Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 298
Filter
1.
Brain Nerve ; 76(4): 343-351, 2024 Apr.
Article in Japanese | MEDLINE | ID: mdl-38589279

ABSTRACT

A definite diagnosis of neurodegenerative diseases is required for neuropathological examination during an autopsy. Each neurodegenerative disease has specific vulnerable regions and affected systems (system degeneration), and is typified by an accumulation of abnormal protein with the formation of characteristic morphological aggregates in the nerve and glial cells, called proteinopathy. The most common neurodegenerative diseases are tauopathy, such as progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), and Pick's disease (PiD); α-synucleinopathy, including multiple system atrophy (MSA); and TAR DNA-binding protein of 43 kDa (TDP-43) proteinopathy, including amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). PSP and CBD show characteristic tau-positive astrocytic inclusions known as tufted astrocytes and astrocytic plaques, respectively. PiD shows tau-positive neuronal inclusions termed Pick bodies. MSA is characterized by α-synuclein-positive oligodendroglial inclusions, called glial cytoplasmic inclusions. ALS- and FTLD-TDP show TDP-43-positive neuronal inclusions, such as skein-like and round inclusions. Huntington's disease shows polyglutamine-positive neuronal inclusions, and Creutzfeldt-Jakob disease shows diffuse deposition of granular prions in the neuropil. The atypical proteins in these diseases have abnormal conformational properties. A comprehensive comparison of the clinical findings and neuropathological observations, including neuroanatomy and images acquired during life, is important to improve the sensitivity of clinical diagnosis.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Frontotemporal Lobar Degeneration , Multiple System Atrophy , Pick Disease of the Brain , Tauopathies , Humans , tau Proteins/metabolism , Amyotrophic Lateral Sclerosis/pathology , Tauopathies/metabolism , Tauopathies/pathology , Pick Disease of the Brain/metabolism , Pick Disease of the Brain/pathology , DNA-Binding Proteins/metabolism
2.
Acta Neuropathol ; 147(1): 58, 2024 Mar 23.
Article in English | MEDLINE | ID: mdl-38520489

ABSTRACT

Neurodegenerative pathologies such as Alzheimer disease neuropathologic change (ADNC), Lewy body disease (LBD), limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC), and cerebrovascular disease (CVD) frequently coexist, but little is known about the exact contribution of each pathology to cognitive decline and dementia in subjects with mixed pathologies. We explored the relative cognitive impact of concurrent common and rare neurodegenerative pathologies employing multivariate logistic regression analysis adjusted for age, gender, and level of education. We analyzed a cohort of 6,262 subjects from the National Alzheimer's Coordinating Center database, ranging from 0 to 6 comorbid neuropathologic findings per individual, where 95.7% of individuals had at least 1 neurodegenerative finding at autopsy and 75.5% had at least 2 neurodegenerative findings. We identified which neuropathologic entities correlate most frequently with one another and demonstrated that the total number of pathologies per individual was directly correlated with cognitive performance as assessed by Clinical Dementia Rating (CDR®) and Mini-Mental State Examination (MMSE). We show that ADNC, LBD, LATE-NC, CVD, hippocampal sclerosis, Pick disease, and FTLD-TDP significantly impact overall cognition as independent variables. More specifically, ADNC significantly affected all assessed cognitive domains, LBD affected attention, processing speed, and language, LATE-NC primarily affected tests related to logical memory and language, while CVD and other less common pathologies (including Pick disease, progressive supranuclear palsy, and corticobasal degeneration) had more variable neurocognitive effects. Additionally, ADNC, LBD, and higher numbers of comorbid neuropathologies were associated with the presence of at least one APOE ε4 allele, and ADNC and higher numbers of neuropathologies were inversely correlated with APOE ε2 alleles. Understanding the mechanisms by which individual and concomitant neuropathologies affect cognition and the degree to which each contributes is an imperative step in the development of biomarkers and disease-modifying therapeutics, particularly as these medical interventions become more targeted and personalized.


Subject(s)
Alzheimer Disease , Cardiovascular Diseases , Dementia , Frontotemporal Dementia , Lewy Body Disease , Pick Disease of the Brain , TDP-43 Proteinopathies , Humans , Pick Disease of the Brain/pathology , Brain/pathology , Alzheimer Disease/pathology , Lewy Body Disease/complications , Lewy Body Disease/pathology , Frontotemporal Dementia/pathology , Cognition
3.
Acta Neuropathol Commun ; 12(1): 31, 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38389095

ABSTRACT

Pick's disease (PiD) is a subtype of the tauopathy form of frontotemporal lobar degeneration (FTLD-tau) characterized by intraneuronal 3R-tau inclusions. PiD can underly various dementia syndromes, including primary progressive aphasia (PPA), characterized by an isolated and progressive impairment of language and left-predominant atrophy, and behavioral variant frontotemporal dementia (bvFTD), characterized by progressive dysfunction in personality and bilateral frontotemporal atrophy. In this study, we investigated the neocortical and hippocampal distributions of Pick bodies in bvFTD and PPA to establish clinicopathologic concordance between PiD and the salience of the aphasic versus behavioral phenotype. Eighteen right-handed cases with PiD as the primary pathologic diagnosis were identified from the Northwestern University Alzheimer's Disease Research Center brain bank (bvFTD, N = 9; PPA, N = 9). Paraffin-embedded sections were stained immunohistochemically with AT8 to visualize Pick bodies, and unbiased stereological analysis was performed in up to six regions bilaterally [middle frontal gyrus (MFG), superior temporal gyrus (STG), inferior parietal lobule (IPL), anterior temporal lobe (ATL), dentate gyrus (DG) and CA1 of the hippocampus], and unilateral occipital cortex (OCC). In bvFTD, peak neocortical densities of Pick bodies were in the MFG, while the ATL was the most affected in PPA. Both the IPL and STG had greater leftward pathology in PPA, with the latter reaching significance (p < 0.01). In bvFTD, Pick body densities were significantly right-asymmetric in the STG (p < 0.05). Hippocampal burden was not clinicopathologically concordant, as both bvFTD and PPA cases demonstrated significant hippocampal pathology compared to neocortical densities (p < 0.0001). Inclusion-to-neuron analyses in a subset of PPA cases confirmed that neurons in the DG are disproportionately burdened with inclusions compared to neocortical areas. Overall, stereological quantitation suggests that the distribution of neocortical Pick body pathology is concordant with salient clinical features unique to PPA vs. bvFTD while raising intriguing questions about the selective vulnerability of the hippocampus to 3R-tauopathies.


Subject(s)
Alzheimer Disease , Frontotemporal Dementia , Frontotemporal Lobar Degeneration , Pick Disease of the Brain , Tauopathies , Humans , Pick Disease of the Brain/pathology , Frontotemporal Dementia/pathology , Alzheimer Disease/pathology , Brain/pathology , Frontotemporal Lobar Degeneration/pathology , Atrophy/pathology , Tauopathies/pathology
4.
J Neuropathol Exp Neurol ; 83(4): 238-244, 2024 03 20.
Article in English | MEDLINE | ID: mdl-38412343

ABSTRACT

The pathobiology of tau is of great importance for understanding the mechanisms of neurodegeneration in aging and age-associated disorders such as Alzheimer disease (AD) and frontotemporal dementias. It is critical to identify neuronal populations and brain regions that are vulnerable or resistant to tau pathological changes. Pick disease (PiD) is a three-repeat (3R) tauopathy that belongs to the group of frontotemporal lobar degenerations. The neuropathologic changes of PiD are characterized by globular tau-positive neuronal intracytoplasmic inclusions, called Pick bodies, in the granule cells of the dentate gyrus and frontal and temporal neocortices, and ballooned neurons, named Pick neurons, in the neocortex. In the present study, we examined 13 autopsy-confirmed cases of PiD. Using immunohistochemistry for phospho-tau (AT8) and 3R tau isoform, all PiD cases demonstrated extensive lesions involving the hippocampus and neocortex. However, the lateral geniculate body (LGB) is spared of significant tau lesions in contrast to the neighboring hippocampus and other thalamic nuclei. Only 1 PiD case (7.7%) had tau-positive neurons, and 4 cases had tau-positive neurites (31%) in the LGB. By contrast, the LGB does consistently harbor tau lesions in other tauopathies including progressive supranuclear palsy, corticobasal degeneration, and AD.


Subject(s)
Alzheimer Disease , Neocortex , Pick Disease of the Brain , Tauopathies , Humans , Pick Disease of the Brain/pathology , tau Proteins/metabolism , Geniculate Bodies/metabolism , Geniculate Bodies/pathology , Tauopathies/pathology , Neocortex/pathology
5.
Brain ; 147(4): 1399-1411, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-37972275

ABSTRACT

The most frequent neurodegenerative proteinopathies include diseases with deposition of misfolded tau or α-synuclein in the brain. Pathological protein aggregates in the PNS are well-recognized in α-synucleinopathies and have recently attracted attention as a diagnostic biomarker. However, there is a paucity of observations in tauopathies. To characterize the involvement of the PNS in tauopathies, we investigated tau pathology in cranial and spinal nerves (PNS-tau) in 54 tauopathy cases [progressive supranuclear palsy (PSP), n = 15; Alzheimer's disease (AD), n = 18; chronic traumatic encephalopathy (CTE), n = 5; and corticobasal degeneration (CBD), n = 6; Pick's disease, n = 9; limbic-predominant neuronal inclusion body 4-repeat tauopathy (LNT), n = 1] using immunohistochemistry, Gallyas silver staining, biochemistry, and seeding assays. Most PSP cases revealed phosphorylated and 4-repeat tau immunoreactive tau deposits in the PNS as follows: (number of tau-positive cases/available cases) cranial nerves III: 7/8 (88%); IX/X: 10/11 (91%); and XII: 6/6 (100%); anterior spinal roots: 10/10 (100%). The tau-positive inclusions in PSP often showed structures with fibrillary (neurofibrillary tangle-like) morphology in the axon that were also recognized with Gallyas silver staining. CBD cases rarely showed fine granular non-argyrophilic tau deposits. In contrast, tau pathology in the PNS was not evident in AD, CTE and Pick's disease cases. The single LNT case also showed tau pathology in the PNS. In PSP, the severity of PNS-tau involvement correlated with that of the corresponding nuclei, although, occasionally, p-tau deposits were present in the cranial nerves but not in the related brainstem nuclei. Not surprisingly, most of the PSP cases presented with eye movement disorder and bulbar symptoms, and some cases also showed lower-motor neuron signs. Using tau biosensor cells, for the first time we demonstrated seeding capacity of tau in the PNS. In conclusion, prominent PNS-tau distinguishes PSP from other tauopathies. The morphological differences of PNS-tau between PSP and CBD suggest that the tau pathology in PNS could reflect that in the central nervous system. The high frequency and early presence of tau lesions in PSP suggest that PNS-tau may have clinical and biomarker relevance.


Subject(s)
Alzheimer Disease , Pick Disease of the Brain , Supranuclear Palsy, Progressive , Tauopathies , Humans , Supranuclear Palsy, Progressive/pathology , tau Proteins/metabolism , Pick Disease of the Brain/pathology , Alzheimer Disease/pathology , Tauopathies/pathology , Spinal Nerves , Biomarkers
6.
Brain ; 147(2): 590-606, 2024 02 01.
Article in English | MEDLINE | ID: mdl-37703311

ABSTRACT

Frontotemporal lobar degeneration with tau (FTLD-tau) is a group of tauopathies that underlie ∼50% of FTLD cases. Identification of genetic risk variants related to innate/adaptive immunity have highlighted a role for neuroinflammation and neuroimmune interactions in FTLD. Studies have shown microglial and astrocyte activation together with T cell infiltration in the brain of THY-Tau22 tauopathy mice. However, this remains to be confirmed in FTLD-tau patients. We conducted a detailed post-mortem study of FTLD-tau cases including 45 progressive supranuclear palsy with clinical frontotemporal dementia, 33 Pick's disease, 12 FTLD-MAPT and 52 control brains to characterize the link between phosphorylated tau (pTau) epitopes and the innate and adaptive immunity. Tau pathology was assessed in the cerebral cortex using antibodies directed against: Tau-2 (phosphorylated and unphosphorylated tau), AT8 (pSer202/pThr205), AT100 (pThr212/pSer214), CP13 (pSer202), PHF1 (pSer396/pSer404), pThr181 and pSer356. The immunophenotypes of microglia and astrocytes were assessed with phenotypic markers (Iba1, CD68, HLA-DR, CD64, CD32a, CD16 for microglia and GFAP, EAAT2, glutamine synthetase and ALDH1L1 for astrocytes). The adaptive immune response was explored via CD4+ and CD8+ T cell quantification and the neuroinflammatory environment was investigated via the expression of 30 inflammatory-related proteins using V-Plex Meso Scale Discovery. As expected, all pTau markers were increased in FTLD-tau cases compared to controls. pSer356 expression was greatest in FTLD-MAPT cases versus controls (P < 0.0001), whereas the expression of other markers was highest in Pick's disease. Progressive supranuclear palsy with frontotemporal dementia consistently had a lower pTau protein load compared to Pick's disease across tau epitopes. The only microglial marker increased in FTLD-tau was CD16 (P = 0.0292) and specifically in FTLD-MAPT cases (P = 0.0150). However, several associations were detected between pTau epitopes and microglia, supporting an interplay between them. GFAP expression was increased in FTLD-tau (P = 0.0345) with the highest expression in Pick's disease (P = 0.0019), while ALDH1L1 was unchanged. Markers of astrocyte glutamate cycling function were reduced in FTLD-tau (P = 0.0075; Pick's disease: P < 0.0400) implying astrocyte reactivity associated with a decreased glutamate cycling activity, which was further associated with pTau expression. Of the inflammatory proteins assessed in the brain, five chemokines were upregulated in Pick's disease cases (P < 0.0400), consistent with the recruitment of CD4+ (P = 0.0109) and CD8+ (P = 0.0014) T cells. Of note, the CD8+ T cell infiltration was associated with pTau epitopes and microglial and astrocytic markers. Our results highlight that FTLD-tau is associated with astrocyte reactivity, remarkably little activation of microglia, but involvement of adaptive immunity in the form of chemokine-driven recruitment of T lymphocytes.


Subject(s)
Frontotemporal Dementia , Frontotemporal Lobar Degeneration , Pick Disease of the Brain , Supranuclear Palsy, Progressive , Tauopathies , Humans , Epitopes , Frontotemporal Dementia/pathology , Frontotemporal Lobar Degeneration/pathology , Glutamates , Pick Disease of the Brain/pathology , Supranuclear Palsy, Progressive/pathology , tau Proteins/metabolism , Tauopathies/pathology
7.
Clin Neuropathol ; 42(6): 212-218, 2023.
Article in English | MEDLINE | ID: mdl-37840526

ABSTRACT

Pick's disease (PiD) is a rare form of frontal temporal lobar degeneration. The pathognomonic feature is atrophy of the frontotemporal lobes and intraneuronal deposits of 3R-τ inclusions, the Pick body. Corticobasal syndrome (CBS) is an atypical parkinsonian syndrome with a heterogeneous spectrum of underlying pathologies. We report a case of clinically diagnosed CBS with a post-mortem diagnosis of PiD and conduct a clinicopathological review of the literature on this unusual presentation.


Subject(s)
Corticobasal Degeneration , Pick Disease of the Brain , Humans , Pick Disease of the Brain/pathology , Atrophy , tau Proteins
8.
Int J Mol Sci ; 24(19)2023 Sep 27.
Article in English | MEDLINE | ID: mdl-37834094

ABSTRACT

Cognitive dysfunction is an important non-motor symptom in amyotrophic lateral sclerosis (ALS) that has a negative impact on survival and caregiver burden. It shows a wide spectrum ranging from subjective cognitive decline to frontotemporal dementia (FTD) and covers various cognitive domains, mainly executive/attention, language and verbal memory deficits. The frequency of cognitive impairment across the different ALS phenotypes ranges from 30% to 75%, with up to 45% fulfilling the criteria of FTD. Significant genetic, clinical, and pathological heterogeneity reflects deficits in various cognitive domains. Modern neuroimaging studies revealed frontotemporal degeneration and widespread involvement of limbic and white matter systems, with hypometabolism of the relevant areas. Morphological substrates are frontotemporal and hippocampal atrophy with synaptic loss, associated with TDP-43 and other co-pathologies, including tau deposition. Widespread functional disruptions of motor and extramotor networks, as well as of frontoparietal, frontostriatal and other connectivities, are markers for cognitive deficits in ALS. Cognitive reserve may moderate the effect of brain damage but is not protective against cognitive decline. The natural history of cognitive dysfunction in ALS and its relationship to FTD are not fully understood, although there is an overlap between the ALS variants and ALS-related frontotemporal syndromes, suggesting a differential vulnerability of motor and non-motor networks. An assessment of risks or the early detection of brain connectivity signatures before structural changes may be helpful in investigating the pathophysiological mechanisms of cognitive impairment in ALS, which might even serve as novel targets for effective disease-modifying therapies.


Subject(s)
Amyotrophic Lateral Sclerosis , Cognition Disorders , Cognitive Dysfunction , Frontotemporal Dementia , Neurodegenerative Diseases , Pick Disease of the Brain , Humans , Amyotrophic Lateral Sclerosis/genetics , Frontotemporal Dementia/genetics , Brain/pathology , Cognition Disorders/pathology , Cognitive Dysfunction/pathology , Neurodegenerative Diseases/pathology , Pick Disease of the Brain/pathology
9.
Acta Neuropathol Commun ; 11(1): 168, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37864255

ABSTRACT

Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) comprise a spectrum of neurodegenerative diseases linked to TDP-43 proteinopathy, which at the cellular level, is characterized by loss of nuclear TDP-43 and accumulation of cytoplasmic TDP-43 inclusions that ultimately cause RNA processing defects including dysregulation of splicing, mRNA transport and translation. Complementing our previous work in motor neurons, here we report a novel model of TDP-43 proteinopathy based on overexpression of TDP-43 in a subset of Drosophila Kenyon cells of the mushroom body (MB), a circuit with structural characteristics reminiscent of vertebrate cortical networks. This model recapitulates several aspects of dementia-relevant pathological features including age-dependent neuronal loss, nuclear depletion and cytoplasmic accumulation of TDP-43, and behavioral deficits in working memory and sleep that occur prior to axonal degeneration. RNA immunoprecipitations identify several candidate mRNA targets of TDP-43 in MBs, some of which are unique to the MB circuit and others that are shared with motor neurons. Among the latter is the glypican Dally-like-protein (Dlp), which exhibits significant TDP-43 associated reduction in expression during aging. Using genetic interactions we show that overexpression of Dlp in MBs mitigates TDP-43 dependent working memory deficits, conistent with Dlp acting as a mediator of TDP-43 toxicity. Substantiating our findings in the fly model, we find that the expression of GPC6 mRNA, a human ortholog of dlp, is specifically altered in neurons exhibiting the molecular signature of TDP-43 pathology in FTD patient brains. These findings suggest that circuit-specific Drosophila models provide a platform for uncovering shared or disease-specific molecular mechanisms and vulnerabilities across the spectrum of TDP-43 proteinopathies.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Pick Disease of the Brain , TDP-43 Proteinopathies , Animals , Humans , Amyotrophic Lateral Sclerosis/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila/metabolism , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Motor Neurons/metabolism , Pick Disease of the Brain/pathology , RNA, Messenger , TDP-43 Proteinopathies/pathology
10.
J Biol Chem ; 299(11): 105252, 2023 11.
Article in English | MEDLINE | ID: mdl-37714465

ABSTRACT

Neurodegenerative tauopathies are caused by the transition of tau protein from a monomer to a toxic aggregate. They include Alzheimer disease (AD), progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), and Pick disease (PiD). We have previously proposed that tau monomer exists in two conformational ensembles: an inert form (Mi), which does not self-assemble, and seed-competent form (Ms), which self-assembles and templates ordered assembly growth. We proposed that cis/trans isomerization of tau at P301, the site of dominant disease-associated S/L missense mutations, might underlie the transition of wild-type tau to a seed-competent state. Consequently, we created monoclonal antibodies using non-natural antigens consisting of fluorinated proline (P∗) at the analogous P270 in repeat 1 (R1), biased toward the trans-configuration at either the R1/R2 (TENLKHQP∗GGGKVQIINKK) or the R1/R3 (TENLKHQP∗GGGKVQIVYK) interfaces. Two antibodies, MD2.2 and MD3.1, efficiently immunoprecipitated soluble seeds from AD and PSP but not CBD or PiD brain samples. The antibodies efficiently stained brain samples of AD, PSP, and PiD, but not CBD. They did not immunoprecipitate or immunostain tau from the control brain. Creation of potent anti-seed antibodies based on the trans-proline epitope implicates local unfolding around P301 in pathogenesis. MD2.2 and MD3.1 may also be useful for therapy and diagnosis.


Subject(s)
Tauopathies , Humans , Alzheimer Disease/metabolism , Antibodies, Monoclonal/metabolism , Brain/metabolism , Epitopes/metabolism , Pick Disease of the Brain/metabolism , Pick Disease of the Brain/pathology , Proline/metabolism , tau Proteins/metabolism , Tauopathies/metabolism
11.
Acta Neuropathol Commun ; 11(1): 130, 2023 08 10.
Article in English | MEDLINE | ID: mdl-37563653

ABSTRACT

Right temporal variant frontotemporal dementia, also called right-predominant semantic dementia, often has an unclear position within the framework of the updated diagnostic criteria for behavioral variant frontotemporal dementia or primary progressive aphasia. Recent studies have suggested that this population may be clinically, neuropathologically, and genetically distinct from those with behavioral variant frontotemporal dementia or left-predominant typical semantic variant primary progressive aphasia. Here we describe a Japanese case of right temporal variant frontotemporal dementia with novel heterozygous MAPT mutation Adenine to Thymidine in intervening sequence (IVS) 9 at position -7 from 3' splicing site of intron 9/exon 10 boundary (MAPT IVS9-7A > T). Postmortem neuropathological analysis revealed a predominant accumulation of 4 repeat tau, especially in the temporal lobe, amygdala, and substantia nigra, but lacked astrocytic plaques or tufted astrocytes. Immunoelectron microscopy of the tau filaments extracted from the brain revealed a ribbon-like structure. Moreover, a cellular MAPT splicing assay confirmed that this novel variant promoted the inclusion of exon 10, resulting in the predominant production of 4 repeat tau. These data strongly suggest that the MAPT IVS9-7 A > T variant found in our case is a novel mutation that stimulates the inclusion of exon 10 through alternative splicing of MAPT transcript and causes predominant 4 repeat tauopathy which clinically presents as right temporal variant frontotemporal dementia.


Subject(s)
Aphasia, Primary Progressive , Frontotemporal Dementia , Pick Disease of the Brain , Tauopathies , Humans , Aphasia, Primary Progressive/pathology , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Introns/genetics , Mutation , Pick Disease of the Brain/pathology , tau Proteins/genetics , tau Proteins/metabolism , Tauopathies/genetics , Tauopathies/pathology , Temporal Lobe/metabolism
12.
Cortex ; 165: 101-118, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37276800

ABSTRACT

In his epoch-making monograph, Wernicke (1874) claimed that atrophy of the brain cannot cause aphasia. Refuting this claim, Pick (1892, 1898, 1901, 1904a) documented in increasing detail several cases of aphasia with circumscribed atrophy of the left temporal lobe, frontal lobe, or both, which persuaded Wernicke (1906). To explain why the atrophy is circumscribed and leads to focal symptoms, Pick (1908a) advanced a functional network account. Behavioral, neuroanatomical, and histopathological studies by Dejerine and Sérieux, Fischer, Alzheimer, Altman, Gans, Onari and Spatz, and Stertz further illuminated the clinical syndromes, the exact spatial distributions of the atrophy, the underlying disease, and its laminar specificity. Unaware of these seminal studies, research from the 1970s until now has independently rediscovered all key findings, and also supports Pick's forgotten functional account of the distribution of atrophy and the focal symptoms. His frontal and temporal forms of aphasia foreshadowed what are now called the nonfluent/agrammatic and semantic variants of primary progressive aphasia. Moreover, aphasic symptoms may occur with frontal degeneration (what used to be called "Pick's disease") that yields personality changes and behavioral disturbances, now called the behavioral variant of frontotemporal dementia.


Subject(s)
Aphasia, Primary Progressive , Aphasia , Frontotemporal Dementia , Pick Disease of the Brain , Humans , Pick Disease of the Brain/pathology , Atrophy/pathology , Brain
13.
Alzheimers Dement ; 19(11): 5253-5263, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37379561

ABSTRACT

INTRODUCTION: The history of frontotemporal dementia (FTD) is both old and new. This study explores its historical roots, dating back to the 19th century, while recognizes it as a distinct neurological entity only a few decades ago. METHODS: This qualitative study and literature review provides an overview of FTD's historical background, birth, evolution, and future directions. RESULTS: Recognition of FTD was hindered by rigid perceptions of dementia, the division between neurology and psychiatry, reliance on IQ-based assessment, limited neuroimaging capabilities, and lack of pathological proof. Overcoming these barriers involved revisiting early pioneers' approaches, focusing on focal impairment, establishing non-Alzheimer's disease cohorts, fostering collaboration, and developing diagnostic criteria. Current gaps include the need for biology-oriented psychiatry education, biological biomarkers, and culturally sensitive, objective clinical instruments predicting underlying pathology. DISCUSSION: Independent multidisciplinary centers are essential. The future of FTD lies in disease-modifying therapies, presenting new opportunities for healthcare professionals and researchers.


Subject(s)
Frontotemporal Dementia , Neurology , Pick Disease of the Brain , Humans , Frontotemporal Dementia/diagnosis , Pick Disease of the Brain/history , Pick Disease of the Brain/pathology , Neuroimaging , Biomarkers
14.
Acta Neuropathol ; 146(1): 77-95, 2023 07.
Article in English | MEDLINE | ID: mdl-37149835

ABSTRACT

Frontotemporal lobar degeneration (FTLD) is an umbrella term describing the neuropathology of a clinically, genetically and pathologically heterogeneous group of diseases, including frontotemporal dementia (FTD) and progressive supranuclear palsy (PSP). Among the major FTLD pathological subgroups, FTLD with TDP-43 positive inclusions (FTLD-TDP) and FTLD with tau-positive inclusions (FTLD-tau) are the most common, representing about 90% of the cases. Although alterations in DNA methylation have been consistently associated with neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease, little is known for FTLD and its heterogeneous subgroups and subtypes. The main goal of this study was to investigate DNA methylation variation in FTLD-TDP and FTLD-tau. We used frontal cortex genome-wide DNA methylation profiles from three FTLD cohorts (142 FTLD cases and 92 controls), generated using the Illumina 450K or EPIC microarrays. We performed epigenome-wide association studies (EWAS) for each cohort followed by meta-analysis to identify shared differentially methylated loci across FTLD subgroups/subtypes. In addition, we used weighted gene correlation network analysis to identify co-methylation signatures associated with FTLD and other disease-related traits. Wherever possible, we also incorporated relevant gene/protein expression data. After accounting for a conservative Bonferroni multiple testing correction, the EWAS meta-analysis revealed two differentially methylated loci in FTLD, one annotated to OTUD4 (5'UTR-shore) and the other to NFATC1 (gene body-island). Of these loci, OTUD4 showed consistent upregulation of mRNA and protein expression in FTLD. In addition, in the three independent co-methylation networks, OTUD4-containing modules were enriched for EWAS meta-analysis top loci and were strongly associated with the FTLD status. These co-methylation modules were enriched for genes implicated in the ubiquitin system, RNA/stress granule formation and glutamatergic synaptic signalling. Altogether, our findings identified novel FTLD-associated loci, and support a role for DNA methylation as a mechanism involved in the dysregulation of biological processes relevant to FTLD, highlighting novel potential avenues for therapeutic development.


Subject(s)
Frontotemporal Dementia , Frontotemporal Lobar Degeneration , Pick Disease of the Brain , Humans , Frontotemporal Dementia/pathology , Frontotemporal Lobar Degeneration/pathology , Brain/pathology , Pick Disease of the Brain/pathology , DNA , tau Proteins/metabolism , Ubiquitin-Specific Proteases/metabolism
15.
Acta Neuropathol Commun ; 11(1): 70, 2023 04 28.
Article in English | MEDLINE | ID: mdl-37118844

ABSTRACT

Loss of function progranulin (GRN) mutations are a major autosomal dominant cause of frontotemporal dementia (FTD). Patients with FTD due to GRN mutations (FTD-GRN) develop frontotemporal lobar degeneration with TDP-43 pathology type A (FTLD-TDP type A) and exhibit elevated levels of lysosomal proteins and storage material in frontal cortex, perhaps indicating lysosomal dysfunction as a mechanism of disease. To investigate whether patients with sporadic FTLD exhibit similar signs of lysosomal dysfunction, we compared lysosomal protein levels, transcript levels, and storage material in patients with FTD-GRN or sporadic FTLD-TDP type A. We analyzed samples from frontal cortex, a degenerated brain region, and occipital cortex, a relatively spared brain region. In frontal cortex, patients with sporadic FTLD-TDP type A exhibited similar increases in lysosomal protein levels, transcript levels, and storage material as patients with FTD-GRN. In occipital cortex of both patient groups, most lysosomal measures did not differ from controls. Frontal cortex from a transgenic mouse model of TDP-opathy had similar increases in cathepsin D and lysosomal storage material, showing that TDP-opathy and neurodegeneration can drive these changes independently of progranulin. To investigate these changes in additional FTLD subtypes, we analyzed frontal cortical samples from patients with sporadic FTLD-TDP type C or Pick's disease, an FTLD-tau subtype. All sporadic FTLD groups had similar increases in cathepsin D activity, lysosomal membrane proteins, and storage material as FTD-GRN patients. However, patients with FTLD-TDP type C or Pick's disease did not have similar increases in lysosomal transcripts as patients with FTD-GRN or sporadic FTLD-TDP type A. Based on these data, accumulation of lysosomal proteins and storage material may be a common aspect of end-stage FTLD. However, the unique changes in gene expression in patients with FTD-GRN or sporadic FTLD-TDP type A may indicate distinct underlying lysosomal changes among FTLD subtypes.


Subject(s)
Frontotemporal Dementia , Frontotemporal Lobar Degeneration , Pick Disease of the Brain , Mice , Animals , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Pick Disease of the Brain/pathology , Progranulins/genetics , Cathepsin D/genetics , Frontotemporal Lobar Degeneration/pathology , Mutation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mice, Transgenic
16.
Lab Invest ; 103(6): 100127, 2023 06.
Article in English | MEDLINE | ID: mdl-36889541

ABSTRACT

Neuropathologic assessment during autopsy is the gold standard for diagnosing neurodegenerative disorders. Neurodegenerative conditions, such as Alzheimer disease (AD) neuropathological change, are a continuous process from normal aging rather than categorical; therefore, diagnosing neurodegenerative disorders is a complicated task. We aimed to develop a pipeline for diagnosing AD and other tauopathies, including corticobasal degeneration (CBD), globular glial tauopathy, Pick disease, and progressive supranuclear palsy. We used a weakly supervised deep learning-based approach called clustering-constrained-attention multiple-instance learning (CLAM) on the whole-slide images (WSIs) of patients with AD (n = 30), CBD (n = 20), globular glial tauopathy (n = 10), Pick disease (n = 20), and progressive supranuclear palsy (n = 20), as well as nontauopathy controls (n = 21). Three sections (A: motor cortex; B: cingulate gyrus and superior frontal gyrus; and C: corpus striatum) that had been immunostained for phosphorylated tau were scanned and converted to WSIs. We evaluated 3 models (classic multiple-instance learning, single-attention-branch CLAM, and multiattention-branch CLAM) using 5-fold cross-validation. Attention-based interpretation analysis was performed to identify the morphologic features contributing to the classification. Within highly attended regions, we also augmented gradient-weighted class activation mapping to the model to visualize cellular-level evidence of the model's decisions. The multiattention-branch CLAM model using section B achieved the highest area under the curve (0.970 ± 0.037) and diagnostic accuracy (0.873 ± 0.087). A heatmap showed the highest attention in the gray matter of the superior frontal gyrus in patients with AD and the white matter of the cingulate gyrus in patients with CBD. Gradient-weighted class activation mapping showed the highest attention in characteristic tau lesions for each disease (eg, numerous tau-positive threads in the white matter inclusions for CBD). Our findings support the feasibility of deep learning-based approaches for the classification of neurodegenerative disorders on WSIs. Further investigation of this method, focusing on clinicopathologic correlations, is warranted.


Subject(s)
Alzheimer Disease , Deep Learning , Neurodegenerative Diseases , Pick Disease of the Brain , Supranuclear Palsy, Progressive , Tauopathies , Humans , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Supranuclear Palsy, Progressive/diagnostic imaging , Supranuclear Palsy, Progressive/pathology , Pick Disease of the Brain/pathology , tau Proteins , Tauopathies/diagnostic imaging , Tauopathies/pathology
17.
J Neuroinflammation ; 20(1): 65, 2023 Mar 08.
Article in English | MEDLINE | ID: mdl-36890594

ABSTRACT

BACKGROUND: Neuroinflammation plays a significant role in the progression of frontotemporal dementia (FTD). However, the association between peripheral inflammatory factors and brain neurodegeneration is poorly understood. We aimed to examine changes in peripheral inflammatory markers in patients with behavioural variant FTD (bvFTD) and explore the potential association between peripheral inflammation and brain structure, metabolism, and clinical parameters. METHODS: Thirty-nine bvFTD patients and 40 healthy controls were enrolled and underwent assessment of plasma inflammatory factors, positron emission tomography/magnetic resonance imaging, and neuropsychological assessments. Group differences were tested using Student's t test, Mann‒Whitney U test, or ANOVA. Partial correlation analysis and multivariable regression analysis were implemented using age and sex as covariates to explore the association between peripheral inflammatory markers, neuroimaging, and clinical measures. The false discovery rate was used to correct for the multiple correlation test. RESULTS: Plasma levels of six factors, including interleukin (IL)-2, IL-12p70, IL-17A, tumour necrosis superfamily member 13B (TNFSF/BAFF), TNFSF12 (TWEAK), and TNFRSF8 (sCD30), were increased in the bvFTD group. Five factors were significantly associated with central degeneration, including IL-2, IL-12p70, IL-17A, sCD30/TNFRSF8, and tumour necrosis factor (TNF)-α; the association between inflammation and brain atrophy was mainly distributed in frontal-limbic-striatal brain regions, whereas the association with brain metabolism was mainly in the frontal-temporal-limbic-striatal regions. BAFF/TNFSF13B, IL-4, IL-6, IL-17A and TNF-α were found to correlate with clinical measures. CONCLUSION: Peripheral inflammation disturbance in patients with bvFTD participates in disease-specific pathophysiological mechanisms, which could be a promising target for diagnosis, treatment, and monitoring therapeutic efficacy.


Subject(s)
Frontotemporal Dementia , Pick Disease of the Brain , Humans , Frontotemporal Dementia/complications , Frontotemporal Dementia/diagnostic imaging , Interleukin-17/metabolism , Brain/metabolism , Pick Disease of the Brain/pathology , Magnetic Resonance Imaging , Neuropsychological Tests , Inflammation/pathology
18.
J Histochem Cytochem ; 71(2): 73-86, 2023 02.
Article in English | MEDLINE | ID: mdl-36861683

ABSTRACT

Tau phosphorylation, aggregation, and toxicity are the main drivers of neurodegeneration in multiple tauopathies, including Alzheimer's disease (AD) and frontotemporal lobar degeneration with tau. Although aggregation and amyloid formation are often assumed to be synonymous, the ability of tau aggregates in different diseases to form amyloids in vivo has not been systematically studied. We used the amyloid dye Thioflavin S to look at tau aggregates in mixed tauopathies such as AD and primary age-related tauopathy, as well as pure 3R or 4R tauopathies such as Pick's disease, progressive supranuclear palsy, and corticobasal degeneration. We found that aggregates of tau protein only form thioflavin-positive amyloids in mixed (3R/4R), but not pure (3R or 4R), tauopathies. Interestingly, neither astrocytic nor neuronal tau pathology was thioflavin-positive in pure tauopathies. As most current positron emission tomography tracers are based on thioflavin derivatives, this suggests that they may be more useful for differential diagnosis than the identification of a general tauopathy. Our findings also suggest that thioflavin staining may have utility as an alternative to traditional antibody staining for distinguishing between tau aggregates in patients with multiple pathologies and that the mechanisms for tau toxicity may differ between different tauopathies.


Subject(s)
Alzheimer Disease , Pick Disease of the Brain , Supranuclear Palsy, Progressive , Tauopathies , Humans , Alzheimer Disease/diagnosis , Alzheimer Disease/pathology , Brain/pathology , Neurons/metabolism , Pick Disease of the Brain/metabolism , Pick Disease of the Brain/pathology , Supranuclear Palsy, Progressive/pathology , Tauopathies/diagnosis , Tauopathies/pathology
19.
Brain ; 146(8): 3221-3231, 2023 08 01.
Article in English | MEDLINE | ID: mdl-36883644

ABSTRACT

Frontotemporal dementia is clinically and neuropathologically heterogeneous, but neuroinflammation, atrophy and cognitive impairment occur in all of its principal syndromes. Across the clinical spectrum of frontotemporal dementia, we assess the predictive value of in vivo neuroimaging measures of microglial activation and grey-matter volume on the rate of future cognitive decline. We hypothesized that inflammation is detrimental to cognitive performance, in addition to the effect of atrophy. Thirty patients with a clinical diagnosis of frontotemporal dementia underwent a baseline multimodal imaging assessment, including [11C]PK11195 PET to index microglial activation and structural MRI to quantify grey-matter volume. Ten people had behavioural variant frontotemporal dementia, 10 had the semantic variant of primary progressive aphasia and 10 had the non-fluent agrammatic variant of primary progressive aphasia. Cognition was assessed at baseline and longitudinally with the revised Addenbrooke's Cognitive Examination, at an average of 7-month intervals (for an average of ∼2 years, up to ∼5 years). Regional [11C]PK11195 binding potential and grey-matter volume were determined, and these were averaged within four hypothesis-driven regions of interest: bilateral frontal and temporal lobes. Linear mixed-effect models were applied to the longitudinal cognitive test scores, with [11C]PK11195 binding potentials and grey-matter volumes as predictors of cognitive performance, with age, education and baseline cognitive performance as covariates. Faster cognitive decline was associated with reduced baseline grey-matter volume and increased microglial activation in frontal regions, bilaterally. In frontal regions, microglial activation and grey-matter volume were negatively correlated, but provided independent information, with inflammation the stronger predictor of the rate of cognitive decline. When clinical diagnosis was included as a factor in the models, a significant predictive effect was found for [11C]PK11195 BPND in the left frontal lobe (-0.70, P = 0.01), but not for grey-matter volumes (P > 0.05), suggesting that inflammation severity in this region relates to cognitive decline regardless of clinical variant. The main results were validated by two-step prediction frequentist and Bayesian estimation of correlations, showing significant associations between the estimated rate of cognitive change (slope) and baseline microglial activation in the frontal lobe. These findings support preclinical models in which neuroinflammation (by microglial activation) accelerates the neurodegenerative disease trajectory. We highlight the potential for immunomodulatory treatment strategies in frontotemporal dementia, in which measures of microglial activation may also improve stratification for clinical trials.


Subject(s)
Aphasia, Primary Progressive , Cognitive Dysfunction , Frontotemporal Dementia , Neurodegenerative Diseases , Pick Disease of the Brain , Humans , Frontotemporal Dementia/metabolism , Neuroinflammatory Diseases , Neurodegenerative Diseases/pathology , Microglia/metabolism , Bayes Theorem , Frontal Lobe/pathology , Pick Disease of the Brain/pathology , Cognitive Dysfunction/metabolism , Magnetic Resonance Imaging/methods , Inflammation/pathology , Atrophy/pathology , Aphasia, Primary Progressive/pathology
20.
J Neuropathol Exp Neurol ; 82(2): 103-109, 2023 01 20.
Article in English | MEDLINE | ID: mdl-36458947

ABSTRACT

Concerns about the costs associated with autopsy assessment of Alzheimer disease and related dementias according to 2012 NIA-AA Guidelines have been expressed since the publication of those guidelines. For this reason, we designed and validated a Condensed Protocol for the neuropathologic diagnoses of Alzheimer disease neuropathologic change, Lewy Body disease neuropathologic change, as well as chronic microvascular lesions, hippocampal sclerosis of aging, and cerebral amyloid angiopathy. In this study, the Condensed Protocol is updated to include frontotemporal lobar degeneration [FTLD] tau (corticobasal degeneration, progressive supranuclear palsy, and Pick disease), FTLD-TDP, and limbic-predominant, age-related TDP-43 encephalopathy. The same 20 brain regions are sampled and processed in 5 tissue cassettes, which reduces reagent costs by approximately 65%. Three board-certified neuropathologists were blinded to the original Northwestern University Alzheimer's Disease Research Center Original Protocol neuropathological diagnoses and all clinical history information. The results yielded near uniform agreement with the original comprehensive Alzheimer's Disease Research Center neuropathologic assessments. Diagnostic sensitivity was not impacted. In summary, our recent results show that our updated Condensed Protocol is also an accurate and less expensive alternative to the comprehensive protocols for the additional neuropathologic diagnoses of FTLD Tau and TDP43 proteinopathies.


Subject(s)
Alzheimer Disease , Frontotemporal Dementia , Frontotemporal Lobar Degeneration , Pick Disease of the Brain , Humans , Alzheimer Disease/diagnosis , Alzheimer Disease/pathology , Neuropathology , Frontotemporal Lobar Degeneration/pathology , Pick Disease of the Brain/pathology , tau Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...