Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Int J Mol Sci ; 24(2)2023 Jan 06.
Article in English | MEDLINE | ID: mdl-36674605

ABSTRACT

Lafora disease is a rare, fatal form of progressive myoclonus epilepsy characterized by continuous neurodegeneration with epileptic seizures, characterized by the intracellular accumulation of aberrant polyglucosan granules called Lafora bodies. Several works have provided numerous evidence of molecular and cellular alterations in neural tissue from experimental mouse models deficient in either laforin or malin, two proteins related to the disease. Oxidative stress, alterations in proteostasis, and deregulation of inflammatory signals are some of the molecular alterations underlying this condition in both KO animal models. Lafora bodies appear early in the animal's life, but many of the aforementioned molecular aberrant processes and the consequent neurological symptoms ensue only as animals age. Here, using small RNA-seq and quantitative PCR on brain extracts from laforin and malin KO male mice of different ages, we show that two different microRNA species, miR-155 and miR-146a, are overexpressed in an age-dependent manner. We also observed altered expression of putative target genes for each of the microRNAs studied in brain extracts. These results open the path for a detailed dissection of the molecular consequences of laforin and malin deficiency in brain tissue, as well as the potential role of miR-155 and miR-146a as specific biomarkers of disease progression in LD.


Subject(s)
Lafora Disease , MicroRNAs , Mice , Male , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Lafora Disease/genetics , Lafora Disease/metabolism , Neuroinflammatory Diseases , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Oxidative Stress/genetics , Ubiquitin-Protein Ligases/metabolism
2.
Transplant Proc ; 53(9): 2721-2723, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34600758

ABSTRACT

BACKGROUND: To analyze the relationship of the antigen carbohydrate 125 (CA125) biomarker with the cellular rejection of the heart graft during the first year after transplantation. METHODS: Retrospective study of consecutive heart transplant (HTx) patients for 1.5 years. The total number of patients included in the study was 23 with a total of 103 follow-ups. In all patients, CA125 was determined before HTx and determined post-HTx in every follow-up. These were performed during months 1, 2, 4, 6, 9, and 12. Endomyocardial biopsy was performed in all revisions to assess the degree of graft rejection in the pathologic study. The biopsy results were grouped into 1. absence of rejection and 2. presence of some degree of rejection. RESULTS: The mean pretransplant CA125 value presented a median of 120 U/mL with an interquartile range of 28.8 U/mL. One month after transplantation, the value was reduced by 20% and at 2 months by 81%. In subsequent reviews, plasma values were always between 10 and 20 U/mL. When comparing the values by periods and according to the presence or absence of rejection, no significant differences were found other than a slight elevation at the 6-month checkup (P = .03) but without clinical relevance, because the CA125 value was slightly higher in biopsy results without rejection. CONCLUSION: The rapid reduction of CA125 corroborates its usefulness as a marker of congestion in heart failure. This biomarker is not useful for predicting rejection. However, in cases of very severe rejections that occurred with systemic congestion, it could be raised. It would be necessary to corroborate this hypothesis in a larger study with a higher number of severe rejections.


Subject(s)
Heart Transplantation , Hematopoietic Stem Cell Transplantation , Biomarkers , Biopsy , Carbohydrates , Graft Rejection/diagnosis , Heart Transplantation/adverse effects , Humans , Retrospective Studies
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
4.
Biochim Biophys Acta Mol Basis Dis ; 1864(10): 3234-3246, 2018 10.
Article in English | MEDLINE | ID: mdl-30006152

ABSTRACT

Circulating histones have been proposed as targets for therapy in sepsis and hyperinflammatory symptoms. However, the proposed strategies have failed in clinical trials. Although different mechanisms for histone-related cytotoxicity are being explored, those mediated by circulating histones are not fully understood. Extracellular histones induce endothelial cell death, thereby contributing to the pathogenesis of complex diseases such as sepsis and septic shock. Therefore, the comprehension of cellular responses triggered by histones is capital to design effective therapeutic strategies. Here we report how extracellular histones induce autophagy and apoptosis in a dose-dependent manner in cultured human endothelial cells. In addition, we describe how histones regulate these pathways via Sestrin2/AMPK/ULK1-mTOR and AKT/mTOR. Furthermore, we evaluate the effect of Toll-like receptors in mediating autophagy and apoptosis demonstrating how TLR inhibitors do not prevent apoptosis and/or autophagy induced by histones. Our results confirm that histones and autophagic pathways can be considered as novel targets to design therapeutic strategies in endothelial damage.


Subject(s)
Histones/pharmacology , Human Umbilical Vein Endothelial Cells/cytology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases/metabolism , Autophagy , Autophagy-Related Protein-1 Homolog/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism
5.
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
6.
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
7.
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.

8.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
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
15.
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.

16.
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
17.
Free Radic Biol Med ; 65: 347-359, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23850970

ABSTRACT

Thioredoxin 1 (Trx1) is a key regulator of cellular redox balance and participates in cellular signaling events. Recent evidence from yeast indicates that members of the Trx family interact with the 20S proteasome, indicating redox regulation of proteasome activity. However, there is little information about the interrelationship of Trx proteins with the proteasome system in mammalian cells, especially in the nucleus. Here, we have investigated this relationship under various cellular conditions in mammalian cells. We show that Trx1 levels and its subcellular localization (cytosol, endoplasmic reticulum, and nucleus) depend on proteasome activity during the cell cycle in NIH3T3 fibroblasts and under stress conditions, when proteasomes are inhibited. In addition, we also studied in these cells how the main cellular antioxidant systems are stimulated when proteasome activity is inhibited. Finally, we describe a reduction in Trx1 levels in Lafora disease fibroblasts and demonstrate that the nuclear colocalization of Trx1 with 20S proteasomes in laforin-deficient cells is altered compared with control cells. Our results indicate a close relationship between Trx1 and the 20S nuclear proteasome and give a new perspective to the study of diseases or physiopathological conditions in which defects in the proteasome system are associated with oxidative stress.


Subject(s)
Fibroblasts/metabolism , Lafora Disease/metabolism , Oxidative Stress/physiology , Proteasome Endopeptidase Complex/metabolism , Thioredoxins/metabolism , Animals , Blotting, Western , Endoplasmic Reticulum Chaperone BiP , Flow Cytometry , Humans , Immunoprecipitation , Mice , Microscopy, Confocal , NIH 3T3 Cells , Oxidation-Reduction , Reverse Transcriptase Polymerase Chain Reaction
18.
PLoS One ; 8(2): e55526, 2013.
Article in English | MEDLINE | ID: mdl-23408996

ABSTRACT

Neuronal ceroid lipofuscinoses (NCL) are lysosomal storage disorders characterized by the accumulation of lipofuscin within lysosomes. Late infantile (LINCL) and juvenile (JNCL) are their most common forms and are caused by loss-of-function mutations in tripeptidyl peptidase 1 (TPP1), a lysosomal endopeptidase, and CLN3 protein (CLN3p), whose location and function is still controversial. LINCL patients suffer more severely from NCL consequences than JNCL patients, in spite of having in common an abnormal accumulation of material with a similar composition in the lysosomes. To identify distinctive characteristics that could explain the differences in the severity of LINCL and JNCL pathologies, we compared the protein degradation mechanisms in patients fibroblasts. Pulse-chase experiments show a significant decrease in protein degradation by macroautophagy in fibroblasts bearing TPP1 (CLN2) and CLN3p (CLN3) mutations. In CLN2 fibroblasts, LC3-II levels and other procedures indicate an impaired formation of autophagosomes, which confirms the pulse-chase experiments. This defect is linked to an accumulation of reactive oxygen species (ROS), an upregulation of the Akt-mTOR signalling pathway and increased activities of the p38α and ERK1/2 MAPKs. In CLN3 fibroblasts, LC3-II analysis indicates impairment in autophagosome maturation and there is also a defect in fluid phase endocytosis, two alterations that can be related to an observed increase of 0.5 units in lysosomal pH. CLN3 fibroblasts also accumulate ROS but to a lower extent than CLN2. TPP1 activity is completely abrogated in CLN2 and partially diminished in CLN3 fibroblasts. TPP1 cleaves small hydrophobic proteins like subunit c of mitochondrial ATP synthase and the lack or a lower activity of this enzyme can contribute to lipofuscin accumulation. These alterations in TPP1 activity lead to an increased ROS production, especially in CLN2 in which it is aggravated by a decrease in catalase activity. This could explain the earlier appearance of the symptoms in the LINCL form.


Subject(s)
Autophagy , Fibroblasts/immunology , Hydrogen-Ion Concentration , Lysosomes/metabolism , Neuronal Ceroid-Lipofuscinoses/immunology , Reactive Oxygen Species/metabolism , Catalase/metabolism , Cells, Cultured , Fibroblasts/enzymology , Fibroblasts/metabolism , Humans , Neuronal Ceroid-Lipofuscinoses/enzymology , Neuronal Ceroid-Lipofuscinoses/metabolism , Oxidative Stress , Tripeptidyl-Peptidase 1
19.
Autophagy ; 8(4): 701-3, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22361617

ABSTRACT

Lafora disease (LD), a fatal neurodegenerative disorder characterized by intracellular inclusions called Lafora bodies (LBs), is caused by recessive loss-of-function mutations in the genes encoding either laforin or malin. Previous studies suggested a role of these proteins in regulating glycogen biosynthesis, in glycogen dephosphorylation and in the modulation of intracellular proteolytic systems. However, the contribution of each of these processes to LD pathogenesis is unclear. Here we review our recent finding that dysfunction of autophagy is a common feature of both laforin- and malin-deficient mice, preceding other pathological manifestations. We propose that autophagy plays a primary role in LD pathogenesis and is a potential target for its treatment.


Subject(s)
Autophagy , Lafora Disease/etiology , Lafora Disease/pathology , Ubiquitin-Protein Ligases/deficiency , Animals , Mice , Mice, Knockout , Models, Biological , Ubiquitin-Protein Ligases/metabolism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...