Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Prion ; 18(1): 40-53, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38627365

ABSTRACT

Prion disease is an infectious and fatal neurodegenerative disease. Western blotting (WB)-based identification of proteinase K (PK)-resistant prion protein (PrPres) is considered a definitive diagnosis of prion diseases. In this study, we aimed to detect PrPres using formalin-fixed paraffin-embedded (FFPE) specimens from cases of sporadic Creutzfeldt-Jakob disease (sCJD), Gerstmann-Sträussler-Scheinker disease (GSS), glycosylphosphatidylinositol-anchorless prion disease (GPIALP), and V180I CJD. FFPE samples were prepared after formic acid treatment to inactivate infectivity. After deparaffinization, PK digestion was performed, and the protein was extracted. In sCJD, a pronounced PrPres signal was observed, with antibodies specific for type 1 and type 2 PrPres exhibited a strong or weak signals depending on the case. Histological examination of serial sections revealed that the histological changes were compatible with the biochemical characteristics. In GSS and GPIALP, prion protein core-specific antibodies presented as PrPres bands at 8-9 kDa and smear bands, respectively. However, an antibody specific for the C-terminus presented as smears in GSS, with no PrPres detected in GPIALP. It was difficult to detect PrPres in V180I CJD. Collectively, our findings demonstrate the possibility of detecting PrPres in FFPE and classifying the prion disease types. This approach facilitates histopathological and biochemical evaluation in the same sample and is safe owing to the inactivation of infectivity. Therefore, it may be valuable for the diagnosis and research of prion diseases.


Subject(s)
Creutzfeldt-Jakob Syndrome , Gerstmann-Straussler-Scheinker Disease , Neurodegenerative Diseases , Prion Diseases , Prions , Humans , Prion Proteins , PrPSc Proteins/metabolism , Paraffin Embedding , Prion Diseases/diagnosis , Prion Diseases/metabolism , Creutzfeldt-Jakob Syndrome/pathology , Prions/metabolism , Gerstmann-Straussler-Scheinker Disease/metabolism , Endopeptidase K , Antibodies , Formaldehyde
2.
Brain Pathol ; 34(2): e13191, 2024 03.
Article in English | MEDLINE | ID: mdl-37586842

ABSTRACT

FUS mutations are one of the major mutations in familial amyotrophic lateral sclerosis (ALS). The pathological hallmark is FUS-positive neuronal cytoplasmic inclusions (FUS-NCI), known as FUS proteinopathy. Human myxovirus resistance protein 1 (MxA) is an IFN-induced dynamin-like GTPase that acts as antiviral factor. In this study, we examined the expression of MxA in neurons bearing FUS-NCI. We performed immunohistochemistry for FUS and MxA to examine the expression of MxA in two autopsy cases with different FUS gene mutations localized at the nuclear localization signal site (Case 1, H517P; Case 2, R521C). MxA. Most neurons bearing FUS-NCI have increased cytoplasmic MxA expression. Increased cytoplasmic MxA showed several distribution patterns in relation to FUS-NCIs such as the following: colocalization with NCI, distribution more widely than NCI, and different distribution peaks from NCI. Our results suggested that antiviral signaling IFNs are involved upstream in the formation of FUS-NCI in ALS-FUS patients.


Subject(s)
Amyotrophic Lateral Sclerosis , Humans , Amyotrophic Lateral Sclerosis/pathology , Antiviral Agents/metabolism , Mutation , Neurons/pathology , RNA-Binding Protein FUS/genetics
3.
Neuropathology ; 44(1): 31-40, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37340992

ABSTRACT

Neuronal intranuclear inclusions (NIIs) are common key structures in polyglutamine (polyQ) diseases such as Huntington disease (HD), spinocerebellar ataxia type 1 (SCA1), and SCA3. Marinesco bodies (MBs) of dopaminergic neurons in the substantia nigra are also intranuclear structures and are frequently seen in normal elderly people. Ribosomal dysfunction is closely related to two differential processes; therefore, we aimed to identify the pathological characteristics of ribosomal protein SA (RPSA), a ribosomal protein, in both states. To this end, we evaluated the autopsy findings in four patients with HD, two SCA3, and five normal elderly cases (NCs). Immunohistochemical studies demonstrated that both NIIs and MBs contain RPSA. In polyQ diseases, RPSA was co-localized with polyQ aggregations, and 3D-reconstructed images revealed their mosaic-like distribution. Assessments of the organization of RPSA and p62 in NIIs showed that RPSA was more localized toward the center than p62 and that this unique organization was more evident in the MBs. Immunoblotting of the temporal cortices revealed that the nuclear fraction of HD patients contained more RPSA than that of NCs. In conclusion, our study revealed that RPSA is a common component of both NIIs and MBs, indicating that a similar mechanism contributes to the formation of polyQ NIIs and MBs.


Subject(s)
Brain , Intranuclear Inclusion Bodies , Aged , Humans , Brain/pathology , Intranuclear Inclusion Bodies/metabolism , Peptides/metabolism , Ribosomal Proteins/metabolism
4.
J Neuropathol Exp Neurol ; 82(3): 231-241, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36592411

ABSTRACT

Fused in sarcoma (FUS), coded by FUS, is a heterogeneous nuclear ribonucleoprotein (hnRNP). FUS mutations are among the major mutations in familial amyotrophic lateral sclerosis (ALS-FUS: ALS6). The pathological hallmarks of ALS-FUS are FUS-positive neuronal cytoplasmic inclusions (NCI). We examined various hnRNPs in FUS NCIs in the hippocampus in ALS-FUS cases with different FUS mutations (Case 1, H517P; Case 2, R521C). We also examined TDP43-positive NCIs in sporadic ALS hippocampi. Immunohistochemistry was performed using primary antibodies against FUS, p-TDP43, TDP43, hnRNPA1, hnRNPD, PCBP1, PCBP2, and p62. Numerous FUS inclusions were found in the hippocampal granule and pyramidal cell layers. Double immunofluorescence revealed colocalization of FUS and p-TDP43, and FUS and PCBP2 (p-TDP43/FUS: 64.3%, PCBP2/FUS: 23.9%). Colocalization of FUS and PCBP1, however, was rare (PCBP1/FUS: 7.6%). In the hippocampi of patients with sporadic ALS, no colocalization was observed between TDP43-positive inclusions and other hnRNPs. This is the first study to show that FUS inclusions colocalize with other hnRNPs, such as TDP43, PCBP2, and PCBP1. These findings suggest that in ALS-FUS, FUS inclusions are the initiators, followed by alterations of multiple other hnRNPs, resulting in impaired RNA metabolism.


Subject(s)
Amyotrophic Lateral Sclerosis , RNA-Binding Protein FUS , Humans , Amyotrophic Lateral Sclerosis/pathology , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Inclusion Bodies/pathology , Mutation/genetics , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
5.
Brain Pathol ; 33(3): e13131, 2023 05.
Article in English | MEDLINE | ID: mdl-36368713

ABSTRACT

The pathological hallmark of multiple system atrophy (MSA) is aberrant accumulation of phosphorylated α-synuclein in oligodendrocytes, forming glial cytoplasmic inclusions (GCIs). Extensive demyelination occurs particularly in the olivopontocerebellar and striatonigral pathways, but its precise mechanism remains elusive. Glial connexins (Cxs), which form gap junction channels between astrocytes and oligodendrocytes, play critical roles in myelin maintenance, and have not been studied in MSA. Therefore, we immunohistochemically investigated glial Cx changes in the cerebellar afferent fibers in 15 autopsied patients with MSA. We classified demyelinating lesions into three stages based on Klüver-Barrera staining: early (Stage I), intermediate (Stage II), and late (Stage III) stages showing subtle, moderate, and severe myelin reduction, respectively. Myelin-associated glycoprotein, but not myelin oligodendrocyte glycoprotein, was preferentially decreased in Stage I, suggesting distal oligodendrogliopathy type demyelination. Accumulation of phosphorylated α-synuclein in oligodendrocytes was frequently seen in Stage I but less frequently observed in Stages II and III. Tubulin polymerization-promoting protein (TPPP/p25α)-positive oligodendrocytes were preserved in Stage I but successively decreased in Stages II and III. Even at Stage I, Cx32 was nearly absent from myelin, despite the relative preservation of other nodal proteins, such as neurofascin, claudin-11/oligodendrocyte-specific protein, and contactin-associated protein 1, which successively decreased in the later stages. Cx32 was re-distributed in the oligodendrocyte cytoplasm and co-localized with GCIs. Cx47 gradually decreased at the oligodendrocyte surface in a stage-dependent manner but was not co-localized with GCIs. Astrocytic Cx43 was down-regulated in Stage I but up-regulated in Stages II and III, reflecting astrogliosis. Cx43/Cx47 gap junctions significantly decreased from Stage I to III. Activated microglia/macrophages and T cells infiltrated in Stage I rather than Stages II and III. Therefore, early and extensive alterations of glial Cxs, particularly Cx32 loss, occur in MSA and may accelerate distal oligodendrogliopathy type demyelination and nodal/paranodal dysfunction through disruption of inter-glial communication.


Subject(s)
Demyelinating Diseases , Multiple System Atrophy , Humans , Connexins/metabolism , Connexin 43/metabolism , alpha-Synuclein
6.
Neuropathology ; 43(1): 117-126, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36003035

ABSTRACT

Here we present the autopsy case of an 80-year-old woman with a 9-year history of motor neuron disease and atypical Parkinsonism. Her initial symptom was gait disturbance, and she subsequently developed limb weakness and Parkinsonism without response to levodopa. Her motor symptoms progressed to bulbar palsy, and she died of respiratory failure. Postmortem examination revealed characteristic findings of amyotrophic lateral sclerosis (ALS), including motor neuronal loss with astrogliosis, corticospinal tract degeneration, and TAR DNA-binding protein of 43 kDa abnormalities, including nuclear loss and skein-like inclusions. In contrast, severe tau pathological changes were seen in the frontotemporal lobes and pallido-nigral system. Tau pathologies affected not only neuronal components, such as neurofibrillary tangles and neuropil threads, but also glial cells (astrocytes and oligodendrocytes). Some glial tau pathologies exhibited peculiar round accumulations, reminiscent of globular glial inclusions (GGIs) in globular glial tauopathy. This unique autopsy case demonstrates that ALS with TDP-43 could be comorbid with globular glial tau inclusions and indicates that common pathological mechanisms exist among ALS and GGI formation.


Subject(s)
Amyotrophic Lateral Sclerosis , Parkinsonian Disorders , Female , Humans , Amyotrophic Lateral Sclerosis/pathology , Neuroglia/pathology , Neurons/pathology , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , DNA-Binding Proteins/metabolism
7.
J Neuropathol Exp Neurol ; 82(1): 29-37, 2022 12 19.
Article in English | MEDLINE | ID: mdl-36331500

ABSTRACT

Myotonic dystrophy type 1 (DM1) is an inherited autosomal-dominant condition that induces altered splicing of transcripts, including MAPT, leading to a distinctive abnormal deposition of tau protein in the CNS. We characterized the tau isoforms of abnormal depositions in the brains of 4 patients with classic DM1 by immunohistochemistry using isoform-specific antibodies. All patients, including those of presenile age, showed numerous neurofibrillary tangles (NFTs) of both 3-repeat and 4-repeat tau in the limbic area and mild involvement in the cerebral cortex. Amyloid-ß deposition was only seen in 1 senile case while cortical tauopathy in all other cases was consistent with primary age-related tauopathy (PART). In the putamen and globus pallidus, only a few tau deposits were observed. Tau deposits in the brainstem frequently showed a DM1-specific pattern with 3-repeat tau dominant NFTs. Additionally, tau-positive astrocytes morphologically similar to tufted astrocytes and astrocytic plaques were occasionally observed in the brainstem; however, they were predominantly composed of 3-repeat tau. Thus, the classic DM1 showed both early onset of PART-like pathology in the limbic areas as a progeroid syndrome of DM1 and an abnormal splicing event in the brainstem leading to 3-repeat tau dominant accumulation with both neuronal and astrocytic involvement.


Subject(s)
Brain Stem , Hippocampus , Myotonic Dystrophy , Tauopathies , tau Proteins , Humans , Brain Stem/metabolism , Brain Stem/pathology , Hippocampus/metabolism , Hippocampus/pathology , Myotonic Dystrophy/metabolism , Myotonic Dystrophy/pathology , Neurofibrillary Tangles/pathology , Protein Isoforms/metabolism , tau Proteins/metabolism , Tauopathies/pathology
8.
J Neuropathol Exp Neurol ; 82(1): 38-48, 2022 12 19.
Article in English | MEDLINE | ID: mdl-36331509

ABSTRACT

GPI anchorless prion diseases (GPIALPs) show numerous coarse prion protein (PrP) deposits in the CNS but neuropil spongiform changes are mild and the incidence of dementia is low. Here, we examined differences in resident microglial phenotypes between GPIALP (D178fs25) and the other prion diseases Gerstmann-Sträussler-Scheinker (GSS) disease and sporadic Creutzfeldt-Jakob disease (sCJD) with respect to homeostasis and activation. Immunohistochemistry was performed on 2 GPIALP (D178fs25), 4 GSS (P102L), and 4 sCJD cases. Homeostatic microglia expressing TMEM119 and P2RY12 were preserved in GPIALP compared to GSS and sCJD. Microglia/macrophage activation in GSS and sCJD was associated with the extent of spongiform change. Immunoelectron microscopy revealed TMEM119 and P2RY12 in PrP plaque cores. Activated microglia/macrophages expressing HLA-DR and CD68 were predominant in GSS and sCJD whereas in GPIALP, homeostatic microglia were retained and activated microglia/macrophages were rarely observed. These data suggest that PrP deposition in GPIALP is less toxic and that microglia may be immune-tolerant to PrP deposition. This may be associated with milder tissue damage and a low incidence of dementia. Whereas microglia/macrophage activation is considered to be a reaction to tissue injury, this study shows that the degree of microglia/macrophage activity might influence the extent of tissue damage.


Subject(s)
Creutzfeldt-Jakob Syndrome , Gerstmann-Straussler-Scheinker Disease , Membrane Proteins , Microglia , Receptors, Purinergic P2Y12 , Humans , Creutzfeldt-Jakob Syndrome/metabolism , Gerstmann-Straussler-Scheinker Disease/genetics , Microglia/metabolism , Prion Proteins/genetics , Prion Proteins/metabolism , Receptors, Purinergic P2Y12/genetics , Receptors, Purinergic P2Y12/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism
9.
J Neuropathol Exp Neurol ; 81(11): 900-909, 2022 10 18.
Article in English | MEDLINE | ID: mdl-36063412

ABSTRACT

The differential effects of sporadic Creutzfeldt-Jakob disease (sCJD) on the hippocampus and other neocortical areas are poorly understood. We aimed to reveal the histological patterns of cellular prion protein (PrPC) and abnormal prion protein (PrPSc) in hippocampi of sCJD patients and normal controls (NCs). Our study examined 18 postmortem sCJD patients (MM1, 14 cases; MM1 + 2c, 3 cases; MM1 + 2t, 1 case) and 12 NCs. Immunohistochemistry was conducted using 4 primary antibodies, of which 3 targeted the N-terminus of the prion protein (PrP), and 1 (EP1802Y) targeted the C-terminal domain. PrPC expression was abundant in the hippocampus of NCs, and the distribution of PrPC at CA3/4 was reminiscent of synaptic complexes. In sCJD cases with a disease history of <2 years, antibodies against the N-terminus could not detect synapse-like PrP expression at CA4; however, EP1802Y could characterize the synapse-like expression. PrPSc accumulation and spongiform changes became evident after 2 years of illness, when PrPSc deposits were more noticeably detected by N-terminal-specific antibodies. Our findings highlighted the chronology of histopathological alterations in the CA4 region in sCJD patients.


Subject(s)
Creutzfeldt-Jakob Syndrome , Prions , Humans , Creutzfeldt-Jakob Syndrome/pathology , Prion Proteins/metabolism , PrPSc Proteins/metabolism , Disease Progression , Hippocampus/pathology , Brain/pathology
10.
Sci Rep ; 12(1): 15289, 2022 Sep 10.
Article in English | MEDLINE | ID: mdl-36088465

ABSTRACT

Prion disease is an infectious and fatal neurodegenerative disease. Human prion disease autopsy studies have revealed abnormal prion protein (PrPSc) deposits in the central nervous system and systemic organs. In deer, chronic wasting disease has also become a global problem, with PrPSc in saliva and feces. Therefore, understanding normal cellular prion proteins (PrPc) characteristics in human systemic organs is important since they could be a PrPSc source. This study used western blotting and immunohistochemistry to investigate endocrine and exocrine tissues, such as the human pituitary, adrenal, submandibular glands and the pancreas. All tissues had 30-40 kDa PrP signals, which is a slightly higher molecular weight than normal brain tissue. Most cytoplasmic PrP-positive adenohypophyseal cells were immunopositive for nuclear pituitary-specific positive transcription factor 1. The adrenal medulla and islet cells of the pancreas were PrP-positive and colocalized with chromogranin A. The duct epithelium in the submandibular gland and pancreas were immunopositive for PrP. This study reports the characteristic molecular properties and detailed tissue localization of PrPc in endocrine and exocrine tissues, which is important for infection control and diagnosis.


Subject(s)
Prion Proteins/chemistry , Animals , Deer , Humans , Organ Specificity , Prion Diseases/metabolism
11.
BMC Neurol ; 21(1): 302, 2021 Aug 05.
Article in English | MEDLINE | ID: mdl-34353291

ABSTRACT

BACKGROUND: Swallowing dysfunction is related to major cause of adverse events and an indicator of shorter survival among patients with neuromuscular disorders (NMD). It is critical to assess the swallowing function during disease progression, however, there are limited tools that can easily evaluate swallowing function without using videofluoroscopic or videoendoscopic examination. Here, we evaluated the longitudinal changes in tongue thickness (TT) and maximum tongue pressure (MTP) among patients with amyotrophic lateral sclerosis (ALS), myotonic dystrophy type 1 (DM1), and Duchenne muscular dystrophy (DMD). METHODS: Between 2010 and 2020, TT and MTP were measured from 21 ALS, 30 DM1, and 14 DMD patients (mean ages of 66.9, 44.5, and 21.4 years, respectively) at intervals of more than half a year. TT was measured, by ultrasonography, as the distance from the mylohyoid muscle raphe to the tongue dorsum, and MTP was determined by measuring the maximum compression on a small balloon when pressing the tongue against the palate. Then we examined the relationship between these evaluations and patient background and swallowing function. RESULTS: Mean follow-up periods were 24.0 months in the ALS group, 47.2 months in the DM1group, and 61.1 months in the DMD group. The DMD group demonstrated larger first TT than the other groups, while the DM1 group had lower first MTP than the ALS group. The ALS group showed a greater average monthly reduction in mean TT than the DM1 group and greater monthly reductions in mean body weight (BW) and MTP than the other groups. Significant differences between the first and last BW, TT, and MTP measures were found only in the ALS group. CONCLUSIONS: This study suggests that ALS is associated with more rapid degeneration of tongue function over several years compared to DMD and DM1.


Subject(s)
Deglutition Disorders , Neuromuscular Diseases , Tongue , Amyotrophic Lateral Sclerosis/complications , Amyotrophic Lateral Sclerosis/diagnostic imaging , Deglutition , Humans , Pressure , Tongue/diagnostic imaging
12.
Neuropathology ; 41(4): 253-265, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34031922

ABSTRACT

Spastic paraplegia type 11 (SPG11) is the most common autosomal recessive hereditary spastic paraplegia with thinning of the corpus callosum. Spatacsin, a protein encoded by the SPG11 gene, is associated with autophagy. SPG11 patients show spastic paraplegia, intellectual disability, dementia, and parkinsonism. A previous neuropathological analysis of SPG11 cases reported neurodegeneration mimicking amyotrophic lateral sclerosis without transactivation response DNA-binding protein of 43 kDa (TDP-43) deposits and unique sequestosome 1 (SQSTM1)-positive neuronal inclusions. We performed a neuropathological examination of two Japanese patients with complicated spastic paraplegia with thinning of the corpus callosum from different families, and one was genetically diagnosed as having SPG11. Both cases showed diffuse atrophy of the brain and spinal cord. Depigmentation of the substantia nigra was also observed. Immunohistochemistry revealed widespread distribution of areas showing TDP-43 aggregation in the central nervous system. The TDP-43 deposits in the thalamus and substantia nigra especially resembled skein-like inclusions. Unique SQSTM1-positive neuronal inclusions, as previously reported, were widespread in the whole central nervous system as well as the dorsal root ganglia. Double-labeling immunofluorescence of the dorsal root ganglia revealed that the unique, large SQSTM1-positive cytoplasmic inclusions of the ganglion cells were labeled with lysosome-associated membrane protein 1 and lysosome-associated membrane protein 2. This is the first report showing TDP-43 pathology in SPG11. The common neuropathological findings of TDP-43-positive inclusions in both the cases imply a causal connection between the TDP-43 proteinopathy and autophagy dysfunction in SPG11.


Subject(s)
DNA-Binding Proteins , Spastic Paraplegia, Hereditary , Humans , Lysosomes , Mutation , Proteins , TDP-43 Proteinopathies , Transcriptional Activation
13.
Neuropathology ; 41(2): 152-158, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33543531

ABSTRACT

We report the general autopsy findings of abnormal prion protein (PrP) deposits with their seeding activities, as assessed by the real-time quaking-induced conversion (RT-QuIC) method, in a 72-year-old female patient with sporadic Creutzfeldt-Jakob disease (sCJD). At 68 years of age, she presented with gait disturbance and visual disorders. Electroencephalography showed periodic synchronous discharge. Myoclonus was also observed. A genetic test revealed that PRNP codon 129 was methionine/methionine (MM). She died of pneumonia three years and four months after disease onset, and a general autopsy was performed. The brain weighed 650 g and appeared markedly atrophic. Immunohistochemistry for PrP revealed synaptic PrP deposits and coarse PrP deposits in the cerebral cortices, basal ganglia, cerebellum, and brainstem. Western blot analysis identified type 1 proteinase-K-resistant PrP in frontal cortex samples. PrP deposits were also observed in systemic organs, including the femoral nerve, psoas major muscle, abdominal skin, adrenal medulla, zona reticularis of the adrenal gland, islet cells of the pancreas, and thyroid gland. The RT-QuIC method revealed positive seeding activities in all examined organs, including the frontal cortex, femoral nerve, psoas major muscle, scalp, abdominal skin, adrenal gland, pancreas, and thyroid gland. The following 50% seeding dose (SD50 ) values were 9.5 (frontal cortex); 8 ± 0.53 (femoral nerve); 7 ± 0.53 (psoas major muscle); and 7.88 ± 0.17 (scalp). The SD50 values for the adrenal gland, dermis, pancreas, and thyroid gland were 6.12 ± 0.53, 5.25, 4.75, and 4.5, respectively. PrP deposits in general organs may be associated with long-term disease duration. This case indicated the necessity for general autopsies in sCJD cases to establish strict infection control procedures for surgical treatment and to examine certain organs.


Subject(s)
Creutzfeldt-Jakob Syndrome/pathology , Femoral Nerve/pathology , Muscle, Skeletal/pathology , Prion Proteins/metabolism , Aged , Autopsy/methods , Cerebral Cortex/pathology , Creutzfeldt-Jakob Syndrome/diagnosis , Female , Femoral Nerve/metabolism , Humans , Scalp/metabolism , Scalp/pathology
14.
Neuropathology ; 41(2): 146-151, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33404144

ABSTRACT

Fibrodysplasia ossificans progressiva (FOP) is a rare autosomal dominant disease, characterized by the progressive ossification of skeletal muscles, fascia, tendons, and ligaments. In most cases, the great toes of patients show symmetrical congenital malformations. The causative gene for FOP has been identified as the activin A receptor, type 1 (ACVR1) gene (ACVR1). The ACVR1 R206H mutation is the most common mutation among FOP patients, and the ACVR1 G356D mutation has been identified as a rare mutation in a Japanese FOP patient with slow progression. In addition to musculoskeletal abnormalities, a series of autopsy studies described one FOP case, without genetic testing to identify ACVR1 mutation, showing nodular heterotopia at the edge of the fourth ventricle. Here, we report the general autopsy findings for a 75-year-old man with FOP, caused by the ACVR1 G356D mutation, including the precise examination of brainstem lesions. Postmortem examination revealed unique symmetrical glial hyperplasia of the pons and medulla oblongata. Microscopically, lesions of the pons involving residual neurons and lesions of the medulla oblongata consisted of subependymal cells. Immunohistochemical analysis of these lesions revealed developmental anomalies, with different cellular components. In this report, for the first time, we present the neuropathological description of a patient with genetically confirmed FOP and symmetrical glial hyperplasia of the pons and medulla oblongata. The presented pathological findings, in conjunction with previous reports implying that the glial hyperplasia of the brainstem is common in FOP, suggest that ACVR1 may play an unclarified developmental role in the human brainstem.


Subject(s)
Brain Stem/pathology , Hyperplasia/pathology , Myositis Ossificans/pathology , Neuroglia/pathology , Aged , Genetic Testing/methods , Humans , Hyperplasia/genetics , Male , Mutation/genetics , Myositis Ossificans/diagnosis , Myositis Ossificans/genetics
15.
J Neuropathol Exp Neurol ; 80(3): 220-228, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33313661

ABSTRACT

Various heterogeneous nuclear ribonucleoproteins (hnRNPs) are deposited in pathological inclusions of amyotrophic lateral sclerosis (ALS) and related diseases, such as frontotemporal lobar degeneration (FTLD). Recently, poly (rC)-binding protein 2 (PCBP2, hnRNP-E2), a member of the hnRNP family, was reported to be colocalized with transactivation-responsive DNA-binding protein 43 kDa (TDP-43)-immunopositive inclusions in cases of FTLD-TDP. Here, we used immunohistochemical methods to investigate PCBP1 and PCBP2 expression in the spinal cords of sporadic ALS patients, with special reference to TDP-43-positive inclusions. Thirty autopsy cases of sporadic ALS were examined by immunohistochemistry using antibodies against PCBP1, PCBP2, sequestosome 1 (p62), and TDP-43. In control subjects without neurological disorders, neurons predominantly expressed PCBP2, rather than PCBP1, in their cytoplasm and nuclei. Anterior horn cells of sporadic ALS patients often had various levels of PCBP2 expression, and motor neurons with skein-like inclusions often had reduced or lost cytoplasmic and nuclear PCBP2 staining. Notably, one case with FTLD-TDP subtype B pathology had marked colocalization of TDP-43 and PCBP2 in the cytoplasmic inclusions and dystrophic neurites of the cerebral cortex, hippocampus, and spinal cord. In conclusion, PCBP2 was reduced in anterior horn cells of sporadic ALS, but its occurrence in TDP-43 inclusions was a rare phenomenon.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , DNA-Binding Proteins/metabolism , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , RNA-Binding Proteins/metabolism , Adolescent , Aged , Aged, 80 and over , Down-Regulation/physiology , Female , Humans , Male , Middle Aged
16.
Clin Neurol Neurosurg ; 198: 106196, 2020 11.
Article in English | MEDLINE | ID: mdl-32980799

ABSTRACT

INTRODUCTION: Patients with neurological and neuromuscular disorders (NNMD) frequently experience swallowing disorders that increase aspiration pneumonia risk and therefore require specialized diets or tube feeding. Diet type level usually is assessed by video fluoroscopic swallowing study (VFSS). To identify a simpler assessment method, we examined the association between diet type (based on the Functional Oral Intake Scale [FOIS]) diet type and maximum tongue pressure (MTP). METHODS: From 2011-2020, FOIS diet type level and MTP were assessed in a sample of 927 patients. Of these patients, 186 had Parkinson's disease (PD), 69 had Parkinson-related disease (PRD), 61 had multiple system atrophy (MSA), 42 had spinocerebellar degeneration (SCD), 147 had amyotrophic lateral sclerosis (ALS), 180 had myotonic dystrophy type 1 (DM1), and 242 had Duchenne muscular dystrophy (DMD). VFSS was conducted while patients swallowed water and foods containing barium. MTP measurements were collected the same day. Participants' diet type level was adjusted based on the VFSS, with some participants requiring multiple examinations. Relationships between diet type level and MTP were tested using univariate and Spearman rank correlation analyses. RESULTS: Mean MTP for the entire NNMD group (25.5 ± 13.1 kPa) was lower than that of healthy elderly individuals, as determined in previous reports. The highest MTP was found in the MSA group (32.2 ± 15.7 kPa) and the lowest in the DM1 group (19.1 ± 9.0 kPa). Diet type level was highest in the MSA group (5.8 ± 1.4) and lowest in the DMD group (5.2 ± 1.7). A significant correlation was observed between diet type level and MTP (R = 0.384, p < 0.001). The optimum MTP cutoff values-detected using ROC curves to predict a requirement to change to a dysphagia diet-was highest in the DMD group (29.0 kPa) and lowest in the ALS group (12.3 kPa). CONCLUSIONS: The decision to change NNMD patients to a dysphagia diet can be made based on MTP. Modifying a patient's oral diet (FOIS level ≤ 5) should be considered for those with a MTP of 10-25 kPa, with the cutoff value varying by disease.


Subject(s)
Deglutition Disorders/diagnostic imaging , Deglutition/physiology , Diet/methods , Nervous System Diseases/diagnostic imaging , Neuromuscular Diseases/diagnostic imaging , Tongue/physiology , Administration, Oral , Adolescent , Adult , Aged , Aged, 80 and over , Child , Cohort Studies , Deglutition Disorders/epidemiology , Deglutition Disorders/physiopathology , Diet/trends , Eating/physiology , Female , Fluoroscopy/methods , Humans , Male , Middle Aged , Nervous System Diseases/epidemiology , Nervous System Diseases/physiopathology , Neuromuscular Diseases/epidemiology , Neuromuscular Diseases/physiopathology , Pressure , Video Recording/methods , Young Adult
17.
Neuropathology ; 39(5): 358-367, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31599037

ABSTRACT

Huntington's disease (HD) is an inherited neurodegenerative disease caused by a polyglutamine repeat expansion in the huntingtin protein. Immunohistochemical studies using the 1C2 antibody for polyglutamine expansion have detected characteristic intranuclear inclusions (INIs) in affected neurons in HD. Further, in vitro and mouse models of HD have shown that the INIs recruit several proteins relating to RNA splicing and translation. In the present study, we immunohistochemically investigated the association of INIs with various heterogeneous nuclear ribonucleoproteins in the cerebral cortex of four autopsy cases of HD. Fused in sarcoma (FUS) was colocalized with 1C2-positive nuclear inclusions in all examined cases. Localization of poly (rC)-binding protein 1 (PCBP1) in 1C2-positive nuclear inclusions was also observed. Double immunofluorescence revealed complete or partial loss of the normal, diffuse nuclear distribution of FUS or PCBP1 in neurons with 1C2-positive nuclear inclusions. This maldistribution of FUS in cortical neurons suggests a severe disturbance of messenger RNA processing, which may be a common pathogenetic mechanism of FUS-related familial amyotrophic lateral sclerosis.


Subject(s)
Cerebral Cortex/pathology , DNA-Binding Proteins/metabolism , Huntington Disease/metabolism , Intranuclear Inclusion Bodies/metabolism , RNA-Binding Protein FUS/metabolism , RNA-Binding Proteins/metabolism , Aged , Female , Humans , Huntingtin Protein , Huntington Disease/pathology , Intranuclear Inclusion Bodies/pathology , Male , Middle Aged , Peptides , Protein Transport/physiology , RNA Processing, Post-Transcriptional/physiology
18.
J Neuropathol Exp Neurol ; 78(10): 922-929, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31504701

ABSTRACT

Human prion diseases including sporadic Creutzfeldt-Jakob disease (sCJD), inherited prion diseases, and acquired human prion diseases are lethal neurodegenerative diseases. One of the major sources of iatrogenic Creutzfeldt-Jakob disease was human growth hormone (hGH-iCJD) derived from contaminated cadaveric pituitaries. The incidence of hGH-iCJD has decreased since changing from growth hormone extracted from human cadaveric pituitaries to recombinant pituitary hormones. However, extensive analysis on the localization and detecting of abnormal prion protein in the pituitary gland are limited. In this study, we examined 9 autopsied brains and pituitary glands from 6 patients with prion disease (3 Gerstmann-Sträussler-Scheinker disease, 2 sCJD, and 1 dura mater graft-associated CJD) and 3 individuals with nonprion diseases. Western blot analysis of pituitary samples demonstrated unique glycoforms of normal cellular prion protein with molecular weights of 30-40 kDa, which was higher than the typical 25-35 kDa prion protein in brains. Proteomic analysis also revealed prion protein approximately the molecular weight of 40 kDa in pituitary samples. Moreover, proteinase K-resistant Prion protein was frequently detected in pituitary samples of the prion diseases. Immunohistochemistry for Prion protein revealed mosaic cellular distribution preferentially in growth hormone- or prolactin-producing cells.


Subject(s)
Brain/pathology , Pituitary Gland/pathology , PrPSc Proteins/metabolism , Prion Diseases/pathology , Aged , Aged, 80 and over , Brain/metabolism , Female , Humans , Male , Middle Aged , Pituitary Gland/metabolism , Prion Diseases/metabolism , Proteomics
19.
Neuropathology ; 38(6): 583-590, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30215870

ABSTRACT

Dynactin forms a protein complex with dynein that retrogradely transports cargo along microtubules. Dysfunction of this dynein-dynactin complex causes several neurodegenerative diseases such as Perry syndrome, motor neuron diseases and progressive supranuclear palsy. Recently, we reported colocalization of phosphorylated α-synuclein (p-SNCA) and the largest subunit of dynactin (DCTN1) in Lewy body (LB)-like structures in Perry syndrome. Previous reports have not focused on the relationship between dynactin and synucleinopathies. Thus, we examined autopsied human brains from patients with Parkinson's disease, dementia with LBs, and multiple system atrophy using immunohistochemistry for p-SNCA, DCTN1, dynactin 2 (DCTN2, dynamitin) and dynein cytoplasmic 1 intermediate chain 1 (DYNC1I1). We also examined microtubule affinity-regulating kinases (MARKs), which phosphorylate microtubule-associated proteins and trigger microtubule disruption. Both brainstem-type and cortical LBs were immunopositive for DCTN1, DCTN2, DYNC1I1 and p-MARK and their staining often overlapped with p-SNCA. Lewy neurites were also immunopositive for DCTN1, DCTN2 and DYNC1I1. However, p-SNCA-positive inclusions of multiple system atrophy, which included both glial and neuronal cytoplasmic inclusions, were immunonegative for DCTN1, DCTN2, DYNC1I1 and p-MARK. Thus, immunohistochemistry for dynein-dynactin complex molecules, especially DCTN1, can clearly distinguish LBs from neuronal cytoplasmic inclusions. Our results suggest that dynactin is closely associated with LB pathology.


Subject(s)
Brain/pathology , Dynactin Complex/metabolism , Lewy Bodies/pathology , Aged , Aged, 80 and over , Female , Humans , Lewy Body Disease/pathology , Male , Middle Aged , Multiple System Atrophy/pathology , Parkinson Disease/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...