Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Neurol Sci ; 367: 349-55, 2016 Aug 15.
Article in English | MEDLINE | ID: mdl-27423618

ABSTRACT

Clinical phenotypes of hereditary diffuse leukoencephalopathy with spheroids (HDLS), a familial progressive neurodegenerative disorder affecting the white matter of the brain, are heterogenous and may include behavioral and personality changes, memory impairment, parkinsonism, seizure, and spasticity. Thus, HDLS is frequently unrecognized and misdiagnosed. Heterozygous mutations located within the kinase domain of the gene encoding the colony-stimulating factor 1 receptor (CSF1R), a cell surface receptor with key roles in development and innate immunity, have been shown in HDLS. These different gene mutations may be related to the various clinical phenotypes. We report here a newly identified family with HDLS harboring a mutation in the CSF1R gene. We examined clinical and neuropathological features in three members of this family. These patients presented with affective incontinence, memory impairment, and executive dysfunction at onset, and revealed nonfluent aphasia, parkinsonism, and seizure as the disease progressed. We identified a novel CSF1R splice site mutation (c.2442+2T>C) in intron 18 for two of the patients. MRI of these patients revealed progressive, frontotemporal-predominant, confluent leukoencephalopathy. We also observed severe myelin loss, axonal degeneration, and abundant axonal spheroids, astrocytes, and microglia in the cerebral white matter, consistent with HDLS neuropathological features. Additionally, we identified atypical neuropathological findings for HDLS, including neuronal loss and gliosis with ballooned neurons and central chromatolysis in the frontal cortex and hippocampus. This report provides further evidence for the clinical and neuropathological heterogeneity of HDLS.


Subject(s)
Leukoencephalopathies/genetics , Mutation , Receptor, Macrophage Colony-Stimulating Factor/genetics , Adult , Family , Fatal Outcome , Frontal Lobe/diagnostic imaging , Frontal Lobe/pathology , Hippocampus/diagnostic imaging , Hippocampus/pathology , Humans , Introns , Leukoencephalopathies/diagnostic imaging , Leukoencephalopathies/pathology , Male , Middle Aged
2.
Neuropathology ; 35(3): 280-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25516199

ABSTRACT

The patient was a 72-year-old Japanese woman. At the age of 57, she started having difficulty performing daily work and developed agraphia. She also exhibited restlessness and loss of interest, and began to speak less. Thereafter, stereotypical behavior, gait disturbance and dysphagia were noted. CT scan demonstrated left-dominant frontal and temporal lobe atrophy. She died at the age of 72, about 16 years after the onset of symptoms. Neuropathologically, the brain weighed 867 g, and showed remarkable cerebral atrophy with degeneration of the white matter, predominantly in the left dorsal frontal lobe and anterior temporal lobe. Microscopically, severe neuronal loss and gliosis with rarefaction were found in the cerebral cortex, and severe destruction of myelin and axons was observed in the cerebral white matter. Moderate neuronal loss with gliosis was also found in the pallidum and substantia nigra. Gallyas-Braak staining and tau immunostaining revealed pretangle neurons, NFTs, ballooned neurons and astrocytic plaques in the cerebral cortex, subcortical nuclei and brainstem, and argyrophilic threads and coiled bodies in the subcortical white matter. Tau isoform-specific immunostaining revealed that most tau-immunoreactive structures were positive for 4-repeat (4R) tau, but some of the NFTs were positive for 3-repeat (3R) tau in the cerebral neocortex. Immunoblotting demonstrated an accumulation of 4R tau in the cerebral cortex and subcortical white matter. The patient was pathologically diagnosed as having corticobasal degeneration. Her long survival course likely accounts for the severe white matter degeneration and accumulation of 3R tau in NFTs.


Subject(s)
Basal Ganglia Diseases/pathology , Frontal Lobe/pathology , Temporal Lobe/pathology , Aged , Atrophy , Basal Ganglia Diseases/metabolism , Disease Progression , Female , Frontal Lobe/metabolism , Humans , Middle Aged , Neurofibrillary Tangles/pathology , Temporal Lobe/metabolism , Time Factors , tau Proteins/metabolism
3.
Neurosci Lett ; 569: 68-73, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24704327

ABSTRACT

Hypocretin (Hcrt) is a neuropeptide synthesized in the lateral hypothalamus (LHT) that plays a key role in maintaining arousal state. In Parkinson's disease (PD), a narcolepsy-like syndrome is commonly seen, and a previous study showed substantial Hcrt neuronal loss in accordance with PD severity. In the present study, we quantitatively examined Hcrt immunoreactivity and α-synuclein and tau pathologies in the LHT and locus coeruleus (LC) in dementia with Lewy bodies (DLB) (n=15), Alzheimer's disease (AD) (n=14), and controls (n=7). In the LHT, substantial Hcrt-positive neurons were detected in controls. In contrast, in DLB and AD, the numbers of both total neurons and Hcrt-positive neurons were significantly reduced. The reduction of the latter was significantly severer in DLB than in AD. In the LC of controls, many Hcrt-positive axonal terminals were found. In contrast, the amount of Hcrt immunoreactivity was significantly reduced both in DLB and AD. In DLB, some Lewy body (LB)-bearing neurons were detected in the LHT, but the Hcrt-positive neurons did not have any LBs. Meanwhile, some tau-positive neurofibrillary tangle (NFT)-bearing neurons were detected in the LHT, and Hcrt-positive neurons occasionally contained NFTs. We observed a significant negative correlation between the number of Hcrt-positive neurons in the LHT and the neurofibrillary stage (r=-0.67, p=0.0067), whereas no significant correlation was found between the number of Hcrt-positive neurons and the Lewy stage (r=-0.47, p=0.077). This is the first report clarifying the substantial loss of Hcrt neurons in the LHT and of Hcrt axonal terminals in the LC in DLB and the correlation between the severity of Hcrt neuronal loss and progression of neurofibrillary pathology.


Subject(s)
Brain/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lewy Body Disease/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Axons/metabolism , Brain/pathology , Case-Control Studies , Female , Humans , Lewy Body Disease/pathology , Male , Middle Aged , Orexins , alpha-Synuclein/metabolism , tau Proteins/metabolism
4.
J Neurol Sci ; 314(1-2): 111-9, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22041339

ABSTRACT

We performed a quantitative neuropathological examination of the hypometabolic regions on FDG PET in dementia with Lewy bodies (DLB), Alzheimer's disease (AD) and control cases. When the DLB cases were divided into two groups according to concomitant AD pathology (ADP), neuronal loss in the temporo-parietal association area was milder in the DLB groups than in the AD group, although there were no differences between the two DLB groups. Tau and Aß immunoreactivities were observed in the AD group and the DLB group with ADP, but were rare in the DLB group without ADP. Tau and Aß immunoreactivities as well as numbers of neurofibrillary tangles (NFTs) and neuritic plaques (NPs) were more common in the AD group than in the DLB group with ADP. There was no difference in neuronal loss in the occipital area among the three groups. α-Synuclein immunoreactivity was observed in the DLB groups but not in the AD group. There were no differences in α-synuclein immunoreactivity and number of Lewy bodies (LBs) between the two DLB groups. These findings indicate that the neuropathological bases of the hypometabolic regions in the temporo-parietal association and occipital area in DLB may be AD pathology and Lewy pathology, respectively.


Subject(s)
Lewy Body Disease/diagnostic imaging , Lewy Body Disease/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Autopsy , Brain/diagnostic imaging , Brain/pathology , Cell Count , Data Interpretation, Statistical , Disease Progression , Female , Fluorodeoxyglucose F18 , Humans , Immunohistochemistry , Lewy Body Disease/pathology , Male , Middle Aged , Neurofibrillary Tangles/pathology , Neurons/pathology , Positron-Emission Tomography , Radiopharmaceuticals , alpha-Synuclein/metabolism , tau Proteins/metabolism
5.
J Neuropathol Exp Neurol ; 70(4): 264-80, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21412173

ABSTRACT

There is emerging evidence implicating a role for the autophagy-lysosome pathway in the pathogenesis of Lewy body disease. We investigated potential neuropathologic and biochemical alterations of autophagy-lysosome pathway-related proteins in the brains of patients with dementia with Lewy bodies (DLB), Alzheimer disease (AD), and control subjects using antibodies against Ras-related protein Rab-7B (Rab7B), lysosomal-associated membrane protein 2 (LAMP2), and microtubule-associated protein 1A/1B light chain 3 (LC3). In DLB, but not in control brains, there were large Rab7B-immunoreactive endosomal granules. LC3 immunoreactivity was increased in vulnerable areas of DLB brains relative to that in control brains; computerized cell counting analysis revealed that LC3 levels were greater in the entorhinal cortex and amygdala of DLB brains than in controls. Rab7B levels were increased, and LAMP2 levels were decreased in the entorhinal cortex of DLB brains. In contrast, only a decrease in LAMP2 levels versus controls was found in AD brains. LC3 widely colocalized with several types of Lewy pathology; LAMP2 localized to the periphery or outside of brainstem-type Lewy bodies; Rab7B did not colocalize with Lewy pathology. Immunoblot analysis demonstrated specific accumulation of the autophagosomal LC3-II isoform in detergent-insoluble fractions from DLB brains. These results support apotential role for the autophagy-lysosome pathway in the pathogenesis of DLB.


Subject(s)
Brain/pathology , Lewy Bodies/metabolism , Lewy Bodies/pathology , Lewy Body Disease/pathology , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Adult , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Autophagy , Blotting, Western , Cell Count , Entorhinal Cortex/pathology , Female , Fluorescent Antibody Technique , HEK293 Cells , Humans , Immunohistochemistry , Lysosomal-Associated Membrane Protein 2 , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/physiology , Male , Middle Aged , Neurofibrillary Tangles/pathology , Plasmids/genetics , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/physiology , rab7 GTP-Binding Proteins
6.
J Neurochem ; 115(2): 423-37, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20670374

ABSTRACT

GIGYF2 has been reported as a candidate gene for PARK11-linked Parkinson's disease (PD). Heterozygous knockout of GIGYF2 results in neurodegeneration, suggesting important roles for GIGYF2 (Grb10 interacting GYF protein 2) in the CNS. In this study, we used novel GIGYF2 antibodies to clarify the distribution and function of GIGYF2. GIGYF2 was widely expressed, most highly in the pancreas and testis, and moderately in brain, lung, liver, kidney and spleen. In the brain, GIGYF2 was tightly associated with membrane in the S3 fraction, and localised in neuronal perikarya and proximal dendrites. Immunohistochemical analysis indicated sites of GIGYF2 localisation throughout the mouse brain, with high levels in the cerebral cortex, hippocampus, cerebellum, olfactory bulb and brainstem nuclei, but low levels in the substantia nigra and striatum. GIGYF2 was present in endosomes immunopositive for Rab4 and Grb10. Expression of GIGYF2 altered insulin-like growth factor-1 (IGF-1) receptor trafficking and enhanced IGF-1-induced extracellular signal-regulated kinase 1/2 phosphorylation, but not IGF-1 receptor or serine/threonine protein kinase Akt phosphorylation. There were no significant differences in signalling activation between cells expressing wild-type and putative PD-associated mutant GIGYF2. In PD brains, GIGYF2 did not localise to Lewy bodies. Our findings indicate a role for GIGYF2 in the regulation of signalling at endosomes, but no contribution of GIGYF2 to the pathogenesis of PD.


Subject(s)
Brain/ultrastructure , Carrier Proteins/metabolism , Endosomes/drug effects , Endosomes/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Animals , Cell Line, Transformed , Cricetinae , Cricetulus , GRB10 Adaptor Protein/metabolism , Gene Expression Regulation/drug effects , Humans , Immunoprecipitation/methods , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Signal Transduction/physiology , Subcellular Fractions/metabolism , Synaptophysin/metabolism , Transfection/methods , rab4 GTP-Binding Proteins/metabolism
7.
J Neuropathol Exp Neurol ; 68(9): 994-1005, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19680143

ABSTRACT

Missense mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common causes of both familial and sporadic forms of Parkinson disease and are also associated with diverse pathological alterations. The mechanisms whereby LRRK2 mutations cause these pathological phenotypes are unknown. We used immunohistochemistry with 3 distinct anti-LRRK2 antibodies to characterize the expression of LRRK2 in the brains of 21 subjects with various neurodegenerative disorders and 7 controls. The immunoreactivity of LRRK2 was localized in a subset of brainstem-type Lewy bodies (LBs) but not in cortical-type LBs, tau-positive inclusions, or TAR-DNA-binding protein-43-positive inclusions. The immunoreactivity of LRRK2 frequently appeared as enlarged granules or vacuoles within neurons of affected brain regions, including the substantia nigra, amygdala, and entorhinal cortex in patients with Parkinson disease or dementia with LBs. The volumes of LRRK2-positive granular structures in neurons of the entorhinal cortex were significantly increased in dementia with LBs brains compared with age-matched control brains (p < 0.05). Double immunolabeling demonstrated that these LRRK2-positive granular structures frequently colocalized with the late-endosomal marker Rab7B and occasionally with the lysosomal marker, the lysosomal-associated membrane protein 2. These results suggest that LRRK2 normally localizes to the endosomal-lysosomal compartment within morphologically altered neurons in neurodegenerative diseases, particularly in the brains of patients with LB diseases.


Subject(s)
Brain/pathology , Endosomes/pathology , Lewy Body Disease/pathology , Lysosomes/pathology , Neurons/pathology , Protein Serine-Threonine Kinases/metabolism , Aged , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Brain/metabolism , Dementia/metabolism , Dementia/pathology , Endosomes/metabolism , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Lewy Body Disease/metabolism , Lysosomes/metabolism , Male , Middle Aged , Neurons/metabolism , Parkinson Disease/metabolism , Parkinson Disease/pathology , Pick Disease of the Brain/metabolism , Pick Disease of the Brain/pathology , Supranuclear Palsy, Progressive/metabolism , Supranuclear Palsy, Progressive/pathology
8.
Dement Geriatr Cogn Disord ; 27(3): 214-23, 2009.
Article in English | MEDLINE | ID: mdl-19225235

ABSTRACT

BACKGROUND/AIMS: Semantic dementia is a subtype of frontotemporal lobar degeneration, of which an initial symptom is semantic aphasia. Semantic dementia pathologically corresponds to atypical Pick's disease (aPiD), showing ubiq- uitin-positive inclusions similar to those in dementia with motor neuron disease (D-MND). Previous studies have not clarified the regions responsible for semantic aphasia in aPiD, and there have been no reported neuropathological studies concerning its pathomechanism. METHODS: We neuropathologically investigated aPiD and D-MND cases with and without semantic aphasia. RESULTS: We determined that the regions involved in the early stage of the disease course of semantic dementia were more restricted to the anterior and inferior portion of the temporal lobe on the side of the dominant hemisphere. CONCLUSION: Degeneration of the temporal pole is most likely to participate in the pathomechanism of SA in semantic dementia.


Subject(s)
Aphasia/pathology , Aphasia/psychology , Dementia/pathology , Dementia/psychology , Aged , Aged, 80 and over , Aphasia/etiology , Brain/pathology , Dementia/complications , Female , Gliosis/pathology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Motor Neuron Disease/pathology , Pick Disease of the Brain/pathology
9.
Brain Res ; 1184: 284-94, 2007 Dec 12.
Article in English | MEDLINE | ID: mdl-17963732

ABSTRACT

TAR-DNA-binding protein 43 (TDP-43) has been identified as a major component protein of ubiquitin-positive inclusions in brains from patients with frontotemporal lobar degeneration with ubiquitin-positive inclusions (FTLD-U) and amyotrophic lateral sclerosis. To obtain the precise prevalence of TDP-43 pathology in neurodegenerative disorders, we examined brains from patients with tauopathies and synucleinopathies as well as FTLD-U using immunohistochemical analysis. Consequently, TDP-43-positive inclusions within neurons and oligodendroglia were found in brains from patients with Alzheimer's disease (AD) and dementia with Lewy bodies (DLB) in addition to FTLD-U, but not with Parkinson's disease, Pick's disease, progressive supranuclear palsy, corticobasal degeneration or FTDP-17. The amygdala and hippocampus that were vulnerable to tau or alpha-synuclein pathology demonstrated more severe TDP-43 pathology in AD and DLB cases than in FTLD-U cases. In contrast, in the frontal cortex and basal ganglia that were vulnerable to TDP-43 pathology in FTLD-U, TDP-43 pathology was not observed in AD and DLB cases. Thus, the neuroanatomical distribution of TDP-43 pathology in AD and DLB cases was obviously different from that in FTLD-U cases. Furthermore, a subset of TDP-43-positive inclusions co-existed with neurofibrillary tangles (NFTs) or Lewy bodies (LBs) in the same neurons. Upon double-immunofluorescent labeling analysis, TDP-43 was hardly superimposed with tau, while TDP-43 was partially superimposed with alpha-synuclein, suggesting that neither NFTs nor LBs themselves show TDP-43 immunoreactivity and that TDP-43 pathology found in this study may be related in some way to AD and LB pathology. This study will provide a more in-depth understanding of the various pathogenic pathways leading to neurodegenerative disorders.


Subject(s)
Alzheimer Disease/pathology , DNA-Binding Proteins/metabolism , Lewy Body Disease/pathology , alpha-Synuclein/metabolism , tau Proteins/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Female , Humans , Lewy Body Disease/metabolism , Male , Middle Aged , Postmortem Changes , Ubiquitin/metabolism
10.
Neurosci Lett ; 419(3): 213-8, 2007 Jun 04.
Article in English | MEDLINE | ID: mdl-17507161

ABSTRACT

TAR-DNA-binding protein 43 (TDP-43) was identified as a major component of ubiquitin-positive intracellular inclusions from brains of patients with frontotemporal lobar degeneration with ubiquitin-positive inclusions (FTLD-U). Here, we immunohistochemically investigated the appearance pattern of TDP-43 to compare the distribution of TDP-43-positive structures with that of ubiquitin-positive structures in brains of seven patients with Japanese FTLD-U, five of atypical Pick's disease (aPiD) and two of dementia with motor neuron disease (D-MND), as well as two patients with PiD as control. TDP-43-immunoreactivity generally colocalized to ubiquitin-immunoreactivity in both neuronal cytoplasmic inclusions and neurites in FTLD-U brains, but TDP-43-immunoreactivity alone or ubiquitin-immunoreactivity alone was also observed. In five aPiD cases, double-immunostaining with TDP-43 and ubiquitin demonstrated that diffuse neuronal cytoplasmic immunostaining for ubiquitin did not always display TDP-43-immunoreactivity. In contrast, ubiquitin-positive neuronal cytoplasmic inclusions usually displayed TDP-43-immunoreactivity in two D-MND cases, although most glial inclusions in one of two cases were immunostained only for TDP-43. TDP-43-positive structures were not detected in two PiD cases. Thus, the ratio in the appearance pattern of TDP-43 and ubiquitin was different between aPiD and D-MND, leading to the hypothesis that this difference may be associated with the two pathogenic variants related to clinical and pathological heterogeneity in FTLD-U.


Subject(s)
DNA-Binding Proteins/metabolism , Dementia/metabolism , Dementia/pathology , Inclusion Bodies/pathology , Ubiquitin/metabolism , Aged , Aged, 80 and over , Asian People , Brain/metabolism , Brain/pathology , Female , Humans , Immunohistochemistry , Inclusion Bodies/metabolism , Male , Middle Aged
11.
Brain Nerve ; 59(3): 263-9, 2007 Mar.
Article in Japanese | MEDLINE | ID: mdl-17370652

ABSTRACT

Dementia with motor neuron disease (D-MND) is characterized clinically by frontal and neurological signs, and pathologically by localized atrophy of the fronto-temporal lobes and neuronal ubiquitin(Ub)-positive inclusions. In this study, we compared the clinico-pathological findings of two patients with D-MND. Case 1 (55-year-old male): At the age of 51, he developed personality change and disinhibition, lacking neurological signs. Brain MRI exhibited localized atrophy of the frontal lobes. At the age of 54, he showed dysphagia and died after a disease duration of 4 years. Neuropathologically, the cerebrum showed localized atrophy of the dorsal area of the frontal lobes. The atrophied cerebral cortex demonstrated moderate neuronal loss with spongy change and gliosis in the superficial layers. The brainstem and spinal cord revealed moderate neuronal loss in the substantia nigra, severe neuronal loss with Bunina bodies in the hypoglossal nucleus, and moderate neuronal loss in the cervical anterior horn. There were some Ub-positive neuronal inclusions in the atrophied cortex and many in the dentate gyrus. Case 2 (68-year-old female): At the age of 64, she developed personality change, and then gait disturbance and dysarthria. Brain MRI exhibited localized atrophy of the fronto-temporal lobes. At the age of 67, she showed dysphagia with Babinski signs and died after a disease duration of 4 years. Neuropathologically, the cerebrum showed localized atrophy of the basal area of the temporal lobes, especially on the right side. The atrophied cerebral cortex demonstrated moderated neuronal loss with spongy change and gliosis in the superficial layers. The pre-central cortex revealed severe loss of Betz cells. The brainstem and spinal cord showed mild neuronal loss without Bunina bodies in the hypoglossal nucleus and cervical anterior horn, accompanied by severe degeneration of the bilateral pyramidal tracts. There were many Ub-positive neuronal inclusions with a few neurites in the atrophied cortex and some in the dentate gyrus. Cases 1 and 2 were clinically diagnosed as Pick's disease (PiD) and D-MND, respectively, although pathological diagnoses were both D-MND. Case 1 showed neuropathological findings typical to D-MND, whereas case 2 showed neuropathological findings common to atypical Pick's disease (aPiD). D-MND and aPiD are should be clinico-pathologically differentiated, although they are included in the frontotemporal lobar degeneration with motor neuron disease-type inclusions.


Subject(s)
Dementia/pathology , Frontal Lobe/pathology , Motor Neuron Disease/pathology , Temporal Lobe/pathology , Aged , Dementia/complications , Dementia/diagnosis , Diagnosis, Differential , Fatal Outcome , Female , Humans , Intranuclear Inclusion Bodies/metabolism , Male , Middle Aged , Motor Neuron Disease/complications , Motor Neuron Disease/diagnosis , Pick Disease of the Brain/diagnosis , Pick Disease of the Brain/pathology , Ubiquitin/metabolism
12.
Neurosci Lett ; 415(3): 219-24, 2007 Mar 30.
Article in English | MEDLINE | ID: mdl-17275187

ABSTRACT

We investigated Lewy pathologies in the claustrum and the related cerebral cortices and subcortical nuclei of dementia with Lewy bodies (DLB) brains using alpha-synuclein-immunohistochemistry to clarify the relationship between Lewy pathology in the claustrum and visual misidentification of DLB patients. The claustrum is known to have strong reciprocal connections with the visual areas. Consequently, the claustrum demonstrated many Lewy bodies (LB) and LB-related neurites. The insular and inferior temporal cortices, amygdala, BA 18, 19, transentohrinal and cingulate cortices showed stronger or similar Lewy pathology as compared with the claustrum, while BA 17, precentral, postcentral and transverse temporal cortices showed weaker Lewy pathology. Comparing the correlation coefficient of Lewy pathology between the clausturm and other regions, BA 18 and 19 as well as the insular and transentorhinal cortices demonstrated a higher correlation coefficient. These findings suggest that Lewy pathology in the claustrum is more closely associated with that in visual areas than in auditory, somatosensory or motor areas, and that dysfunction of the visuo-claustral pathway participates in visual misidentification in addition to the visuo-amygdaloid pathway. The paralimbic cortices including the insular and transentorhinal cortices may connect visual areas with limbic areas by relay of the visuo-claustral or visuo-amygdaloid pathway.


Subject(s)
Amygdala/pathology , Basal Ganglia/pathology , Lewy Body Disease/pathology , Visual Cortex/pathology , Visual Pathways/pathology , Aged , Entorhinal Cortex/pathology , Female , Gyrus Cinguli/pathology , Humans , Immunohistochemistry , Lewy Bodies/metabolism , Lewy Bodies/pathology , Male , Temporal Lobe/pathology , alpha-Synuclein/analysis , alpha-Synuclein/metabolism
13.
Neurosci Lett ; 405(1-2): 29-33, 2006 Sep 11.
Article in English | MEDLINE | ID: mdl-16859829

ABSTRACT

Limbic neurofibrillary tangle dementia (LNTD) is a subset of senile dementia characterized by numerous neurofibrillary tangles (NFT) in the hippocampal area, although there is an absence or scarcity of amyloid deposits (AM) throughout the brain. In the present study, we immunohistochemically investigated regional numbers and tau isoforms of NFT in the hippocampal area of nine LNTD patients with anti-three-repeat (3R) tau-specific and anti-four-repeat (4R) tau-specific antibodies, differentiating NFT into three developmental stages of pretangles (PT), NFT and ghost tangles (GT). Consequently, most PT were 4R tau-positive, most GT were 3R tau-positive, and NFT were 3R tau-, 4R tau- or double-positive, suggesting that composition of tau isoforms may shift from a 4R tau-predominant pattern to a 3R tau-predominant pattern during the development of NFT. In addition, a large number of NFT showing different developmental stages and different rates of 3R tau- and 4R tau-positive neurons according to the region were found in the hippocampal area, suggesting that regions undergoing earlier NFT formation may show higher ratio of 3R tau-positive neurons to 4R tau-positive neurons, and that NFT formation may begin in the entorhinal and transentorhinal cortices, subsequently progress to the subiculum and CA1, and further to the CA2, amygdala and CA3-4, although progression to the neocortex is limited. Furthermore, 4R tau-positive astrocytes and grains were found in several patients, suggesting that LNTD is a form of tauopathy.


Subject(s)
Dementia/metabolism , Hippocampus/metabolism , Neurofibrillary Tangles/metabolism , tau Proteins/metabolism , Aged, 80 and over , Female , Humans , Immunohistochemistry , Male , Protein Isoforms/metabolism
14.
J Neurol Sci ; 246(1-2): 95-101, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16624323

ABSTRACT

We examined 19 autopsied cases of dementia with Lewy bodies (DLB) using pathological and alpha-synuclein-immunohistochemical methods, and investigated Lewy pathology in the primary visual pathway (lateral geniculate body and Brodmann's area 17), secondary visual pathway (pulvinar, Brodmann's areas 18 and 19, and inferior temporal cortex), amygdala and substantia nigra, to clarify the relationship between visual misidentification and Lewy pathology in the visual pathway. Consequently, the secondary visual pathway revealed significantly severer Lewy pathology than the primary visual pathway, suggesting that the degeneration of the secondary visual pathway induces dysfunction in the recognition of objects shape and color. In addition, the amygdala revealed significantly severer Lewy pathology and neuronal loss than the primary and secondary visual pathways, suggesting that the degeneration of the amygdala, which receives the afferent connections from the substantia nigra, fails to modulate the visual processing according to cognition and emotion. These findings suggest that Lewy pathologies in the secondary visual pathway and amygdala may cause the dysfunction of the visuo-amygdaloid pathway and participate in visual misidentification in DLB patients. In addition, we compared Lewy pathology between cases with and without visual hallucinations, and showed no significant differences between the two groups.


Subject(s)
Brain/pathology , Lewy Bodies/pathology , Lewy Body Disease/pathology , Visual Pathways/pathology , Aged , Aged, 80 and over , Amygdala/pathology , Autopsy , Cell Count , Disease Progression , Female , Hallucinations/pathology , Humans , Immunohistochemistry , Lewy Body Disease/metabolism , Lewy Body Disease/psychology , Male , Middle Aged , Neurons/pathology , Substantia Nigra/pathology , Visual Pathways/metabolism , alpha-Synuclein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...