Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 141
Filter
1.
J Prev Alzheimers Dis ; 6(2): 112-120, 2019.
Article in English | MEDLINE | ID: mdl-30756118

ABSTRACT

The study of individuals with autosomal dominant Alzheimer's disease affords one of the best opportunities to characterize the biological and cognitive changes of Alzheimer's disease that occur over the course of the preclinical and symptomatic stages. Unifying the knowledge gained from the past three decades of research in the world's largest single-mutation autosomal dominant Alzheimer's disease kindred - a family in Antioquia, Colombia with the E280A mutation in the Presenilin1 gene - will provide new directions for Alzheimer's research and a framework for generalizing the findings from this cohort to the more common sporadic form of Alzheimer's disease. As this specific mutation is virtually 100% penetrant for the development of the disease by midlife, we use a previously defined median age of onset for mild cognitive impairment for this cohort to examine the trajectory of the biological and cognitive markers of the disease as a function of the carriers' estimated years to clinical onset. Studies from this cohort suggest that structural and functional brain abnormalities - such as cortical thinning and hyperactivation in memory networks - as well as differences in biofluid and in vivo measurements of Alzheimer's-related pathological proteins distinguish Presenilin1 E280A mutation carriers from non-carriers as early as childhood, or approximately three decades before the median age of onset of clinical symptoms. We conclude our review with discussion on future directions for Alzheimer's disease research, with specific emphasis on ways to design studies that compare the generalizability of research in autosomal dominant Alzheimer's disease to the larger sporadic Alzheimer's disease population.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Brain/diagnostic imaging , Peptide Fragments/metabolism , Presenilin-1/genetics , Adolescent , Adult , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/cerebrospinal fluid , Aniline Compounds , Asymptomatic Diseases , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Biomarkers/metabolism , Brain/metabolism , Brain/physiopathology , Child , Colombia , Diffusion Tensor Imaging , Disease Progression , Electroencephalography , Ethylene Glycols , Female , Functional Neuroimaging , Humans , Magnetic Resonance Imaging , Male , Mental Status and Dementia Tests , Middle Aged , Peptide Fragments/blood , Peptide Fragments/cerebrospinal fluid , Positron-Emission Tomography , Radiopharmaceuticals , Tomography, Emission-Computed, Single-Photon , Young Adult
3.
Mol Psychiatry ; 20(11): 1294-300, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26324103

ABSTRACT

We have sequenced the complete genomes of 72 individuals affected with early-onset familial Alzheimer's disease caused by an autosomal dominant, highly penetrant mutation in the presenilin-1 (PSEN1) gene, and performed genome-wide association testing to identify variants that modify age at onset (AAO) of Alzheimer's disease. Our analysis identified a haplotype of single-nucleotide polymorphisms (SNPs) on chromosome 17 within a chemokine gene cluster associated with delayed onset of mild-cognitive impairment and dementia. Individuals carrying this haplotype had a mean AAO of mild-cognitive impairment at 51.0 ± 5.2 years compared with 41.1 ± 7.4 years for those without these SNPs. This haplotype thus appears to modify Alzheimer's AAO, conferring a large (~10 years) protective effect. The associated locus harbors several chemokines including eotaxin-1 encoded by CCL11, and the haplotype includes a missense polymorphism in this gene. Validating this association, we found plasma eotaxin-1 levels were correlated with disease AAO in an independent cohort from the University of California San Francisco Memory and Aging Center. In this second cohort, the associated haplotype disrupted the typical age-associated increase of eotaxin-1 levels, suggesting a complex regulatory role for this haplotype in the general population. Altogether, these results suggest eotaxin-1 as a novel modifier of Alzheimer's disease AAO and open potential avenues for therapy.


Subject(s)
Alzheimer Disease/genetics , Chemokine CCL11/genetics , Polymorphism, Single Nucleotide/genetics , Adult , Age of Onset , Aged , Alzheimer Disease/blood , Alzheimer Disease/complications , Chemokine CCL11/blood , Chromosomes, Human, Pair 17/genetics , Cognition Disorders/etiology , Cognition Disorders/genetics , Cohort Studies , Enzyme-Linked Immunosorbent Assay , Female , Genome-Wide Association Study , Genotype , Humans , Male , Middle Aged
4.
Mol Psychiatry ; 18(5): 568-75, 2013 May.
Article in English | MEDLINE | ID: mdl-22710270

ABSTRACT

The literature on GWAS (genome-wide association studies) data suggests that very large sample sizes (for example, 50,000 cases and 50,000 controls) may be required to detect significant associations of genomic regions for complex disorders such as Alzheimer's disease (AD). Because of the challenges of obtaining such large cohorts, we describe here a novel sequential strategy that combines pooling of DNA and bootstrapping (pbGWAS) in order to significantly increase the statistical power and exponentially reduce expenses. We applied this method to a very homogeneous sample of patients belonging to a unique and clinically well-characterized multigenerational pedigree with one of the most severe forms of early onset AD, carrying the PSEN1 p.Glu280Ala mutation (often referred to as E280A mutation), which originated as a consequence of a founder effect. In this cohort, we identified novel loci genome-wide significantly associated as modifiers of the age of onset of AD (CD44, rs187116, P=1.29 × 10⁻¹²; NPHP1, rs10173717, P=1.74 × 10⁻¹²; CADPS2, rs3757536, P=1.54 × 10⁻¹°; GREM2, rs12129547, P=1.69 × 10⁻¹³, among others) as well as other loci known to be associated with AD. Regions identified by pbGWAS were confirmed by subsequent individual genotyping. The pbGWAS methodology and the genes it targeted could provide important insights in determining the genetic causes of AD and other complex conditions.


Subject(s)
Alanine/genetics , Alzheimer Disease/genetics , Genetic Predisposition to Disease , Glutamic Acid/genetics , Presenilin-1/genetics , Age of Onset , Alzheimer Disease/epidemiology , Cohort Studies , Databases, Factual/statistics & numerical data , Female , Founder Effect , Gene Frequency , Genome-Wide Association Study , Genotype , Humans , Male , Mutation/genetics , Polymorphism, Single Nucleotide , Quantitative Trait Loci
5.
Oncogene ; 31(15): 1884-95, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-21874051

ABSTRACT

MicroRNAs (miRNAs) carry out post-transcriptional control of a multitude of cellular processes. Aberrant expression of miRNA can lead to diseases, including cancer. Gliomas are aggressive brain tumors that are thought to arise from transformed glioma-initiating neural stem cells (giNSCs). With the use of giNSCs and human glioblastoma cells, we investigated the function of miRNAs in gliomas. We identified pro-neuronal miR-128 as a candidate glioma tumor suppressor miRNA. Decreased expression of miR-128 correlates with aggressive human glioma subtypes. With a combination of molecular, cellular and in vivo approaches, we characterize miR-128's tumor suppressive role. miR-128 represses giNSC growth by enhancing neuronal differentiation. miR-128 represses growth and mediates differentiation by targeting oncogenic receptor tyrosine kinases (RTKs) epithelial growth factor receptor and platelet-derived growth factor receptor-α. Using an autochthonous glioma mouse model, we demonstrated that miR-128 repressed gliomagenesis. We identified miR-128 as a glioma tumor suppressor that targets RTK signaling to repress giNSC self-renewal and enhance differentiation.


Subject(s)
Brain Neoplasms/genetics , ErbB Receptors/genetics , Genes, Tumor Suppressor , Glioma/genetics , MicroRNAs/physiology , Receptor, Platelet-Derived Growth Factor alpha/genetics , Animals , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Mice, SCID , Neural Stem Cells/physiology
6.
Mol Biol Evol ; 27(5): 1058-69, 2010 May.
Article in English | MEDLINE | ID: mdl-20026484

ABSTRACT

PDZ domains are protein-protein interaction modules widely used to assemble membranous signaling complexes including those found in the neuronal synapse. PDZ-containing genes encoded in metazoan genomes vastly outnumber those in prokaryotes, plants, and fungi. By comparing 40 proteomes to track the evolutionary history of the PDZ domain, we observed that the variety of associations between PDZ and other domains expands greatly along the stem leading to metazoans and choanoflagellates. We asked whether the expansion of PDZ domains was due to random or specific sequence changes. Studying the sequence signatures of 58 PDZ lineages that are common to bilaterian animals, we showed that six common amino acid residues are able to classify 96% of PDZ domains to their correct evolutionary lineage. In PDZ domain-ligand cocrystals, four of these "classifying positions" lie in direct contact with the -1 and -3 residues of the ligand. This suggests coevolution of the more flexible regions of the binding interaction as a central mechanism of specialization inherent within the PDZ domain. To identify these positions, we devised two independent algorithms--a metric termed within-clade entropy (WCE) and an average mutual information (AvgMI) score--that both reached similar results. Extending these tools to the choanoflagellate, Monosiga brevicollis, we compared its PDZ domains with their putative metazoan orthologs. Interestingly, the M. brevicollis genes lack conservation at the classifying positions suggesting dissociation between domain organization in multidomain proteins and specific changes within the PDZ domain.


Subject(s)
Evolution, Molecular , PDZ Domains/genetics , Amino Acid Sequence , Amino Acids/genetics , Animals , Choanoflagellata/metabolism , Computer Simulation , Conserved Sequence , Entropy , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Protein Binding , Sequence Alignment
7.
Neurology ; 59(2): 277-9, 2002 Jul 23.
Article in English | MEDLINE | ID: mdl-12136071

ABSTRACT

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is caused by mutations in the notch3 epidermal growth factor-like repeats. A Colombian kindred carries a novel C455R mutation located in the predicted ligand-binding domain. Stroke occurred in the patients at an unusually early age (median age: 31 years) in comparison to the more frequent onset in the fourth decade of life in other CADASIL populations, including a second Colombian kindred with an R1031C mutation.


Subject(s)
Dementia, Multi-Infarct/genetics , Mutation , Stroke/genetics , Adult , Age of Onset , Aged , Arginine/metabolism , Colombia , Cysteine/metabolism , DNA Mutational Analysis , Female , Heterozygote , Humans , Male , Middle Aged
8.
J Neurosci Res ; 67(5): 618-24, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11891774

ABSTRACT

delta-Catenin associates with adhesive junctions and facilitates cellular morphogenesis (Lu et al., 1999). Here we show that delta-catenin colocalizes with actin filaments and Abl tyrosine kinase in the growth cones of cultured hippocampal neurons. PC12 cells induced to express delta-catenin show accelerated neurite extension upon nerve growth factor (NGF) stimulation. STI571, an Abl family kinase inhibitor, further accentuates these stimulatory effects. delta-Catenin is a potent substrate for Abl in vitro using an immunocomplex assay and most of the Abl-induced tyrosine phosphorylation within cells is present in the N-terminus of delta-catenin. When delta-catenin-expressing epithelial cells are induced to scatter in response to hepatocyte growth factor (HGF), STI571 leads to the rapid redistribution of delta-catenin and changes in cellular morphology. We suggest that delta-catenin is a possible Abl substrate and acts downstream of Abl to orchestrate actin-based cellular morphogenesis.


Subject(s)
Brain/embryology , Brain/enzymology , Cell Differentiation/physiology , Cytoskeletal Proteins/metabolism , Growth Cones/metabolism , Growth Substances/pharmacology , Proto-Oncogene Proteins c-abl/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Animals , Armadillo Domain Proteins , Brain/cytology , Catenins , Cell Adhesion Molecules , Cell Differentiation/drug effects , Cytoskeletal Proteins/genetics , Enzyme Inhibitors/pharmacology , Fetus , Growth Cones/drug effects , Growth Cones/ultrastructure , Immunohistochemistry , Mice , Nucleic Acid Synthesis Inhibitors/pharmacology , PC12 Cells , Phosphoproteins , Phosphorylation/drug effects , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Rats , Delta Catenin
9.
Neuron ; 32(4): 683-96, 2001 Nov 20.
Article in English | MEDLINE | ID: mdl-11719208

ABSTRACT

RNA granules are a macromolecular structure observed in neurons, where they serve as motile units that translocate mRNAs. Isolated RNA granules are highly enriched in Staufen protein and ultrastructurally contain densely packed clusters of ribosomes. With depolarization, many mRNAs, including those involved in plasticity, rapidly shift from the RNA granule fraction to polysomes. Depolarization reorganizes granules and induces a less compact organization of their ribosomes. RNA granules are not translationally competent, as indicated by the failure to incorporate radioactive amino acids and the absence of eIF4E, 4G, and tRNAs. We concluded that RNA granules are a local storage compartment for mRNAs under translational arrest but are poised for release to actively translated pools. Local release of mRNAs and ribosomes from granules may serve as a macromolecular mechanism linking RNA localization to translation and synaptic plasticity.


Subject(s)
Cytoplasmic Granules/metabolism , Neurons/physiology , Protein Biosynthesis/physiology , RNA, Messenger/metabolism , Animals , Cell Fractionation , Cells, Cultured , Cerebral Cortex/cytology , Cytoplasmic Granules/ultrastructure , Female , Microscopy, Electron , Neuronal Plasticity/physiology , Neurons/cytology , Neurons/drug effects , Potassium Chloride/pharmacology , Pregnancy , Rats , Rats, Sprague-Dawley , Ribosomes/metabolism , Stimulation, Chemical , Synapses/metabolism
10.
Neurology ; 57(6): 957-64, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11571317

ABSTRACT

OBJECTIVES: To estimate annual transition probabilities (i.e., the likelihood that a patient will move from one disease stage to another in a given time period) for AD progression. Transition probabilities are estimated by disease stages (mild, moderate, severe) and settings of care (community, nursing home), accounting for differences in age, gender, and behavioral symptoms as well as the length of time a patient has been in a disease stage. METHODS: Using data from the Consortium to Establish a Registry for Alzheimer's Disease (CERAD), the authors employed a modified survival analysis to estimate stage-to-stage and stage-to-nursing home transition probabilities. To account for individual variability, a Cox proportional hazards model was fit to the CERAD data to estimate hazard ratios for gender, age (50 to 64, 65 to 74, and more than 75 years), and level of behavioral symptoms (low/high, according to responses to the Behavioral Rating Scale for Dementia) for each of the key stage-to-stage and stage-to-nursing home transitions. RESULTS: The transition probabilities underscore the rapid progression of patients into more severe disease stages and into nursing homes and the differences among population subgroups. In general, male gender, age under 65, and high level of behavioral symptoms were associated with higher transition probabilities to more severe disease stages. Disease progression is roughly constant as a function of the time a patient has spent in a particular stage. CONCLUSIONS: Transition probabilities provide a useful means of characterizing AD progression. Economic models of interventions for AD should consider the varied course of progression for different population subgroups, particularly those defined by high levels of behavioral symptoms.


Subject(s)
Alzheimer Disease/diagnosis , Registries/statistics & numerical data , Aged , Aged, 80 and over , Alzheimer Disease/classification , Alzheimer Disease/economics , Alzheimer Disease/epidemiology , Disease Progression , Female , Homes for the Aged/economics , Humans , Male , Models, Economic , Nursing Homes/economics , Probability , Proportional Hazards Models , United States/epidemiology
11.
Health Aff (Millwood) ; 20(5): 252-64, 2001.
Article in English | MEDLINE | ID: mdl-11558711

ABSTRACT

In a general population survey (N = 314), 79 percent of respondents stated that they would take a hypothetical genetic test to predict whether they will eventually develop Alzheimer's disease. The proportion fell to 45 percent for a "partially predictive" test (which had a one in ten chance of being incorrect). Inclination to obtain testing was similar across age groups. Respondents were willing to pay $324 for the completely predictive test. Respondents stated that if they tested positive, they would sign advance directives (84 percent), get their finances in order (74 percent), and purchase long-term care insurance (69 percent). Only a third of respondents expressed concern about confidentiality. The results suggest that people value genetic testingfor personal and financial reasons, but they also underscore the need to counsel potential recipients carefully about the accuracy and implications of test information.


Subject(s)
Alzheimer Disease/genetics , Attitude to Health , Genetic Testing , Public Opinion , Adult , Aged , Female , Genetic Testing/economics , Humans , Male , Middle Aged , Motivation , Socioeconomic Factors , United States
12.
Science ; 293(5528): 263-9, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11431533

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the progressive accumulation in selected neurons of protein inclusions containing alpha-synuclein and ubiquitin. Rare inherited forms of PD are caused by autosomal dominant mutations in alpha-synuclein or by autosomal recessive mutations in parkin, an E3 ubiquitin ligase. We hypothesized that these two gene products interact functionally, namely, that parkin ubiquitinates alpha-synuclein normally and that this process is altered in autosomal recessive PD. We have now identified a protein complex in normal human brain that includes parkin as the E3 ubiquitin ligase, UbcH7 as its associated E2 ubiquitin conjugating enzyme, and a new 22-kilodalton glycosylated form of alpha-synuclein (alphaSp22) as its substrate. In contrast to normal parkin, mutant parkin associated with autosomal recessive PD failed to bind alphaSp22. In an in vitro ubiquitination assay, alphaSp22 was modified by normal but not mutant parkin into polyubiquitinated, high molecular weight species. Accordingly, alphaSp22 accumulated in a non-ubiquitinated form in parkin-deficient PD brains. We conclude that alphaSp22 is a substrate for parkin's ubiquitin ligase activity in normal human brain and that loss of parkin function causes pathological alphaSp22 accumulation. These findings demonstrate a critical biochemical reaction between the two PD-linked gene products and suggest that this reaction underlies the accumulation of ubiquitinated alpha-synuclein in conventional PD.


Subject(s)
Brain/metabolism , Ligases/metabolism , Nerve Tissue Proteins/metabolism , Parkinson Disease/metabolism , Ubiquitin-Conjugating Enzymes , Ubiquitin-Protein Ligases , Ubiquitins/metabolism , Brain/enzymology , Brain Stem/enzymology , Brain Stem/metabolism , Cell Line , Detergents , Freezing , Glycosylation , Humans , Lewy Bodies/enzymology , Lewy Bodies/metabolism , Ligases/genetics , Mutation, Missense , Parkinson Disease/enzymology , Parkinson Disease/genetics , Parkinsonian Disorders/enzymology , Parkinsonian Disorders/genetics , Parkinsonian Disorders/metabolism , Substrate Specificity , Synucleins , alpha-Synuclein
13.
Neurology ; 56(11): 1545-51, 2001 Jun 12.
Article in English | MEDLINE | ID: mdl-11402113

ABSTRACT

OBJECTIVE: To investigate the influence of the presenilin-1 gene (PS-1) mutation on regional cerebral perfusion, SPECT was evaluated in 57 individuals. The subjects were members of a large pedigree from Colombia, South America, many of whom carry a PS-1 mutation for early-onset AD. METHODS: Members of this large kindred who were cognitively normal and did not carry the PS-1 mutation (n = 23) were compared with subjects who were carriers of the mutation but were asymptomatic (n = 18) and with individuals with the mutation and a clinical diagnosis of AD (n = 16). Cerebral perfusion was measured in each subject using hexamethylpropyleneamine oxime SPECT. The data were analyzed in two ways: 1) Mean cerebral perfusion in each of 4320 voxels in the brain was compared among the groups using t-tests (t-maps); and 2) each individual received a weighted score on 20 vectors (factors), based on a large normative sample (n = 200), using a method known as singular value decomposition (SVD). RESULTS: Based on t-maps, subjects with the PS-1 mutation who were asymptomatic demonstrated reduced perfusion in comparison with the normal control subjects in the hippocampal complex, anterior and posterior cingulate, posterior parietal lobe, and anterior frontal lobe. The AD patients demonstrated decreased perfusion in the posterior parietal and superior frontal cortex in comparison with the normal control subjects. Discriminant function analysis of the vector scores derived from SVD (adjusted for age and gender) accurately discriminated 86% of the subjects in the three groups (p < 0.0005). CONCLUSION: Regional cerebral perfusion abnormalities based on SPECT are detectable before development of the clinical symptoms of AD in carriers of the PS-1 mutation.


Subject(s)
Alzheimer Disease/physiopathology , Cerebrovascular Circulation/genetics , Heterozygote , Membrane Proteins/genetics , Adult , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics , Amygdala/blood supply , Family Health , Female , Gyrus Cinguli/blood supply , Hippocampus/blood supply , Humans , Male , Middle Aged , Mutation/physiology , Parietal Lobe/blood supply , Presenilin-1 , Temporal Lobe/blood supply , Tomography, Emission-Computed, Single-Photon
14.
Mol Biol Cell ; 12(1): 171-84, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11160831

ABSTRACT

How tau mutations lead to neurodegeneration is unknown but may be related to altered microtubule binding properties of mutant tau protein. The tendency for the mutations to cluster around the microtubule-binding domain of tau or to alter the ratios of those splice isoforms that affect binding supports the view that the tau/microtubule interaction is critical and finely regulated. In cells transfected with both mutant and wild-type tau isoforms fused to either yellow fluorescent protein or cyan fluorescent protein we can observe tau fusion proteins that differ by a single amino acid or by the inclusion or exclusion of exon 10. With coexpression of mutant and wild-type tau, the mutant isoform appears diffuse throughout the cytoplasm; however, when mutant tau is expressed alone, it appears mostly bound to the microtubules. Dual imaging of the three- and four-repeat tau isoforms indicated that the expression of four-repeat tau displaced three-repeat tau from the microtubules. These results suggest that altered kinetic competition among the isoforms for microtubule binding could be a disease precipitant.


Subject(s)
Binding, Competitive , Microtubules/metabolism , tau Proteins/metabolism , 3T3 Cells , Animals , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Mice , Microscopy, Fluorescence , Microscopy, Video , Mutation, Missense , Protein Binding , Protein Isoforms/metabolism , Recombinant Fusion Proteins/genetics , Transfection , tau Proteins/genetics
15.
Bioessays ; 23(2): 203-5, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11169594
16.
Dev Biol ; 227(2): 450-64, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11071766

ABSTRACT

Presenilin is an essential gene for development that when disrupted leads to a neurogenic phenotype that closely resembles Notch loss of function in Drosophila. In humans, many naturally occurring mutations in Presenilin 1 or 2 cause early onset Alzheimer's disease. Both loss of expression and overexpression of Presenilin suggested a role for this protein in the localization of Armadillo/beta-catenin. In blastoderm stage Presenilin mutants, Arm is aberrantly distributed, often in Ubiquitin-immunoreactive cytoplasmic inclusions predominantly located basally in the cell. These inclusions were not observed in loss of function Notch mutants, suggesting that failure to process Notch is not the only consequence of the loss of Presenilin function. Human presenilin 1 expressed in Drosophila produces embryonic phenotypes resembling those associated with mutations in Armadillo and exhibited reduced Armadillo at the plasma membrane that is likely due to retention of Armadillo in a complex with Presenilin. The interaction between Armadillo/beta-catenin and Presenilin 1 requires a third protein which may be delta-catenin. Our results suggest that Presenilin may regulate the delivery of a multiprotein complex that regulates Armadillo trafficking between the adherens junction and the proteasome.


Subject(s)
Cytoskeletal Proteins/metabolism , Drosophila Proteins , Drosophila/genetics , Drosophila/metabolism , Insect Proteins/metabolism , Membrane Proteins/metabolism , Trans-Activators , Animals , Animals, Genetically Modified , Armadillo Domain Proteins , Catenins , Cell Adhesion Molecules , Cytoskeletal Proteins/genetics , Drosophila/embryology , Female , Gene Expression Regulation, Developmental , Genes, Insect , Humans , In Vitro Techniques , Insect Proteins/genetics , Membrane Proteins/genetics , Models, Biological , Mutation , Phenotype , Phosphoproteins , Presenilin-1 , Receptors, Notch , Transcription Factors , beta Catenin , Delta Catenin
17.
J Neurosci ; 20(17): 6385-93, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10964944

ABSTRACT

The CaMKIIalpha mRNA extends into distal hippocampal dendrites, and the 3' untranslated region (3'UTR) is sufficient to mediate this localization. We labeled the 3'UTR of the CaMKIIalpha mRNA in hippocampal cultures by using a green fluorescent protein (GFP)/MS2 bacteriophage tagging system. The CaMKIIalpha 3'UTR formed discrete granules throughout the dendrites of transfected cells. The identity of the fluorescent granules was verified by in situ hybridization. Over 30 min time periods these granules redistributed without a net increase in granule number; with depolarization there is a tendency toward increased numbers of granules in the dendrites. These observations suggest that finer time resolution of granule motility might reveal changes in the motility characteristics of granules after depolarization. So that motile granules could be tracked, shorter periods of observation were required. The movements of motile granules can be categorized as oscillatory, unidirectional anterograde, or unidirectional retrograde. Colocalization of CaMKIIalpha 3'UTR granules and synapses suggested that oscillatory movements allowed the granules to sample several local synapses. Neuronal depolarization increased the number of granules in the anterograde motile pool. Based on the time frame over which the granule number increased, the translocation of granules may serve to prepare the dendrite for mounting an adequate local translation response to future stimuli. Although the resident pool of granules can respond to signals that induce local translation, the number of granules in a dendrite might reflect its activation history.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/genetics , Hippocampus/metabolism , Neurons/enzymology , RNA, Messenger/genetics , 3' Untranslated Regions/genetics , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/analysis , Cells, Cultured , Cytoplasmic Granules/enzymology , Cytoplasmic Granules/physiology , Dendrites/enzymology , Embryo, Mammalian , Green Fluorescent Proteins , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Models, Neurological , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/biosynthesis
18.
J Clin Exp Neuropsychol ; 22(4): 483-95, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10923058

ABSTRACT

Few longitudinal studies have been carried out to investigate the cognitive decline in early onset of familial Alzheimer's disease (FAD). In this study 12 patients with FAD (M age = 49.61 years, SD = 4.99), 10 patients with sporadic Alzheimer's disease (SAD) (M age = 71.40, SD =10.00), and 15 matched normal controls (M age = 45.01, SD = 7.24) were selected. A comprehensive neuropsychological battery was administered three times over a period of 18 months. Individuals designated as FAD met the criteria for dementia and were positive for the E280A presenilin 1 mutation. Participants with SAD met the criteria for dementia and were negative for the E280A presenilin 1 mutation. Normal control participants were the FAD patients' relatives, who were negative for the mutation. Two groups of neuropsychological instruments were administered: (1) The Consortium to Establish a Registry for Alzheimer's Disease (CERAD) neuropsychological test battery, and (2) additional neuropsychological tests of abstraction and constructional abilities. Patients with FAD were significantly impaired on all measures at the first examination except for reading of words. While the performance of the normal controls remained unchanged over the 18 months for most neuropsychological tests, the patients with FAD displayed a decline in verbal memory, language, constructional and abstraction tests. The greatest decline was observed on the Mini-Mental State Exam scores. Patients with SAD demonstrated a similar pattern of cognitive decline, but the decline was faster in FAD than in SAD participants.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/psychology , Cognition Disorders/genetics , Cognition Disorders/psychology , Membrane Proteins/genetics , Mutation , Aged , Aged, 80 and over , Case-Control Studies , Disease Progression , Female , Genetic Predisposition to Disease , Humans , Longitudinal Studies , Male , Memory , Middle Aged , Neuropsychological Tests , Presenilin-1
19.
Genomics ; 63(2): 157-64, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10673328

ABSTRACT

Delta-catenin is an adherens junction protein involved in cell motility and expressed early in neuronal development. It was discovered as an interactor with presenilin-1. The genomic structure of the human delta-catenin gene (Human Gene Nomenclature Committee-approved symbol CTNND2) was determined and mapped to 5p15.2. A deletion of this chromosomal region has been associated with the cri-du-chat syndrome (CDCS), a segmental aneusomy syndrome of 5p that is associated with an unusual high-pitched cry at birth, facial dysmorphology, poor growth, and severe mental retardation. delta-catenin maps to a specific region in 5p15.2 that has been implicated in the mental retardation phenotype. The breakpoints in patients with 5p terminal deletions were characterized with respect to the severity of mental retardation and the physical location of the delta-catenin gene. A strong correlation was found between the hemizygous loss of delta-catenin and severe mental retardation. These findings and the properties of delta-catenin as a neuronal-specific protein, expressed early in development and involved in cell motility, support its role in the mental retardation of CDCS when present in only one copy.


Subject(s)
Cri-du-Chat Syndrome/genetics , Cytoskeletal Proteins/genetics , Intellectual Disability/genetics , Armadillo Domain Proteins , Base Sequence , Catenins , Cell Adhesion Molecules , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Cloning, Molecular , DNA, Complementary/genetics , Exons , Genotype , Humans , Introns , Phenotype , Phosphoproteins , Physical Chromosome Mapping , Delta Catenin
20.
Chem Biol ; 7(1): R9-R12, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10662685

ABSTRACT

The rare neurological disorders frontotemporal dementia and British dementia have been linked to two mutant genes whose products constitute the fibrils that define the two disease pathologies. Two recent studies add to the mounting circumstantial case that protein fibrillization, inside (neurofibrillary tangles) or outside (amyloid plaques) of the neuron, may be pathogenic and suggest that either or both of these mechanisms could initiate Alzheimer's disease.


Subject(s)
Inclusion Bodies/pathology , Nerve Degeneration/pathology , Amino Acid Sequence , Dementia/genetics , Dementia/pathology , Humans , Inclusion Bodies/genetics , Molecular Sequence Data , Nerve Degeneration/genetics , Neurofibrillary Tangles/genetics , Neurofibrillary Tangles/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...