Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
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
2.
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
3.
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
5.
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
6.
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
7.
Hum Mol Genet ; 21(7): 1521-33, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22186026

ABSTRACT

Lafora disease (LD), a fatal neurodegenerative disorder characterized by the presence of intracellular inclusions called Lafora bodies (LBs), is caused by loss-of-function mutations in laforin or malin. Previous studies suggested a role of these proteins in the regulation of glycogen biosynthesis, in glycogen dephosphorylation and in the modulation of the intracellular proteolytic systems. However, the contribution of each of these processes to LD pathogenesis is unclear. We have generated a malin-deficient (Epm2b-/-) mouse with a phenotype similar to that of LD patients. By 3-6 months of age, Epm2b-/- mice present neurological and behavioral abnormalities that correlate with a massive presence of LBs in the cortex, hippocampus and cerebellum. Sixteen-day-old Epm2b-/- mice, without detectable LBs, show an impairment of macroautophagy (hereafter called autophagy), which remains compromised in adult animals. These data demonstrate similarities between the Epm2a-/- and Epm2b-/- mice that provide further insights into LD pathogenesis. They illustrate that the dysfunction of autophagy is a consequence of the lack of laforin-malin complexes and a common feature of both mouse models of LD. Because this dysfunction precedes other pathological manifestations, we propose that decreased autophagy plays a primary role in the formation of LBs and it is critical in LD pathogenesis.


Subject(s)
Autophagy , Lafora Disease/pathology , Ubiquitin-Protein Ligases/genetics , Animals , Behavior, Animal , Brain/metabolism , Brain/pathology , Dual-Specificity Phosphatases/analysis , Dual-Specificity Phosphatases/metabolism , Glucans/chemistry , Lafora Disease/genetics , Lafora Disease/physiopathology , Mice , Mice, Knockout , Motor Skills Disorders/genetics , Myocardium/ultrastructure , Protein Tyrosine Phosphatases, Non-Receptor , Ubiquitin/analysis , Ubiquitin-Protein Ligases/deficiency
8.
J Cell Sci ; 125(Pt 1): 92-107, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22266906

ABSTRACT

Macroautophagy is a major lysosomal catabolic process activated particularly under starvation in eukaryotic cells. A new organelle, the autophagosome, engulfs cytoplasmic substrates, which are degraded after fusion with endosomes and/or lysosomes. During a shotgun proteome analysis of purified lysosomal membranes from mouse fibroblasts, a Ca(2+)-dependent phospholipid-binding protein, annexin A5, was found to increase on lysosomal membranes under starvation. This suggests a role for this protein, an abundant annexin with a still unknown intracellular function, in starvation-induced lysosomal degradation. Transient overexpression and silencing experiments showed that annexin A5 increased lysosomal protein degradation, and colocalisation experiments, based on GFP sensitivity to lysosomal acidic pH, indicated that this was mainly the result of inducing autophagosome-lysosome fusion. Annexin A5 also inhibited the endocytosis of a fluid-phase marker and cholera toxin, but not receptor-mediated endocytosis. Therefore, we propose a double and opposite role of annexin A5 in regulating the endocytic and autophagic pathways and the fusion of autophagosomes with lysosomes and endosomes.


Subject(s)
Annexin A5/metabolism , Autophagy , Endocytosis , Animals , Annexin A5/deficiency , Annexin A5/genetics , Calcium Signaling , Cholera Toxin/metabolism , Electrophoresis, Gel, Two-Dimensional , Endosomes/metabolism , Fibroblasts , Food Deprivation , Golgi Apparatus/metabolism , HEK293 Cells , Humans , Intracellular Membranes/metabolism , Lysosomes/metabolism , Membrane Fusion , Mice , NIH 3T3 Cells , Phagosomes/metabolism , Protein Transport , Proteolysis , Proteomics
9.
Brain ; 136(Pt 3): 891-904, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23436506

ABSTRACT

Oxidative damage is a pivotal aetiopathogenic factor in X-linked adrenoleukodystrophy. This is a neurometabolic disease characterized by the accumulation of very-long-chain fatty acids owing to the loss of function of the peroxisomal transporter Abcd1. Here, we used the X-linked adrenoleukodystrophy mouse model and patient's fibroblasts to detect malfunctioning of the ubiquitin-proteasome system resulting from the accumulation of oxidatively modified proteins, some involved in bioenergetic metabolism. Furthermore, the immunoproteasome machinery appears upregulated in response to oxidative stress, in the absence of overt inflammation. i-Proteasomes are recruited to mitochondria when fibroblasts are exposed to an excess of very-long-chain fatty acids in response to oxidative stress. Antioxidant treatment regulates proteasome expression, prevents i-proteasome induction and translocation of i-proteasomes to mitochondria. Our findings support a key role of i-proteasomes in quality control in mitochondria during oxidative damage in X-linked adrenoleukodystrophy, and perhaps in other neurodegenerative conditions with similar pathogeneses.


Subject(s)
Adrenoleukodystrophy/metabolism , Oxidative Stress/physiology , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Adrenoleukodystrophy/immunology , Adrenoleukodystrophy/physiopathology , Animals , Disease Models, Animal , Fibroblasts/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteasome Endopeptidase Complex/immunology , Reverse Transcriptase Polymerase Chain Reaction , Ubiquitin/immunology
10.
Biochem J ; 449(2): 497-506, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23116132

ABSTRACT

Autophagy is a natural process of 'self-eating' that occurs within cells and can be either pro-survival or can cause cell death. As a pro-survival mechanism, autophagy obtains energy by recycling cellular components such as macromolecules or organelles. In response to nutrient deprivation, e.g. depletion of amino acids or serum, autophagy is induced and most of these signals converge on the kinase mTOR (mammalian target of rapamycin). It is commonly accepted that glucose inhibits autophagy, since its deprivation from cells cultured in full medium induces autophagy by a mechanism involving AMPK (AMP-activated protein kinase), mTOR and Ulk1. However, we show in the present study that under starvation conditions addition of glucose produces the opposite effect. Specifically, the results of the present study demonstrate that the presence of glucose induces an increase in the levels of LC3 (microtubule-associated protein 1 light chain)-II, in the number and volume density of autophagic vacuoles and in protein degradation by autophagy. Addition of glucose also increases intracellular ATP, which is in turn necessary for the induction of autophagy because the glycolysis inhibitor oxamate inhibits it, and there is also a good correlation between LC3-II and ATP levels. Moreover, we also show that, surprisingly, the induction of autophagy by glucose is independent of AMPK and mTOR and mainly relies on p38 MAPK (mitogen-activated protein kinase).


Subject(s)
Autophagy/drug effects , Glucose/pharmacology , MAP Kinase Signaling System/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Amino Acids/pharmacology , Animals , Blotting, Western , Culture Media, Serum-Free/pharmacology , Enzyme Activation/drug effects , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Knockout , Microscopy, Electron , Microtubule-Associated Proteins/metabolism , NIH 3T3 Cells , Phagosomes/drug effects , Phagosomes/metabolism , Phagosomes/ultrastructure , Proteolysis/drug effects , RNA Interference , Vacuoles/drug effects , Vacuoles/metabolism , Vacuoles/ultrastructure , p38 Mitogen-Activated Protein Kinases/genetics
11.
J Biol Chem ; 287(46): 38625-36, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-23027865

ABSTRACT

Autophagy is the main lysosomal catabolic process that becomes activated under stress conditions, such as amino acid starvation and cytosolic Ca(2+) upload. However, the molecular details on how both conditions control autophagy are still not fully understood. Here we link essential amino acid starvation and Ca(2+) in a signaling pathway to activate autophagy. We show that withdrawal of essential amino acids leads to an increase in cytosolic Ca(2+), arising from both extracellular medium and intracellular stores, which induces the activation of adenosine monophosphate-activated protein kinase (AMPK) via Ca(2+)/calmodulin-dependent kinase kinase-ß (CaMKK-ß). Furthermore, we show that autophagy induced by amino acid starvation requires AMPK, as this induction is attenuated in its absence. Subsequently, AMPK activates UNC-51-like kinase (ULK1), a mammalian autophagy-initiating kinase, through phosphorylation at Ser-555 in a process that requires CaMKK-ß. Finally, the mammalian target of rapamycin complex C1 (mTORC1), a negative regulator of autophagy downstream of AMPK, is inhibited by amino acid starvation in a Ca(2+)-sensitive manner, and CaMKK-ß appears to be important for mTORC1 inactivation, especially in the absence of extracellular Ca(2+). All these results highlight that amino acid starvation regulates autophagy in part through an increase in cellular Ca(2+) that activates a CaMKK-ß-AMPK pathway and inhibits mTORC1, which results in ULK1 stimulation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , 3T3 Cells , Amino Acids, Essential/chemistry , Animals , Autophagy , Autophagy-Related Protein-1 Homolog , Cytosol/metabolism , HeLa Cells , Humans , Insulin/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Models, Biological , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Signal Transduction
12.
Hum Mol Genet ; 19(14): 2867-76, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20453062

ABSTRACT

Lafora disease (LD) is an autosomal recessive, progressive myoclonus epilepsy, which is characterized by the accumulation of polyglucosan inclusion bodies, called Lafora bodies, in the cytoplasm of cells in the central nervous system and in many other organs. However, it is unclear at the moment whether Lafora bodies are the cause of the disease, or whether they are secondary consequences of a primary metabolic alteration. Here we describe that the major genetic lesion that causes LD, loss-of-function of the protein laforin, impairs autophagy. This phenomenon is confirmed in cell lines from human patients, mouse embryonic fibroblasts from laforin knockout mice and in tissues from such mice. Conversely, laforin expression stimulates autophagy. Laforin regulates autophagy via the mammalian target of rapamycin kinase-dependent pathway. The changes in autophagy mediated by laforin regulate the accumulation of diverse autophagy substrates and would be predicted to impact on the Lafora body accumulation and the cell stress seen in this disease that may eventually contribute to cell death.


Subject(s)
Autophagy/genetics , Lafora Disease/genetics , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/physiology , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Lafora Disease/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutant Proteins/genetics , Mutant Proteins/physiology , Phagosomes/genetics , Phagosomes/metabolism , Protein Processing, Post-Translational/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Signal Transduction/genetics , Signal Transduction/physiology , TOR Serine-Threonine Kinases
13.
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
14.
Exp Cell Res ; 316(16): 2618-29, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20599945

ABSTRACT

In recent years, the function of different tumour suppressors in the regulation of macroautophagy has been studied. We show here that BRCA1, unlike other tumour suppressors, negatively regulates formation of autophagosomes and lysosomal mass under conditions of both basal and enhanced autophagy. In MCF-7 breast cancer cells, increased formation of autophagic vacuoles after inactivation of BRCA1 by siRNAs is associated with an increase in reactive oxygen species, such as superoxide anion and hydrogen peroxide. This allows one to propose an antioxidant function for BRCA1 and suggests that dysfunctional mitochondria and the generated reactive oxygen species excess could explain the increased macroautophagy observed in the absence of BRCA1. In addition, a quick decrease in BRCA1 levels occurs when MCF-7 cells are switched to a nutrient-poor environment that stimulates macroautophagy and that is also reminiscent of certain phases of tumour growth. Inhibition of BRCA1 synthesis has an important role in this reduction, while there are almost no changes in BRCA1 degradation by lysosomes and proteasomes. Therefore, BRCA1 produces macroautophagy inhibition by reducing the formation of autophagic vacuoles, and this, together with the other results presented here, shows new functional aspects of BRCA1 that could help to clarify the role of autophagy in cancer development.


Subject(s)
Autophagy , BRCA1 Protein/physiology , Breast Neoplasms/pathology , Mitochondria/metabolism , Vacuoles/metabolism , Blotting, Western , Breast Neoplasms/metabolism , Female , Flow Cytometry , Humans , Lysosomes , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
15.
Hum Mol Genet ; 17(5): 667-78, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18029386

ABSTRACT

Lafora progressive myoclonus epilepsy (LD) is a fatal autosomal recessive neurodegenerative disorder characterized by the presence of glycogen-like intracellular inclusions called Lafora bodies. LD is caused by mutations in two genes, EPM2A and EPM2B, encoding respectively laforin, a dual-specificity protein phosphatase, and malin, an E3 ubiquitin ligase. Previously, we and others have suggested that the interactions between laforin and PTG (a regulatory subunit of type 1 protein phosphatase) and between laforin and malin are critical in the pathogenesis of LD. Here, we show that the laforin-malin complex downregulates PTG-induced glycogen synthesis in FTO2B hepatoma cells through a mechanism involving ubiquitination and degradation of PTG. Furthermore, we demonstrate that the interaction between laforin and malin is a regulated process that is modulated by the AMP-activated protein kinase (AMPK). These findings provide further insights into the critical role of the laforin-malin complex in the control of glycogen metabolism and unravel a novel link between the energy sensor AMPK and glycogen metabolism. These data advance our understanding of the functional role of laforin and malin, which hopefully will facilitate the development of appropriate LD therapies.


Subject(s)
Carrier Proteins/genetics , Glycogen/biosynthesis , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/genetics , AMP-Activated Protein Kinases , Adenoviridae/genetics , Amino Acid Sequence , Animals , Binding Sites , Carrier Proteins/metabolism , Cell Line , Cell Line, Tumor , Cell Physiological Phenomena , Escherichia coli/genetics , Glycogen/analysis , Green Fluorescent Proteins/metabolism , Humans , Kidney/cytology , Models, Biological , Molecular Sequence Data , Mutation , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Statistics as Topic , Transfection , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases , Ubiquitination
16.
Cell Mol Life Sci ; 66(15): 2427-43, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19399586

ABSTRACT

In higher organisms, dietary proteins are broken down into amino acids within the digestive tract but outside the cells, which incorporate the resulting amino acids into their metabolism. However, under certain conditions, an organism loses more nitrogen than is assimilated in the diet. This additional loss was found in the past century to come from intracellular proteins and started an intensive research that produced an enormous expansion of the field and a dispersed literature. Therefore, our purpose is to provide an updated summary of the current knowledge on the proteolytic machinery involved in intracellular protein degradation and its physiological and pathological relevance, especially addressed to newcomers in the field who may find further details in more specialized reviews. However, even providing a general overview, this is an extremely wide field and, therefore, we mainly focus on mammalian cells, while other cells will be mentioned only for comparison purposes.


Subject(s)
Proteins/metabolism , Amino Acids/metabolism , Animals , Autophagy/physiology , Humans , Lysosomes/metabolism , Phagosomes/metabolism , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism
17.
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
18.
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
19.
FEBS Lett ; 582(17): 2650-4, 2008 Jul 23.
Article in English | MEDLINE | ID: mdl-18593584

ABSTRACT

In this work we present evidence that A769662, a novel activator of AMP-activated protein kinase (AMPK), is able to inhibit the function of the 26S proteasome by an AMPK-independent mechanism. Contrary to the mechanism of action of most proteasome inhibitors, A769662 does not affect the proteolytic activities of the 20S core subunit, defining in this way a novel mechanism of inhibition of 26S proteasome activity. Inhibition of proteasome activity by A769662 is reversible and leads to an arrest of cell cycle progression. These side effects of this new activator of AMPK should be taken into account when this compound is used as an alternative activator of the kinase.


Subject(s)
Multienzyme Complexes/metabolism , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Protein Serine-Threonine Kinases/metabolism , Pyrones/pharmacology , Thiophenes/pharmacology , AMP-Activated Protein Kinases , Biphenyl Compounds , Cell Line , Humans , Multienzyme Complexes/genetics , Proteasome Endopeptidase Complex , Protein Serine-Threonine Kinases/genetics
20.
Biochem Biophys Res Commun ; 369(3): 964-8, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18328803

ABSTRACT

In this work, we have examined the possible role of AMP-activated protein kinase (a key energy sensor) in regulating intracellular protein degradation. We have found that AICAR, a known activator of AMPK, has a dual effect. On one hand, it inhibits autophagy by a mechanism independent of AMPK activity; AICAR decreases class III PI3-kinase binding to beclin-1 and this effect counteracts and reverses the known positive effect of AMPK activity on autophagy. On the other hand, AICAR inhibits the proteasomal degradation of proteins by an AMPK-dependent mechanism. This is a novel function of AMPK that allows the regulation of proteasomal activity under conditions of energy demand.


Subject(s)
Autophagy , Multienzyme Complexes/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Beclin-1 , Cells, Cultured , Down-Regulation , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/metabolism , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proteasome Inhibitors , Ribonucleosides/pharmacology , Vacuoles/drug effects , Vacuoles/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL