Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters








Publication year range
1.
Parkinsonism Relat Disord ; 128: 107134, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39293154

ABSTRACT

INTRODUCTION: Magnetic resonance imaging (MRI)-determined atrophy of the nucleus basalis of Meynert (Ch4) predicts cognitive decline in Parkinson's disease (PD). However, interactions with other brain regions causing the decline remain unclear. This study aimed to describe how MRI-determined Ch4 atrophy leads to cognitive decline in patients with PD. METHODS: We evaluated 137 patients with PD and 39 healthy controls using neuropsychological examinations, MRI, and 123I-ioflupane single-photon emission computed tomography. First, we explored brain areas with regional gray matter loss correlated with Ch4 volume reduction using voxel-based morphometry (VBM). We then assessed the correlation between Ch4 volume reduction and cognitive impairments in PD using partial correlation coefficients (rpar). Finally, we examined whether the regional gray matter loss mediated the association between Ch4 volume reduction and cognitive impairments using mediation analysis. RESULTS: Our PD cohort was "advanced-stage enriched." VBM analyses revealed that Ch4 volume loss was correlated with volume reduction in the medial temporal lobe in PD (P < 0.05, family-wise error corrected, >29 voxels). Ch4 volume reduction was significantly correlated with verbal memory deficits in PD when adjusted for age, sex, total brain volume, and 123I-ioflupane uptake in the caudate (rpar = 0.28, P < 0.001). The mediation analysis revealed that the hippocampus mediated the effects of Ch4 volumes on verbal memory (average causal mediation effect = 0.013, 95 % CI = 0.006-0.020, P < 0.001). CONCLUSION: Particularly in advanced-stage PD, Ch4 atrophy was associated with medial temporal lobe atrophy, which played an intermediary role in the relationship between Ch4 atrophy and verbal memory impairments.

2.
Mov Disord ; 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39092513

ABSTRACT

BACKGROUND: Patient-rated motor symptoms (PRMS) and clinician-rated motor symptoms (CRMS) often differ in Parkinson's disease (PD). OBJECTIVE: Our goal was to investigate the determinants and clinical implications of PRMS compared with CRMS in PD. METHODS: This retrospective, observational cohort study analyzed the cross-sectional associations and longitudinal impacts of PRMS as assessed by the Movement Disorders Society-sponsored Unified PD Rating Scale (MDS-UPDRS) part 2, while controlling for CRMS measured by MDS-UPDRS part 3. Longitudinal analyses used Cox proportional hazards models and multiple linear mixed-effects random intercepts/slope models, adjusting for many clinical predictors. We conducted propensity score matching (PSM) to reinforce our analyses' robustness and surface-based morphometry to investigate neural correlates. RESULTS: We enrolled 442 patients with early-stage PD. At baseline, regardless of CRMS, PRMS were associated with the severity of postural instability and gait disturbance (PIGD). Notably, PRMS independently and more accurately predicted faster long-term deterioration in motor function than CRMS (Hoehn and Yahr 4, adjusted hazard ratio per +1 point = 1.19 [95% confidence intervals, 1.08-1.32]), particularly in PIGD (PIGD subscore, ß-interaction = 0.052 [95% confidence intervals, 0.018-0.086]). PSM confirmed these findings' robustness. Surface-based morphometry suggested that enhanced sensory processing was distinctively associated with PRMS. CONCLUSIONS: In early-stage PD, PRMS weighed different aspects of symptoms and more effectively predicted motor deterioration compared to CRMS, with distinctive brain structural characteristics. The superior sensitivity of PRMS to subtle declines in drug-refractory symptoms like PIGD likely underlie our results, highlighting the importance of understanding the differential clinical implications of PRMS to prevent long-term motor deterioration. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.

3.
Neurology ; 103(3): e209606, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-38976821

ABSTRACT

BACKGROUND AND OBJECTIVES: Neural computations underlying gait disorders in Parkinson disease (PD) are multifactorial and involve impaired expression of stereotactic locomotor patterns and compensatory recruitment of cognitive functions. This study aimed to clarify the network mechanisms of cognitive contribution to gait control and its breakdown in patients with PD. METHODS: Patients with PD were instructed to walk at a comfortable pace on a mat with pressure sensors. The characterization of cognitive-motor interplay was enhanced by using a gait with a secondary cognitive task (dual-task condition) and a gait without additional tasks (single-task condition). Participants were scanned using 3-T MRI and 123I-ioflupane SPECT. RESULTS: According to gait characteristics, cluster analysis assisted by a nonlinear dimensionality reduction technique, t-distributed stochastic neighbor embedding, categorized 56 patients with PD into 3 subpopulations. The preserved gait (PG) subgroup (n = 23) showed preserved speed and variability during gait, both with and without additional cognitive load. Compared with the PG subgroup, the mildly impaired gait (MIG) subgroup (n = 16) demonstrated deteriorated gait variability with additional cognitive load and impaired speed and gait variability without additional cognitive load. The severely impaired gait (SIG) subgroup (n = 17) revealed the slowest speed and highest gait variability. In addition, group differences were found in attention/working memory and executive function domains, with the lowest performance in the SIG subgroup than in the PG and MIG subgroups. Using resting-state functional MRI, the SIG subgroup demonstrated lower functional connectivity of the left and right frontoparietal network (FPN) with the caudate than the PG subgroup did (left FPN, d = 1.21, p < 0.001; right FPN, d = 1.05, p = 0.004). Cortical thickness in the FPN and 123I-ioflupane uptake in the striatum did not differ among the 3 subgroups. By contrast, the severity of Ch4 density loss was significantly correlated with the level of functional connectivity degradation of the FPN and caudate (left FPN-caudate, r = 0.27, p = 0.04). DISCUSSION: These findings suggest that the functional connectivity of the FPN with the caudate, as mediated by the cholinergic Ch4 projection system, underlies the compensatory recruitment of attention and executive function for damaged automaticity in gait in patients with PD.


Subject(s)
Gait Disorders, Neurologic , Magnetic Resonance Imaging , Parkinson Disease , Tomography, Emission-Computed, Single-Photon , Humans , Parkinson Disease/physiopathology , Parkinson Disease/diagnostic imaging , Parkinson Disease/complications , Male , Female , Aged , Gait Disorders, Neurologic/etiology , Gait Disorders, Neurologic/physiopathology , Gait Disorders, Neurologic/diagnostic imaging , Middle Aged , Frontal Lobe/diagnostic imaging , Frontal Lobe/physiopathology , Corpus Striatum/diagnostic imaging , Corpus Striatum/physiopathology , Parietal Lobe/diagnostic imaging , Parietal Lobe/physiopathology , Nerve Net/diagnostic imaging , Nerve Net/physiopathology , Neural Pathways/physiopathology , Neural Pathways/diagnostic imaging , Basal Nucleus of Meynert/physiopathology , Basal Nucleus of Meynert/diagnostic imaging , Nortropanes
4.
Parkinsonism Relat Disord ; 112: 105440, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37267818

ABSTRACT

INTRODUCTION: Pathological α-synuclein (α-Syn) propagation may cause Parkinson's disease progression. We aimed to verify whether single-dose intranasal administration of α-Syn preformed fibrils (PFFs) induces α-Syn pathology in the olfactory bulb (OB). METHODS: A single dose of α-Syn PFFs was administered to the left nasal cavity of wild-type mice. The untreated right side served as a control. The α-Syn pathology of the OBs was examined up to 12 months after the injection. RESULTS: Lewy neurite-like aggregates were observed in the OB 6 and 12 months after the treatment. CONCLUSIONS: These findings suggest that pathological α-Syn can propagate from the olfactory mucosa to the OB and reveal the potential dangers of α-Syn PFFs inhalation.


Subject(s)
Parkinson Disease , Synucleinopathies , Mice , Animals , Lewy Bodies/pathology , Olfactory Bulb/metabolism , Olfactory Bulb/pathology , Administration, Intranasal , alpha-Synuclein/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/etiology , Parkinson Disease/pathology , Synucleinopathies/pathology
5.
Mov Disord ; 38(6): 1056-1067, 2023 06.
Article in English | MEDLINE | ID: mdl-37066491

ABSTRACT

BACKGROUND: The intercellular transmission of pathogenic proteins plays a crucial role in the progression of neurodegenerative diseases. Previous research has shown that the neuronal uptake of such proteins is activity-dependent; however, the detailed mechanisms underlying activity-dependent α-synuclein transmission in Parkinson's disease remain unclear. OBJECTIVE: To examine whether α-synuclein transmission is affected by Ca2+ -calmodulin-calcineurin signaling in cultured cells and mouse models of Parkinson's disease. METHODS: Mouse primary hippocampal neurons were used to examine the effects of the modulation of Ca2+ -calmodulin-calcineurin signaling on the neuronal uptake of α-synuclein preformed fibrils. The effects of modulating Ca2+ -calmodulin-calcineurin signaling on the development of α-synuclein pathology were examined using a mouse model injected with α-synuclein preformed fibrils. RESULTS: Modulation of Ca2+ -calmodulin-calcineurin signaling by inhibiting voltage-gated Ca2+ channels, calmodulin, and calcineurin blocked the neuronal uptake of α-synuclein preformed fibrils via macropinocytosis. Different subtypes of voltage-gated Ca2+ channel differentially contributed to the neuronal uptake of α-synuclein preformed fibrils. In wild-type mice inoculated with α-synuclein preformed fibrils, we found that inhibiting calcineurin ameliorated the development of α-synuclein pathology. CONCLUSION: Our data suggest that Ca2+ -calmodulin-calcineurin signaling modulates α-synuclein transmission and has potential as a therapeutic target for Parkinson's disease. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Parkinson Disease , Synucleinopathies , Humans , Animals , Mice , alpha-Synuclein/metabolism , Parkinson Disease/pathology , Calmodulin/metabolism , Calcineurin/metabolism , Neurons/metabolism
6.
Intern Med ; 62(20): 3043-3046, 2023 Oct 15.
Article in English | MEDLINE | ID: mdl-36889711

ABSTRACT

The pathophysiology of idiopathic intracranial hypertension (IIH) and idiopathic normal-pressure hydrocephalus (iNPH) differs in terms of cerebrospinal fluid (CSF) pressure and imaging-related characteristics. A 51-year-old man presented with optic nerve papillary edema, visual disturbance, bilateral abducens nerve palsy, and a wide-based gait. Imaging showed characteristic findings of IIH and disproportionately enlarged subarachnoid space hydrocephalus (DESH) - characteristic of iNPH. A CSF examination revealed marked CSF hypertension. IIH with iNPH-like imaging features (DESH) was diagnosed, and ventriculoperitoneal shunt surgery was performed. Postoperatively, the visual acuity and visual field improved. This report also describes the distinct and overlapping pathophysiological mechanisms of IIH and iNPH.


Subject(s)
Hydrocephalus, Normal Pressure , Pseudotumor Cerebri , Male , Humans , Middle Aged , Pseudotumor Cerebri/complications , Pseudotumor Cerebri/diagnostic imaging , Pseudotumor Cerebri/surgery , Hydrocephalus, Normal Pressure/complications , Hydrocephalus, Normal Pressure/diagnostic imaging , Hydrocephalus, Normal Pressure/surgery , Ventriculoperitoneal Shunt , Magnetic Resonance Imaging/methods , Subarachnoid Space/diagnostic imaging , Subarachnoid Space/surgery
10.
Mov Disord ; 37(10): 2033-2044, 2022 10.
Article in English | MEDLINE | ID: mdl-35989519

ABSTRACT

BACKGROUND: Lewy body diseases (LBDs), which are pathologically defined as the presence of intraneuronal α-synuclein (α-Syn) inclusions called Lewy bodies, encompass Parkinson's disease, Parkinson's disease with dementia, and dementia with Lewy bodies. Autopsy studies have shown that the olfactory bulb (OB) is one of the regions where Lewy pathology develops and initiates its spread in the brain. OBJECTIVE: This study aims to clarify how Lewy pathology spreads from the OB and affects brain functions using nonhuman primates. METHODS: We inoculated α-Syn preformed fibrils into the unilateral OBs of common marmosets (Callithrix jacchus) and performed pathological analyses, manganese-enhanced magnetic resonance imaging, and 18 F-fluoro-2-deoxy-d-glucose positron emission tomography up to 6 months postinoculation. RESULTS: Severe α-Syn pathology was observed within the olfactory pathway and limbic system, while mild α-Syn pathology was seen in a wide range of brain regions, including the substantia nigra pars compacta, locus coeruleus, and even dorsal motor nucleus of the vagus nerve. The brain imaging analyses showed reduction in volume of the OB and progressive glucose hypometabolism in widespread brain regions, including the occipital lobe, and extended beyond the pathologically affected regions. CONCLUSIONS: We generated a novel nonhuman primate LBD model with α-Syn propagation from the OB. This model suggests that α-Syn propagation from the OB is related to OB atrophy and cerebral glucose hypometabolism in LBDs. © 2022 International Parkinson and Movement Disorder Society.


Subject(s)
Lewy Body Disease , Parkinson Disease , Animals , Callithrix/metabolism , Deoxyglucose/metabolism , Glucose/metabolism , Lewy Body Disease/pathology , Manganese/metabolism , Olfactory Bulb/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/metabolism
11.
Sci Rep ; 12(1): 12636, 2022 07 25.
Article in English | MEDLINE | ID: mdl-35879519

ABSTRACT

Mutations within Superoxide dismutase 1 (SOD1) cause amyotrophic lateral sclerosis (ALS), accounting for approximately 20% of familial cases. The pathological feature is a loss of motor neurons with enhanced formation of intracellular misfolded SOD1. Homozygous SOD1-D90A in familial ALS has been reported to show slow disease progression. Here, we reported a rare case of a slowly progressive ALS patient harboring a novel SOD1 homozygous mutation D92G (homD92G). The neuronal cell line overexpressing SOD1-D92G showed a lower ratio of the insoluble/soluble fraction of SOD1 with fine aggregates of the misfolded SOD1 and lower cellular toxicity than those overexpressing SOD1-G93A, a mutation that generally causes rapid disease progression. Next, we analyzed spinal motor neurons derived from induced pluripotent stem cells (iPSC) of a healthy control subject and ALS patients carrying SOD1-homD92G or heterozygous SOD1-L144FVX mutation. Lower levels of misfolded SOD1 and cell loss were observed in the motor neurons differentiated from patient-derived iPSCs carrying SOD1-homD92G than in those carrying SOD1-L144FVX. Taken together, SOD1-homD92G has a lower propensity to aggregate and induce cellular toxicity than SOD1-G93A or SOD1-L144FVX, and these cellular phenotypes could be associated with the clinical course of slowly progressive ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Amyotrophic Lateral Sclerosis/metabolism , Animals , Disease Models, Animal , Disease Progression , Mice , Mice, Transgenic , Motor Neurons/metabolism , Mutation , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
12.
Ann Neurol ; 92(1): 110-121, 2022 07.
Article in English | MEDLINE | ID: mdl-35428994

ABSTRACT

OBJECTIVE: The motor severity in Parkinson disease (PD) is believed to parallel dopaminergic terminal degeneration in the striatum, although the terminal was reported to be virtually absent by 4 years postdiagnosis. Meanwhile, neuromelanin-laden dopamine neuron loss in the substantia nigra (SN) elucidated a variability at early stages and gradual loss with less variability 10 years postdiagnosis. Here, we aimed to clarify the correlation between motor impairments and striatal dopaminergic terminal degeneration and nigral neuromelanin-laden dopamine neuron loss at early to advanced stages of PD. METHODS: Ninety-three PD patients were divided into early and advanced subgroups based on motor symptom duration and whether motor fluctuation was present. Striatal dopaminergic terminal degeneration was evaluated using a presynaptic dopamine transporter tracer, 123 I-ioflupane single photon emission computed tomography (SPECT). Nigral neuromelanin-laden dopamine neuron density was assessed by neuromelanin-sensitive magnetic resonance imaging (NM-MRI). RESULTS: In patients with early stage PD (motor symptoms for ≤8 or 10 years), motor dysfunction during the drug-off state was paralleled by a decline in 123 I-ioflupane uptake in the striatum despite the absence of a correlation with reductions in NM-MRI signals in SN. Meanwhile, in patients with advanced stage PD (motor symptoms for >8 or 10 years and with fluctuation), the degree of motor deficits during the drug-off state was not correlated with 123 I-ioflupane uptake in the striatum, despite its significant negative correlation with NM-MRI signals in SN. INTERPRETATION: We propose striatal dopaminergic terminal loss measured using 123 I-ioflupane SPECT and nigral dopamine neuron loss assessed with NM-MRI as early stage and advanced stage motor impairment biomarkers, respectively. ANN NEUROL 2022;92:110-121.


Subject(s)
Parkinson Disease , Corpus Striatum/metabolism , Dopamine , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopaminergic Neurons/pathology , Humans , Magnetic Resonance Imaging/methods , Nerve Degeneration/diagnostic imaging , Nerve Degeneration/pathology , Parkinson Disease/complications , Parkinson Disease/diagnostic imaging , Substantia Nigra/pathology , Tomography, Emission-Computed, Single-Photon/methods
13.
Neurotherapeutics ; 19(1): 289-304, 2022 01.
Article in English | MEDLINE | ID: mdl-34935120

ABSTRACT

Parkinson's disease (PD), the most common neurodegenerative movement disorder, is characterized by dopaminergic neuron loss in the substantia nigra pars compacta (SNpc) and intraneuronal α-synuclein (α-syn) inclusions. It is highly needed to establish a rodent model that recapitulates the clinicopathological features of PD within a short period to efficiently investigate the pathological mechanisms and test disease-modifying therapies. To this end, we analyzed three mouse lines, i.e., wild-type mice, wild-type human α-syn bacterial artificial chromosome (BAC) transgenic (BAC-SNCA Tg) mice, and A53T human α-syn BAC transgenic (A53T BAC-SNCA Tg) mice, receiving dorsal striatum injections of human and mouse α-syn preformed fibrils (hPFFs and mPFFs, respectively). mPFF injections induced more severe α-syn pathology in most brain regions, including the ipsilateral SNpc, than hPFF injections in all genotypes at 1-month post-injection. Although these Tg mouse lines expressed a comparable amount of α-syn in the brains, the mPFF-injected A53T BAC-SNCA Tg mice exhibited the most severe α-syn pathology as early as 0.5-month post-injection. The mPFF-injected A53T BAC-SNCA Tg mice showed a 38% reduction in tyrosine hydroxylase (TH)-positive neurons in the ipsilateral SNpc, apomorphine-induced rotational behavior, and motor dysfunction at 2 months post-injection. These data indicate that the extent of α-syn pathology induced by α-syn PFF injection depends on the types of α-syn PFFs and exogenously expressed α-syn in Tg mice. The mPFF-injected A53T BAC-SNCA Tg mice recapitulate the key features of PD more rapidly than previously reported mouse models, suggesting their usefulness for testing disease-modifying therapies as well as analyzing the pathological mechanisms.


Subject(s)
Parkinson Disease , alpha-Synuclein , Animals , Chromosomes, Artificial, Bacterial/genetics , Dopamine , Dopaminergic Neurons/pathology , Lewy Bodies/pathology , Mice , Mice, Transgenic , Parkinson Disease/genetics , Parkinson Disease/pathology , alpha-Synuclein/genetics
14.
Methods Mol Biol ; 2322: 119-130, 2021.
Article in English | MEDLINE | ID: mdl-34043198

ABSTRACT

Parkinson's disease (PD) is pathologically characterized by intraneuronal α-synuclein (α-Syn) inclusions called Lewy bodies (LBs) and the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). Autopsy studies have suggested that Lewy pathology initially occurs in the olfactory bulb and enteric nervous system, subsequently spreading in the brain stereotypically. Recent studies have demonstrated that templated fibrillization and intercellular dissemination of misfolded α-Syn underlie this pathological progression. Injection of animals with α-Syn preformed fibrils (PFFs) can recapitulate LB-like inclusions and the subsequent intercellular transmission of α-Syn pathology. Moreover, targeting specific brain regions or body parts enables the generation of unique models depending on the injection sites. These features of α-Syn PFF-injected animal models provide a platform to explore disease mechanisms and to test disease modifying therapies in PD research. Here, we describe a methodology for the generation of α-Syn PFFs and the surgery on mice.


Subject(s)
Parkinson Disease/metabolism , Parkinson Disease/pathology , alpha-Synuclein/metabolism , Animals , Disease Models, Animal , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Enteric Nervous System/metabolism , Enteric Nervous System/pathology , Humans , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Mice , Substantia Nigra/metabolism , Substantia Nigra/pathology
15.
Mov Disord ; 36(7): 1554-1564, 2021 07.
Article in English | MEDLINE | ID: mdl-33813737

ABSTRACT

BACKGROUND: The intercellular transmission of pathogenic proteins plays a key role in the clinicopathological progression of neurodegenerative diseases. Previous studies have demonstrated that this uptake and release process is regulated by neuronal activity. OBJECTIVE: The objective of this study was to examine the effect of perampanel, an antiepileptic drug, on α-synuclein transmission in cultured cells and mouse models of Parkinson's disease. METHODS: Mouse primary hippocampal neurons were transduced with α-synuclein preformed fibrils to examine the effect of perampanel on the development of α-synuclein pathology and its mechanisms of action. An α-synuclein preformed fibril-injected mouse model was used to validate the effect of oral administration of perampanel on the α-synuclein pathology in vivo. RESULTS: Perampanel inhibited the development of α-synuclein pathology in mouse hippocampal neurons transduced with α-synuclein preformed fibrils. Interestingly, perampanel blocked the neuronal uptake of α-synuclein preformed fibrils by inhibiting macropinocytosis in a neuronal activity-dependent manner. We confirmed that oral administration of perampanel ameliorated the development of α-synuclein pathology in wild-type mice inoculated with α-synuclein preformed fibrils. CONCLUSION: Modulation of neuronal activity could be a promising therapeutic target for Parkinson's disease, and perampanel could be a novel disease-modifying drug for Parkinson's disease. © 2021 International Parkinson and Movement Disorder Society.


Subject(s)
Parkinson Disease , Synucleinopathies , Animals , Mice , Nitriles , Parkinson Disease/drug therapy , Pyridones/pharmacology , alpha-Synuclein/genetics
16.
Acta Neuropathol Commun ; 8(1): 120, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32727582

ABSTRACT

Multiple system atrophy (MSA) is pathologically characterized by the presence of fibrillar α-synuclein-immunoreactive inclusions in oligodendrocytes. Although the myelinating process of oligodendrocytes can be observed in adult human brains, little is known regarding the presence of α-synuclein pathology in immature oligodendrocytes and how their maturation and myelination are affected in MSA brains. Recently, breast carcinoma amplified sequence 1 (BCAS1) has been found to be specifically expressed in immature oligodendrocytes undergoing maturation and myelination. Here, we analyzed the altered dynamics of oligodendroglial maturation in both MSA brains and primary oligodendroglial cell cultures which were incubated with α-synuclein pre-formed fibrils. The numbers of BCAS1-expressing oligodendrocytes that displayed a matured morphology negatively correlated with the density of pathological inclusions in MSA brains but not with that in Parkinson's disease and diffuse Lewy body disease. In addition, a portion of the BCAS1-expressing oligodendrocyte population showed cytoplasmic inclusions, which were labeled with antibodies against phosphorylated α-synuclein and cleaved caspase-9. Further in vitro examination indicated that the α-synuclein pre-formed fibrils induced cytoplasmic inclusions in the majority of BCAS1-expressing oligodendrocytes. In contrast, the majority of BCAS1-non-expressing mature oligodendrocytes did not develop inclusions on day 4 after maturation induction. Furthermore, exposure of α-synuclein pre-formed fibrils in the BCAS1-positive phase caused a reduction in oligodendroglial cell viability. Our results indicated that oligodendroglial maturation and myelination are impaired in the BCAS1-positive phase of MSA brains, which may lead to the insufficient replacement of defective oligodendrocytes. In vitro, the high susceptibility of BCAS1-expressing primary oligodendrocytes to the extracellular α-synuclein pre-formed fibrils suggests the involvement of insufficient oligodendroglial maturation in MSA disease progression and support the hypothesis that the BCAS1-positive oligodendrocyte lineage cells are prone to take up aggregated α-synuclein in vivo.


Subject(s)
Multiple System Atrophy/pathology , Oligodendrocyte Precursor Cells/drug effects , Oligodendrocyte Precursor Cells/pathology , alpha-Synuclein/metabolism , alpha-Synuclein/toxicity , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Humans , Neoplasm Proteins , Nerve Tissue Proteins , Oligodendroglia/drug effects , Oligodendroglia/pathology , Rats , Rats, Sprague-Dawley
17.
J Stroke Cerebrovasc Dis ; 29(5): 104755, 2020 May.
Article in English | MEDLINE | ID: mdl-32171626

ABSTRACT

BACKGROUND: Determining stroke subtypes on initial clinical evaluation is a prerequisite for the selection of appropriate initial treatment. Although diffusion-weighted imaging (DWI) is a powerful tool for detection of acute cerebral infarction, its diagnostic accuracy is not always sufficient particularly in the hyperacute phase. METHODS: Patients admitted within 2 weeks from the symptom onset with the diagnosis of acute ischemic strokes were analyzed with thin-section iso-voxel DWI, namely 3-dimension DWI (3D-DWI), to obtain axial, coronal, and sagittal sections in order to elucidate stroke characteristics. In this case series, we introduce the effectiveness of 3D-DWI. RESULTS: 3D-DWI uncovered stroke subtypes and distribution more precisely compared with conventional DWI. While previous studies indicated the utility of thin section DWI in detecting infratentrial infarctions, 3D-DWI is beneficial for the detection of not only infratentrial but also supratentorial lesions. Furthermore, since both 3D-DWI and magnetic resonance angiography (MRA) are multiplanar reconstruction images, the fusion image of 3D-DWI with MRA is available, enabling cross-reference of spatial cerebrovascular configuration and ischemic lesions. CONCLUSIONS: 3D-DWI is applicable to standard 1.5 T MRI by slight modification of data acquisition protocols, and becomes a key modality to solve the diagnostic puzzle of acute ischemic strokes.


Subject(s)
Diffusion Magnetic Resonance Imaging , Image Interpretation, Computer-Assisted , Stroke/diagnostic imaging , Aged , Aged, 80 and over , Cerebral Angiography , Female , Humans , Magnetic Resonance Angiography , Male , Middle Aged , Multimodal Imaging , Predictive Value of Tests , Reproducibility of Results , Stroke/classification , Stroke/therapy
18.
Brain ; 143(1): 249-265, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31816026

ABSTRACT

Parkinson's disease is one of the most common movement disorders and is characterized by dopaminergic cell loss and the accumulation of pathological α-synuclein, but its precise pathogenetic mechanisms remain elusive. To develop disease-modifying therapies for Parkinson's disease, an animal model that recapitulates the pathology and symptoms of the disease, especially in the prodromal stage, is indispensable. As subjects with α-synuclein gene (SNCA) multiplication as well as point mutations develop familial Parkinson's disease and a genome-wide association study in Parkinson's disease has identified SNCA as a risk gene for Parkinson's disease, the increased expression of α-synuclein is closely associated with the aetiology of Parkinson's disease. In this study we generated bacterial artificial chromosome transgenic mice harbouring SNCA and its gene expression regulatory regions in order to maintain the native expression pattern of α-synuclein. Furthermore, to enhance the pathological properties of α-synuclein, we inserted into SNCA an A53T mutation, two single-nucleotide polymorphisms identified in a genome-wide association study in Parkinson's disease and a Rep1 polymorphism, all of which are causal of familial Parkinson's disease or increase the risk of sporadic Parkinson's disease. These A53T SNCA bacterial artificial chromosome transgenic mice showed an expression pattern of human α-synuclein very similar to that of endogenous mouse α-synuclein. They expressed truncated, oligomeric and proteinase K-resistant phosphorylated forms of α-synuclein in the regions that are specifically affected in Parkinson's disease and/or dementia with Lewy bodies, including the olfactory bulb, cerebral cortex, striatum and substantia nigra. Surprisingly, these mice exhibited rapid eye movement (REM) sleep without atonia, which is a key feature of REM sleep behaviour disorder, at as early as 5 months of age. Consistent with this observation, the REM sleep-regulating neuronal populations in the lower brainstem, including the sublaterodorsal tegmental nucleus, nuclei in the ventromedial medullary reticular formation and the pedunculopontine nuclei, expressed phosphorylated α-synuclein. In addition, they also showed hyposmia at 9 months of age, which is consistent with the significant accumulation of phosphorylated α-synuclein in the olfactory bulb. The dopaminergic neurons in the substantia nigra pars compacta degenerated, and their number was decreased in an age-dependent manner by up to 17.1% at 18 months of age compared to wild-type, although the mice did not show any related locomotor dysfunction. In conclusion, we created a novel mouse model of prodromal Parkinson's disease that showed RBD-like behaviour and hyposmia without motor symptoms.


Subject(s)
Brain/metabolism , Disease Models, Animal , Mice , Olfaction Disorders/genetics , Parkinson Disease/genetics , Prodromal Symptoms , REM Sleep Behavior Disorder/genetics , alpha-Synuclein/genetics , Animals , Cell Count , Chromosomes, Artificial, Bacterial , Electroencephalography , Electromyography , Endopeptidase K/metabolism , Mice, Transgenic , Olfaction Disorders/physiopathology , Parkinson Disease/physiopathology , Polymorphism, Single Nucleotide , REM Sleep Behavior Disorder/physiopathology , Sleep , alpha-Synuclein/metabolism
19.
Intern Med ; 59(4): 587-588, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31666470
20.
Brain Nerve ; 70(1): 41-54, 2018 Jan.
Article in Japanese | MEDLINE | ID: mdl-29348374

ABSTRACT

Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is an illness characterized by fatigue lasting for at least six months, post-exertional malaise, unrefreshing sleep, cognitive impairment and orthostatic intolerance. ME/CFS has been a controversial illness because it is defined exclusively by subjective complaints. However, recent studies of neuroimaging as well as analysis of blood markers, energy metabolism and mitochondrial function have revealed many objective biological abnormalities. Specifically, it is suspected that the symptoms of ME/CFS may be triggered by immune activation - either inside or outside the brain - through release of inflammatory cytokines. In this review, we summarize potentially important recent findings on ME/CFS, focusing on objective evidence.


Subject(s)
Fatigue Syndrome, Chronic , Central Nervous System Diseases , Cytokines/immunology , Energy Metabolism , Humans , Pain
SELECTION OF CITATIONS
SEARCH DETAIL