leads to the death of the patients around 10 years from the onset of the first symptoms. The hallmark of LD is the accumulation of insoluble glycogen-like inclusions in the brain and peripheral tissues, as a consequence of altered glycogen homeostasis. In addition, other determinants in the pathophysiology of LD have been suggested, such as proteostasis impairment, with reduction in autophagy, and oxidative stress, among others. In order to gain a general view of the genes involved in the pathophysiology of LD, in this work, we have performed RNA-Seq transcriptome analyses of whole-brain tissue from two independent mouse models of the disease, namely Epm2a-/- and Epm2b-/- mice, at different times of age. Our results provide strong evidence for three major facts: first, in both models of LD, we found a common set of upregulated genes, most of them encoding mediators of inflammatory response; second, there was a progression with the age in the appearance of these inflammatory markers, starting at 3 months of age; and third, reactive glia was responsible for the expression of these inflammatory genes. These results clearly indicate that neuroinflammation is one of the most important traits to be considered in order to fully understand the pathophysiology of LD, and define reactive glia as novel therapeutic targets in the disease.


Subject(s)
Age Factors , Lafora Disease/metabolism , Myoclonic Epilepsies, Progressive/metabolism , Neuroglia/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Animals , Disease Models, Animal , Disease Progression , Glycogen/metabolism , Inclusion Bodies/metabolism , Lafora Disease/genetics , Mice, Knockout , Myoclonic Epilepsies, Progressive/genetics , Oxidative Stress/physiology
5.
Cells ; 7(8)2018 Jul 26.
Article in English | MEDLINE | ID: mdl-30050012

ABSTRACT

Lafora disease (LD, OMIM254780) is a rare and fatal form of progressive myoclonus epilepsy (PME). Among PMEs, LD is unique because of the rapid neurological deterioration of the patients and the appearance in brain and peripheral tissues of insoluble glycogen-like (polyglucosan) inclusions, named Lafora bodies (LBs). LD is caused by mutations in the EPM2A gene, encoding the dual phosphatase laforin, or the EPM2B gene, encoding the E3-ubiquitin ligase malin. Laforin and malin form a functional complex that is involved in the regulation of glycogen synthesis. Thus, in the absence of a functional complex glycogen accumulates in LBs. In addition, it has been suggested that the laforin-malin complex participates in alternative physiological pathways, such as intracellular protein degradation, oxidative stress, and the endoplasmic reticulum unfolded protein response. In this work we review the possible cellular functions of laforin and malin with a special focus on their role in the ubiquitination of specific substrates. We also discuss here the pathological consequences of defects in laforin or malin functions, as well as the therapeutic strategies that are being explored for LD.

6.
FEBS J ; 285(11): 2071-2090, 2018 06.
Article in English | MEDLINE | ID: mdl-29645350

ABSTRACT

Lafora disease (LD) is a fatal neurodegenerative disorder caused mostly by mutations in either of two genes encoding laforin and malin. LD is characterized by accumulation of a poorly branched form of glycogen in the cytoplasm of neurons and other cells. We previously reported dysfunctional mitochondria in different LD models. Now, using mitochondrial uncouplers and respiratory chain inhibitors, we have investigated with human fibroblasts a possible alteration in the selective degradation of damaged mitochondria (mitophagy) in LD. By flow cytometry of MitoTracker-labelled cells and measuring the levels of various mitochondrial proteins by western blot, we found in LD fibroblasts a partial impairment in the increased mitochondrial degradation produced by these treatments. In addition, colocalization of mitochondrial and lysosomal markers decreased in LD fibroblasts. All these results are consistent with a partial impairment in the induced autophagic degradation of dysfunctional mitochondria in LD fibroblasts. However, canonical recruitment of Parkin to mitochondria under these conditions remained unaffected in LD fibroblasts, and also in SH-SY5Y cells after malin and laforin overexpression. Neither mitochondrial localization nor protein levels of Bcl-2-like protein 13, another component of the mitophagic machinery that operates under these conditions, were affected in LD fibroblasts. In contrast, although these treatments raised autophagy in both control and LD fibroblasts, this enhanced autophagy was clearly lower in the latter cells. Therefore, the autophagic degradation of altered mitochondria is impaired in LD, which is due to a partial defect in the autophagic response and not in the canonical mitophagy signalling pathways.


Subject(s)
Autophagy/genetics , Lafora Disease/genetics , Mitochondria/genetics , Mitophagy/genetics , Carrier Proteins/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Flow Cytometry , Gene Expression Regulation/genetics , Humans , Lafora Disease/metabolism , Lafora Disease/pathology , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Ubiquitin-Protein Ligases
7.
Neurogenetics ; 18(4): 245-250, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28975462

ABSTRACT

In two siblings, who suffer from an early childhood-onset axonal polyneuropathy with exclusive involvement of motor fibers, the c.629T>C (p.F210S) mutation was identified in the X-linked AIFM1 gene, which encodes for the apoptosis-inducing factor (AIF). The mutation was predicted as deleterious, according to in silico analysis. A decreased expression of the AIF protein, altered cellular morphology, and a fragmented mitochondrial network were observed in the proband's fibroblasts. This new form of motor neuropathy expands the phenotypic spectrum of AIFM1 mutations and therefore, the AIFM1 gene should be considered in the diagnosis of hereditary motor neuropathies.


Subject(s)
Apoptosis Inducing Factor/genetics , Muscular Atrophy, Spinal/genetics , Mutation/genetics , Female , Genes, X-Linked/genetics , Humans , Infant , Male , Muscular Atrophy, Spinal/diagnosis , Pedigree , Phenotype , Proteins/genetics
8.
Ann Transplant ; 22: 285-295, 2017 May 09.
Article in English | MEDLINE | ID: mdl-28484204

ABSTRACT

BACKGROUND Cardioplegic arrest is a common procedure for many types of cardiac surgery, and different formulations have been proposed to enhance its cardio-protective effect. Hydrogen sulfide is an important signaling molecule that has cardio-protective properties. We therefore studied the cardio-protective effect of hydrogen sulfide in cardiac cell culture and its potential therapeutic use in combination with cardioplegia formulations. MATERIAL AND METHODS We added hydrogen sulfide donor GYY4137 to HL-1 cells to study its protective effect in nutrient starved conditions. In addition, we tested the potential use of GYY4137 when it is added into two different cardioplegia formulations: Cardi-Braun® solution and del Nido solution in an ex vivo Langendorff perfused rat hearts model. RESULTS We observed that eight-hour pre-treatment with GYY4137 significantly suppressed apoptosis in nutrient-starved HL-1 cells (28% less compared to untreated cells; p<0.05), maintained ATP content, and reduced protein synthesis. In ex vivo experiments, Cardi-Braun® and del Nido cardioplegia solutions supplemented with GYY4137 significantly reduced the pro-apoptotic protein caspase-3 content and preserved ATP content. Furthermore, GYY4137 supplemented cardioplegia solutions decreased the S-(5-adenosyl)-L-methionine/S-(adenosyl)-L-homocysteine ratio, reducing the oxidative stress in cardiac tissue. Finally, heart beating analysis revealed the preservation of the inter-beat interval and the heart rate in del Nido cardioplegia solution supplemented with GYY4137. CONCLUSIONS GYY4137 preconditioning preserved energetic state during starved conditions, attenuating the cardiomyocytes apoptosis in vitro. The addition of GYY4137 to cardioplegia solutions prevented apoptosis, ATP consumption, and oxidative stress in perfused rat hearts, restoring its electrophysiological status after cardiac arrest. These findings suggested that GYY4137 sulfide donor may improve the cardioplegia solution performance during cardiac surgery.


Subject(s)
Apoptosis/drug effects , Heart Arrest/metabolism , Heart/drug effects , Hydrogen Sulfide/pharmacology , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Adenosine Triphosphate/metabolism , Animals , Cardioplegic Solutions , Caspase 3/metabolism , Cell Line , Cells, Cultured , Male , Myocytes, Cardiac/metabolism , Rats , Rats, Wistar
9.
Acta Neuropathol ; 133(2): 283-301, 2017 02.
Article in English | MEDLINE | ID: mdl-28004277

ABSTRACT

The activation of the highly conserved unfolded protein response (UPR) is prominent in the pathogenesis of the most prevalent neurodegenerative disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD) and amyotrophic lateral sclerosis (ALS), which are classically characterized by an accumulation of aggregated or misfolded proteins. This activation is orchestrated by three endoplasmic reticulum (ER) stress sensors: PERK, ATF6 and IRE1. These sensors transduce signals that induce the expression of the UPR gene programme. Here, we first identified an early activator of the UPR and investigated the role of a chronically activated UPR in the pathogenesis of X-linked adrenoleukodystrophy (X-ALD), a neurometabolic disorder that is caused by ABCD1 malfunction; ABCD1 transports very long-chain fatty acids (VLCFA) into peroxisomes. The disease manifests as inflammatory demyelination in the brain or and/or degeneration of corticospinal tracts, thereby resulting in spastic paraplegia, with the accumulation of intracellular VLCFA instead of protein aggregates. Using X-ALD mouse model (Abcd1 - and Abcd1 - /Abcd2 -/- mice) and X-ALD patient's fibroblasts and brain samples, we discovered an early engagement of the UPR. The response was characterized by the activation of the PERK and ATF6 pathways, but not the IRE1 pathway, showing a difference from the models of AD, PD or ALS. Inhibition of PERK leads to the disruption of homeostasis and increased apoptosis during ER stress induced in X-ALD fibroblasts. Redox imbalance appears to be the mechanism that initiates ER stress in X-ALD. Most importantly, we demonstrated that the bile acid tauroursodeoxycholate (TUDCA) abolishes UPR activation, which results in improvement of axonal degeneration and its associated locomotor impairment in Abcd1 - /Abcd2 -/- mice. Altogether, our preclinical data provide evidence for establishing the UPR as a key drug target in the pathogenesis cascade. Our study also highlights the potential role of TUDCA as a treatment for X-ALD and other axonopathies in which similar molecular mediators are implicated.


Subject(s)
Adrenoleukodystrophy/physiopathology , Axons/drug effects , Nerve Degeneration/physiopathology , Taurochenodeoxycholic Acid/pharmacology , Unfolded Protein Response/physiology , Animals , Axons/pathology , Humans , Mice , Mice, Knockout
10.
Front Mol Biosci ; 3: 81, 2016.
Article in English | MEDLINE | ID: mdl-28018906

ABSTRACT

Distal hereditary motor neuropathies (dHMN) are a group of rare hereditary neuromuscular disorders characterized by an atrophy that affects peroneal muscles in the absence of sensory symptoms. To date, 23 genes are thought to be responsible for dHMN, four of which encode chaperones: DNAJB2, which encodes a member of the HSP40/DNAJ co-chaperone family; and HSPB1, HSPB3, and HSPB8, encoding three members of the small heat shock protein family. While around 30 different mutations in HSPB1 have been identified, the remaining three genes are altered in many fewer cases. Indeed, a mutation of HSPB3 has only been described in one case, whereas a few cases have been reported carrying mutations in DNAJB2 and HSPB8, most of them caused by a founder c.352+1G>A mutation in DNAJB2 and by mutations affecting the K141 residue in the HSPB8 chaperone. Hence, their rare occurrence makes it difficult to understand the pathological mechanisms driven by such mutations in this neuropathy. Chaperones can assemble into multi-chaperone complexes that form an integrated chaperone network within the cell. Such complexes fulfill relevant roles in a variety of processes, such as the correct folding of newly synthesized proteins, in which chaperones escort them to precise cellular locations, and as a response to protein misfolding, which includes the degradation of proteins that fail to refold properly. Despite this range of functions, mutations in some of these chaperones lead to diseases with a similar clinical profile, suggesting common pathways. This review provides an overview of the genetics of those dHMNs that share a common disease mechanism and that are caused by mutations in four genes encoding chaperones: DNAJB2, HSPB1, HSPB3, and HSPB8.

11.
Methods Mol Biol ; 1449: 299-311, 2016.
Article in English | MEDLINE | ID: mdl-27613045

ABSTRACT

Lysosomes participate within the cells in the degradation of organelles, macromolecules, and a wide variety of substrates. In any study on specific roles of lysosomes, both under physiological and pathological conditions, it is advisable to include methods that allow their reproducible and reliable isolation. However, purification of lysosomes is a difficult task, particularly in the case of cultured cells. This is mainly because of the heterogeneity of these organelles, along with their low number and high fragility. Also, isolation methods, while disrupting plasma membranes, have to preserve the integrity of lysosomes, as the breakdown of their membranes releases enzymes that could damage all cell organelles, including themselves. The protocols described below have been routinely used in our laboratory for the specific isolation of lysosomes from rat liver, NIH/3T3, and other cultured cells, but can be adapted to other mammalian tissues or cell lines.


Subject(s)
Cell Fractionation/methods , Organelles/metabolism , Animals , Cell Line , Cell Membrane , Cells, Cultured , Centrifugation, Density Gradient/methods , Liver/cytology , Lysosomes , Mice , Rats , Subcellular Fractions/metabolism
12.
Proteomics ; 16(1): 150-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26507101

ABSTRACT

Niemann-Pick type C (NPC) disease is an inherited lysosomal storage disorder, characterized by severe neurodegeneration. It is mostly produced by mutations in the NPC1 gene, encoding for a protein of the late endosomes/lysosomes membrane, involved in cholesterol metabolism. However, the specific role of this protein in NPC disease still remains unknown. We aimed to identify Npc1-binding proteins in order to define new putative NPC1 lysosomal functions. By affinity chromatography using an Npc1 peptide (amino acids 1032-1066 of loop I), as bait, we fished 31 lysosomal proteins subsequently identified by LC-MS/MS. Most of them were involved in proteolysis and lipid catabolism and included the protease cathepsin D. Cathepsin D and NPC1 interaction was validated by immunoprecipitation and the functional relevance of this interaction was studied. We found that fibroblasts from NPC patients with low levels of NPC1 protein have high amounts of procathepsin D but reduced quantities of the mature protein, thus showing a diminished cathepsin D activity. The increase of NPC1 protein levels in NPC cells by treatment with the proteasome inhibitor bortezomib, induced an elevation of cathepsin D activity. All these results suggest a new lysosomal function of NPC1 as a regulator of cathepsin D processing and activity.


Subject(s)
Carrier Proteins/metabolism , Cathepsin D/metabolism , Enzyme Precursors/metabolism , Membrane Glycoproteins/metabolism , Niemann-Pick Diseases/metabolism , Proteins/metabolism , Amino Acid Sequence , Carrier Proteins/analysis , Cathepsin D/analysis , Cell Line , Chromatography, Liquid , Enzyme Precursors/analysis , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/analysis , Molecular Sequence Data , Niemann-Pick C1 Protein , Protein Interaction Maps , Proteins/analysis , Tandem Mass Spectrometry
13.
PLoS One ; 10(12): e0144273, 2015.
Article in English | MEDLINE | ID: mdl-26642043

ABSTRACT

GTPBP3 is an evolutionary conserved protein presumably involved in mitochondrial tRNA (mt-tRNA) modification. In humans, GTPBP3 mutations cause hypertrophic cardiomyopathy with lactic acidosis, and have been associated with a defect in mitochondrial translation, yet the pathomechanism remains unclear. Here we use a GTPBP3 stable-silencing model (shGTPBP3 cells) for a further characterization of the phenotype conferred by the GTPBP3 defect. We experimentally show for the first time that GTPBP3 depletion is associated with an mt-tRNA hypomodification status, as mt-tRNAs from shGTPBP3 cells were more sensitive to digestion by angiogenin than tRNAs from control cells. Despite the effect of stable silencing of GTPBP3 on global mitochondrial translation being rather mild, the steady-state levels and activity of Complex I, and cellular ATP levels were 50% of those found in the controls. Notably, the ATPase activity of Complex V increased by about 40% in GTPBP3 depleted cells suggesting that mitochondria consume ATP to maintain the membrane potential. Moreover, shGTPBP3 cells exhibited enhanced antioxidant capacity and a nearly 2-fold increase in the uncoupling protein UCP2 levels. Our data indicate that stable silencing of GTPBP3 triggers an AMPK-dependent retrograde signaling pathway that down-regulates the expression of the NDUFAF3 and NDUFAF4 Complex I assembly factors and the mitochondrial pyruvate carrier (MPC), while up-regulating the expression of UCP2. We also found that genes involved in glycolysis and oxidation of fatty acids are up-regulated. These data are compatible with a model in which high UCP2 levels, together with a reduction in pyruvate transport due to the down-regulation of MPC, promote a shift from pyruvate to fatty acid oxidation, and to an uncoupling of glycolysis and oxidative phosphorylation. These metabolic alterations, and the low ATP levels, may negatively affect heart function.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Anion Transport Proteins/metabolism , GTP-Binding Proteins/genetics , Ion Channels/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Adenosine Triphosphate/metabolism , Anion Transport Proteins/genetics , Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/metabolism , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Escherichia coli/genetics , Fatty Acids/genetics , Fatty Acids/metabolism , GTP-Binding Proteins/metabolism , Gene Expression Regulation , Glycolysis/genetics , HEK293 Cells , Humans , Ion Channels/genetics , Mitochondria/genetics , Mitochondrial Membrane Transport Proteins , Mitochondrial Proteins/genetics , Monocarboxylic Acid Transporters , Oxidative Phosphorylation , RNA, Transfer, Lys/metabolism , Ribonuclease, Pancreatic/chemistry , Ribonuclease, Pancreatic/metabolism , Uncoupling Protein 2
14.
Free Radic Biol Med ; 88(Pt A): 30-41, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25680286

ABSTRACT

Lafora disease (LD; OMIM 254780, ORPHA501) is a devastating neurodegenerative disorder characterized by the presence of glycogen-like intracellular inclusions called Lafora bodies and caused, in most cases, by mutations in either the EPM2A or the EPM2B gene, encoding respectively laforin, a phosphatase with dual specificity that is involved in the dephosphorylation of glycogen, and malin, an E3-ubiquitin ligase involved in the polyubiquitination of proteins related to glycogen metabolism. Thus, it has been reported that laforin and malin form a functional complex that acts as a key regulator of glycogen metabolism and that also plays a crucial role in protein homeostasis (proteostasis). Regarding this last function, it has been shown that cells are more sensitive to ER stress and show defects in proteasome and autophagy activities in the absence of a functional laforin-malin complex. More recently, we have demonstrated that oxidative stress accompanies these proteostasis defects and that various LD models show an increase in reactive oxygen species and oxidative stress products together with a dysregulated antioxidant enzyme expression and activity. In this review we discuss possible connections between the multiple defects in protein homeostasis present in LD and oxidative stress.


Subject(s)
Lafora Disease/etiology , Oxidative Stress , Proteostasis Deficiencies/metabolism , Antioxidants/metabolism , Carrier Proteins/genetics , Glycogen/metabolism , Humans , Lafora Disease/metabolism , Lafora Disease/physiopathology , Mutation , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Ubiquitin-Protein Ligases
15.
Acta Neuropathol ; 129(3): 399-415, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25549970

ABSTRACT

X-linked adrenoleukodystrophy (X-ALD) is a rare neurometabolic disease characterized by the accumulation of very long chain fatty acids (VLCFAs) due to a loss of function of the peroxisomal transporter ABCD1. Here, using in vivo and in vitro models, we demonstrate that autophagic flux was impaired due to elevated mammalian target of rapamycin (mTOR) signaling, which contributed to X-ALD pathogenesis. We also show that excess VLCFAs downregulated autophagy in human fibroblasts. Furthermore, mTOR inhibition by a rapamycin derivative (temsirolimus) restored autophagic flux and inhibited the axonal degenerative process as well as the associated locomotor impairment in the Abcd1 (-) /Abcd2 (-/-) mouse model. This process was mediated through the restoration of proteasome function and redox as well as metabolic homeostasis. These findings provide the first evidence that links impaired autophagy to X-ALD, which may yield a therapy based on autophagy activators for adrenomyeloneuropathy patients.


Subject(s)
Adrenoleukodystrophy/pathology , Adrenoleukodystrophy/physiopathology , Autophagy/physiology , Nerve Degeneration/physiopathology , Adult , Animals , Blotting, Western , Cells, Cultured , Disease Models, Animal , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Middle Aged , Nerve Degeneration/pathology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism
16.
Mol Neurobiol ; 51(3): 932-46, 2015.
Article in English | MEDLINE | ID: mdl-24838580

ABSTRACT

Lafora disease (LD, OMIM 254780, ORPHA501) is a fatal neurodegenerative disorder characterized by the presence of glycogen-like intracellular inclusions called Lafora bodies and caused, in the vast majority of cases, by mutations in either EPM2A or EPM2B genes, encoding respectively laforin and malin. In the last years, several reports have revealed molecular details of these two proteins and have identified several processes affected in LD, but the pathophysiology of the disease still remains largely unknown. Since autophagy impairment has been reported as a characteristic treat in both Lafora disease cell and animal models, and as there is a link between autophagy and mitochondrial performance, we sought to determine if mitochondrial function could be altered in those models. Using fibroblasts from LD patients, deficient in laforin or malin, we found mitochondrial alterations, oxidative stress and a deficiency in antioxidant enzymes involved in the detoxification of reactive oxygen species (ROS). Similar results were obtained in brain tissue samples from transgenic mice deficient in either the EPM2A or EPM2B genes. Furthermore, in a proteomic analysis of brain tissue obtained from Epm2b-/- mice, we observed an increase in a modified form of peroxiredoxin-6, an antioxidant enzyme involved in other neurological pathologies, thus corroborating an alteration of the redox condition. These data support that oxidative stress produced by an increase in ROS production and an impairment of the antioxidant enzyme response to this stress play an important role in development of LD.


Subject(s)
Antioxidants/metabolism , Lafora Disease/genetics , Oxidative Stress/genetics , Reactive Oxygen Species/metabolism , Animals , Autophagy/genetics , Humans , Lafora Disease/metabolism , Lafora Disease/pathology , Mice , Mice, Knockout , Mice, Transgenic , Mutation/genetics , Proteomics
17.
PLoS One ; 9(2): e87898, 2014.
Article in English | MEDLINE | ID: mdl-24498393

ABSTRACT

The function of CERKL (CERamide Kinase Like), a causative gene of retinitis pigmentosa and cone-rod dystrophy, still awaits characterization. To approach its cellular role we have investigated the subcellular localization and interaction partners of the full length CERKL isoform, CERKLa of 532 amino acids, in different cell lines, including a photoreceptor-derived cell line. We demonstrate that CERKLa is a main component of compact and untranslated mRNPs and that associates with other RNP complexes such as stress granules, P-bodies and polysomes. CERKLa is a protein that binds through its N-terminus to mRNAs and interacts with other mRNA-binding proteins like eIF3B, PABP, HSP70 and RPS3. Except for eIF3B, these interactions depend on the integrity of mRNAs but not of ribosomes. Interestingly, the C125W CERKLa pathological mutant does not interact with eIF3B and is absent from these complexes. Compact mRNPs containing CERKLa also associate with microtubules and are found in neurites of neural differentiated cells. These localizations had not been reported previously for any member of the retinal disorders gene family and should be considered when investigating the pathogenic mechanisms and therapeutical approaches in these diseases.


Subject(s)
Carrier Proteins/metabolism , Microtubules/metabolism , Neuroblastoma/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , RNA, Messenger/metabolism , Retina/metabolism , Ribonucleoproteins/metabolism , Alternative Splicing , Blotting, Western , Carrier Proteins/genetics , Cells, Cultured , Chromatography, Liquid , Electrophoretic Mobility Shift Assay , Fluorescent Antibody Technique , Humans , Immunoprecipitation , In Situ Hybridization, Fluorescence , Microscopy, Confocal , Microtubules/pathology , Neuroblastoma/pathology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Polyribosomes/metabolism , Protein Isoforms , RNA, Messenger/genetics , Retina/pathology , Tandem Mass Spectrometry , Tumor Cells, Cultured
18.
Brain ; 137(Pt 3): 806-18, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24430976

ABSTRACT

Lafora progressive myoclonus epilepsy (Lafora disease) is a fatal autosomal recessive neurodegenerative disorder characterized by the presence of glycogen-like intracellular inclusions called Lafora bodies. The vast majority of patients carry mutations in either the EPM2A or EPM2B genes, encoding laforin, a glucan phosphatase, and malin, an E3 ubiquitin ligase, respectively. Although the precise physiological role of these proteins is not fully understood, work in past years has established a link between glycogen synthesis, Lafora bodies formation and Lafora disease development. To determine the role of the phosphatase activity of laforin in disease development we generated two Epm2a(-/-) mouse lines expressing either wild-type laforin or a mutant (C265S) laforin lacking only the phosphatase activity. Our results demonstrate that expression of either transgene blocks formation of Lafora bodies and restores the impairment in macroautophagy, preventing the development of Lafora bodies in Epm2a(-/-) mice. These data indicate that the critical pathogenic process is the control of abnormal glycogen accumulation through intracellular proteolytic systems by the laforin-malin complex, and not glycogen dephosphorylation by laforin. Understanding which is the essential process leading to Lafora disease pathogenesis represents a critical conceptual advance that should facilitate development of appropriate therapeutics.


Subject(s)
Dual-Specificity Phosphatases/deficiency , Dual-Specificity Phosphatases/metabolism , Lafora Disease/metabolism , Animals , Autophagy/genetics , Disease Models, Animal , Dual-Specificity Phosphatases/genetics , Female , Lafora Disease/enzymology , Lafora Disease/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutation , Phosphorylation/genetics , Protein Tyrosine Phosphatases, Non-Receptor
19.
Free Radic Biol Med ; 75 Suppl 1: S47, 2014 Oct.
Article in English | MEDLINE | ID: mdl-26461389

ABSTRACT

Lafora Disease (LD, OMIM 254780, ORPHA501) is a fatal neurodegenerative disorder characterized by the presence of glycogen-like intracellular inclusions called Lafora bodies and caused, in the vast majority of cases, by mutations in either EPM2A or EPM2B genes, encoding respectively laforin and malin. In the last years, several reports have revealed molecular details of these two proteins and have identified several processes affected in LD, but the pathophysiology of the disease still remains largely unknown. Since autophagy impairment has been reported as a characteristic treat in both Lafora disease cell and animal models, and as there is a link between autophagy and mitochondrial performance, we sought to determine if mitochondrial function could be altered in those models. Using fibroblasts from LD patients, deficient in laforin or malin, we found mitochondrial alterations, oxidative stress and a deficiency in antioxidant enzymes involved in the detoxification of reactive oxygen species (ROS). Similar results were obtained in brain tissue samples from transgenic mice deficient in either the EPM2A or EPM2B genes. Furthermore, in a proteomic analysis of brain tissue obtained from Epm2b-/- mice, we observed an increase in a modified form of peroxirredoxin-6, an antioxidant enzyme involved in other neurological pathologies, thus corroborating an alteration of the redox condition. These data support that oxidative stress produced by an increase in ROS production and an impairment of the antioxidant enzyme response to this stress play an important role in development of LD.

20.
PLoS One ; 8(12): e83318, 2013.
Article in English | MEDLINE | ID: mdl-24349488

ABSTRACT

Two major mechanisms of intracellular protein degradation, autophagy and the ubiquitin-proteasome pathway, operate in mammalian cells. PTEN, which is frequently mutated in glioblastomas, is a tumor suppressor gene that encodes a dual specificity phosphatase that antagonizes the phosphatidylinositol 3-kinase class I/AKT/mTOR pathway, which is a key regulator of autophagy. Here, we investigated in U87MG human glioma cells the role of PTEN in the regulation of autophagy and the ubiquitin-proteasome pathway, because both are functionally linked and are relevant in cancer progression. Since U87MG glioma cells lack a functional PTEN, we used stable clones that express, under the control of a tetracycline-inducible system (Tet-on), wild-type PTEN and two of its mutants, G129E-PTEN and C124S-PTEN, which, respectively, lack the lipid phosphatase activity only and both the lipid and the protein phosphatase activities of this protein. Expression of PTEN in U87MG glioma cells decreased proteasome activity and also reduced protein ubiquitination. On the contrary, expression of PTEN increased the autophagic flux and the lysosomal mass. Interestingly, and although PTEN negatively regulates the phosphatidylinositol 3-kinase class I/AKT/mTOR signaling pathway by its lipid phosphatase activity, both effects in U87MG cells were independent of this activity. These results suggest a new mTOR-independent signaling pathway by which PTEN can regulate in opposite directions the main mechanisms of intracellular protein degradation.


Subject(s)
Autophagy , Glioma/metabolism , PTEN Phosphohydrolase/metabolism , Phosphatidate Phosphatase/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin/metabolism , Cell Line, Tumor , Glioma/genetics , Glioma/pathology , Humans , Lysosomes/genetics , Lysosomes/metabolism , PTEN Phosphohydrolase/genetics , Phosphatidate Phosphatase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proteasome Endopeptidase Complex/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Ubiquitin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...