Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Carbohydr Polym ; 331: 121815, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38388067

ABSTRACT

Bacterial nanocellulose (BNC) is a promising dietary fiber with potential as a functional food additive. We evaluated BNC fibers (BNCf) in the Caenorhabditis elegans model to obtain insight into the BNCf's biointeraction with its gastrointestinal tract while reducing the variables of higher complex animals. BNCf were uptaken and excreted by worms without crossing the intestinal barrier, confirming its biosafety regarding survival rate, reproduction, and aging for concentrations up to 34 µg/ml BNCf. However, a slight decrease in the worms' length was detected. A possible nutrient shortage or stress produced by BNCf was discarded by measuring stress and chemotactic response pathways. Besides, we detected a lipid-lowering effect of BNCf in N2 C. elegans in normal and high-caloric diets. Oxidative damage was computed in N2 worms and Rac1/ced-10 mutants. The GTPase Rac1 is involved in neurological diseases, where its dysregulation enhances ROS production and neuronal damage. BNCf reduced the lipid oxidative markers produced by ROS species in this worm strain. Finally, we detected that BNCf activated the genetic expression of the immunological response and lipid catabolic process. These results strengthen the use of BNCf as a functional dietary fiber and encourage the potential treatment of neurological disease by modulating diet.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/pharmacology , Reactive Oxygen Species/metabolism , Oxidative Stress , Bacteria/metabolism , Dietary Fiber/pharmacology , Dietary Fiber/metabolism , Lipids
2.
MicroPubl Biol ; 20212021 Jan 14.
Article in English | MEDLINE | ID: mdl-33474532

ABSTRACT

X-linked Adrenoleukodystrophy (X-ALD) is a neurometabolic disorder caused by a defective peroxisomal ABCD1 transporter of very long-chain fatty acids (VLCFAs). We have characterized a nematode model of X-ALD with loss of the pmp-4 gene, the worm orthologue of ABCD1. These mutants recapitulated the key hallmarks of X-ALD and importantly mitochondria targeted antioxidant MitoQ prevented axonal degeneration and locomotor disability. In this study, we further demonstrated that the AWB chemosensory neuron of the pmp-4 mutant worm is defective, both in morphology and function. Interestingly, MitoQ could rescue both the phenotypes. Collectively, our results suggest that C. elegans' chemosensation might provide a novel setting for exploring peroxisomal disease related disorders.​.

3.
Front Bioeng Biotechnol ; 8: 588947, 2020.
Article in English | MEDLINE | ID: mdl-33178678

ABSTRACT

Synucleinopathies are a group of disorders characterized by the accumulation of α-Synuclein amyloid inclusions in the brain. Preventing α-Synuclein aggregation is challenging because of the disordered nature of the protein and the stochastic nature of fibrillogenesis, but, at the same time, it is a promising approach for therapeutic intervention in these pathologies. A high-throughput screening initiative allowed us to discover ZPDm, the smallest active molecule in a library of more than 14.000 compounds. Although the ZPDm structure is highly related to that of the previously described ZPD-2 aggregation inhibitor, we show here that their mechanisms of action are entirely different. ZPDm inhibits the aggregation of wild-type, A30P, and H50Q α-Synuclein variants in vitro and interferes with α-Synuclein seeded aggregation in protein misfolding cyclic amplification assays. However, ZPDm distinctive feature is its strong potency to dismantle preformed α-Synuclein amyloid fibrils. Studies in a Caenorhabditis elegans model of Parkinson's Disease, prove that these in vitro properties are translated into a significant reduction in the accumulation of α-Synuclein inclusions in ZPDm treated animals. Together with previous data, the present work illustrates how different chemical groups on top of a common molecular scaffold can result in divergent but complementary anti-amyloid activities.

4.
Free Radic Biol Med ; 152: 797-809, 2020 05 20.
Article in English | MEDLINE | ID: mdl-32017990

ABSTRACT

Adrenoleukodystrophy is a neurometabolic disorder caused by a defective peroxisomal ABCD1 transporter of very long-chain fatty acids (VLCFAs). Its pathogenesis is incompletely understood. Here we characterize a nematode model of X-ALD with loss of the pmp-4 gene, the worm orthologue of ABCD1. These mutants recapitulate the hallmarks of X-ALD: i) VLCFAs accumulation and impaired mitochondrial redox homeostasis and ii) axonal damage coupled to locomotor dysfunction. Furthermore, we identify a novel role for PMP-4 in modulating lipid droplet dynamics. Importantly, we show that the mitochondria targeted antioxidant MitoQ normalizes lipid droplets size, and prevents axonal degeneration and locomotor disability, highlighting its therapeutic potential. Moreover, PMP-4 acting solely in the hypodermis rescues axonal and locomotion abnormalities, suggesting a myelin-like role for the hypodermis in providing essential peroxisomal functions for the nematode nervous system.


Subject(s)
Adrenoleukodystrophy , ATP Binding Cassette Transporter, Subfamily D, Member 1/genetics , ATP-Binding Cassette Transporters/genetics , Adrenoleukodystrophy/drug therapy , Adrenoleukodystrophy/genetics , Animals , Caenorhabditis elegans/genetics , Fatty Acids , Mice , Mice, Knockout , Subcutaneous Tissue
5.
Front Mol Neurosci ; 12: 306, 2019.
Article in English | MEDLINE | ID: mdl-31920537

ABSTRACT

α-Synuclein (α-Syn) forms toxic intracellular protein inclusions and transmissible amyloid structures in Parkinson's disease (PD). Preventing α-Syn self-assembly has become one of the most promising approaches in the search for disease-modifying treatments for this neurodegenerative disorder. Here, we describe the capacity of a small molecule (ZPD-2), identified after a high-throughput screening, to inhibit α-Syn aggregation. ZPD-2 inhibits the aggregation of wild-type α-Syn and the A30P and H50Q familial variants in vitro at substoichiometric compound:protein ratios. In addition, the molecule prevents the spreading of α-Syn seeds in protein misfolding cyclic amplification assays. ZPD-2 is active against different α-Syn strains and blocks their seeded polymerization. Treating with ZPD-2 two different PD Caenorhabditis elegans models that express α-Syn either in muscle or in dopaminergic (DA) neurons substantially reduces the number of α-Syn inclusions and decreases synuclein-induced DA neurons degeneration. Overall, ZPD-2 is a hit compound worth to be explored in order to develop lead molecules for therapeutic intervention in PD.

6.
Proc Natl Acad Sci U S A ; 115(41): 10481-10486, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30249646

ABSTRACT

Parkinson's disease (PD) is characterized by a progressive loss of dopaminergic neurons, a process that current therapeutic approaches cannot prevent. In PD, the typical pathological hallmark is the accumulation of intracellular protein inclusions, known as Lewy bodies and Lewy neurites, which are mainly composed of α-synuclein. Here, we exploited a high-throughput screening methodology to identify a small molecule (SynuClean-D) able to inhibit α-synuclein aggregation. SynuClean-D significantly reduces the in vitro aggregation of wild-type α-synuclein and the familiar A30P and H50Q variants in a substoichiometric molar ratio. This compound prevents fibril propagation in protein-misfolding cyclic amplification assays and decreases the number of α-synuclein inclusions in human neuroglioma cells. Computational analysis suggests that SynuClean-D can bind to cavities in mature α-synuclein fibrils and, indeed, it displays a strong fibril disaggregation activity. The treatment with SynuClean-D of two PD Caenorhabditis elegans models, expressing α-synuclein either in muscle or in dopaminergic neurons, significantly reduces the toxicity exerted by α-synuclein. SynuClean-D-treated worms show decreased α-synuclein aggregation in muscle and a concomitant motility recovery. More importantly, this compound is able to rescue dopaminergic neurons from α-synuclein-induced degeneration. Overall, SynuClean-D appears to be a promising molecule for therapeutic intervention in Parkinson's disease.


Subject(s)
Amyloid/drug effects , Caenorhabditis elegans/drug effects , Dopaminergic Neurons/drug effects , Parkinson Disease/drug therapy , Protein Aggregation, Pathological/drug therapy , Small Molecule Libraries/pharmacology , alpha-Synuclein/antagonists & inhibitors , Amyloid/metabolism , Animals , Caenorhabditis elegans/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , High-Throughput Screening Assays , Humans , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Neuroblastoma/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Tumor Cells, Cultured , alpha-Synuclein/metabolism
8.
Mol Neurobiol ; 55(9): 7533-7552, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29429047

ABSTRACT

Parkinson's disease is associated with intracellular α-synuclein accumulation and ventral midbrain dopaminergic neuronal death in the Substantia Nigra of brain patients. The Rho GTPase pathway, mainly linking surface receptors to the organization of the actin and microtubule cytoskeletons, has been suggested to participate to Parkinson's disease pathogenesis. Nevertheless, its exact contribution remains obscure. To unveil the participation of the Rho GTPase family to the molecular pathogenesis of Parkinson's disease, we first used C elegans to demonstrate the role of the small GTPase RAC1 (ced-10 in the worm) in maintaining dopaminergic function and survival in the presence of alpha-synuclein. In addition, ced-10 mutant worms determined an increase of alpha-synuclein inclusions in comparison to control worms as well as an increase in autophagic vesicles. We then used a human neuroblastoma cells (M17) stably over-expressing alpha-synuclein and found that RAC1 function decreased the amount of amyloidogenic alpha-synuclein. Further, by using dopaminergic neurons derived from patients of familial LRRK2-Parkinson's disease we report that human RAC1 activity is essential in the regulation of dopaminergic cell death, alpha-synuclein accumulation, participates in neurite arborization and modulates autophagy. Thus, we determined for the first time that RAC1/ced-10 participates in Parkinson's disease associated pathogenesis and established RAC1/ced-10 as a new candidate for further investigation of Parkinson's disease associated mechanisms, mainly focused on dopaminergic function and survival against α-synuclein-induced toxicity.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/physiology , Dopaminergic Neurons/enzymology , alpha-Synuclein/toxicity , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Amyloid/metabolism , Animals , Autophagy/drug effects , Behavior, Animal/drug effects , Biomarkers/metabolism , Caenorhabditis elegans/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dopamine/metabolism , Humans , Inclusion Bodies/metabolism , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Mesencephalon/pathology , Mutation/genetics , Neurites/drug effects , Neurites/metabolism , Neuroprotection/drug effects , Parkinson Disease/pathology
9.
Hum Genet ; 135(12): 1343-1354, 2016 12.
Article in English | MEDLINE | ID: mdl-27541642

ABSTRACT

Classical Rett syndrome (RTT) is a neurodevelopmental disorder where most of cases carry MECP2 mutations. Atypical RTT variants involve mutations in CDKL5 and FOXG1. However, a subset of RTT patients remains that do not carry any mutation in the described genes. Whole exome sequencing was carried out in a cohort of 21 female probands with clinical features overlapping with those of RTT, but without mutations in the customarily studied genes. Candidates were functionally validated by assessing the appearance of a neurological phenotype in Caenorhabditis elegans upon disruption of the corresponding ortholog gene. We detected pathogenic variants that accounted for the RTT-like phenotype in 14 (66.6 %) patients. Five patients were carriers of mutations in genes already known to be associated with other syndromic neurodevelopmental disorders. We determined that the other patients harbored mutations in genes that have not previously been linked to RTT or other neurodevelopmental syndromes, such as the ankyrin repeat containing protein ANKRD31 or the neuronal acetylcholine receptor subunit alpha-5 (CHRNA5). Furthermore, worm assays demonstrated that mutations in the studied candidate genes caused locomotion defects. Our findings indicate that mutations in a variety of genes contribute to the development of RTT-like phenotypes.


Subject(s)
Carrier Proteins/genetics , High-Throughput Nucleotide Sequencing , Nerve Tissue Proteins/genetics , Receptors, Nicotinic/genetics , Rett Syndrome/genetics , Adolescent , Adult , Animals , Caenorhabditis elegans/genetics , Cell Cycle Proteins , Child , Child, Preschool , DNA Mutational Analysis , Exome/genetics , Female , Forkhead Transcription Factors/genetics , Genetic Variation , Humans , Methyl-CpG-Binding Protein 2/genetics , Mutation , Protein Serine-Threonine Kinases/genetics , Rett Syndrome/physiopathology
10.
J Neuropathol Exp Neurol ; 73(1): 81-97, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24335532

ABSTRACT

Tauopathies are degenerative diseases characterized by the accumulation of phosphorylated tau in neurons and glial cells. With some exceptions, tau deposits in neurons are mainly manifested as pretangles and tangles unrelated to the tauopathy. It is thought that abnormal tau deposition in neurons occurs following specific steps, but little is known about the progression of tau pathology in glial cells in tauopathies. We compared tau pathology in different astrocyte phenotypes and oligodendroglial inclusions with that in neurons in a large series of tauopathies, including progressive supranuclear palsy, corticobasal degeneration, argyrophilic grain disease, Pick disease, frontotemporal lobar degenerations (FTLD) associated with mutations in the tau gene, globular glial tauopathy (GGT), and tauopathy in the elderly. Our findings indicate that disease-specific astroglial phenotypes depend on i) the primary amino acid sequence of tau (mutated tau, 3Rtau, and 4Rtau); ii) phospho-specific sites of tau phosphorylation, tau conformation, tau truncation, and ubiquitination in that order (which parallel tau modifications related to pretangle and tangle stages in neurons); and iii) modifications of the astroglial cytoskeleton. In contrast to astrocytes, coiled bodies in oligodendrocytes have similar characteristics whatever the tauopathy, except glial globular inclusions in GGT, and coiled bodies and globular oligodendroglial inclusions in FTLD-tau/K317M. These observations indicate that tau pathology in glial cells largely parallels, but is not identical to, that in neurons in many tauopathies.


Subject(s)
Disease Progression , Neuroglia/pathology , Neurons/pathology , Phenotype , Tauopathies/pathology , tau Proteins , Brain/pathology , Humans , Tauopathies/genetics , tau Proteins/genetics
11.
Ann N Y Acad Sci ; 1126: 315-9, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18079479

ABSTRACT

Nonenzymatic protein modifications are generated from direct oxidation of amino acid side chains and from reaction of the nucleophilic side chains of specific amino acids with reactive carbonyl species. These reactions give rise to specific markers that have been analyzed in different neurodegenerative diseases sharing protein aggregation, such as Alzheimer's disease, Pick's disease, Parkinson's disease, dementia with Lewy bodies, Creutzfeldt-Jakob disease, and amyotrophic lateral sclerosis. Collectively, available data demonstrate that oxidative stress homeostasis, mitochondrial function, and energy metabolism are key factors in determining the disease-specific pattern of protein molecular damage. In addition, these findings suggest the lack of a "gold marker of oxidative stress," and, consequently, they strengthen the need for a molecular dissection of the nonenzymatic reactions underlying neurodegenerative processes.


Subject(s)
Maillard Reaction , Neurodegenerative Diseases/physiopathology , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Humans , Lewy Body Disease/pathology , Lewy Body Disease/physiopathology , Neurodegenerative Diseases/pathology , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Pick Disease of the Brain/pathology , Pick Disease of the Brain/physiopathology , Protein Folding , Proteins/metabolism
12.
Neurobiol Aging ; 29(3): 408-17, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17166629

ABSTRACT

Previous studies in Lewy body diseases (LBDs), including Parkinson's disease (PD) and Dementia with Lewy bodies (DLB), have shown oxidative stress damage more extended than the expected for the distribution of Lewy pathology. Since malondialdehyde (MDA) can form adducts with lysine residues of proteins, MDA-Lys immunoprecipitation and alpha-synuclein immunoblotting has been carried out in frontal cortex and substantia nigra homogenates from five patients with PD, five DLB, three iPD and seven aged-matched controls to decipher the extent of lipoxidized alpha-synuclein in LBDs. MDA-Lys-lipoxidation of alpha-synuclein in the substantia nigra and frontal cortex has been found in all DLB and PD cases examined, but also in the frontal cortex in 3/3 and in the substantia nigra in 2/3 cases with iPD. In addition, one control case had MDA-Lys-modified alpha-synuclein in the frontal cortex, and another in the substantia nigra. This work provides evidence of extended lipoxidative modification of alpha-synuclein in LBDs. Moreover, it demonstrates that alpha-synuclein lipoxidation is an early event in LBDs which precedes alpha-synuclein solubility modification and aggregation, and formation of Lewy bodies and neurites.


Subject(s)
Lewy Body Disease/pathology , Neocortex/metabolism , alpha-Synuclein/metabolism , Aged , Aged, 80 and over , Female , Humans , Immunoprecipitation , Lysine/analogs & derivatives , Lysine/pharmacology , Male , Middle Aged , Oxidation-Reduction/drug effects , Parkinson Disease/metabolism , Parkinson Disease/pathology
13.
Brain ; 130(Pt 12): 3111-23, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17716997

ABSTRACT

The occurrence of endoplasmic reticulum (ER) stress in the sporadic form of amyotrophic lateral sclerosis (ALS) is unknown, despite it has been recently documented in experimental models of the familial form. Here we show that spinal cord from patients with sporadic ALS showed signs of ER stress, such as increased levels of ER chaperones such as protein-disulfide isomerase, and increased phosphorylation of eukaryotic initiation factor 2alpha (eIF2alpha). Among the potential causes of such ER stress proteasomal impairment was confirmed in the same samples by demonstrating increased ubiquitin immunoreactivity and increased protein lipoxidative (125%), glycoxidative (55%) and direct oxidative damage (62%) over control values, as evidenced by mass-spectrometry and immunological methods. We found that protein oxidative damage was strongly associated to ALS-specific changes in fatty acid concentrations, specifically of n-3 series (as docosahexaenoic acid), and in the amount of mitochondrial components as respiratory complexes I and III, suggesting a mitochondrial dysfunction leading to increased free radical production. Oxidative stress was also evidenced in frontal cortex, suggesting that this region is affected early in ALS. As those events were partially reproduced by threohydroxyaspartate exposure in organotypic spinal cord cultures, we concluded that changes in fatty acid composition, mitochondrial function and proteasome activity, which may be driven by excitotoxicity, lead to oxidative stress and finally contribute to ER stress in sporadic ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Endoplasmic Reticulum/physiology , Adult , Aged , Amyotrophic Lateral Sclerosis/metabolism , Fatty Acids/analysis , Female , Frontal Lobe/chemistry , Humans , Male , Middle Aged , Mitochondria/physiology , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/chemistry , Organ Culture Techniques , Oxidation-Reduction , Oxidative Stress , Proteasome Endopeptidase Complex/physiology , Spinal Cord/chemistry , Spinal Cord/physiopathology , Ubiquitin/metabolism
14.
Neurobiol Dis ; 24(2): 403-18, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16963267

ABSTRACT

Accumulating evidence has suggested that neurotrophins participate in the pathophysiology of mood disorders. We have developed transgenic mice overexpressing the full-length neurotrophin-3 receptor TrkC (TgNTRK3) in the central nervous system. TgNTRK3 mice show increased anxiety-like behavior and enhancement of panic reaction in the mouse defense test battery, along with an increase in the number and density of catecholaminergic (tyrosine hydroxylase positive) neurons in locus coeruleus and substantia nigra. Furthermore, treatment of TgNTRK3 mice with diazepam significantly attenuated the anxiety-like behaviors in the plus maze. These results provide evidence for the involvement of TrkC in the development of noradrenergic neurons in the central nervous system with consequences on anxiety-like behavior and panic reaction. Thus, changes in TrkC expression levels could contribute to the phenotypic expression of panic disorder through a trophic effect on noradrenergic neurons in the locus coeruleus. Our results demonstrate that the elevated NT3-TrkC tone via overexpression of TrkC in the brain may constitute a molecular mechanism for the expression of anxiety and anxiety.


Subject(s)
Anxiety Disorders/metabolism , Brain/metabolism , Brain/physiopathology , Catecholamines/metabolism , Neurons/metabolism , Panic Disorder/metabolism , Receptor, trkC/metabolism , Animals , Anxiety Disorders/genetics , Anxiety Disorders/physiopathology , Autonomic Nervous System Diseases/genetics , Autonomic Nervous System Diseases/metabolism , Autonomic Nervous System Diseases/physiopathology , Behavior, Animal/physiology , Brain/pathology , Cell Count , Cell Proliferation , Disease Models, Animal , Female , Genetic Predisposition to Disease/genetics , Locus Coeruleus/metabolism , Locus Coeruleus/pathology , Locus Coeruleus/physiopathology , Male , Mice , Mice, Transgenic , Neural Pathways/metabolism , Neural Pathways/pathology , Neural Pathways/physiopathology , Neuropsychological Tests , Norepinephrine/metabolism , Panic Disorder/genetics , Panic Disorder/physiopathology , Receptor, trkC/genetics , Substantia Nigra/metabolism , Substantia Nigra/pathology , Substantia Nigra/physiopathology , Up-Regulation/genetics
15.
Neurosci Lett ; 400(1-2): 125-9, 2006 May 29.
Article in English | MEDLINE | ID: mdl-16516382

ABSTRACT

Abnormal solubility and aggregation of alpha-synuclein have been observed in the frontal cortex in three cases with Pick's disease (PiD) when compared with age-matched controls. Bands of 45 kDa and higher molecular weight were detected in the SDS-soluble fractions only in PiD. Patterns in PiD differed from that observed in the cerebral cortex in Lewy body diseases which were examined in parallel. Immunoblots to alpha-synuclein nitrated in tyrosines revealed bands of 45 and 60 kDa in Dxc- and SDS-soluble fractions in the frontal cortex (which is vulnerable to PiD) but not in the occipital cortex (which is resistant to this degenerative disease). Moreover, nitrated alpha-synuclein was found in Lewy bodies and neurites in synucleinopathies but diffusely in the cytoplasm of scattered neurons in PiD. These findings demonstrate abnormal and distinct alpha-synuclein solubility and aggregation, and alpha-synuclein nitration without formation of Lewy bodies in the frontal cortex in PiD.


Subject(s)
Frontal Lobe/metabolism , Pick Disease of the Brain/metabolism , Pick Disease of the Brain/pathology , alpha-Synuclein/metabolism , Aged , Blotting, Western/methods , Case-Control Studies , Female , Humans , Immunohistochemistry/methods , Male , Molecular Weight , Parkinson Disease/metabolism , Postmortem Changes , Tyrosine/metabolism
16.
Neurobiol Aging ; 27(6): 848-56, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16006012

ABSTRACT

In addition to genetic factors, environmental factors have long been suspected to contribute to the pathogenesis of Parkinson's disease (PD). We investigated the possible interaction between genetic factors and neurotoxins by testing whether alpha-synuclein A30P Tg5093 transgenic mice show increased sensitivity to secondary toxic insults like 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or rotenone. While sensitivity to chronic treatment with rotenone was not enhanced in the Tg5093 line, chronic treatment with 80 or 150 mg/kg MPTP resulted in increased deterioration of the nigrostriatal dopaminergic system as assessed by quantitation of nigral tyrosine hydroxylase (TH) positive neurons and striatal dopamine (DA) levels in Tg5093 mice when compared to non-transgenic littermate controls. Thus, the results of this study demonstrate a role for the overexpression of mutant human alpha-synuclein A30P in increased vulnerability of DA neurons to MPTP.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Alanine/genetics , Neurotoxins/pharmacology , Proline/genetics , Substantia Nigra , alpha-Synuclein/genetics , Age Factors , Animals , Behavior, Animal/drug effects , Dopamine/metabolism , Dose-Response Relationship, Drug , Humans , Immunohistochemistry/methods , Insecticides/pharmacology , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Rotenone/pharmacology , Substantia Nigra/cytology , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Time Factors , Tyrosine 3-Monooxygenase/metabolism
17.
Neurobiol Dis ; 22(2): 265-73, 2006 May.
Article in English | MEDLINE | ID: mdl-16380264

ABSTRACT

Parkinson disease (PD) and dementia with Lewy bodies (DLB) are characterized by the accumulation of abnormal alpha-synuclein and ubiquitin in protein aggregates conforming Lewy bodies and Lewy neurites. Ubiquitin C-terminal hydrolase-1 (UCHL-1) disassembles polyubiquitin chains to increase the availability of free monomeric ubiquitin to the ubiquitin proteasome system (UPS) thus favoring protein degradation. Since mutations in the UCHL-1 gene, reducing UPS activity by 50%, have been reported in autosomal dominant PD, and UCHL-1 inhibition results in the formation of alpha-synuclein aggregates in mesencephalic cultured neurons, the present study was initiated to test UCHL-1 mRNA and protein levels in post-mortem frontal cortex (area 8) of PD and DLB cases, compared with age-matched controls. TaqMan PCR assays, and Western blots demonstrated down-regulation of UCHL-1 mRNA and UCHL-1 protein in the cerebral cortex in DLB (either in pure forms, not associated with Alzheimer disease: AD, and in common forms, with accompanying AD changes), but not in PD, when compared with age-matched controls. Interestingly, UCHL-1 mRNA and protein expressions were reduced in the medulla oblongata in the same PD cases. Moreover, UCHL-1 protein was decreased in the substantia nigra in cases with Lewy body pathology. UCHL-1 down-regulation was not associated with reduced protein levels of several proteasomal subunits, including 20SX, 20SY, 19S and 11Salpha. Yet UCHL-3 expression was reduced in the cerebral cortex of PD and DLB patients. Together, these observations show reduced UCHL-1 expression as a contributory factor in the abnormal protein aggregation in DLB, and points UCHL-1 as a putative therapeutic target in the treatment of DLB.


Subject(s)
Brain/enzymology , Down-Regulation/physiology , Lewy Body Disease/enzymology , Neurons/enzymology , Ubiquitin Thiolesterase/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/enzymology , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Brain/pathology , Brain/physiopathology , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Female , Humans , Lewy Bodies/enzymology , Lewy Bodies/genetics , Lewy Body Disease/genetics , Lewy Body Disease/physiopathology , Male , Medulla Oblongata/enzymology , Medulla Oblongata/pathology , Medulla Oblongata/physiopathology , Middle Aged , Neurons/pathology , Parkinson Disease/enzymology , Parkinson Disease/genetics , Parkinson Disease/physiopathology , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/metabolism , Signal Transduction/physiology , Substantia Nigra/enzymology , Substantia Nigra/pathology , Substantia Nigra/physiopathology , Ubiquitin/metabolism , Ubiquitin Thiolesterase/genetics
18.
J Neuropathol Exp Neurol ; 64(9): 816-30, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16141792

ABSTRACT

Oxidative stress has been well documented in the substantia nigra in Parkinson disease (PD), but little is known about oxidative damage, particularly lipoxidation, advanced glycation (AGE), and AGE receptors (RAGE) in other structures, including the cerebral cortex, in early stages of diseases with Lewy bodies. The present study was undertaken to analyze these parameters in the frontal cortex (area 8), amygdala, and substantia nigra in selected cases with no neurologic symptoms and with neuropathologically verified incidental Lewy body disease-related changes, comparing them with healthy age-matched individuals. Results of the present study have shown mass spectrometric and immunologic evidences of increased lipoxidative damage by the markers malondialdehyde-lysine (MDAL) and 4-hydroxynonenal-lysine (HNE), increased expression of AGE in the substantia nigra, amygdala, and frontal cortex, and increased and heterogeneous RAGE cellular expression in the substantia nigra and frontal cortex in cases with early stages of parkinsonian neuropathology. In addition, increased content of the highly peroxidizable docosahexaenoic acid in the amygdala and frontal cortex. These changes were not associated to alpha-synuclein aggregation in cortex, contrasting with aggregates found in SDS-soluble fractions of frontal cortex in dementia with Lewy bodies (DLB) cases. The pattern of lipidic abnormalities differed in DLB and incidental Lewy body disease. Furthermore, although AGE and RAGE expression were raised in DLB, no increase in the total amount of HNE and MDAL adducts was found in the cerebral cortex in DLB. Preliminary analyses have identified 2 proteins with lipoxidative damage, alpha-synuclein and manganese superoxide dismutase (SOD2), in incidentally Lewy body disease cortex. This study demonstrates abnormal fatty acid profiles, increased and selective lipoxidative damage, and increased AGE and RAGE expression in the frontal cortex in cases with early stages of parkinsonian neuropathology without treatment. These findings further support antioxidant therapy in the treatment of PD to reduce cortical damage associated with oxidative stress.


Subject(s)
Lewy Body Disease/pathology , Neocortex/pathology , Oxidative Stress/physiology , Aged , Aged, 80 and over , Aldehydes/metabolism , Blotting, Western , Docosahexaenoic Acids/metabolism , Electrophoresis, Gel, Two-Dimensional , Fatty Acids/analysis , Fatty Acids/metabolism , Female , Glycation End Products, Advanced/metabolism , Humans , Immunohistochemistry , Lewy Body Disease/metabolism , Male , Malondialdehyde/metabolism , Mass Spectrometry , Neocortex/metabolism , Nerve Tissue Proteins/metabolism , Parkinson Disease/metabolism , Parkinson Disease/pathology , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Synucleins , alpha-Synuclein
19.
J Neuropathol Exp Neurol ; 64(7): 638-47, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16042316

ABSTRACT

Group I metabotropic glutamate receptors (mGluR1) regulate synaptic transmission through the stimulation of phospholipase Cbeta1 (PLCbeta1) and then by the activation of protein kinase C (PKC). Considering these properties, it is conceivable that major cortical functional deficits may be attributed to abnormal mGluR processing and signaling. The present work examines mGluRI expression and signaling in the frontal cortex (area 8) of 3 cases with Pick disease (PiD), a neurodegenerative disease with abnormal phospho-tau accumulation, in comparison with 3 age-matched controls by means of glutamate binding assays, enzymatic activity, gel electrophoresis and Western blotting, solubility and immunoprecipitation assays, and confocal microscopy. Reduced expression levels of PLCbeta1 and reduced PLCbeta1 activity have been found in PiD. The expression levels of the nonrelated phospholipase PLCgamma, a substrate of tyrosine kinase, are also reduced in PiD. This is accompanied by a marked decrease in the expression of cPKCalpha and increased expression of the inner band (76 kDa) of the nPKCdelta doublet at the expense of a decrease of the phosphorylated (active) form (78 kDa). In contrast, L-[3H]glutamate-specific binding to mGluRs is augmented in PiD cases, mainly because of the higher mGluR1 and mGluRs expression levels detected. No modifications in PLCbeta1 solubility have been observed in PiD and no interactions between PLCbeta1 and tau have been demonstrated in diseased and control cases. Moreover, double-labeling immunofluorescence and confocal microscopy have shown no colocalization of phospho-tau (AT8 antibody) and PLCbeta1 in phospho-tau inclusions, including Pick bodies. These results demontrate for the first time abnormal mGluR signaling in the cerebral cortex in PiD and selective vulnerability of phospholipases and PKC to PiD.


Subject(s)
Frontal Lobe/metabolism , Pick Disease of the Brain/metabolism , Receptors, Metabotropic Glutamate/metabolism , Aged , Blotting, Western , Female , Fluorescent Antibody Technique , Frontal Lobe/pathology , Glutamic Acid/metabolism , Humans , Immunoprecipitation , Isoenzymes/metabolism , Male , Microscopy, Confocal , Phospholipase C beta , Phospholipase C gamma , Pick Disease of the Brain/pathology , Protein Kinase C/metabolism , Protein Kinase C-alpha , Protein Kinase C-delta , Type C Phospholipases/metabolism
20.
Neurobiol Dis ; 20(3): 685-93, 2005 Dec.
Article in English | MEDLINE | ID: mdl-15949941

ABSTRACT

The aim of the present work was to analyze the status of metabotropic glutamate receptors (mGluRs) in the frontal cortex (area 8) from ten cases with common form DLB (cDLB) and eleven cases with pure AD in comparison with five age-matched controls. mGluRs, determined by radioligand binding assays, were significantly decreased in cerebral cortex in cDLB. This decrease was already present in cases with early AD changes not involving the frontal cortex, but dramatically correlated with AD neuropathological changes, at its greatest in isocortical stages, which was associated with a decrease in the expression levels of mGluR1 detected by Western blotting. Moreover, mGluRs analyzed in pure AD were lower than those obtained in cDLB and also correlated with progression of illness. On the other hand, the expression levels of phospholipase Cbeta1 (PLCbeta1) isoform, which is the effector of group I mGluRs, was decreased in parallel in cDLB cases. Finally, the PLCbeta1 decrease was associated with reduced GTP- and l-glutamate-stimulated PLC activity in both cDLB and AD cases. These results show that group I mGluRs/PLC signaling are down-regulated and desensitized in the frontal cortex in cDLB and AD cases and that these modifications worsen with progression of AD changes in the cerebral neocortex. Therefore, group I mGluR dysfunction may be implicated in the pathogenesis of cognitive impairment and dementia in common form of DLB and pure AD.


Subject(s)
Alzheimer Disease/enzymology , Frontal Lobe/enzymology , Isoenzymes/metabolism , Lewy Body Disease/enzymology , Neurons/enzymology , Receptors, Metabotropic Glutamate/metabolism , Type C Phospholipases/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Binding, Competitive/physiology , Disease Progression , Down-Regulation/physiology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Female , Frontal Lobe/pathology , Frontal Lobe/physiopathology , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Guanosine Triphosphate/metabolism , Guanosine Triphosphate/pharmacology , Humans , Isoenzymes/drug effects , Lewy Body Disease/pathology , Lewy Body Disease/physiopathology , Male , Middle Aged , Neurons/pathology , Phospholipase C beta , Radioligand Assay , Signal Transduction/physiology , Type C Phospholipases/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...