Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Neurobiol Dis ; 137: 104739, 2020 04.
Article in English | MEDLINE | ID: mdl-31927145

ABSTRACT

The toxic conformer of amyloid ß-protein (Aß) ending at 42 (Aß42), which contains a unique turn conformation at amino acid residue positions 22 and 23 and tends to form oligomers that are neurotoxic, was reported to play a critical role in the pathomechanisms of Alzheimer's disease (AD), in which diabetes mellitus (DM)-like mechanisms are also suggested to be operative. It remains to be established whether the attenuation of insulin signaling is involved in an increase of toxic Aß42 conformer levels. The present study investigated the association between impaired insulin metabolism and formation of toxic Aß42 conformers in the brains of an AD mouse model. In particular, we studied whether insulin deficiency or resistance affected the formation of toxic Aß42 conformers in vivo. We induced insulin deficiency and resistance in 3xTg-AD mice, a mouse AD model harboring two familial AD-mutant APP (KM670/671NL) and PS1 (M146 V) genes and a mutant TAU (P301L) gene, by streptozotocin (STZ) injection and a high fructose diet (HFuD), respectively. Cognitive impairment was significantly worsened by STZ injection but not by HFuD. Dot blot analysis revealed significant increases in total Aß42 levels and the ratio of toxic Aß42 conformer/total Aß42 in STZ-treated mice compared with control and HFuD-fed mice. Immunostaining showed the accumulation of toxic Aß42 conformers and hyper-phosphorylated tau protein (p-tau), which was more prominent in the cortical and hippocampal neurons of STZ-treated mice compared with HFuD-fed and control mice. HFuD-fed mice showed only a mild-to-moderate increase of these proteins compared with controls. Toxic Aß42 conformers were co-localized with p-tau oligomers (Pearson's correlation coefficient = 0.62) in the hippocampus, indicating their co-aggregation. Toxic Aß42 conformer levels were inversely correlated with pancreatic insulin secretion capacity as shown by fasting immunoreactive insulin levels in STZ-treated mice (correlation coefficient = -0.5879, p = .04441), but not HFuD-fed mice, suggesting a decrease in serum insulin levels correlates with toxic Aß42 conformer formation. Levels of p-Akt and phosphorylated glycogen synthase kinase-3ß measured by a homogeneous time-resolved fluorescence assay were significantly lower in STZ-treated mice than in HFuD-fed mice, suggesting a greater inhibition of brain insulin signaling by STZ than HFuD, although both levels were significantly decreased in these groups compared with controls. Iba1-positive and NOS2-positive areas in the cortex and hippocampus were significantly increased in STZ-treated mice and to a lesser extent in HFuD-fed mice compared with controls. These findings suggest that insulin deficiency rather than insulin resistance and the resultant impairment of brain insulin signaling facilitates the formation of toxic Aß42 conformer and its co-aggregation with p-tau oligomers, and that insulin deficiency is an important pathogenic factor in the progression of AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Cognitive Dysfunction/metabolism , tau Proteins/metabolism , Alzheimer Disease/genetics , Animals , Brain/metabolism , Cognitive Dysfunction/genetics , Disease Models, Animal , Insulin/metabolism , Mice, Transgenic , Neurons/metabolism , Peptide Fragments/metabolism
2.
Nat Commun ; 7: 10332, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26755131

ABSTRACT

The etiology of idiopathic Parkinson's disease (idPD) remains enigmatic despite recent successes in identification of genes (PARKs) that underlie familial PD. To find new keys to this incurable neurodegenerative disorder we focused on the poorly understood PARK14 disease locus (Pla2g6 gene) and the store-operated Ca(2+) signalling pathway. Analysis of the cells from idPD patients reveals a significant deficiency in store-operated PLA2g6-dependent Ca(2+) signalling, which we can mimic in a novel B6.Cg-Pla2g6(ΔEx2-VB) (PLA2g6 ex2(KO)) mouse model. Here we demonstrate that genetic or molecular impairment of PLA2g6-dependent Ca(2+) signalling is a trigger for autophagic dysfunction, progressive loss of dopaminergic (DA) neurons in substantia nigra pars compacta and age-dependent L-DOPA-sensitive motor dysfunction. Discovery of this previously unknown sequence of pathological events, its association with idPD and our ability to mimic this pathology in a novel genetic mouse model opens new opportunities for finding a cure for this devastating neurodegenerative disease.


Subject(s)
Brain/metabolism , Calcium Signaling/genetics , Dopaminergic Neurons/metabolism , Group VI Phospholipases A2/genetics , Movement , Parkinson Disease/genetics , Aged , Animals , Blotting, Western , Brain/pathology , Dopaminergic Neurons/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Group VI Phospholipases A2/metabolism , Humans , Induced Pluripotent Stem Cells , Mice , Mice, Transgenic , Microscopy, Confocal , Microscopy, Fluorescence , Middle Aged , Motor Skills , Parkinson Disease/metabolism , Parkinson Disease/pathology , Reverse Transcriptase Polymerase Chain Reaction , Skin/cytology , Young Adult
3.
Nat Neurosci ; 18(11): 1584-93, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26436904

ABSTRACT

Accumulation of pathological tau protein is a major hallmark of Alzheimer's disease. Tau protein spreads from the entorhinal cortex to the hippocampal region early in the disease. Microglia, the primary phagocytes in the brain, are positively correlated with tau pathology, but their involvement in tau propagation is unknown. We developed an adeno-associated virus-based model exhibiting rapid tau propagation from the entorhinal cortex to the dentate gyrus in 4 weeks. We found that depleting microglia dramatically suppressed the propagation of tau and reduced excitability in the dentate gyrus in this mouse model. Moreover, we demonstrate that microglia spread tau via exosome secretion, and inhibiting exosome synthesis significantly reduced tau propagation in vitro and in vivo. These data suggest that microglia and exosomes contribute to the progression of tauopathy and that the exosome secretion pathway may be a therapeutic target.


Subject(s)
Exosomes/metabolism , Hippocampus/metabolism , Microglia/metabolism , Neurons/metabolism , tau Proteins/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Disease Models, Animal , Entorhinal Cortex/pathology , Hippocampus/pathology , Male , Mice, Transgenic
4.
J Neurosci ; 35(26): 9764-81, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26134658

ABSTRACT

Peripheral and CNS inflammation leads to aberrations in developmental and postnatal neurogenesis, yet little is known about the mechanism linking inflammation to neurogenic abnormalities. Specific miRs regulate peripheral and CNS inflammatory responses. miR-155 is the most significantly upregulated miR in primary murine microglia stimulated with lipopolysaccharide (LPS), a proinflammatory Toll-Like Receptor 4 ligand. Here, we demonstrate that miR-155 is essential for robust IL6 gene induction in microglia under LPS stimulation in vitro. LPS-stimulated microglia enhance astrogliogenesis of cocultured neural stem cells (NSCs), whereas blockade of IL6 or genetic ablation of microglial miR-155 restores neural differentiation. miR-155 knock-out mice show reversal of LPS-induced neurogenic deficits and microglial activation in vivo. Moreover, mice with transgenic elevated expression of miR-155 in nestin-positive neural and hematopoietic stem cells, including microglia, show increased cell proliferation and ectopically localized doublecortin-positive immature neurons and radial glia-like cells in the hippocampal dentate gyrus (DG) granular cell layer. Microglia have proliferative and neurogenic effects on NSCs, which are significantly altered by microglial miR-155 overexpression. In addition, miR-155 elevation leads to increased microglial numbers and amoeboid morphology in the DG. Our study demonstrates that miR-155 is essential for inflammation-induced neurogenic deficits via microglial activation and induction of IL6 and is sufficient for disrupting normal hippocampal development.


Subject(s)
Gene Expression Regulation/genetics , Hippocampus/pathology , Inflammation/genetics , Inflammation/pathology , MicroRNAs/metabolism , Neurogenesis/genetics , Animals , Calcium-Binding Proteins/metabolism , Cell Culture Techniques , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Coculture Techniques , Disease Models, Animal , Doxycycline/pharmacology , Embryo, Mammalian , Female , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/drug effects , Inflammation/chemically induced , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-6/metabolism , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Microfilament Proteins/metabolism , Nestin/genetics , Nestin/metabolism , Neurogenesis/drug effects , Pregnancy
5.
Ann Neurol ; 76(3): 379-92, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25043464

ABSTRACT

OBJECTIVE: Much of the genetic basis for Alzheimer disease (AD) is unexplained. We sought to identify novel AD loci using a unique family-based approach that can detect robust associations with infrequent variants (minor allele frequency < 0.10). METHODS: We conducted a genome-wide association study in the Framingham Heart Study (discovery) and NIA-LOAD (National Institute on Aging-Late-Onset Alzheimer Disease) Study (replication) family-based cohorts using an approach that accounts for family structure and calculates a risk score for AD as the outcome. Links between the most promising gene candidate and AD pathogenesis were explored in silico as well as experimentally in cell-based models and in human brain. RESULTS: Genome-wide significant association was identified with a PLXNA4 single nucleotide polymorphism (rs277470) located in a region encoding the semaphorin-3A (SEMA3A) binding domain (meta-analysis p value [meta-P] = 4.1 × 10(-8) ). A test for association with the entire region was also significant (meta-P = 3.2 × 10(-4) ). Transfection of SH-SY5Y cells or primary rat neurons with full-length PLXNA4 (TS1) increased tau phosphorylation with stimulated by SEMA3A. The opposite effect was observed when cells were transfected with shorter isoforms (TS2 and TS3). However, transfection of any isoform into HEK293 cells stably expressing amyloid ß (Aß) precursor protein (APP) did not result in differential effects on APP processing or Aß production. Late stage AD cases (n = 9) compared to controls (n = 5) had 1.9-fold increased expression of TS1 in cortical brain tissue (p = 1.6 × 10(-4) ). Expression of TS1 was significantly correlated with the Clinical Dementia Rating score (ρ = 0.75, p = 2.2 × 10(-4) ), plaque density (ρ = 0.56, p = 0.01), and Braak stage (ρ = 0.54, p = 0.02). INTERPRETATION: Our results indicate that PLXNA4 has a role in AD pathogenesis through isoform-specific effects on tau phosphorylation.


Subject(s)
Alzheimer Disease/genetics , Frontal Lobe/metabolism , Receptors, Cell Surface/metabolism , tau Proteins/metabolism , Aged , Aged, 80 and over , Amyloid beta-Protein Precursor/metabolism , Animals , Cohort Studies , Female , Frontal Lobe/pathology , Genetic Predisposition to Disease , Genome-Wide Association Study , HEK293 Cells , Hippocampus/cytology , Hippocampus/metabolism , Humans , Male , Pedigree , Phosphorylation/genetics , Phosphorylation/physiology , Polymorphism, Single Nucleotide/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Receptors, Cell Surface/genetics
6.
Am J Pathol ; 184(3): 808-18, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24418258

ABSTRACT

Tau-tubulin kinase-1 (TTBK1) is a central nervous system (CNS)-specific protein kinase implicated in the pathological phosphorylation of tau. TTBK1-transgenic mice show enhanced neuroinflammation in the CNS. Double-transgenic mice expressing TTBK1 and frontotemporal dementia with parkinsonism-17-linked P301L (JNPL3) tau mutant (TTBK1/JNPL3) show increased accumulation of oligomeric tau protein in the CNS and enhanced loss of motor neurons in the ventral horn of the lumbar spinal cord. To determine the role of TTBK1-induced neuroinflammation in tauopathy-related neuropathogenesis, age-matched TTBK1/JNPL3, JNPL3, TTBK1, and non-transgenic littermates were systematically characterized. There was a striking switch in the activation phenotype and population of mononuclear phagocytes (resident microglia and infiltrating macrophages) in the affected spinal cord region: JNPL3 mice showed accumulation of alternatively activated microglia, whereas TTBK1 and TTBK1/JNPL3 mice showed accumulation of classically activated infiltrating peripheral monocytes. In addition, expression of chemokine ligand 2, a chemokine important for the recruitment of peripheral monocytes, was enhanced in TTBK1 and TTBK1/JNPL3 but not in other groups in the spinal cord. Furthermore, primary cultured mouse motor neurons showed axonal degeneration after transient expression of the TTBK1 gene or treatment with conditioned media derived from lipopolysaccharide-stimulated microglia; this was partially blocked by silencing of the endogenous TTBK1 gene in neurons. These data suggest that TTBK1 accelerates motor neuron neurodegeneration by recruiting proinflammatory monocytes and enhancing sensitivity to neurotoxicity in inflammatory conditions.


Subject(s)
Frontotemporal Dementia/genetics , Nerve Degeneration/genetics , Protein Serine-Threonine Kinases/genetics , tau Proteins/genetics , Animals , Disease Models, Animal , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Mice , Mice, Transgenic , Mutation , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurons/metabolism , Neurons/pathology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Spinal Cord/pathology , Tauopathies/genetics , Tauopathies/metabolism , Tauopathies/pathology , tau Proteins/metabolism
7.
Biochem Biophys Res Commun ; 391(1): 800-5, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19945426

ABSTRACT

Intranuclear events due to mutations in the Parkin gene remain elusive in autosomal recessive juvenile parkinsonism (ARJP). We identified a mutant PARKIN protein in fibroblast cultures from a pair of siblings with ARJP who were homozygous for the exon 4-deleted Parkin gene. Disease was mild in one patient and debilitating in the other. The detected mutant, encoded by a transcript lacking exon 3 as well as exon 4, is an in-frame deletion that removes 121 aa, resulting in a 344-aa protein (PaDel3,4). Cell culture and transfection studies revealed negative correlations between expression levels of PaDel3,4 and those of cell cycle proteins, including cyclin E, CDK2, ppRb, and E2F-1, and demonstrated that GFP-PaDel3,4 entered nucleus and ubiquitinated cyclin E as a part of SCF(hSel-10) ligase complex in the patient cells. In addition, nuclear localization signal-tagged PaDel3,4 expressed in the transfected patient cells most effectively ubiquitinated cyclin E and reduced DNA damage, protecting cells from oxidative stress. Antisense-oligonucleotide treatment promoted skipping of exon 3 and thus generated PaDel3,4, increasing cell survival. Collectively, we propose that naturally- and experimentally-induced exon skipping at least partly restores the mutant Parkin gene deficit, providing a molecular basis for the development of therapeutic exon skipping.


Subject(s)
Exons/genetics , Fibroblasts/metabolism , Parkinsonian Disorders/genetics , Ubiquitin-Protein Ligases/genetics , Aged , Buthionine Sulfoximine/pharmacology , Cell Survival , Cells, Cultured , Cyclin E/metabolism , DNA Damage/genetics , Female , Fibroblasts/drug effects , Gene Deletion , Humans , Male , Oligonucleotides, Antisense/genetics , Open Reading Frames/genetics , Oxidative Stress/genetics , SKP Cullin F-Box Protein Ligases , Transfection
8.
Hum Mol Genet ; 18(19): 3533-43, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19561170

ABSTRACT

Spinocerebellar ataxia type 14 (SCA14) is an autosomal dominant disease caused by mutations in the gene encoding protein kinase C gamma (PKC gamma). We report an SCA14 family with a novel deletion of a termination-codon-containing region, resulting in a missense change and a C-terminal 13-amino-acid extension with increased kinase activity. Notably, one patient with a severe phenotype is the first homozygote for the mutation causing SCA14. We show the novel molecular consequences of increased kinase activities of mutants: aprataxin (APTX), a DNA repair protein causative for autosomal recessive ataxia, was found to be a preferential substrate of mutant PKC gamma, and phosphorylation inhibited its nuclear entry. The phosphorylated residue was Thr111, located adjacent to the nuclear localization signal, and disturbed interactions with importin alpha, a nuclear import adaptor. Decreased nuclear APTX increased oxidative stress-induced DNA damage and cell death. Phosphorylation-resistant APTX, kinase inhibitors, and antioxidants may be therapeutic options for SCA14.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Down-Regulation , Nuclear Proteins/metabolism , Protein Kinase C/metabolism , Spinocerebellar Ataxias/metabolism , Active Transport, Cell Nucleus , Aged, 80 and over , Amino Acid Sequence , Base Sequence , Cell Line , Cell Nucleus/chemistry , Cell Nucleus/genetics , DNA Damage , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Female , Humans , Male , Middle Aged , Molecular Sequence Data , Mutation, Missense , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Pedigree , Phosphorylation , Protein Kinase C/chemistry , Protein Kinase C/genetics , Protein Transport , Spinocerebellar Ataxias/genetics , Young Adult
9.
Clin Neurol Neurosurg ; 111(4): 341-4, 2009 May.
Article in English | MEDLINE | ID: mdl-19097685

ABSTRACT

OBJECTIVES: Sleep attacks (SAs) in Parkinson's disease (PD) are rare, but clinically important because they significantly impair the daily lives of patients. Causes of SAs include long-term activation of dopaminergic (especially D3) receptors. Recent studies suggest that SAs in PD may be related to impairment of hypothalamic orexin neurons, similar to narcolepsy. Whether orexin is associated with long-term activation of dopaminergic receptors remains uncertain. PATIENTS AND METHODS: We measured levels of orexin in samples of spinal cerebrospinal fluid (CSF) from 25 patients with PD, including 9 with excessive daytime sleepiness and 4 with SAs. Furthermore, in the four patients with SAs, the selective dopamine D1/D2 agonist pergolide was substituted for the causative drugs with D3 stimulatory activity, and CSF-orexin levels were measured before and after switching treatment. RESULTS: In the 25 patients with PD, including the 4 patients with SAs, lower CSF-orexin levels were associated with a longer disease duration, which has been linked to a higher incidence of SAs. Switching treatment to pergolide significantly increased CSF-orexin levels and completely resolved SAs in the four patients with PD. CONCLUSION: Despite the small number of patients studied, our results suggest that orexin transmission is most likely involved in SAs in PD and that abrogation of D3 receptor stimulation may increase orexin and thereby inhibit SAs.


Subject(s)
Dopamine Agonists/pharmacology , Intracellular Signaling Peptides and Proteins/cerebrospinal fluid , Neuropeptides/cerebrospinal fluid , Parkinson Disease/cerebrospinal fluid , Pergolide/pharmacology , Sleep Wake Disorders/cerebrospinal fluid , Sleep Wake Disorders/etiology , Adult , Aged , Female , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Middle Aged , Neuropeptides/metabolism , Orexins , Parkinson Disease/drug therapy , Parkinson Disease/physiopathology , Pergolide/administration & dosage , Radioimmunoassay , Receptors, Dopamine D1/agonists , Receptors, Dopamine D2/agonists , Receptors, Dopamine D3/metabolism , Severity of Illness Index , Sleep Wake Disorders/metabolism
10.
Clin Neurol Neurosurg ; 111(1): 102-4, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18938027

ABSTRACT

Bardet-Biedl syndrome (BBS) is an autosomal recessive disorder characterized by rod-cone dystrophy, polydactyly, central obesity, mental retardation, and hypogonadism. Although many organs are involved in BBS, hyperammonemia caused by portal hypertension has been reported previously in only a single patient. We describe the second such patient with BBS and hyperammonemia, associated with fluctuating mental impairment. The patient was a 17-year-old boy with BBS. Esophageal, gastric, and rectal varices and mild hepatic dysfunction started to develop at 5 years of age. A liver biopsy showed dilated portal veins with mild fibrosis in portal tract. From the age of 17 years, he often had forced laughter with apparently normal consciousness. Laboratory examinations revealed hyperammonemia (112.2mg/ml). Oral medication lowered the blood ammonia level to 69.9 mg/ml, reduced the frequency of forced laughter, and improved his IQ. Patients with BBS may have additional diseases or conditions that affect mental status, such as hyperammonemia. Physicians should explore the underlying causes of these conditions and treat such patients, who already have a compromised quality of life.


Subject(s)
Bardet-Biedl Syndrome/complications , Hyperammonemia/complications , Intellectual Disability/physiopathology , Adolescent , Humans , Intellectual Disability/drug therapy , Intellectual Disability/etiology , Laughter/physiology , Laughter/psychology , Male , Treatment Outcome
11.
Biochem Biophys Res Commun ; 374(4): 631-4, 2008 Oct 03.
Article in English | MEDLINE | ID: mdl-18662670

ABSTRACT

Triple A syndrome is an autosomal recessive neurological disease, mimicking motor neuron disease, and is caused by mutant ALADIN, a nuclear-pore complex component. We recently discovered that the pathogenesis involved impaired nuclear import of DNA repair proteins, including DNA ligase I and the cerebellar ataxia causative protein aprataxin. Such impairment was overcome by fusing classical nuclear localization signal (NLS) and 137-aa downstream sequence of XRCC1, designated stretched NLS (stNLS). We report here that the minimum essential sequence of stNLS (mstNLS) is residues 239-276, downsized by more than 100 aa. mstNLS enabled efficient nuclear import of DNA repair proteins in patient fibroblasts, functioned under oxidative stress, and reduced oxidative-stress-induced cell death, more effectively than stNLS. The stress-tolerability of mstNLS was also exerted in control fibroblasts and neuroblastoma cells. These findings may help develop treatments for currently intractable triple A syndrome and other oxidative-stress-related neurological diseases, and contribute to nuclear compartmentalization study.


Subject(s)
Adrenal Insufficiency/metabolism , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Dry Eye Syndromes/metabolism , Esophageal Achalasia/metabolism , Nuclear Localization Signals , Active Transport, Cell Nucleus/genetics , Adrenal Insufficiency/therapy , Amino Acid Sequence , Cells, Cultured , DNA Ligase ATP , DNA Ligases/genetics , DNA Ligases/metabolism , DNA-Binding Proteins/genetics , Dry Eye Syndromes/therapy , Esophageal Achalasia/therapy , Fibroblasts/metabolism , Humans , Molecular Sequence Data , Nuclear Localization Signals/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oxidative Stress , Syndrome , X-ray Repair Cross Complementing Protein 1
12.
Clin Neurol Neurosurg ; 110(5): 500-1, 2008 May.
Article in English | MEDLINE | ID: mdl-18329165

ABSTRACT

Odor is the only sensation thought to be unrelated to the thalamus. However, accumulating evidence suggests that the dorsomedial nucleus (DM) of the thalamus is associated with odor. Although the thalamus is prone to ischemia, only a single patient with bilateral DM infarctions was reported to have odor abnormalities. We describe a second such patient with infarctions involving the left DM and the right ventral posterior nucleus and ventral lateral nucleus, nuclei adjacent to the DM, associated with transient edema. In contrast to the previous case, our patient had transient odor abnormality. These observations suggested that direct and/or indirect bilateral involvement of the DM might be associated with odor abnormalities in patients with thalamic infarction.


Subject(s)
Brain Infarction/complications , Olfaction Disorders/etiology , Olfactory Pathways/physiopathology , Taste Disorders/etiology , Thalamus/blood supply , Aged , Brain Infarction/diagnosis , Brain Infarction/pathology , Functional Laterality , Humans , Magnetic Resonance Imaging , Male , Olfaction Disorders/pathology , Olfactory Pathways/pathology , Taste Disorders/pathology , Thalamus/pathology
14.
Clin Neurol Neurosurg ; 109(6): 526-30, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17507156

ABSTRACT

We describe the findings on single photon emission computed tomography (SPECT) in a patient who had genetically definite megalencephalic leukoencephalopathy with subcortical cysts. Technetium-99m-ethyl cysteinate dimer SPECT revealed hypoperfusion in the cerebral white matter, which had shown high signal intensity on magnetic resonance imaging (MRI) T2 images. Hypoperfusion was also unexpectedly found in the frontal cortices, which showed no abnormalities on MRI. This frontal abnormality corresponded clinically to a low score on the frontal assessment battery. Decreased GABA receptor density as suggested by (123)I-Iomazenil SPECT provided further evidence of cortical neuron dysfunction. Although confirmation must await future larger-scale SPECT and functional studies, our findings suggest that SPECT can be used to non-invasively monitor in vivo cortical function in this disease.


Subject(s)
Central Nervous System Cysts/diagnostic imaging , Cephalometry , Cerebral Cortex/diagnostic imaging , Dementia, Vascular/diagnostic imaging , Membrane Proteins/genetics , Tomography, Emission-Computed, Single-Photon , Adult , Astrocytes/metabolism , Central Nervous System Cysts/genetics , Cerebellum/blood supply , Cerebellum/diagnostic imaging , Cerebral Cortex/blood supply , Chromosome Aberrations , Codon , Consanguinity , Cysteine/analogs & derivatives , DNA Mutational Analysis , Dementia, Vascular/genetics , Genes, Recessive , Homozygote , Humans , Leucine/genetics , Magnetic Resonance Imaging , Male , Neurologic Examination , Organotechnetium Compounds , Serine/genetics
15.
Neurosci Lett ; 419(2): 184-7, 2007 May 29.
Article in English | MEDLINE | ID: mdl-17485165

ABSTRACT

Early-onset ataxia with ocular motor apraxia and hypoalbuminemia (EAOH)/ataxia with oculomotor apraxia type 1 (AOA1) is caused by mutations in the gene encoding aprataxin (APTX). Although several in vitro findings proposed that impaired enzymatic activities of APTX are responsible for EAOH/AOA1, potential instability of mutant proteins has also been suggested as the pathogenesis based on in vivo finding that mutant proteins are almost undetectable in EAOH/AOA1 tissues or cells. The present study aimed to experimentally prove instability of mutant proteins in neuronal cells, the cell type preferentially affected by this disease. Results of pulse-chase experiments demonstrated that all of the disease-associated mutants had extremely shorter half-lives than the WT. We further found that mutants were targeted for rapid proteasome-mediated degradation. These results help establish pathogenic and physiological protein characteristics of APTX in neuronal cells.


Subject(s)
Brain/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Neurons/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Spinocerebellar Degenerations/metabolism , Biomarkers/analysis , Biomarkers/metabolism , Brain/physiopathology , Cell Line, Tumor , DNA-Binding Proteins/genetics , Enzyme Stability/genetics , Genetic Predisposition to Disease/genetics , Half-Life , Humans , Mutation/genetics , Nuclear Proteins/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Spinocerebellar Degenerations/genetics , Spinocerebellar Degenerations/physiopathology , Time Factors
16.
J Neurol Sci ; 254(1-2): 78-83, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17303172

ABSTRACT

BACKGROUND: ALS exclusively involves motor neurons, however, accumulating evidence suggests involvement of sympathetic neurons, as in other diseases including Parkinson's disease and multiple system atrophy. In these diseases increased risk of sudden cardiac arrest is established, while that in ALS remains uncertain. METHODS: The authors retrospectively studied 12 pathologically confirmed sporadic ALS patients who received no assisted ventilation. Among them, two patients died of sudden cardiac arrest. Changes in QTc interval and dispersion, indices of sympathetic activities obtainable by routine electrocardiograms, were evaluated at the early stage and the terminal stage. Pathologically, intermediolateral nucleus (IML) sympathetic neurons in the upper thoracic cord were examined. RESULTS: The QTc intervals and dispersion were significantly increased at the terminal stage compared with that at the early stage (p<0.01). The numbers of IML neurons were significantly lower in ALS patients than in controls (p=0.017), and had linear inverse correlation with the rate of increases in maximum QTc interval and QTc dispersion (p=0.01, r=-0.915 and p=0.02, r=-0.884). Notably, two patients with sudden cardiac arrest showed longer QTc interval, larger QTc dispersion, and lower number of IML neurons than most of others. CONCLUSIONS: Patients with ALS had reduced sympathetic activities at the terminal stage of disease, presumably due to neuronal loss in IML, which may increase risk of sudden cardiac arrest. Thus, prolonged QTc intervals and increased QTc dispersion may suggest an increased risk of sudden death in ALS, as in other neurodegenerative diseases.


Subject(s)
Amyotrophic Lateral Sclerosis/complications , Autonomic Nervous System Diseases/etiology , Autonomic Nervous System Diseases/physiopathology , Death, Sudden, Cardiac/etiology , Heart/physiopathology , Sympathetic Nervous System/physiopathology , Aged , Amyotrophic Lateral Sclerosis/physiopathology , Autonomic Nervous System Diseases/pathology , Electrocardiography , Female , Heart/innervation , Humans , Long QT Syndrome/etiology , Long QT Syndrome/physiopathology , Male , Middle Aged , Nerve Degeneration/etiology , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Neurons/pathology , Retrospective Studies , Risk Factors , Spinal Cord/pathology , Spinal Cord/physiopathology , Sympathetic Nervous System/pathology
17.
Ann Neurol ; 61(2): 162-74, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17315206

ABSTRACT

OBJECTIVE: Early-onset ataxia with ocular motor apraxia and hypoalbuminemia (EAOH)/ataxia with oculomotor apraxia type 1 (AOA1) is an autosomal recessive form of cerebellar ataxia. The causative protein for EAOH/AOA1, aprataxin (APTX), interacts with X-ray repair cross-complementing 1 (XRCC1), a scaffold DNA repair protein for single-strand breaks (SSBs). The goal of this study was to prove the functional involvement of APTX in SSB repair (SSBR). METHODS: We visualized the SSBR process with a recently developed laser irradiation system that allows real-time observation of SSBR proteins and with a local ultraviolet-irradiation system using a XPA-UVDE cell line that repairs DNA lesions exclusively via SSBR. APTX was knocked down using small interference RNA in the cells. Oxidative stress-induced DNA damage and cell death were assessed in EAOH fibroblasts and cerebellum. RESULTS: Our systems showed the XRCC1-dependent recruitment of APTX to SSBs. SSBR was impaired in APTX-knocked-down cells. Oxidative stress in EAOH fibroblasts readily induced SSBs and cell death, which were blocked by antioxidants. Accumulated oxidative DNA damage was confirmed in EAOH cerebellum. INTERPRETATION: This study provides the first direct evidence for the functional involvement of APTX in SSBR and in vivo DNA damage in EAOH/AOA1, and suggests a benefit of antioxidant treatment.


Subject(s)
Cerebellar Ataxia/genetics , DNA Breaks, Single-Stranded , DNA Repair , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Adult , Animals , Antibodies, Monoclonal , Cell Death , Cells, Cultured , Cerebellar Ataxia/metabolism , Cerebellar Ataxia/physiopathology , Cerebellum/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Drug Interactions , Drug Stability , Female , Fibroblasts/metabolism , Genes, Recessive , Humans , Lasers , Male , Mutation , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Oxidative Stress , Proliferating Cell Nuclear Antigen/metabolism , Proteasome Endopeptidase Complex/metabolism , RNA, Small Interfering/pharmacology , Ultraviolet Rays , X-ray Repair Cross Complementing Protein 1
18.
Proc Natl Acad Sci U S A ; 103(7): 2298-303, 2006 Feb 14.
Article in English | MEDLINE | ID: mdl-16467144

ABSTRACT

Triple A syndrome is an autosomal recessive neuroendocrinological disease caused by mutations in a gene that encodes 546 amino acid residues. The encoded protein is the nucleoporin ALADIN, a component of nuclear pore complex (NPC). We identified a mutant ALADIN(I482S) that fails to target NPC and investigated the consequences of mistargeting using cultured fibroblasts (I482Sf) from a patient with triple A syndrome. ALADIN(I482S) affected a karyopherin-alpha/beta-mediated import pathway and decreased nuclear accumulations of aprataxin (APTX), a repair protein for DNA single-strand breaks (SSBs), and of DNA ligase I in I482Sf. This decrease was restored by wild-type ALADIN. ALADIN(I482S) had no effect on imports of M9/kap-beta2, BIB/kap-beta3, histone H1/importin 7, the ubiquitin conjugating enzyme UbcM2/importin 11, or the spliceosome protein U1A, indicating that ALADIN(I482S) selectively impaired transport of discrete import complexes through NPC. Cell survival assay showed hypersensitivity of I482Sf to l-buthionine-(S,R)-sulfoximine (BSO), a glutathione-depleting agent. BSO decreased nuclear APTX and ligase I levels in I482Sf and normal control fibroblasts, but increased SSBs only in I482Sf. These observations implied that I482Sf are hypersensitive to BSO and no longer sufficiently repair SSBs. Consistent with this notion, I482Sf transfected with both APTX and ligase I had increased resistance to BSO, whereas I482Sf transfected with LacZ vector remained hypersensitive to BSO. We propose that oxidative stress aggravates nuclear import failure, which is already compromised in patient cells. Consequent DNA damage, beyond the limited capacity of DNA repair proteins, i.e., APTX and ligase I, may participate in triggering cell death.


Subject(s)
Cell Nucleus/metabolism , Endocrine System Diseases/metabolism , Nervous System Diseases/metabolism , Nuclear Pore/metabolism , Oxidative Stress/genetics , Proteins/genetics , Active Transport, Cell Nucleus/genetics , Adult , Buthionine Sulfoximine/pharmacology , Cell Nucleus/chemistry , Cytoplasm/chemistry , Cytoplasm/metabolism , DNA Damage , DNA Repair Enzymes/metabolism , Endocrine System Diseases/genetics , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Male , Middle Aged , Mutation , Nerve Tissue Proteins , Nervous System Diseases/genetics , Nuclear Pore/chemistry , Nuclear Pore Complex Proteins , Proteins/analysis , Syndrome
19.
Neurosci Lett ; 392(3): 213-5, 2006 Jan 16.
Article in English | MEDLINE | ID: mdl-16199124

ABSTRACT

Dentatorubral-pallidoluysian atrophy (DRPLA) is an autosomal dominant neurological disorder caused by a CAG repeat expansion in the DRPLA gene encoding polyglutamine (polyQ). Although previous experimental studies have demonstrated that histone deacetylase (HDAC) inhibitors are therapeutically active, known HDAC inhibitors have considerable adverse effects clinically. To identify new HDAC inhibitors for the treatment of DRPLA, we evaluated a new series of HDAC inhibitors, N-hydroxycarboxamides, with our drug screening system, which uses neuronal PC12 cells stably transfected with a part of the DRPLA gene. We found that two of four N-hydroxycarboxamides significantly reduced polyQ-induced cell death. The essential structure of these compounds is a hydroxamic acid residue, which is shared with trichostatin A, a known HDAC inhibitor. Although our study showed mild neuroprotective effects, further structural modification of compounds that retain this residue may decrease cytotoxicity and increase protective activity against polyQ toxicity.


Subject(s)
Cantharidin/analogs & derivatives , Histone Deacetylase Inhibitors , Neuroprotective Agents/pharmacology , Peptides/antagonists & inhibitors , Animals , Cantharidin/chemistry , Cantharidin/pharmacology , Cell Count/methods , Cell Death/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Histone Deacetylases/chemistry , Nerve Tissue Proteins/genetics , PC12 Cells , Peptides/toxicity , Rats , Transfection/methods
20.
Parkinsonism Relat Disord ; 11(8): 499-502, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16263322

ABSTRACT

We investigated the clinical efficacy and tolerability of 45 mg/day mosapride, a selective 5-hydroxytryptamine type 4 (5-HT4) agonist, in an open-label study involving five patients with Parkinson's disease (PD) who had response fluctuations (RFs). 'On' time and motor function scores were determined, and gastric motility was measured by a radionuclide gastric emptying (GE) test, the most reliable quantitative method available. We found that mosapride therapy significantly shortened GE half-time, reduced RFs, and improved motor functions in all patients. There were no adverse reactions. We conclude that selective 5-HT4 agonist therapy is beneficial for patients with PD who have RFs.


Subject(s)
Benzamides/therapeutic use , Gastrointestinal Motility/physiology , Morpholines/therapeutic use , Parkinson Disease/drug therapy , Parkinson Disease/physiopathology , Receptors, Serotonin, 5-HT4/drug effects , Serotonin Receptor Agonists/therapeutic use , Aged , Antiparkinson Agents/blood , Antiparkinson Agents/pharmacokinetics , Benzamides/adverse effects , Female , Gastric Emptying/drug effects , Humans , Levodopa/blood , Levodopa/pharmacokinetics , Morpholines/adverse effects , Parkinson Disease/diagnostic imaging , Positron-Emission Tomography , Stomach/diagnostic imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...