Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Clin Pharmacol Ther ; 105(5): 1234-1243, 2019 05.
Article in English | MEDLINE | ID: mdl-30347431

ABSTRACT

ß-site amyloid precursor protein cleaving enzyme 1 (BACE1) is required for the production of ß-amyloid (Aß) peptides and is considered a potential treatment target for Alzheimer's disease (AD). To support Japan's participation in the global clinical development program, we characterized the safety, pharmacokinetics (PKs), and pharmacodynamics of the BACE1 inhibitor verubecestat (MK-8931) in 24 healthy Japanese adults in a two-part, single-center, randomized, placebo-controlled phase I trial (protocol MK-8931-007) and compared the results with historical data from non-Japanese subjects. Both single (20, 100, and 450 mg) and multiple (80 and 150 mg once daily for 14 days) doses of verubecestat were well tolerated. Verubecestat's PK profile was similar in Japanese and non-Japanese subjects. Verubecestat also reduced mean cerebrospinal fluid concentrations of the Aß proteins Aß40, Aß42, and soluble ß fragment of amyloid precursor protein; the level of reduction was comparable between Japanese and non-Japanese subjects. These results support the continued global development of verubecestat as a potential disease-modifying agent for Japanese and non-Japanese subjects who are at risk for developing AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides , Aspartic Acid Endopeptidases/antagonists & inhibitors , Cyclic S-Oxides , Thiadiazines , Adult , Alzheimer Disease/ethnology , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Protein Precursor/blood , Amyloid beta-Protein Precursor/metabolism , Cyclic S-Oxides/administration & dosage , Cyclic S-Oxides/pharmacokinetics , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Monitoring/methods , Female , Healthy Volunteers , Humans , Japan , Male , Thiadiazines/administration & dosage , Thiadiazines/pharmacokinetics
2.
Sci Transl Med ; 8(363): 363ra150, 2016 11 02.
Article in English | MEDLINE | ID: mdl-27807285

ABSTRACT

ß-Amyloid (Aß) peptides are thought to be critically involved in the etiology of Alzheimer's disease (AD). The aspartyl protease ß-site amyloid precursor protein cleaving enzyme 1 (BACE1) is required for the production of Aß, and BACE1 inhibition is thus an attractive target for the treatment of AD. We show that verubecestat (MK-8931) is a potent, selective, structurally unique BACE1 inhibitor that reduced plasma, cerebrospinal fluid (CSF), and brain concentrations of Aß40, Aß42, and sAPPß (a direct product of BACE1 enzymatic activity) after acute and chronic administration to rats and monkeys. Chronic treatment of rats and monkeys with verubecestat achieved exposures >40-fold higher than those being tested in clinical trials in AD patients yet did not elicit many of the adverse effects previously attributed to BACE inhibition, such as reduced nerve myelination, neurodegeneration, altered glucose homeostasis, or hepatotoxicity. Fur hypopigmentation was observed in rabbits and mice but not in monkeys. Single and multiple doses were generally well tolerated and produced reductions in Aß40, Aß42, and sAPPß in the CSF of both healthy human subjects and AD patients. The human data were fit to an amyloid pathway model that provided insight into the Aß pools affected by BACE1 inhibition and guided the choice of doses for subsequent clinical trials.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/chemistry , Aspartic Acid Endopeptidases/antagonists & inhibitors , Central Nervous System/metabolism , Cyclic S-Oxides/pharmacology , Thiadiazines/pharmacology , Administration, Oral , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Brain/metabolism , Catalytic Domain , Crystallography, X-Ray , Drug Design , Female , Glucose/metabolism , Macaca fascicularis , Magnetic Resonance Spectroscopy , Mice , Myelin Sheath/chemistry , Peptides/chemistry , Rabbits , Rats
3.
Sci Rep ; 6: 27147, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27256922

ABSTRACT

Orexin neuropeptides regulate sleep/wake through orexin receptors (OX1R, OX2R); OX2R is the predominant mediator of arousal promotion. The potential for single OX2R antagonism to effectively promote sleep has yet to be demonstrated in humans. MK-1064 is an OX2R-single antagonist. Preclinically, MK-1064 promotes sleep and increases both rapid eye movement (REM) and non-REM (NREM) sleep in rats at OX2R occupancies higher than the range observed for dual orexin receptor antagonists. Similar to dual antagonists, MK-1064 increases NREM and REM sleep in dogs without inducing cataplexy. Two Phase I studies in healthy human subjects evaluated safety, tolerability, pharmacokinetics and sleep-promoting effects of MK-1064, and demonstrated dose-dependent increases in subjective somnolence (via Karolinska Sleepiness Scale and Visual Analogue Scale measures) and sleep (via polysomnography), including increased REM and NREM sleep. Thus, selective OX2R antagonism is sufficient to promote REM and NREM sleep across species, similarly to that seen with dual orexin receptor antagonism.


Subject(s)
Orexin Receptor Antagonists/administration & dosage , Orexin Receptors/metabolism , Sleep Aids, Pharmaceutical/administration & dosage , Sleep Stages/drug effects , Animals , Dogs , Dose-Response Relationship, Drug , Female , Humans , Male , Mice , Orexin Receptor Antagonists/pharmacology , Polysomnography , Rats , Sleep Aids, Pharmaceutical/pharmacology , Sleep, REM/drug effects
4.
J Neurosci ; 30(22): 7729-39, 2010 Jun 02.
Article in English | MEDLINE | ID: mdl-20519548

ABSTRACT

Inclusion body myopathy associated with Paget's disease of bone and frontotemporal dementia (IBMPFD) is a dominantly inherited degenerative disorder caused by mutations in the valosin-containing protein (VCP7) gene. VCP (p97 in mouse, TER94 in Drosophila melanogaster, and CDC48 in Saccharomyces cerevisiae) is a highly conserved AAA(+) (ATPases associated with multiple cellular activities) ATPase that regulates a wide array of cellular processes. The mechanism of IBMPFD pathogenesis is unknown. To elucidate the pathogenic mechanism, we developed and characterized a Drosophila model of IBMPFD (mutant-VCP-related degeneration). Based on genetic screening of this model, we identified three RNA-binding proteins that dominantly suppressed degeneration; one of these was TBPH, the Drosophila homolog of TAR (trans-activating response region) DNA-binding protein 43 (TDP-43). Here we demonstrate that VCP and TDP-43 interact genetically and that disease-causing mutations in VCP lead to redistribution of TDP-43 to the cytoplasm in vitro and in vivo, replicating the major pathology observed in IBMPFD and other TDP-43 proteinopathies. We also demonstrate that TDP-43 redistribution from the nucleus to the cytoplasm is sufficient to induce cytotoxicity. Furthermore, we determined that a pathogenic mutation in TDP-43 promotes redistribution to the cytoplasm and enhances the genetic interaction with VCP. Together, our results show that degeneration associated with VCP mutations is mediated in part by toxic gain of function of TDP-43 in the cytoplasm. We suggest that these findings are likely relevant to the pathogenic mechanism of a broad array of TDP-43 proteinopathies, including frontotemporal lobar degeneration and amyotrophic lateral sclerosis.


Subject(s)
Cell Cycle Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Frontotemporal Dementia/genetics , Mutation/genetics , Osteitis Deformans/genetics , Aminopeptidases/metabolism , Animals , Animals, Genetically Modified , Binding Sites/genetics , Cell Line, Transformed , Central Nervous System/metabolism , Cytoplasm/genetics , Cytoplasm/metabolism , DNA-Binding Proteins/genetics , Disease Models, Animal , Drosophila , Drosophila Proteins/metabolism , Frontotemporal Dementia/complications , Frontotemporal Dementia/pathology , Gene Expression Regulation/genetics , Glycoproteins/metabolism , Humans , Indoles , Models, Biological , Osteitis Deformans/complications , Promoter Regions, Genetic/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae/genetics , Transfection/methods , Valosin Containing Protein
5.
J Biol Chem ; 284(18): 12384-98, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19237541

ABSTRACT

Frontotemporal lobar degeneration (FTLD) with inclusion body myopathy and Paget disease of bone is a rare, autosomal dominant disorder caused by mutations in the VCP (valosin-containing protein) gene. The disease is characterized neuropathologically by frontal and temporal lobar atrophy, neuron loss and gliosis, and ubiquitin-positive inclusions (FTLD-U), which are distinct from those seen in other sporadic and familial FTLD-U entities. The major component of the ubiquitinated inclusions of FTLD with VCP mutation is TDP-43 (TAR DNA-binding protein of 43 kDa). TDP-43 proteinopathy links sporadic amyotrophic lateral sclerosis, sporadic FTLD-U, and most familial forms of FTLD-U. Understanding the relationship between individual gene defects and pathologic TDP-43 will facilitate the characterization of the mechanisms leading to neurodegeneration. Using cell culture models, we have investigated the role of mutant VCP in intracellular trafficking, proteasomal function, and cell death and demonstrate that mutations in the VCP gene 1) alter localization of TDP-43 between the nucleus and cytosol, 2) decrease proteasome activity, 3) induce endoplasmic reticulum stress, 4) increase markers of apoptosis, and 5) impair cell viability. These results suggest that VCP mutation-induced neurodegeneration is mediated by several mechanisms.


Subject(s)
Adenosine Triphosphatases/metabolism , Apoptosis , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Dementia/metabolism , Models, Biological , Mutation , Adenosine Triphosphatases/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cell Survival/genetics , Cytoplasm/genetics , Cytoplasm/metabolism , Cytoplasm/pathology , DNA-Binding Proteins/genetics , Dementia/genetics , Dementia/pathology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Female , Humans , Male , Middle Aged , Protein Transport/genetics , Ubiquitination/genetics , Valosin Containing Protein
6.
Acta Neuropathol ; 117(2): 137-49, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19125255

ABSTRACT

Accumulation of hyperphosphorylated, ubiquitinated and N-terminally truncated TAR DNA-binding protein (TDP-43) is the pathological hallmark lesion in most familial and sporadic forms of FTLD-U and ALS, which can be subsumed as TDP-43 proteinopathies. In order to get more insight into the role of abnormal phosphorylation in the disease process, the identification of specific phosphorylation sites and the generation of phosphorylation-specific antibodies are mandatory. Here, we developed and characterized novel rat monoclonal antibodies (1D3 and 7A9) raised against phosphorylated S409/410 of TDP-43. These antibodies were used to study the presence of S409/410 phosphorylation by immunohistochemistry and biochemical analysis in a large series of 64 FTLD-U cases with or without motor neuron disease including familial cases with mutations in progranulin (n = 5), valosin-containing protein (n = 4) and linkage to chromosome 9p (n = 4), 18 ALS cases as well as other neurodegenerative diseases with concomitant TDP-43 pathology (n = 5). Our data demonstrate that phosphorylation of S409/410 of TDP-43 is a highly consistent feature in pathologic inclusions in the whole spectrum of sporadic and familial forms of TDP-43 proteinopathies. Physiological nuclear TDP-43 was not detectable with these mAbs by immunohistochemistry and by immunoblot analyses. While the accumulation of phosphorylated C-terminal fragments was a robust finding in the cortical brain regions of FTLD-U and ALS, usually being much more abundant than the phosphorylated full-length TDP-43 band, spinal cord samples revealed a predominance of full-length TDP-43 over C-terminal fragments. This argues for a distinct TDP-43 species composition in inclusions in cortical versus spinal cord cells. Overall, these mAbs are powerful tools for the highly specific detection of disease-associated abnormal TDP-43 species and will be extremely useful for the neuropathological routine diagnostics of TDP-43 proteinopathies and for the investigation of emerging cellular and animal models for TDP-43 proteinopathies.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Brain/metabolism , DNA-Binding Proteins/metabolism , Dementia/metabolism , Adult , Aged , Aged, 80 and over , Amyotrophic Lateral Sclerosis/complications , Amyotrophic Lateral Sclerosis/pathology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Brain/pathology , DNA-Binding Proteins/immunology , Dementia/complications , Dementia/pathology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoblotting , Immunohistochemistry , Male , Middle Aged , Motor Neuron Disease/complications , Motor Neuron Disease/metabolism , Neurons/metabolism , Neurons/pathology , Phosphorylation , Rats , Spinal Cord/metabolism
7.
J Neuropathol Exp Neurol ; 67(6): 555-64, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18520774

ABSTRACT

Pathologic TAR-DNA-binding protein 43 (TDP-43) is a disease protein in frontotemporal lobar degeneration with ubiquitin-positive inclusions (FTLD-U) and amyotrophic lateral sclerosis. We studied the presence, frequency, and distribution of TDP-43 pathology by immunohistochemistry and biochemistry in a series of clinically well-characterized tauopathy patient brains, including 182 Alzheimer disease (AD), 39 corticobasal degeneration, 77 progressive supranuclear palsy, and 12 Pick disease cases and investigated the clinical impact of concomitant TDP-43 pathology in these cases. TAR-DNA-binding protein 43 pathology was found in 25.8% of AD cases. It was restricted to the dentate gyrus and entorhinal cortex in approximately 75% of cases; approximately 25% showed more widespread TDP-43 pathology in frontal and temporal cortices, resembling the FTLD-U subtype associated with progranulin mutations. TAR-DNA-binding protein 43 pathology in AD was associated with significantly longer disease duration, but there was no association with the clinical presentation (148 cases diagnosed as AD and 34 cases diagnosed as frontotemporal lobar degeneration). Progressive supranuclear palsy and Pick disease cases showed no TDP-43 inclusions and no biochemical alterations of TDP-43. There was, however, a unique, predominantly glial TDP-43 pathology with staining of astrocytic plaque-like structures and coiled bodies in 15.4% of corticobasal degeneration cases; this was associated with biochemical TDP-43 changes similar to those in FTLD-U. These findings provide further insight into the burden and clinical significance of TDP-43 pathology in disorders other than FTLD-U and amyotrophic lateral sclerosis.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , DNA-Binding Proteins/metabolism , Neurons/pathology , Tauopathies/pathology , Aged , Alzheimer Disease/metabolism , Brain/metabolism , Female , Humans , Immunoblotting , Immunohistochemistry , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Male , Neurons/metabolism , Tauopathies/metabolism
8.
Am J Med Genet A ; 146A(6): 745-57, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18260132

ABSTRACT

Inclusion body myopathy with Paget disease of the bone (PDB) and/or frontotemporal dementia (IBMPFD, OMIM 167320), is a progressive autosomal dominant disorder caused by mutations in the Valousin-containing protein (VCP, p97 or CDC48) gene. IBMPFD can be difficult to diagnose. We assembled data on a large set of families to illustrate the number and type of misdiagnoses that occurred. Clinical analysis of 49 affected individuals in nine families indicated that 42 (87%) of individuals had muscle disease. The majority were erroneously diagnosed with limb girdle muscular dystrophy (LGMD), facioscapular muscular dystrophy, peroneal muscular dystrophy, late adult onset distal myopathy, spinal muscular atrophy, scapuloperoneal muscular dystrophy, or amyotrophic lateral sclerosis (ALS) among others. Muscle biopsies showed rimmed vacuoles characteristic of an inclusion body myopathy in 7 of 18 patients (39%), however, inclusion body myopathy was correctly diagnosed among individuals in only families 5 and 15. Frontotemporal dementia (FTD) was diagnosed in 13 individuals (27%) at a mean age of 57 years (range 48.9-60.2 years); however, several individuals had been diagnosed with Alzheimer disease. Histopathological examination of brains of three affected individuals revealed a pattern of ubiquitin positive neuronal intranuclear inclusions and dystrophic neurites. These families expand the clinical phenotype in IBMPFD, a complex disorder caused by mutations in VCP. The presence of PDB in 28 (57%) individuals suggests that measuring serum alkaline phosphatase (ALP) activity may be a useful screen for IBMPFD in patients with myopathy.


Subject(s)
Adenosine Triphosphatases/genetics , Cell Cycle Proteins/genetics , Dementia/complications , Dementia/genetics , Family , Muscular Diseases/complications , Muscular Diseases/genetics , Osteitis Deformans/complications , Osteitis Deformans/genetics , Adult , Cross-Sectional Studies , Female , Humans , Inclusion Bodies/ultrastructure , Male , Middle Aged , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , Pedigree , Retrospective Studies , Valosin Containing Protein
9.
Arch Neurol ; 64(11): 1601-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17998442

ABSTRACT

BACKGROUND: Clinical-pathologic studies are crucial to understanding brain-behavior relations and improving diagnostic accuracy in neurodegenerative diseases. OBJECTIVE: To establish clinical, neuropsychological, and imaging features of clinically diagnosed patients with frontotemporal dementia (FTD) that help discriminate between pathologically determined tau-positive FTD, tau-negative FTD, and frontal-variant Alzheimer disease. DESIGN: Retrospective clinical-pathologic survey. SETTING: Academic medical center. Patients Sixty-one participants with the clinical diagnosis of a frontotemporal spectrum disorder who underwent a neuropsychological evaluation and had an autopsy-confirmed disease. MAIN OUTCOME MEASURES: Neuropsychological performance and high-resolution structural magnetic resonance imaging (MRI). RESULTS: Distinguishing features of patients with tau-positive FTD include visual perceptual-spatial difficulty and an extrapyramidal disorder significantly more often than other patients, significant cortical atrophy in the frontal and parietal regions as evidenced on MRI, and the burden of pathology is greatest in the frontal and parietal regions. Patients with tau-negative FTD are distinguished by their greater difficulties with social, language, and verbally mediated executive functions, significant cortical atrophy in the frontal and temporal regions as evidenced on MRI, and significant frontal and temporal pathology. Patients with Alzheimer disease at autopsy have significantly impaired delayed recall during episodic memory testing; atrophy that involves temporal areas, including the hippocampus, as evidenced on MRI; and widely distributed pathology including the medial temporal structures. A discriminant function analysis grouped patients on the basis of clinical and neuropsychological features with 87.5% accuracy. CONCLUSION: Clinical, neuropsychological, and imaging profiles can contribute to accurate antemortem diagnosis in FTD.


Subject(s)
Dementia/pathology , Inpatients , tau Proteins/metabolism , Aged , Aged, 80 and over , Analysis of Variance , Autopsy/methods , Brain Mapping , Dementia/physiopathology , Female , Health Surveys , Humans , Magnetic Resonance Imaging/methods , Male , Middle Aged , Neuropsychological Tests , Retrospective Studies
10.
Curr Opin Neurobiol ; 17(5): 548-55, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17936612

ABSTRACT

Over the past decade, it has become clear that there is a significant overlap in the clinical spectrum of frontotemporal lobar degeneration and amyotrophic lateral sclerosis (ALS). The identification of TDP-43 as the major disease protein in the pathology of both frontotemporal lobar degeneration with ubiquitin inclusions and ALS provides the first molecular link for these diseases. Pathological TDP-43 is abnormally phosphorylated, ubiquitinated, and cleaved to generate carboxy-terminal fragments in affected brain regions. The normal nuclear expression of TDP-43 is also reduced leading to the hypothesis that sequestration of TDP-43 in pathological inclusions contributes to disease pathogenesis. Thus, TDP-43 is the newest member of the growing list of neurodegenerative proteinopathies, but unique in that it lacks features of brain amyloidosis.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , DNA-Binding Proteins/metabolism , Dementia/metabolism , Amyotrophic Lateral Sclerosis/pathology , Dementia/pathology , Humans , Ubiquitin/metabolism
11.
Arch Neurol ; 64(10): 1449-54, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17923628

ABSTRACT

BACKGROUND: TDP-43 is a major ubiquitinated disease protein in the pathologic condition of frontotemporal lobar degeneration with ubiquitin-positive inclusions (FTLD-U). OBJECTIVE: To investigate the demographic, clinical, and neuropsychological features associated with subtypes of FTLD-U with TDP-43 inclusions (FTLD-U/TDP-43). DESIGN: Retrospective clinical-pathologic study. SETTING: Academic medical center. Patients Twenty-three patients with histopathologically proven FTLD-U. MAIN OUTCOME MEASURES: Demographic, symptom, neuropsychological, and autopsy characteristics. RESULTS: There are notably different clinical and neuropsychological patterns of impairment in FTLD-U subtypes. Patients with FTLD-U/TDP-43 characterized by numerous neuronal intracytoplasmic inclusions have shorter survival; patients with FTLD-U/TDP-43 featuring numerous neurites have difficulty with object naming; and patients with FTLD-U/TDP-43 in whom neuronal intranuclear inclusions are present have substantial executive deficits. There are also different anatomical distributions of ubiquitin pathologic features in FTLD-U subgroups, consistent with their cognitive deficits. CONCLUSION: Distinct TDP-43 profiles may affect clinical phenotypes differentially in patients with FTLD-U.


Subject(s)
DNA-Binding Proteins/genetics , Dementia/genetics , Dementia/pathology , Inclusion Bodies/genetics , Inclusion Bodies/pathology , Ubiquitin/metabolism , Aged , Autopsy , Brain/pathology , Cognition/physiology , Data Interpretation, Statistical , Dementia/psychology , Female , Humans , Immunohistochemistry , Male , Middle Aged , Neuropsychological Tests , Phenotype , Psychomotor Performance/physiology , Retrospective Studies
12.
Arch Neurol ; 64(8): 1148-53, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17698705

ABSTRACT

BACKGROUND: Patients with frontotemporal dementia due to mutation of progranulin may have a distinct phenotype. OBJECTIVE: To identify distinct clinical and pathologic features of patients with frontotemporal dementia who have mutations of progranulin (GRN). DESIGN: Retrospective clinical-pathologic study. SETTING: Academic medical center. PATIENTS: Twenty-eight patients with frontotemporal dementia, including 9 with GRN mutations (4 autopsy cases and 5 with only clinical information) and 19 with the identical pathologic diagnosis--frontotemporal lobar degeneration with ubiquitin-positive and tau-negative inclusions (FTLD-U)--and no GRN mutations. MAIN OUTCOME MEASURES: Demographic, symptom, neuropsychological, and autopsy characteristics. RESULTS: Patients with and without a GRN mutation have similar demographic features, although family history is significantly more common in patients with frontotemporal dementia and a GRN mutation. Both patient groups have frequent social and personality complaints. Neuropsychological evaluation reveals a significant recognition memory deficit in patients with a GRN mutation but a significant language deficit only in patients without a GRN mutation. At autopsy, the semiquantitative burden of ubiquitin abnormality is relatively modest in both groups of patients. CONCLUSION: Patients with a GRN mutation differ clinically from those with the same pathologic diagnosis but no GRN mutation.


Subject(s)
Dementia/genetics , Dementia/psychology , Frontal Lobe , Intercellular Signaling Peptides and Proteins/genetics , Mutation , Temporal Lobe , Aged , Brain/metabolism , Brain/pathology , Dementia/metabolism , Dementia/pathology , Female , Humans , Interpersonal Relations , Language Disorders/etiology , Male , Memory Disorders/etiology , Memory Disorders/psychology , Middle Aged , Neuropsychological Tests , Personality Disorders/etiology , Progranulins , Recognition, Psychology , Retrospective Studies , Ubiquitin/metabolism
13.
Am J Pathol ; 171(1): 227-40, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17591968

ABSTRACT

TAR DNA-binding protein 43 (TDP-43) is a major pathological protein of sporadic and familial frontotemporal lobar degeneration with ubiquitin-positive, tau-negative inclusions (FTLD-U) with or without motor neuron disease (MND). Thus, TDP-43 defines a novel class of neurodegenerative diseases called TDP-43 proteinopathies. We performed ubiquitin and TDP-43 immunohistochemistry on 193 cases of familial and sporadic FTLD with or without MND. On selected cases, immunoelectron microscopy and biochemistry were performed. Clinically defined frontotemporal dementias (FTDs) included four groups: 1) familial FTD with mutations in progranulin (n = 36), valosin-containing protein (n = 5), charged multivesicular body protein 2B (n = 4), and linked to chromosome 9p (n = 7); 2) familial cases of FTD with unknown gene association (n = 29); 3) sporadic FTD (n = 72); and 4) familial and sporadic FTD with MND (n = 40). Our studies confirm that the spectrum of TDP-43 proteinopathies includes most cases of sporadic and familial FTLD-U with and without MND and expand this disease spectrum to include reported families with FTD linked to chromosome 9p but not FTD with charged multivesicular body protein 2B mutations. Thus, despite significant clinical, genetic, and neuropathological heterogeneity of FTLD-U, TDP-43 is a common pathological substrate underlying a large subset of these disorders, thereby implicating TDP-43 in novel and unifying mechanisms of FTLD pathogenesis.


Subject(s)
DNA-Binding Proteins/physiology , Dementia/genetics , Ubiquitin/metabolism , Chromosomes, Human, Pair 3 , Chromosomes, Human, Pair 9 , DNA-Binding Proteins/genetics , Dementia/metabolism , Female , Humans , Male , Motor Neuron Disease/genetics
14.
Ann Neurol ; 61(5): 427-34, 2007 May.
Article in English | MEDLINE | ID: mdl-17469116

ABSTRACT

OBJECTIVE: Amyotrophic lateral sclerosis (ALS) is a common, fatal motor neuron disorder with no effective treatment. Approximately 10% of cases are familial ALS (FALS), and the most common genetic abnormality is superoxide dismutase-1 (SOD1) mutations. Most ALS research in the past decade has focused on the neurotoxicity of mutant SOD1, and this knowledge has directed therapeutic strategies. We recently identified TDP-43 as the major pathological protein in sporadic ALS. In this study, we investigated TDP-43 in a larger series of ALS cases (n = 111), including familial cases with and without SOD1 mutations. METHODS: Ubiquitin and TDP-43 immunohistochemistry was performed on postmortem tissue from sporadic ALS (n = 59), ALS with SOD1 mutations (n = 15), SOD-1-negative FALS (n = 11), and ALS with dementia (n = 26). Biochemical analysis was performed on representative cases from each group. RESULTS: All cases of sporadic ALS, ALS with dementia, and SOD1-negative FALS had neuronal and glial inclusions that were immunoreactive for both ubiquitin and TDP-43. Cases with SOD1 mutations had ubiquitin-positive neuronal inclusions; however, no cases were immunoreactive for TDP-43. Biochemical analysis of postmortem tissue from sporadic ALS and SOD1-negative FALS demonstrated pathological forms of TDP-43 that were absent in cases with SOD1 mutations. INTERPRETATION: These findings implicate pathological TDP-43 in the pathogenesis of sporadic ALS. In contrast, the absence of pathological TDP-43 in cases with SOD1 mutations implies that motor neuron degeneration in these cases may result from a different mechanism, and that cases with SOD1 mutations may not be the familial counterpart of sporadic ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , DNA-Binding Proteins/genetics , Superoxide Dismutase/genetics , Humans , Immunohistochemistry , Medulla Oblongata/pathology , Motor Cortex/pathology , Mutation/genetics , Spinal Cord/pathology , Superoxide Dismutase-1 , Ubiquitin/genetics
15.
Acta Neuropathol ; 114(1): 55-61, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17457594

ABSTRACT

Frontotemporal dementia with inclusion body myopathy and Paget's disease of bone (IBMPFD) is a rare, autosomal dominant disorder caused by mutations in the gene valosin-containing protein (VCP). The CNS pathology is characterized by a novel pattern of ubiquitin pathology distinct from sporadic and familial frontotemporal lobar degeneration with ubiquitin-positive inclusions without VCP mutations. Yet, the ubiquitin-positive inclusions in IBMPFD also stain for TAR DNA binding protein, a feature that links this rare disease with the pathology associated with the majority of sporadic FTD as well as disease resulting from different genetic alterations. VCP, a member of the AAA-ATPase gene family, associates with a plethora of protein adaptors to perform a variety of cellular processes including Golgi assembly/disassembly and regulation of the ubiquitin-proteasome system. However, the mechanism whereby mutations in VCP lead to CNS, muscle, and bone disease is largely unknown. In this report, we review current literature on IBMPFD, focusing on the pathology of the disease and the biology of VCP with respect to IBMPFD.


Subject(s)
Adenosine Triphosphatases/genetics , Cell Cycle Proteins/genetics , Dementia/genetics , Inclusion Bodies/pathology , Dementia/complications , Dementia/pathology , Humans , Inclusion Bodies/metabolism , Mutation/genetics , Myositis, Inclusion Body/complications , Myositis, Inclusion Body/genetics , Myositis, Inclusion Body/pathology , Osteitis Deformans/complications , Osteitis Deformans/genetics , Osteitis Deformans/pathology , Valosin Containing Protein
16.
J Neuropathol Exp Neurol ; 66(2): 152-7, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17279000

ABSTRACT

Frontotemporal dementia with inclusion body myopathy and Paget disease of bone is a rare, autosomal-dominant disorder caused by mutations in the gene valosin-containing protein (VCP). The CNS pathology is characterized by a novel pattern of ubiquitin pathology distinct from sporadic and familial frontotemporal lobar degeneration with ubiquitin-positive inclusions (FTLD-U) without VCP mutations. TAR DNA binding protein 43 (TDP-43) was recently identified as a major disease protein in the ubiquitin-positive inclusions of sporadic and familial FTLD-U. To determine whether the ubiquitin pathology associated with mutations in VCP is characterized by the accumulation of TDP-43, we analyzed TDP-43 in the CNS pathology of five patients with VCP gene mutations. Accumulations of TDP-43 colocalized with ubiquitin pathology in inclusion body myopathy and Paget disease of bone, including both intranuclear inclusions and dystrophic neurites. Similar to FTLD-U, phosphorylated TDP-43 was detected only in insoluble brain extracts from affected brain regions. Identification of TDP-43, but not VCP, within ubiquitin-positive inclusions supports the hypothesis that VCP gene mutations lead to a dominant negative loss or alteration of VCP function culminating in impaired degradation of TDP-43. TDP-43 is a common pathologic substrate linking a variety of distinct patterns of FTLD-U pathology caused by different genetic alterations.


Subject(s)
Adenosine Triphosphatases/genetics , Cell Cycle Proteins/genetics , Cerebral Cortex/metabolism , DNA-Binding Proteins/metabolism , Dementia/genetics , Dementia/metabolism , Ubiquitin/metabolism , Aged , Cerebral Cortex/pathology , Dementia/pathology , Female , Humans , Inclusion Bodies/genetics , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Male , Middle Aged , Mutation/genetics , Myositis, Inclusion Body/genetics , Myositis, Inclusion Body/metabolism , Myositis, Inclusion Body/pathology , Neurons/metabolism , Neurons/pathology , Osteitis Deformans/genetics , Osteitis Deformans/metabolism , Osteitis Deformans/pathology , Valosin Containing Protein
17.
Acta Neuropathol ; 113(4): 471-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17072625

ABSTRACT

Frontotemporal dementia (FTD) is a clinically heterogeneous disorder characterized by alterations in language and/or behavior, often in association with Parkinsonism or motor neuron disease. A familial form of FTD is associated with mutations in the microtubule-associated protein tau (MAPT) gene. We report here on the clinical, neuroimaging, cerebral spinal fluid biomarker, genetic, biochemical and postmortem neuropathological analyses of a case of familial FTD with a Leu266Val MAPT mutation which results in a very early age of onset and a rapid course of disease. This is also the first reported case of any MAPT mutation in an individual of African American ethnicity.


Subject(s)
Dementia/genetics , Leucine/genetics , Mutation , Valine/genetics , tau Proteins/genetics , Adult , Black or African American/ethnology , DNA-Binding Proteins/metabolism , Dementia/metabolism , Dementia/pathology , Dementia/physiopathology , Family Health , Female , Humans , Magnetic Resonance Imaging , Neuropsychological Tests , Polycomb-Group Proteins , Transcription Factors/metabolism
18.
Neurobiol Dis ; 25(3): 561-70, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17174556

ABSTRACT

Previously we have shown that the H1c haplotype on the background of the H1 clade of haplotypes at the MAPT locus is associated with increased risk for progressive supranuclear palsy (PSP), corticobasal degeneration (CBD) and Alzheimer's disease (AD). Here we replicated the association with AD in an additional autopsy confirmed series. We show that this haplotype increases both the expression of total MAPT transcript as well as specifically increasing the proportion of 4 microtubule binding repeat containing transcripts. We discuss these findings both in terms of the problems facing the dissection of the etiologies of complex traits and the pathogenesis of the tauopathies.


Subject(s)
Alzheimer Disease/genetics , Tauopathies/genetics , tau Proteins/genetics , Age of Onset , Alleles , Alzheimer Disease/epidemiology , Cell Line, Tumor , Gene Expression Regulation , Genetic Predisposition to Disease/epidemiology , Haplotypes , Heterozygote , Homozygote , Humans , Neuroblastoma , RNA, Messenger/genetics , Repetitive Sequences, Nucleic Acid , Reverse Transcriptase Polymerase Chain Reaction , Risk Factors , Tauopathies/epidemiology
19.
J Neuropathol Exp Neurol ; 65(6): 571-81, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16783167

ABSTRACT

Frontotemporal dementia (FTD) with inclusion body myopathy and Paget disease of bone (IBMPFD) is a rare, autosomal-dominant disorder caused by mutations in the valosin-containing protein (VCP) gene, a member of the AAA-ATPase gene superfamily. The neuropathology associated with sporadic FTD is heterogeneous and includes tauopathies and frontotemporal lobar degeneration with ubiquitin-positive inclusions (FTLD-U). However, there is limited information on the neuropathology in IBMPFD. We performed a detailed, systematic analysis of the neuropathologic changes in 8 patients with VCP mutations. A novel pattern of ubiquitin pathology was identified in IBMPFD that was distinct from sporadic and familial FTLD-U without VCP gene mutations. This was characterized by ubiquitin-positive neuronal intranuclear inclusions and dystrophic neurites. In contrast to FTLD-U, only rare intracytoplasmic inclusions were identified. The ubiquitin pathology was abundant in the neocortex, less robust in limbic and subcortical nuclei, and absent in the dentate gyrus. Only rare inclusions were detected with antibodies to VCP and there was no biochemical alteration in the VCP protein. VCP is associated with a variety of cellular activities, including regulation of the ubiquitin-proteasome system. Our findings are consistent with the hypothesis that the pathology associated with VCP gene mutations is the result of impairment of ubiquitin-based degradation pathways.


Subject(s)
Cell Cycle Proteins/genetics , Dementia/genetics , Dementia/metabolism , Mutation/genetics , Ubiquitin/metabolism , Adenosine Triphosphatases , Blotting, Western/methods , Dementia/pathology , Female , Humans , Immunohistochemistry/methods , Male , Myositis, Inclusion Body/genetics , Myositis, Inclusion Body/pathology , Osteitis Deformans/genetics , Osteitis Deformans/pathology , Valosin Containing Protein
20.
Ann Neurol ; 59(6): 952-62, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16718704

ABSTRACT

OBJECTIVE: Frontotemporal lobar degeneration (FTLD) is characterized by impairments in social, behavioral, and/or language function, but postmortem studies indicate that multiple neuropathological entities lead to FTLD. This study assessed whether specific clinical features predict the underlying pathology. METHODS: A clinicopathological correlation was performed on 90 consecutive patients with a pathological diagnosis of frontotemporal dementia and was compared with an additional 24 cases accrued during the same time period with a clinical diagnosis of FTLD, but with pathology not typically associated with frontotemporal dementia. RESULTS: Postmortem examination showed multiple pathologies including tauopathies (46%), FTLD with ubiquitin-positive inclusions (29%), and Alzheimer's disease (17%). The pathological groups manifested some distinct demographic, clinical, and neuropsychological features, although these attributes showed only a statistical association with the underlying pathology. FTLD with ubiquitin-positive inclusions was more likely to present with both social and language dysfunction, and motor neuron disease was more likely to emerge in these patients. Tauopathies were more commonly associated with an extrapyramidal disorder. Alzheimer's disease was associated with relatively greater deficits in memory and executive function. INTERPRETATION: Clinical and neuropsychological features contribute to delineating the spectrum of pathology underlying a patient diagnosed with FTLD, but biomarkers are needed that, together with the clinical phenotype, can predict the underlying neuropathology.


Subject(s)
Brain/pathology , Dementia/pathology , Dementia/physiopathology , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology , Adult , Age of Onset , Aged , Aged, 80 and over , Alzheimer Disease/complications , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Benzothiazoles , Brain/metabolism , Brain/physiopathology , Dementia/complications , Diagnosis, Differential , Female , Humans , Immunohistochemistry , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Male , Middle Aged , Neurodegenerative Diseases/complications , Neuropsychological Tests , Tauopathies/complications , Tauopathies/pathology , Tauopathies/physiopathology , Thiazoles/metabolism , Ubiquitin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...