Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 150
Filter
1.
Neuropathol Appl Neurobiol ; 45(5): 430-440, 2019 08.
Article in English | MEDLINE | ID: mdl-30635947

ABSTRACT

A quarter of a century ago, we proposed an innovative approach to study the pathogenesis of prion disease, one of the most intriguing biomedical problems that remains unresolved. The synthesis of a peptide homologous to residues 106-126 of the human prion protein (PrP106-126), a sequence present in the PrP amyloid protein of Gerstmann-Sträussler-Scheinker syndrome patients, provided a tractable tool for investigating the mechanisms of neurotoxicity. Together with several other discoveries at the beginning of the 1990s, PrP106-126 contributed to underpin the role of amyloid in the pathogenesis of protein-misfolding neurodegenerative disorders. Later, the role of oligomers on one hand and of prion-like spreading of pathology on the other further clarified mechanisms shared by different neurodegenerative conditions. Our original report on PrP106-126 neurotoxicity also highlighted a role for programmed cell death in CNS diseases. In this review, we analyse the prion research context in which PrP106-126 first appeared and the advances in our understanding of prion disease pathogenesis and therapeutic perspectives 25 years later.


Subject(s)
Peptide Fragments , Prion Diseases , Prions , Animals , Humans
2.
Neurology ; 74(10): 798-806, 2010 Mar 09.
Article in English | MEDLINE | ID: mdl-20164095

ABSTRACT

OBJECTIVE: Large kindreds segregating familial Alzheimer disease (FAD) offer the opportunity of studying clinical variability as observed for presenilin 1 (PSEN1) mutations. Two early-onset FAD (EOFAD) Calabrian families with PSEN1 Met146Leu (ATG/CTG) mutation constitute a unique population descending from a remote common ancestor. Recently, several other EOFAD families with the same mutation have been described worldwide. METHODS: We searched for a common founder of the PSEN1 Met146Leu mutation in families with different geographic origins by genealogic and molecular analyses. We also investigated the phenotypic variability at onset in a group of 50 patients (mean age at onset 40.0 +/- 4.8 years) by clinical, neuropsychological, and molecular methodologies. RESULTS: EOFAD Met146Leu families from around the world resulted to be related and constitute a single kindred originating from Southern Italy before the 17th century. Phenotypic variability at onset is broad: 4 different clinical presentations may be recognized, 2 classic for AD (memory deficits and spatial and temporal disorientation), whereas the others are expressions of frontal impairment. The apathetic and dysexecutive subgroups could be related to orbital-medial prefrontal cortex and dorsolateral prefrontal cortex dysfunction. CONCLUSIONS: Genealogic and molecular findings provided evidence that the PSEN1 Met146Leu families from around the world analyzed in this study are related and represent a single kindred originating from Southern Italy. The marked phenotypic variability might reflect early involvement by the pathologic process of different cortical areas. Although the clinical phenotype is quite variable, the neuropathologic and biochemical characteristics of the lesions account for neurodegenerative processes unmistakably of Alzheimer nature.


Subject(s)
Alzheimer Disease/genetics , Leucine/genetics , Methionine/genetics , Mutation/genetics , Presenilin-1/genetics , Adult , Alzheimer Disease/complications , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/history , Brain/diagnostic imaging , Brain/pathology , Cognition Disorders/etiology , Cognition Disorders/genetics , Family Health , Female , Fluorodeoxyglucose F18 , Gene Frequency , Genetic Predisposition to Disease , Genetic Testing , Genotype , Global Health , History, 17th Century , History, 21st Century , Humans , International Cooperation , Italy , Male , Memory Disorders/etiology , Memory Disorders/genetics , Middle Aged , Phenotype , Positron-Emission Tomography
3.
Neurobiol Aging ; 29(12): 1864-73, 2008 Dec.
Article in English | MEDLINE | ID: mdl-17560687

ABSTRACT

Cerebral accumulation of hyperphosphorylated tau (phospho-tau) occurs in several neurodegenerative conditions including Alzheimer disease. In prion diseases, phospho-tau deposition has been described in a rare genetic form, Gerstmann-Sträussler-Scheinker disease, but is not considered part of the neuropathological picture of Creutzfeldt-Jakob disease. Aim of this study was to investigate whether changes related to phospho-tau accumulation are present in the brain of patients with variant Creutzfeldt-Jakob disease (vCJD) that shares with Gerstmann-Sträussler-Scheinker disease abundant prion protein (PrP) deposition in amyloid form. The analysis was extended to experimental mouse models of vCJD. We detected a large number of phospho-tau-immunoreactive neuritic profiles, often clustered around PrP amyloid deposits, not only in the cerebral cortex, but also in the cerebellum of all vCJD patients examined, in the absence of Abeta. Although less constantly, phospho-tau was localized in some perikaria and dendrites. The biochemical counterpart was the presence of phospho-tau in the detergent-insoluble fraction of cerebral cortex. Phospho-tau-immunoreactive neuronal profiles were also found in association with PrP deposits in mouse models of vCJD. These findings suggest that the abnormal forms of PrP associated with vCJD trigger a tauopathy, and provide a paradigm for the early stages of tau pathology associated with cerebral amyloidoses, including Alzheimer disease.


Subject(s)
Cerebellum/metabolism , Cerebral Cortex/metabolism , Creutzfeldt-Jakob Syndrome/metabolism , Disease Models, Animal , tau Proteins/metabolism , Adult , Animals , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Tissue Distribution
4.
Neurol Sci ; 28(5): 241-4, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17972037

ABSTRACT

BACKGROUND: Frontotemporal degeneration (FTD) is the most common cause of dementia after Alzheimer's disease. To date, it has been addressed with intensive and intense research. OBJECTIVE: To report on the most recent findings in the biology of FTD. METHODS: Review of FTD literature. RESULTS: FTD presents with many phenotypes that span from prefrontal syndromes to lower motor neuron disease passing through temporal, parietal and extrapyramidal syndromes. FTD includes the frontotemporal lobar atrophies clinically characterised by abnormal behaviour, progressive aphasia or semantic dementia, as well as corticobasal degeneration, progressive supranuclear palsy, progressive subcortical gliosis and FTD with motor neuron disease. The molecular classification of FTD can be traced following the immunocytochemical properties of the material accumulated in neuroectodermic cells. This procedure allows the separation of FTD with tau-positive inclusions from FTD with ubiquitin-positive inclusions, and from FTD with inclusions negative for both. Genetically, seven loci (chromosomes 3p, 9q and 17q24, one locus each; 9p and 17q21, two loci each) and four genes (MAPT, PRGN, VCP, CHMP2B) have been identified. Proteins involved are tau, progranulin, VCP, CHMP2B, Progranulin TDP43, ubiquitin and the intermediate neurofilament system. Neurodegeneration is most likely due to changes in cytoskeletal structure and in ubiquitin-dependent protein degradation activity.


Subject(s)
Dementia , Chromosomes, Human, Pair 17 , Chromosomes, Human, Pair 9 , Dementia/genetics , Dementia/metabolism , Dementia/physiopathology , Humans , Ubiquitin/metabolism , tau Proteins/metabolism
5.
J Neurol Neurosurg Psychiatry ; 78(12): 1379-82, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18024694

ABSTRACT

An atypical case of sporadic Creutzfeldt-Jakob disease (CJD) is described in a 78-year-old woman homozygous for methionine at codon 129 of the prion protein (PrP) gene. The neuropathological signature was the presence of PrP immunoreactive plaque-like deposits in the cerebral cortex, striatum and thalamus. Western blot analysis showed a profile of the pathological form of PrP (PrP(Sc)) previously unrecognised in sporadic CJD, marked by the absence of diglycosylated protease resistant species. These features define a novel neuropathological and molecular CJD phenotype.


Subject(s)
Creutzfeldt-Jakob Syndrome/genetics , PrPSc Proteins/genetics , Aged , Antibodies/immunology , Antibodies, Monoclonal/immunology , Antiparkinson Agents/therapeutic use , Blotting, Western , Brain/immunology , Brain/pathology , Codon/genetics , Creutzfeldt-Jakob Syndrome/diagnosis , Creutzfeldt-Jakob Syndrome/immunology , Female , Humans , Immunohistochemistry , Levodopa/therapeutic use , Magnetic Resonance Imaging , Methionine/genetics , Parkinsonian Disorders/drug therapy , Phenotype , Polymorphism, Genetic/genetics , PrPSc Proteins/immunology , Valine/genetics
6.
Acta Neurochir Suppl ; 97(Pt 2): 399-406, 2007.
Article in English | MEDLINE | ID: mdl-17691328

ABSTRACT

Chronic high frequency stimulation (HFS) of the posteromedial hypothalamus (PMH) has been the first direct therapeutic application of functional neuroimaging data in a restorative reversible procedure for the treatment of an otherwise refractory neurological condition; in fact, the target coordinates for the stereotactic implantation of the electrodes have been provided by positron emission tomography (PET) studies, which were performed during cluster headache attacks. HFS of PMH produced a significant and marked reduction of pain attacks in patients with chronic cluster headache (CCH) and in one patient with short-lasting unilateral neuralgiform headache attacks with conjunctival injection and tearing (SUNCT). The episodes of violent behaviour and psychomotor agitation during the attacks of CCH supported the idea that the posteromedial hypothalamus could be also involved in the control of aggressiveness; this has been previously suggested, in the seventies, by the results obtained in Sano's hypothalamotomies for the treatment of abnormal aggression and disruptive behaviour. On the basis of these considerations, we have performed HFS of the PMH and controlled successfully violent and disruptive behaviour in patients refractory to the conventional sedative drugs. Finally, we also tested the same procedure in three patients with refractory atypical facial pain, but unfortunately, they did not respond to this treatment.


Subject(s)
Behavioral Symptoms/surgery , Deep Brain Stimulation/methods , Facial Neuralgia/therapy , Hypothalamus, Posterior/surgery , Adult , Aged , Behavioral Symptoms/pathology , Behavioral Symptoms/physiopathology , Dose-Response Relationship, Radiation , Facial Neuralgia/pathology , Facial Neuralgia/physiopathology , Female , Functional Laterality , Humans , Hypothalamus, Posterior/physiopathology , Male , Middle Aged , SUNCT Syndrome/pathology , SUNCT Syndrome/physiopathology , SUNCT Syndrome/surgery , Time Factors , Treatment Outcome
7.
J Neural Transm (Vienna) ; 114(7): 947-50, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17318302

ABSTRACT

In 9 patients with frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) with a P301S tau mutation, the predominant phenotype was frontotemporal dementia in 3 and parkinsonism in 6. Comparison of the tau genotype/haplotype carrying the mutation and the initial clinical sign showed association between H1/H1 and parkinsonism and between H1/H2 and personality change. Thus, the tau haplotype carrying the mutation and the tau genotype may be related to the clinical phenotype throughout the disease course.


Subject(s)
Chromosomes, Human, Pair 17/genetics , Frontotemporal Lobar Degeneration/genetics , Genetic Linkage , Mutation/genetics , Parkinsonian Disorders/genetics , tau Proteins/genetics , Adult , Female , Frontotemporal Lobar Degeneration/pathology , Genotype , Haplotypes/genetics , Humans , Male , Parkinsonian Disorders/pathology , Young Adult
8.
AJNR Am J Neuroradiol ; 27(5): 1037-9, 2006 May.
Article in English | MEDLINE | ID: mdl-16687539

ABSTRACT

CT and MR imaging showed diffuse changes of the frontal white matter and genu of the corpus callosum with minimal atrophy and no contrast enhancement in a 41-year-old woman with progressive dementia. Brain biopsy disclosed axonal spheroids and gliosis in the white matter without macrophage or inflammatory infiltration or vessel abnormalities consistent with neuroaxonal leukodystrophy. This disease can be suspected on CT and MR imaging findings but requires neuropathologic examination to be diagnosed.


Subject(s)
Axons/pathology , Brain Diseases/complications , Brain Diseases/diagnosis , Dementia/etiology , Magnetic Resonance Imaging , Tomography, X-Ray Computed , Adult , Female , Humans
9.
Neurol Sci ; 27 Suppl 1: S44-6, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16708184

ABSTRACT

Dementia is due to lesions destroying a large amount of circuits anatomically connected with, or functionally related to, the associative areas of the cortex and the limbic structures. Dementia is not only the clinical hallmark of neurodegenerative diseases, most often proteinopathies, primarily involving the cerebral cortex, but also a symptom frequently associated to movement disorders, or secondary to systemic or neurological, non-degenerative diseases. In the elderly, it is often the consequence of the cumulative effect of different diseases.


Subject(s)
Alzheimer Disease/diagnosis , Brain/pathology , Dementia/diagnosis , Diagnosis, Differential , Humans
10.
Clin Neurophysiol ; 116(4): 895-904, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15792899

ABSTRACT

OBJECTIVE: To characterise flash visual evoked potentials (FVEPs) in 20 patients with Creutzfeldt-Jacob disease (CJD), and assess the relationships between spontaneous EEG patterns and the responses to individual stimuli. METHODS: We analysed the shape and time course of periodic sharp wave complexes (PSWCs) and responses to 1 Hz flashes. In nine patients, we applied an algorithm based on an autoregressive model with exogenous input (ARX) to estimate responses to individual random flashes and their interaction with PSWCs. RESULTS: The FVEPs included P1 and N1 components in all patients, and the P2 peak in 18. Eight patients showed giant FVEPs (N1-P2>60 V), all of whom had an MM polymorphism in codon 129 of the prion protein gene; in seven cases, the presence of giant FVEPs correlated with a prominent and almost continuous periodic EEG pattern. Giant N1-P2 abnormally spread on the anterior scalp regions, and had a different waveform distribution from that of the PSWCs. In five patients with a normal or slightly enlarged average N1-P2 amplitude, single sweep (ARX) analysis revealed a period of relative refractoriness following individual PSWCs. In four patients with 'giant' FVEPs, the individual responses occurred regardless of the interval between the stimulus and previous PSWC, but their amplitude had an inverse relationship with the interval length. CONCLUSIONS: Giant responses to flash stimuli are a common finding in CJD patients (40% of our cases). Single sweep ARX analysis showed that PSWCs were followed by a period of partial refractoriness, which prevented most of the individual responses to flashes, but not giant FVEPs. The association between prominent spontaneous paroxysms and giant FVEPs suggests that both are due to a common hyperexcitable change favouring neuronal synchronisation. SIGNIFICANCE: Our data contribute to clarifying the debated problem of the occurrence of giant FVEPs in CJD and their relationships with the spontaneous periodic EEG pattern.


Subject(s)
Creutzfeldt-Jakob Syndrome/physiopathology , Electroencephalography/methods , Evoked Potentials, Visual/physiology , Photic Stimulation/methods , Aged , Female , Humans , Male , Middle Aged
11.
Neurology ; 63(5): 910-2, 2004 Sep 14.
Article in English | MEDLINE | ID: mdl-15365148

ABSTRACT

Three members of an Italian family with autosomal dominant dementia and multiple strokes had the A713T mutation of the APP gene. The neuropathologic examination of the proband disclosed Alzheimer disease (AD) with severe cerebral amyloid angiopathy and multiple infarcts. This indicates that the A713T mutation of the APP gene, lying at the gamma-secretase cleavage site, can be responsible for AD with symptomatic cerebral amyloid angiopathy.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Cerebral Amyloid Angiopathy/genetics , Cerebral Infarction/genetics , Mutation, Missense , Point Mutation , Alzheimer Disease/pathology , Amino Acid Substitution , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor/chemistry , Aspartic Acid Endopeptidases , Brain/pathology , Cerebral Amyloid Angiopathy/pathology , Cerebral Infarction/pathology , Codon/genetics , DNA Mutational Analysis , Disease Progression , Endopeptidases/metabolism , Female , Genes, Dominant , Humans , Italy , Magnetic Resonance Imaging , Male , Middle Aged , Pedigree
12.
Neurol Sci ; 25 Suppl 1: S1-2, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15045608

ABSTRACT

Cerebral amyloid angiopathy is due to beta-protein accumulation in the vessel walls and occurs in normal aging, Alzheimer's disease and hereditary cerebral hemorrhage with amyloidosis. It causes intraparenchimal and subarachnoid bleeding with hemosiderin deposits and multiple infarcts presenting with headache, stroke and epilepsy. Such lesions may contribute to cognitive impairment. So far, no therapy is available. In future, amyloid in the vessel wall might be addressed by amyloid disaggregating drugs and amyloid antibodies.


Subject(s)
Aging/pathology , Alzheimer Disease/complications , Cerebral Amyloid Angiopathy/physiopathology , Cerebral Arteries/pathology , Cerebral Arteries/physiopathology , Aging/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Cerebral Amyloid Angiopathy/etiology , Cerebral Amyloid Angiopathy/therapy , Cerebral Arteries/metabolism , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/physiopathology , Humans , Risk Factors
13.
Neurology ; 61(9): 1288-91, 2003 Nov 11.
Article in English | MEDLINE | ID: mdl-14610142

ABSTRACT

The authors investigated two unrelated patients with Creutzfeldt-Jakob disease (CJD) with clinical features of sporadic CJD (sCJD) carrying one extra octapeptide repeat in the prion protein (PrP) gene (PRNP). A synaptic type PrP distribution throughout the cerebral gray matter and plaque-like PrP deposits in the subcortical gray structures were detected immunocytochemically. The different patterns of PrP deposition were associated with distinct types of protease-resistant PrP, similar to type 1 and type 2 of sCJD. The features suggest that this insertion is a pathogenic mutation.


Subject(s)
Amyloid/genetics , Creutzfeldt-Jakob Syndrome/genetics , Prions/genetics , Protein Precursors/genetics , 14-3-3 Proteins , Brain/pathology , Brain Chemistry , Creutzfeldt-Jakob Syndrome/cerebrospinal fluid , Creutzfeldt-Jakob Syndrome/diagnosis , Electroencephalography , Endopeptidases/chemistry , Heterozygote , Homozygote , Humans , Immunoblotting , Immunohistochemistry , Male , Middle Aged , Mutation , Polymerase Chain Reaction , Prion Proteins , Prions/chemistry , Tyrosine 3-Monooxygenase/cerebrospinal fluid , tau Proteins/cerebrospinal fluid
15.
Am J Pathol ; 158(6): 2201-7, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11395398

ABSTRACT

Gerstmann-Sträussler-Scheinker disease (GSS) is characterized by the accumulation of proteinase K (PK)-resistant prion protein fragments (PrP(sc)) of approximately 7 to 15 kd in the brain. Purified GSS amyloid is composed primarily of approximately 7-kd PrP peptides, whose N terminus corresponds to residues W(81) and G(88) to G(90) in patients with the A117V mutation and to residue W(81) in patients with the F198S mutation. The aim of this study was to characterize PrP in brain extracts, microsomal preparations, and purified fractions from A117V patients and to determine the N terminus of PrP(sc) species in both GSS A117V and F198S. In all GSS A117V patients, the approximately 7-kd PrP(sc) fragment isolated from nondigested and PK-digested samples had the major N terminus at residue G(88) and G(90), respectively. Conversely, in all patients with GSS F198S, an approximately 8-kd PrP(sc) fragment was isolated having the major N terminus start at residue G(74). It is possible that a further degradation of this fragment generates the amyloid subunit starting at W(81). The finding that patients with GSS A117V and F198S accumulate PrP(sc) fragments of different size and N-terminal sequence, suggests that these mutations generate two distinct PrP conformers.


Subject(s)
Amyloid/genetics , Gerstmann-Straussler-Scheinker Disease/genetics , Point Mutation , PrPSc Proteins/chemistry , Protein Precursors/genetics , Binding Sites , Brain/metabolism , Cell Extracts/analysis , Endopeptidase K/chemistry , Gerstmann-Straussler-Scheinker Disease/metabolism , Glycosylation , Humans , Peptide Fragments/chemistry , PrPSc Proteins/isolation & purification , PrPSc Proteins/metabolism , Prion Proteins , Prions , Protein Conformation , Subcellular Fractions/metabolism
16.
J Biol Chem ; 276(28): 25692-6, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11316802

ABSTRACT

A synthetic peptide consisting of amino acid residues 106-126 of the human prion protein (PrP-(106--126)) has been previously demonstrated to be neurotoxic and to induce microglial activation. The present study investigated the expression of the inducible form of the nitric-oxide synthase (NOS-II) in human microglial cells treated with PrP-(106--126). Using reverse transcriptase-polymerase chain reaction, we found that PrP-(106--126) induces NOS-II gene expression after 24 h of treatment and that this effect is accompanied by a peak of nuclear factor kappa B (NF-kappa B) binding at 30 min as evaluated by electrophoretic mobility shift assay. Since our previous data demonstrated tumor necrosis factor-alpha (TNF-alpha) to be a potent inducer of NOS-II in these cells, we analyzed the expression of this cytokine in PrP-(106--126)-treated microglia. PrP-(106--126) caused the release of TNF-alpha as detected by enzyme-linked immunosorbent assay, and a blocking antibody, anti-TNF-alpha, abolished NOS-II induction elicited by this peptide. Moreover, PrP-(106-126) activates p38 mitogen-activated protein kinase, and the inhibition of this pathway determines the ablation of NF-kappa B binding induced by this fragment peptide.


Subject(s)
Microglia/metabolism , Mitogen-Activated Protein Kinases/metabolism , Nitric Oxide Synthase/biosynthesis , Peptide Fragments/pharmacology , Prions/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Cells, Cultured , Gene Expression Regulation/drug effects , Humans , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Signal Transduction/drug effects , Signal Transduction/genetics , p38 Mitogen-Activated Protein Kinases
17.
J Biol Chem ; 276(8): 6009-15, 2001 Feb 23.
Article in English | MEDLINE | ID: mdl-11087738

ABSTRACT

Gerstmann-Sträussler-Scheinker disease (GSS) is a cerebral amyloidosis associated with mutations in the prion protein (PrP) gene (PRNP). The aim of this study was to characterize amyloid peptides purified from brain tissue of a patient with the A117V mutation who was Met/Val heterozygous at codon 129, Val(129) being in coupling phase with mutant Val117. The major peptide extracted from amyloid fibrils was a approximately 7-kDa PrP fragment. Sequence analysis and mass spectrometry showed that this fragment had ragged N and C termini, starting mainly at Gly88 and Gly90 and ending with Arg148, Glu152, or Asn153. Only Val was present at positions 117 and 129, indicating that the amyloid protein originated from mutant PrP molecules. In addition to the approximately 7-kDa peptides, the amyloid fraction contained N- and C-terminal PrP fragments corresponding to residues 23-41, 191-205, and 217-228. Fibrillogenesis in vitro with synthetic peptides corresponding to PrP fragments extracted from brain tissue showed that peptide PrP-(85-148) readily assembled into amyloid fibrils. Peptide PrP-(191-205) also formed fibrillary structures although with different morphology, whereas peptides PrP-(23-41) and PrP-(217-228) did not. These findings suggest that the processing of mutant PrP isoforms associated with Gerstmann-Sträussler-Scheinker disease may occur extracellularly. It is conceivable that full-length PrP and/or large PrP peptides are deposited in the extracellular compartment, partially degraded by proteases and further digested by tissue endopeptidases, originating a approximately 7-kDa protease-resistant core that is similar in patients with different mutations. Furthermore, the present data suggest that C-terminal fragments of PrP may participate in amyloid formation.


Subject(s)
Amyloid/genetics , Gerstmann-Straussler-Scheinker Disease/etiology , Peptide Fragments/isolation & purification , Prions/pathogenicity , Protein Precursors/genetics , Adult , Alleles , Cerebral Cortex/pathology , Gerstmann-Straussler-Scheinker Disease/genetics , Heterozygote , Humans , Male , Methionine/genetics , Prion Proteins , Prions/isolation & purification , Sequence Analysis, Protein , Syndrome , Valine/genetics
18.
Clin Neuropsychol ; 15(4): 508-15, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11935452

ABSTRACT

C.B., a right-handed 33-year-old man, presented with anterograde amnesia after acute heart block. Cognitive abilities were normal except for serious impairment of long-term episodic memory. The access to semantic information was fully preserved. Magnetic resonance showed high signal intensity and marked volume loss in the hippocampus bilaterally; the left and right parahippocampal gyrus, lateral occipito-temporal gyrus, inferior temporal gyrus, and lateral temporal cortex were normal. This case underlines that global amnesia associated with hippocampal damage does not affect semantic memory. Although the hippocampus is important in retrieving context-linked information, its role is not so crucial in retrieving semantic contents. Cortical areas surrounding the hippocampus and lateral temporal areas might guide the recall of semantic information.


Subject(s)
Amnesia, Anterograde/etiology , Brain Injuries/complications , Hippocampus/injuries , Semantics , Adult , Hippocampus/pathology , Humans , Magnetic Resonance Imaging , Male , Neuropsychological Tests , Severity of Illness Index
19.
J Neuropathol Exp Neurol ; 60(12): 1137-52, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11764087

ABSTRACT

We report clinical, neuropathologic and molecular genetic data from an individual affected by a familial Alzheimer disease (AD) variant. The proband had an onset of dementia at age 29 followed by generalized seizures a year later. He died at age 40. Neuropathologically, he had severe brain atrophy and characteristic histopathologic lesions of AD. Three additional neuropathologic features need to be emphasized: 1) severe deposition of Abeta in the form of diffuse deposits in the cerebral and cerebellar cortices, 2) numerous Abeta deposits in the subcortical white matter and in the centrum semiovale, and 3) numerous ectopic neurons, often containing tau-immunopositive neurofibrillary tangles, in the white maner of the frontal and temporal lobes. A molecular genetic analysis of DNA extracted from brain tissue of the proband revealed a S169L mutation in the Presenilin 1 (PSEN1) gene. The importance of this case lies in the presence of ectopic neurons in the white matter, early-onset seizures, and a PSEN1 mutation. We hypothesize that the PSEN1 mutation may have a causal relationship with an abnormality in neuronal development.


Subject(s)
Alzheimer Disease/genetics , Choristoma/genetics , Membrane Proteins/genetics , Mutation , Myoclonus/genetics , Neurons/pathology , Seizures/genetics , Adult , Alzheimer Disease/pathology , Amino Acid Substitution/genetics , Amyloid beta-Protein Precursor/genetics , Brain/pathology , Choristoma/pathology , Fatal Outcome , Female , Humans , Leucine/genetics , Male , Myoclonus/pathology , Pedigree , Presenilin-1 , Seizures/pathology , Serine/genetics
20.
Neurol Sci ; 21(3 Suppl): S57-61, 2000.
Article in English | MEDLINE | ID: mdl-11073229

ABSTRACT

The neuronal ceroid lipofuscinoses (NCL) are progressive neurodegenerative diseases occurring in infancy and adulthood. Atypical forms of these diseases have been described and are particularly represented in the late-infantile and juvenile onset groups. Recent progress in biochemistry and molecular genetics has identified some of these variants as separate disease entities while disclosing the phenotypic variability of some classic forms. We report the results of a retrospective analysis performed on a series of 27 NCL patients, 15 of which were atypical as to clinical and/or pathological findings. Most of such patients, belonging to the late-infantile onset group and displaying homogeneous clinical-pathological features, were suggestive for CLN6. The two atypical juvenile NCL patients had features which resembled the "protracted form" of the disease. Given their relative frequency, strict clinical and pathological criteria are still the most useful tools for identifying and characterizing atypical forms and for defining phenotype-genotype correlations.


Subject(s)
Membrane Glycoproteins , Molecular Chaperones , Neuronal Ceroid-Lipofuscinoses/classification , Neuronal Ceroid-Lipofuscinoses/diagnosis , Adult , Age of Onset , Child , Child, Preschool , Electroencephalography , Epilepsies, Myoclonic/diagnosis , Epilepsies, Myoclonic/etiology , Evoked Potentials, Somatosensory , Evoked Potentials, Visual , Female , Genotype , Humans , Male , Middle Aged , Neuronal Ceroid-Lipofuscinoses/genetics , Phenotype , Proteins/genetics , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...