Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Biochim Biophys Acta ; 1862(8): 1423-32, 2016 08.
Article in English | MEDLINE | ID: mdl-27130438

ABSTRACT

Danon disease, a condition characterized by cardiomyopathy, myopathy, and intellectual disability, is caused by mutations in the LAMP-2 gene. Lamp-2A protein, generated by alternative splicing from the Lamp-2 pre-mRNA, is reported to be the lysosomal membrane receptor essential for the chaperone-mediated autophagic pathway (CMA) aimed to selective protein targeting and translocation into the lysosomal lumen for degradation. To study the relevance of Lamp-2 in protein degradation, a lymphoblastoid cell line was obtained by EBV transformation of B-cells from a Danon patient. The derived cell line showed no significant expression of Lamp-2 protein. The steady-state mRNA and protein levels of alpha-synuclein, IΚBα, Rcan1, and glyceraldehyde-3-phosphate dehydrogenase, four proteins reported to be selective substrates of the CMA pathway, were similar in control and Lamp-2-deficient cells. Inhibition of protein synthesis showed that the half-life of alpha-synuclein, IΚBα, and Rcan1 was similar in control and Lamp-2-deficient cells, and its degradation prevented by proteasome inhibitors. Both in control and Lamp-2-deficient cells, induction of CMA and macroautophagy by serum and aminoacid starvation of cells for 8h produced a similar decrease in IΚBα and Rcan1 protein levels and was prevented by the addition of lysosome and autophagy inhibitors. In conclusion, the results presented here showed that Lamp-2 deficiency in human lymphoblastoid cells did not modify the steady-state levels or the degradation of several protein substrates reported as selective substrates of the CMA pathway.


Subject(s)
Autophagy , B-Lymphocytes/metabolism , Lysosomal-Associated Membrane Protein 2/genetics , Proteolysis , B-Lymphocytes/pathology , Cell Line, Transformed , DNA-Binding Proteins , Glycogen Storage Disease Type IIb , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lysosomal-Associated Membrane Protein 2/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , NF-KappaB Inhibitor alpha/genetics , NF-KappaB Inhibitor alpha/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
2.
Int J Mol Sci ; 18(12)2017 Dec 08.
Article in English | MEDLINE | ID: mdl-29292768

ABSTRACT

Spinal muscular atrophy is due to mutations affecting the SMN1 gene coding for the full-length protein (survival motor neuron; SMN) and the SMN2 gene that preferentially generates an exon 7-deleted protein (SMNΔ7) by alternative splicing. To study SMN and SMNΔ7 degradation in the cell, we have used tagged versions at the N- (Flag) or C-terminus (V5) of both proteins. Transfection of those constructs into HeLa cells and treatment with cycloheximide showed that those protein constructs were degraded. Proteasomal degradation usually requires prior lysine ubiquitylation. Surprisingly, lysine-less variants of both proteins tagged either at N- (Flag) or C-terminus (V5) were also degraded. The degradation of the endogenous SMN protein, and the protein constructs mentioned above, was mediated by the proteasome, as it was blocked by lactacystin, a specific and irreversible proteasomal inhibitor. The results obtained allowed us to conclude that SMN and SMNΔ7 proteasomal degradation did not absolutely require internal ubiquitylation nor N-terminal ubiquitylation (prevented by N-terminal tagging). While the above conclusions are firmly supported by the experimental data presented, we discuss and justify the need of deep proteomic techniques for the study of SMN complex components (orphan and bound) turn-over to understand the physiological relevant mechanisms of degradation of SMN and SMNΔ7 in the cell.


Subject(s)
Muscular Atrophy, Spinal/genetics , Proteomics , Survival of Motor Neuron 1 Protein/genetics , Alternative Splicing/genetics , Exons/genetics , HeLa Cells , Humans , Metabolic Networks and Pathways , Muscular Atrophy, Spinal/pathology , Mutation , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Survival of Motor Neuron 2 Protein/genetics
3.
Biochim Biophys Acta ; 1843(2): 352-65, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24315858

ABSTRACT

Alpha-synuclein is a small protein implicated in the pathophysiology of Parkinson's disease (PD). We have investigated the mechanism of cleavage of alpha-synuclein by the 20S proteasome. Alpha-synuclein interacts with the C8 (α7) subunit of the proteasome. The N-terminal part of alpha-synuclein (amino acids 1-60) is essential for its proteasomal degradation and analysis of peptides released from proteasomal digestion allows concluding that initial cleavages occur within the N-terminal region of the molecule. Aggregated alpha-synucleins are also degraded by the proteasome with a reduced rate, likely due to Met oxidation. In fact, mild oxidation of alpha-synuclein with H2O2 resulted in the inhibition of its degradation by the proteasome, mainly due to oxidation of Met 1 and 5 of alpha-synuclein. The inhibition was reversed by treatment of the oxidized protein with methionine sulfoxide reductases (MsrA plus MsrB). Similarly, treatment with H2O2 of N2A cells transfected with alpha-synuclein resulted in the inhibition of its degradation that was also reverted by co-transfection of MsrA plus MsrB. These results clearly indicate that oxidative stress, a common feature of PD and other synucleinopathies, promotes a RedOx change in the proteostasis of alpha-synuclein due to Met oxidation and reduced proteasomal degradation; compromised reversion of those oxidative changes would result in the accumulation of oxidative damaged alpha-synuclein likely contributing to the pathogenesis of PD.


Subject(s)
Methionine/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , alpha-Synuclein/metabolism , Amino Acid Sequence , Animals , Humans , Hydrogen Peroxide/pharmacology , Immunoblotting , Methionine Sulfoxide Reductases/metabolism , Mice , Molecular Sequence Data , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Peptides/chemistry , Peptides/metabolism , Protein Binding/drug effects , Protein Interaction Mapping , Protein Structure, Quaternary , Protein Subunits/metabolism , Proteolysis/drug effects , Rats , Silver Staining , alpha-Synuclein/chemistry
4.
Biochim Biophys Acta ; 1823(2): 524-33, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22173095

ABSTRACT

Parkinson's disease (PD) is characterized by dopaminergic dysfunction and degeneration. DJ-1/PARK7 mutations have been linked with a familial form of early onset PD. In this study, we found that human DJ-1 wild type and the missense mutants M26I, R98Q, A104T and D149A were stable proteins in cells, only the L166P mutant was unstable. In parallel, the former were not degraded and the L166P mutant was directly degraded in vitro by proteasome-mediated endoproteolytic cleavage. Furthermore, genetic evidence in fission yeast showed the direct involvement of proteasome in the degradation of human DJ-1 L166P and the corresponding L169P mutant of SPAC22E12.03c, the human orthologue of DJ-1 in Schizosaccharomyces Pombe, as their protein levels were increased at restrictive temperature in fission yeast (mts4 and pts1-732) harboring temperature sensitive mutations in proteasomal subunits. In total, our results provide evidence that direct proteasomal endoproteolytic cleavage of DJ-1 L166P is the mechanism of degradation contributing to the loss-of-function of the mutant protein, a property not shared by other DJ-1 missense mutants associated with PD.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mutation, Missense , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Parkinsonian Disorders/genetics , Parkinsonian Disorders/metabolism , Proteasome Endopeptidase Complex/metabolism , Amino Acid Sequence , Animals , Cell Line , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Mice , Molecular Sequence Data , Oncogene Proteins/chemistry , Peptides/genetics , Peptides/metabolism , Proteasome Endopeptidase Complex/chemistry , Protein Deglycase DJ-1 , Protein Structure, Secondary , Protein Subunits/chemistry , Protein Subunits/metabolism , Rats , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism
6.
Cell Mol Life Sci ; 68(15): 2643-54, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21103907

ABSTRACT

Intracellular deposits of aggregated alpha-synuclein are a hallmark of Parkinson's disease. Protein-protein interactions are critical in the regulation of cell proteostasis. Synphilin-1 interacts both in vitro and in vivo with alpha-synuclein promoting its aggregation. We report here that synphilin-1 specifically inhibits the degradation of alpha-synuclein wild-type and its missense mutants by the 20S proteasome due at least in part by the interaction of the ankyrin and coiled-coil domains of synphilin-1 (amino acids 331-555) with the N-terminal region (amino acids 1-60) of alpha-synuclein. Co-expression of synphilin-1 and alpha-synuclein wild-type in HeLa and N2A cells produces a specific increase in the half-life of alpha-synuclein, as degradation of unstable fluorescent reporters is not affected. Synphilin-1 inhibition can be relieved by co-expression of Siah-1 that targets synphilin-1 to degradation. Synphilin-1 inhibition of the proteasomal pathway of degradation of alpha-synuclein may help to understand the pathophysiological changes occurring in PD and other synucleinopathies.


Subject(s)
Carrier Proteins/physiology , Nerve Tissue Proteins/physiology , Proteasome Endopeptidase Complex/metabolism , alpha-Synuclein/metabolism , Animals , Carrier Proteins/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Down-Regulation/drug effects , HeLa Cells , Humans , Mutation, Missense/physiology , Nerve Tissue Proteins/pharmacology , Proteasome Inhibitors , Protein Processing, Post-Translational/genetics , Rats , Substrate Specificity , alpha-Synuclein/genetics
7.
Sci Rep ; 11(1): 7320, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33795807

ABSTRACT

DJ-1/PARK7 mutations are linked with familial forms of early-onset Parkinson's disease (PD). We have studied the degradation of untagged DJ-1 wild type (WT) and missense mutants in mouse embryonic fibroblasts obtained from DJ-1-null mice, an approach closer to the situation in patients carrying homozygous mutations. The results showed that the mutants L10P, M26I, A107P, P158Δ, L166P, E163K, and L172Q are unstable proteins, while A39S, E64D, R98Q, A104T, D149A, A171S, K175E, and A179T are as stable as DJ-1 WT. Inhibition of proteasomal and autophagic-lysosomal pathways had little effect on their degradation. Immunofluorescence and biochemical fractionation studies indicated that M26I, A107P, P158Δ, L166P, E163K, and L172Q mutants associate with mitochondria. Silencing of mitochondrial matrix protease LonP1 produced a strong reduction of the degradation of the mitochondrial-associated DJ-1 mutants A107P, P158Δ, L166P, E163K, and L172Q but not of mutant L10P. These results demonstrated a mitochondrial pathway of degradation of those DJ-1 missense mutants implicated in PD pathogenesis.


Subject(s)
ATP-Dependent Proteases/biosynthesis , Mitochondria/enzymology , Mitochondrial Proteins/biosynthesis , Mutation, Missense , Parkinson Disease/enzymology , Parkinson Disease/genetics , Protein Deglycase DJ-1/genetics , Animals , Fibroblasts/metabolism , Gene Silencing , Homozygote , Humans , Mice , Microscopy, Fluorescence , Proteasome Endopeptidase Complex , Subcellular Fractions
8.
J Biol Chem ; 284(41): 28253-28262, 2009 Oct 09.
Article in English | MEDLINE | ID: mdl-19679666

ABSTRACT

Fluorescent unstable proteins obtained by the fusion of a fluorescent protein coding sequence with specific amino acid sequences that promote its fast degradation have become popular to gauge the activity of the ubiquitin/proteasome system in living cells. The steady-state levels of expression of these unstable proteins is low in agreement with their short half-lives, and they accumulate in the cell upon treatment with proteasome inhibitors. We show here that this accumulation is mainly due to transcriptional up-regulation of the cytomegalovirus promoter by proteasome inhibitors and mediated, at least in part, by AP1 transactivation. These simple facts put under quarantine conclusions reached about the activity of the ubiquitin/proteasome pathway in animal cells in culture or in transgenic mice, where popular cytomegalovirus-driven constructs are used, as transcriptional regulation of the expression of the reporter protein construct and not degradation of the unstable protein by the ubiquitin/proteasome system may contribute significantly to the interpretation of the results observed.


Subject(s)
Cytomegalovirus , Promoter Regions, Genetic , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Recombinant Fusion Proteins , Animals , Cell Line , Cytomegalovirus/drug effects , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Genes, Reporter , Humans , Mice , Mice, Transgenic , Proteasome Endopeptidase Complex/metabolism , Protein Biosynthesis , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcription, Genetic , Up-Regulation
10.
J Neurochem ; 110(5): 1523-37, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19549073

ABSTRACT

Parkin mutations in humans produce parkinsonism whose pathogenesis is related to impaired protein degradation, increased free radicals and abnormal neurotransmitter release. In this study, we have investigated whether partial proteasomal inhibition by epoxomicin, an ubiquitin proteasomal system (UPS) irreversible inhibitor, further aggravates the cellular effects of parkin suppression in midbrain neurons and glia. We observed that parkin null (PK-KO) midbrain neuronal cultures are resistant to epoxomicin-induced cell death. This resistance is due to increased GSH and DJ-1 protein levels in PK-KO mice. The treatment with epoxomicin increases, in wild type (WT) cultures, the pro-apoptotic Bax/Bcl-2 ratio, the phosphorylation of tau, and the levels of chaperones heat-shock protein 70 and C-terminal Hsc-interacting protein, but none of these effects took place in epoxomicin-treated PK-KO cultures. Poly-ubiquitinated proteins increased more in WT than in PK-KO-treated neuronal cultures. Parkin accumulated in WT neuronal cultures treated with epoxomicin. Markers of autophagy, such as LC3II/I, were increased in naïve PK-KO cultures, and further increased after treatment with epoxomicin, implying that the blockade of the proteasome in PK-KO neurons triggers the enhancement of autophagy. The treatment with l-buthionine-S,R-sulfoximine and the inhibition of autophagy, however, reverted the increase resistance to epoxomicin of the PK-KO cultures. We also found that PK-KO glial cells, stressed by growth in defined medium and depleted of GSH, were more susceptible to epoxomicin induced cell death than WT glia treated similarly. This susceptibility was linked to reduced GSH levels and less heat-shock protein 70 response, and to activation of p-serine/threonine kinase protein signaling pathway as well as to increased poly-ubiquitinated proteins. These data suggest that mild UPS inhibition is compensated by other mechanisms in PK-KO midbrain neurons. However the depletion of GSH, as happens in stressed glia, suppresses the protection against UPS inhibition-induced cell death. Furthermore, GSH inhibition regulated differentially UPS activity and in old PK-KO mice, which have depletion of GSH, UPS activity is decreased in comparison with that of old-WT.


Subject(s)
Autophagy/physiology , Glutathione/physiology , Homeostasis/physiology , Neuroglia/metabolism , Neurons/metabolism , Proteasome Inhibitors , Ubiquitin-Protein Ligases/deficiency , Animals , Autophagy/drug effects , Cell Line, Tumor , Cells, Cultured , Dose-Response Relationship, Drug , Glutathione/drug effects , Homeostasis/drug effects , Humans , Mesencephalon/drug effects , Mesencephalon/enzymology , Mesencephalon/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neuroglia/drug effects , Neuroglia/enzymology , Neurons/drug effects , Neurons/enzymology , Oligopeptides/pharmacology , Proteasome Endopeptidase Complex/metabolism
11.
PLoS One ; 13(7): e0201152, 2018.
Article in English | MEDLINE | ID: mdl-30048497

ABSTRACT

Mutations in PARK7/DJ-1 gene are associated with familial autosomal recessive Parkinson disease. Recently, lysosomes and chaperone mediated autophagy (CMA) has been reported to participate in the degradation of DJ-1/PARK7 protein. Lamp-2A isoform is considered as the lysosomal receptor for the uptake of proteins being degraded by the CMA pathway. We have used several cell lines with disrupted LAMP2 gene expression and their respective control cells to test the possible role of lysosomal degradation and in particular CMA in DJ-1 /PARK7 degradation. Interruption of LAMP-2 expression did not result in an increase of the steady-state protein levels of DJ-1 /PARK7, as it would have been expected. Furthermore, no change in DJ-1 /PARK7 protein levels were observed upon inhibition of lysosomal function with NH4Cl or NH4Cl plus leupeptin, or after activation of CMA by serum starvation for 24h. Accordingly, we have not found any evidence that DJ-1 /PARK7 protein levels are regulated via lysosomal degradation or the CMA pathway.


Subject(s)
Lysosomal-Associated Membrane Protein 2/metabolism , Lysosomes/metabolism , Protein Deglycase DJ-1/metabolism , Animals , Cell Line , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Silencing , Humans , Lysosomes/drug effects , Mice, Knockout , Protein Synthesis Inhibitors/pharmacology , Serum/metabolism
12.
Mol Cell Biol ; 23(20): 7377-90, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14517305

ABSTRACT

Cot, initially identified as an oncogene in a truncated form, is a mitogen-activated protein kinase kinase kinase implicated in cellular activation and proliferation. Here, we show that this truncation of Cot results in a 10-fold increase in its overall kinase activity through two different mechanisms. Truncated Cot protein exhibits a lower turnover rate (half-life, 95 min) than wild-type Cot (half-life, 35 min). The degradation of wild-type and truncated Cot can be specifically inhibited by proteasome inhibitors in situ. The 20S proteasome also degrades wild-type Cot more efficiently than the truncated protein. Furthermore, the amino acid 435 to 457 region within the wild-type Cot COOH-terminal domain confers instability when transferred to the yellow fluorescent protein and targets this fusion protein to degradation via the proteasome. On the other hand, the kinase specific activity of wild-type Cot is 3.8-fold lower than that of truncated Cot, and it appears that the last 43 amino acids of the wild-type Cot COOH-terminal domain are those responsible for this inhibition of kinase activity. In conclusion, these data demonstrate that the oncogenic activity of truncated Cot is the result of its prolonged half-life and its higher kinase specific activity with respect to wild-type Cot.


Subject(s)
MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Cell Line , Cysteine Endopeptidases/metabolism , Dose-Response Relationship, Drug , Humans , Mice , Molecular Sequence Data , Multienzyme Complexes/metabolism , NIH 3T3 Cells , Plasmids/metabolism , Precipitin Tests , Proteasome Endopeptidase Complex , Protein Binding , Protein Structure, Tertiary , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Time Factors , Transcription, Genetic , Transfection
13.
Mol Biol Cell ; 13(8): 2771-82, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12181345

ABSTRACT

Nuclear bodies represent a heterogeneous class of nuclear structures. Herein, we describe that a subset of nuclear bodies is highly enriched in components of the ubiquitin-proteasome pathway of proteolysis. We coined the term clastosome (from the Greek klastos, broken and soma, body) to refer to this type of nuclear body. Clastosomes contain a high concentration of 1) ubiquitin conjugates, 2) the proteolytically active 20S core and the 19S regulatory complexes of the 26S proteasome, and 3) protein substrates of the proteasome. Although detected in a variety of cell types, clastosomes are scarce under normal conditions; however, they become more abundant when proteasomal activity is stimulated. In contrast, clastosomes disappear when cells are treated with proteasome inhibitors. Protein substrates of the proteasome that are found concentrated in clastosomes include the short-lived transcription factors c-Fos and c-Jun, adenovirus E1A proteins, and the PML protein. We propose that clastosomes are sites where proteolysis of a variety of protein substrates is taking place.


Subject(s)
Cell Nucleus Structures/metabolism , Cysteine Endopeptidases/metabolism , Multienzyme Complexes/metabolism , Nuclear Proteins/metabolism , Protein Subunits/metabolism , Ubiquitin/metabolism , Animals , Animals, Newborn , Cell Nucleus Structures/chemistry , Cell Nucleus Structures/ultrastructure , Cells, Cultured , Enzyme Inhibitors/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Immunohistochemistry , Male , Multienzyme Complexes/antagonists & inhibitors , Neoplasm Proteins/metabolism , Promyelocytic Leukemia Protein , Proteasome Endopeptidase Complex , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Rats , Rats, Sprague-Dawley , Schwann Cells/cytology , Schwann Cells/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins
14.
Sci Rep ; 7(1): 13526, 2017 10 19.
Article in English | MEDLINE | ID: mdl-29051532

ABSTRACT

The CCAAT/Enhancer binding protein ß (C/EBPß) is a transcription factor involved in numerous physiological as well as pathological conditions in the brain. However, little is known regarding its possible role in neurodegenerative disorders. We have previously shown that C/EBPß regulates the expression of genes involved in inflammatory processes and brain injury. Here, we have analyzed the effects of C/EBPß interference in dopaminergic cell death and glial activation in the 6-hydroxydopamine model of Parkinson's disease. Our results showed that lentivirus-mediated C/EBPß deprivation conferred marked in vitro and in vivo neuroprotection of dopaminergic cells concomitant with a significant attenuation of the level of the inflammatory response and glial activation. Additionally, C/EBPß interference diminished the induction of α-synuclein in the substantia nigra pars compacta of animals injected with 6-hydroxydopamine. Taking together, these results reveal an essential function for C/EBPß in the pathways leading to inflammatory-mediated brain damage and suggest novel roles for C/EBPß in neurodegenerative diseases, specifically in Parkinson's disease, opening the door for new therapeutic interventions.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Parkinson Disease/pathology , Animals , Apoptosis/drug effects , CCAAT-Enhancer-Binding Protein-beta/antagonists & inhibitors , CCAAT-Enhancer-Binding Protein-beta/genetics , Cells, Cultured , Disease Models, Animal , Dopaminergic Neurons/cytology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Humans , Male , Mesencephalon/cytology , Mesencephalon/drug effects , Mesencephalon/metabolism , Oxidopamine/pharmacology , Parkinson Disease/metabolism , Pars Compacta/drug effects , Pars Compacta/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , alpha-Synuclein/metabolism
15.
J Mol Biol ; 351(5): 995-1006, 2005 Sep 02.
Article in English | MEDLINE | ID: mdl-16051267

ABSTRACT

We have reported the existence in rat nuclear extracts of a specific cleavage activity on a DNA fragment containing the human minisatellite MsH42 region (minisatellite plus its flanking sequences). Here, we have developed a system to analyse the nature of the cleavage products from the MsH42 region generated by the nuclear extracts. Our results demonstrated the formation of DNA double-strand breaks (DSB) in the MsH42 region by two different enzymatic activities, and that their distribution along this fragment changes depending on the presence of Mg2+. In the assays with Mg2+, the DSB were located in the minisatellite and its 3'-flanking region, showing preference for G-rich stretches. Oligonucleotide mutagenesis analysis confirmed that this enzymatic activity has a strong preference for G-tracts and that the recognition site is polarized towards the 3' end. Moreover, this activity cuts GC palindromes efficiently. In contrast, in the experiments without Mg2+, most DSB were mapped within the minisatellite flanking sequences. The analysis with oligonucleotides showed that G-tracts are recognized by this endonuclease activity, but with differences in the cleavage behaviour with respect to the reactions observed with Mg2+. The existence of two separate activities (Mg2+-dependent and Mg2+-independent) for the production of DSB was confirmed by analysing the effect of EGTA, N-ethyl maleimide, ionic strength, and by preincubations of the nuclear extracts at different temperatures. The tissue distribution of both DSB-producing activities was also different. The in vitro system used in the present work may be a useful tool for studying the formation of DSB and for investigation of the mechanisms of DNA repair.


Subject(s)
Cell Nucleus/metabolism , DNA Damage , Amino Acid Motifs , Animals , Base Sequence , Binding Sites , Cell-Free System , Chickens , Cloning, Molecular , DNA/chemistry , DNA Repair , Egtazic Acid/chemistry , Electrophoresis, Agar Gel , Magnesium/chemistry , Molecular Sequence Data , Mutagenesis , Mutagenesis, Site-Directed , Oligonucleotides/chemistry , Plasmids/metabolism , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Recombination, Genetic , Sequence Analysis, DNA
16.
J Neurosci ; 23(37): 11653-61, 2003 Dec 17.
Article in English | MEDLINE | ID: mdl-14684867

ABSTRACT

Huntington's disease (HD) inclusions are stained with anti-ubiquitin and anti-proteasome antibodies. This, together with proteasome activity studies on transfected cells, suggest that an impairment of the ubiquitin-proteasome system (UPS) may be key in HD pathogenesis. To test whether proteasome activity is impaired in vivo, we performed enzymatic assays for the three peptidase activities of the proteasome in brain extracts from the HD94 conditional mouse model of HD. We found no inhibition of any of the activities, suggesting that if UPS impairment happens in vivo, it is not at the level of the proteasome catalytic core. Intriguingly, the chymotrypsin- and trypsin-like activities increased selectively in the affected and aggregate-containing regions: cortex and striatum. Western blot analysis revealed no difference in total proteasome content whereas an increase in the interferon-inducible subunits of the immunoproteasome, LMP2 and LMP7, was observed. These subunits confer to the proteasome catalytic properties that are optimal for MHC-I peptide presentation. Immunohistochemistry in control mouse brain revealed LMP2 and LMP7 mainly in neurons. Accordingly, their increase in HD94 mice predominantly took place in neurons, and 5% of the ubiquitin-positive cortical aggregates were also LMP2-positive. Ultrastructural analysis of neurons with high level of immunoproteasome subunits revealed signs of neurodegeneration like nuclear indentation or fragmentation and dark cell appearance. The neuronal induction of LMP2 and LMP7 and the associated signs of neurodegeneration were also found in HD postmortem brains. Our results indicate that LMP2 and LMP7 participate in normal neuronal physiology and suggest a role in HD neurodegeneration.


Subject(s)
Cysteine Endopeptidases/biosynthesis , Huntington Disease/enzymology , Multienzyme Complexes/biosynthesis , Neurons/enzymology , Aged , Animals , Brain/pathology , Cerebral Cortex/enzymology , Chymotrypsin/metabolism , Cysteine Endopeptidases/immunology , Cysteine Endopeptidases/metabolism , Female , Humans , Huntington Disease/pathology , Immunohistochemistry , Male , Mice , Mice, Transgenic , Multienzyme Complexes/immunology , Multienzyme Complexes/metabolism , Neostriatum/enzymology , Neurons/ultrastructure , Proteasome Endopeptidase Complex , Trypsin/metabolism , Ubiquitins/analysis
17.
FEBS Lett ; 579(21): 4797-802, 2005 Aug 29.
Article in English | MEDLINE | ID: mdl-16098527

ABSTRACT

IkappaBalpha regulates activation of the transcription factor NF-kappaB. NF-kappaB is activated in response to several stimuli, i.e. proinflamatory cytokines, infections, and physical stress. This signal dependent pathway involves IkappaBalpha phosphorylation, ubiquitylation, and degradation by 26S proteasome. A signal independent (basal) turnover of IkappaBalpha has also been described. Here, we show that IkappaBalpha can be directly degraded by 20S proteasomes. Deletion constructs of IkappaBalpha allow us to the determine that N-terminal (DeltaN 1-70) and C-terminal regions (DeltaC 280-327, removing the PEST region) of IkappaBalpha are not required for IkappaBalpha degradation, while a further C-terminal deletion including part of the arm repeats (DeltaC2 245-327) almost completely suppress the degradation by 20S proteasome. Binding and competition experiments demonstrate that the degradation of IkappaBalpha involves specific interactions with alpha2(C3) subunit of the proteasome. Finally, p65/relA (not itself a substrate for 20S proteasome) inhibits the degradation of IkappaBalpha by the proteasome. These results recapitulate in vitro the main characteristics of signal independent (basal) turnover of IkappaBalpha demonstrated in intact cells.


Subject(s)
I-kappa B Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Recombinant Proteins/metabolism , Animals , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Recombinant Proteins/genetics , Transcription Factor RelA
18.
J Neuropathol Exp Neurol ; 63(5): 484-98, 2004 May.
Article in English | MEDLINE | ID: mdl-15198127

ABSTRACT

Inclusion body myositis (IBM) and myofibrillar myopathy (MM) are diseases characterized by the abnormal accumulation of proteins in muscle fibers, including desmin, alphaB-crystallin, gelsolin, actin, kinases, and phospho-tau, along with ubiquitin in muscle fibers, suggesting abnormal protein degradation as a possible cause of the surplus myopathy. Since the ubiquitin-proteasome system plays a crucial role in non-lysosomal protein degradation, the present study has examined by immunohistochemistry the expression of components of the catalytic core of 20S proteasomes and its regulators: 19S and PA28alpha/beta, and the expression of immunoproteasome subunits LMP2, LMP7, and MECL1 in 8 patients with MM and 10 patients with IBM. The patients with MM were from 6 unrelated families, 2 sporadic cases, I with autosomal recessive and 5 with autosomal dominant inheritance. One sporadic patient had a de novo R406W mutation in the desmin gene, and 1 patient with autosomal dominant MM had a single amino acid deletion at position 366 in the desmin gene. Increased immunoreactivity to 20S, 19S, and PA28alpha/beta colocalizing abnormal protein deposits, as revealed in consecutive serial sections, was seen in all cases with MM and IBM. In all cases, the subunits of the immunoproteasome LMP2, LMP7, and MECL1 colocalized with proteasomal immunoreactivity and abnormal protein accumulation. Immunohistochemistry revealed focal MHC class I immunoreactivity in the cytoplasmic membrane of muscle fibers in IBM and in association with protein aggregates in IBM, and to a lesser degree, in MM. The present findings provide a link between abnormal protein accumulation and altered proteasomal expression in IBM and MM.


Subject(s)
Cysteine Endopeptidases/immunology , Histocompatibility Antigens Class I/immunology , Multienzyme Complexes/immunology , Myopathies, Structural, Congenital/immunology , Myopathies, Structural, Congenital/pathology , Myositis, Inclusion Body/immunology , Myositis, Inclusion Body/pathology , Adenosine Triphosphatases/immunology , Adenosine Triphosphatases/metabolism , Adult , Aged , Aged, 80 and over , Antigen Presentation/immunology , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Membrane/pathology , Cysteine Endopeptidases/metabolism , DNA Mutational Analysis , Desmin/deficiency , Desmin/genetics , Endopeptidases/immunology , Endopeptidases/metabolism , Female , Genetic Testing , Histocompatibility Antigens Class I/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , Multienzyme Complexes/metabolism , Muscle Proteins/immunology , Muscle Proteins/metabolism , Muscle, Skeletal/immunology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Mutation/genetics , Myofibrils/immunology , Myofibrils/metabolism , Myofibrils/pathology , Myopathies, Structural, Congenital/metabolism , Myositis, Inclusion Body/metabolism , Proteasome Endopeptidase Complex , Protein Subunits/genetics , Protein Subunits/immunology , Protein Subunits/metabolism , Protein Transport/genetics , Proteins/immunology , Proteins/metabolism
19.
Biomolecules ; 4(4): 1140-54, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25534281

ABSTRACT

The mammalian 20S proteasome is a heterodimeric cylindrical complex (α7ß7ß7α7), composed of four rings each composed of seven different α or ß subunits with broad proteolytic activity. We review the mammalian proteins shown to directly interact with specific 20S proteasomal subunits and those subjected to ubiquitin-independent proteasomal degradation (UIPD). The published reports of proteins that interact with specific proteasomal subunits, and others found on interactome databases and those that are degraded by a UIPD mechanism, overlap by only a few protein members. Therefore, systematic studies of the specificity of the interactions, the elucidation of the protein regions implicated in the interactions (that may or may not be followed by degradation) and competition experiments between proteins known to interact with the same proteasomal subunit, are needed. Those studies should provide a coherent picture of the molecular mechanisms governing the interactions of cellular proteins with proteasomal subunits, and their relevance to cell proteostasis and cell functioning.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Proteins/metabolism , Animals , Humans , Mammals/metabolism , Protein Subunits , Ubiquitin/metabolism
20.
Front Aging Neurosci ; 6: 169, 2014.
Article in English | MEDLINE | ID: mdl-25076905

ABSTRACT

Alpha-synuclein (Snca) plays a major role in Parkinson disease (PD). Circulating anti-Snca antibodies has been described in PD patients and healthy controls, but they have been poorly characterized. This study was designed to assess the prevalence of anti-Snca reactivity in human subjects carrying the LRRK2 mutation, idiopathic PD (iPD) patients, and healthy controls and to map the epitopes of the anti-Snca antibodies. Antibodies to Snca were detected by ELISA and immunoblotting using purified recombinant Snca in plasma from individuals carrying LRRK2 mutations (104), iPD patients (59), and healthy controls (83). Epitopes of antibodies were mapped using recombinant protein constructs comprising different regions of Snca. Clear positive anti-Snca reactivity showed no correlation with age, sex, years of evolution, or the disability scores for PD patients and anti-Snca reactivity was not prevalent in human patients with other neurological or autoimmune diseases. Thirteen of the positive individuals were carriers of LRRK2 mutations either non-manifesting (8 out 49 screened) or manifesting (5 positive out 55), three positive (out of 59) were iPD patients, and five positive (out of 83) were healthy controls. Epitope mapping showed that antibodies against the N-terminal (a.a. 1-60) or C-terminal (a.a. 109-140) regions of Snca predominate in LRRK2 mutation carriers and iPD patients, being N122 a critical amino acid for recognition by the anti-C-terminal directed antibodies. Anti-Snca circulating antibodies seem to cluster within families carrying the LRRK2 mutation indicating possible genetic or common environmental factors in the generation of anti-Snca antibodies. These results suggest that case-controls' studies are insufficient and further studies in family cohorts of patients and healthy controls should be undertaken, to progress in the understanding of the possible relationship of anti-Snca antibodies and PD pathology.

SELECTION OF CITATIONS
SEARCH DETAIL