Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
1.
PLoS One ; 13(11): e0206547, 2018.
Article in English | MEDLINE | ID: mdl-30383799

ABSTRACT

A large body of literature links risk of cognitive decline, mild cognitive impairment (MCI) and dementia with Type 2 Diabetes (T2D) or pre-diabetes. Accumulating evidence implicates a close relationship between the brain insulin receptor signaling pathway (IRSP) and the accumulation of amyloid beta and hyperphosphorylated and conformationally abnormal tau. We showed previously that the neuropathological features of Alzheimer's disease (AD were reduced in patients with diabetes who were treated with insulin and oral antidiabetic medications. To understand better the neurobiological substrates of T2D and T2D medications in AD, we examined IRSP and endothelial cell markers in the parahippocampal gyrus of controls (N = 30), of persons with AD (N = 19), and of persons with AD and T2D, who, in turn, had been treated with anti-diabetic drugs (insulin and or oral agents; N = 34). We studied the gene expression of selected members of the IRSP and selective endothelial cell markers in bulk postmortem tissue from the parahippocampal gyrus and in endothelial cell enriched isolates from the same brain region. The results indicated that there are considerable abnormalities and reductions in gene expression (bulk tissue homogenates and endothelial cell isolates) in the parahippocampal gyri of persons with AD that map directly to genes associated with the microvasculature and the IRSP. Our results also showed that the numbers of abnormally expressed microvasculature and IRSP associated genes in diabetic AD donors who had been treated with anti-diabetic agents were reduced significantly. These findings suggest that anti-diabetic treatments may reduce or normalize compromised microvascular and IRSP functions in AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Endothelial Cells/metabolism , Hypoglycemic Agents/therapeutic use , Parahippocampal Gyrus/drug effects , Parahippocampal Gyrus/metabolism , Aged, 80 and over , Cohort Studies , Endothelial Cells/drug effects , Female , Gene Expression/drug effects , Humans , Male , Microvessels/drug effects , Microvessels/metabolism , RNA, Messenger/metabolism , Receptor, Insulin
3.
Sci Rep ; 6: 19393, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26786552

ABSTRACT

Non-fibrillar soluble oligomeric forms of amyloid-ß peptide (oAß) and tau proteins are likely to play a major role in Alzheimer's disease (AD). The prevailing hypothesis on the disease etiopathogenesis is that oAß initiates tau pathology that slowly spreads throughout the medial temporal cortex and neocortices independently of Aß, eventually leading to memory loss. Here we show that a brief exposure to extracellular recombinant human tau oligomers (oTau), but not monomers, produces an impairment of long-term potentiation (LTP) and memory, independent of the presence of high oAß levels. The impairment is immediate as it raises as soon as 20 min after exposure to the oligomers. These effects are reproduced either by oTau extracted from AD human specimens, or naturally produced in mice overexpressing human tau. Finally, we found that oTau could also act in combination with oAß to produce these effects, as sub-toxic doses of the two peptides combined lead to LTP and memory impairment. These findings provide a novel view of the effects of tau and Aß on memory loss, offering new therapeutic opportunities in the therapy of AD and other neurodegenerative diseases associated with Aß and tau pathology.


Subject(s)
Long-Term Potentiation , Memory , Protein Aggregates , Protein Aggregation, Pathological , Protein Multimerization , tau Proteins/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Extracellular Space/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Mice , Neurons/metabolism , tau Proteins/chemistry
5.
Mol Psychiatry ; 21(6): 823-30, 2016 06.
Article in English | MEDLINE | ID: mdl-26416546

ABSTRACT

Excitatory amino-acid transporters (EAATs) bind and transport glutamate, limiting spillover from synapses due to their dense perisynaptic expression primarily on astroglia. Converging evidence suggests that abnormalities in the astroglial glutamate transporter localization and function may underlie a disease mechanism with pathological glutamate spillover as well as alterations in the kinetics of perisynaptic glutamate buffering and uptake contributing to dysfunction of thalamo-cortical circuits in schizophrenia. We explored this hypothesis by performing cell- and region-level studies of EAAT1 and EAAT2 expression in the mediodorsal nucleus of the thalamus in an elderly cohort of subjects with schizophrenia. We found decreased protein expression for the typically astroglial-localized glutamate transporters in the mediodorsal and ventral tier nuclei. We next used laser-capture microdissection and quantitative polymerase chain reaction to assess cell-level expression of the transporters and their splice variants. In the mediodorsal nucleus, we found lower expression of transporter transcripts in a population of cells enriched for astrocytes, and higher expression of transporter transcripts in a population of cells enriched for relay neurons. We confirmed expression of transporter protein in neurons in schizophrenia using dual-label immunofluorescence. Finally, the pattern of transporter mRNA and protein expression in rodents treated for 9 months with antipsychotic medication suggests that our findings are not due to the effects of antipsychotic treatment. We found a compensatory increase in transporter expression in neurons that might be secondary to a loss of transporter expression in astrocytes. These changes suggest a profound abnormality in astrocyte functions that support, nourish and maintain neuronal fidelity and synaptic activity.


Subject(s)
Astrocytes/metabolism , Glutamate Plasma Membrane Transport Proteins/metabolism , Glutamic Acid/metabolism , Aged , Amino Acid Transport System X-AG/metabolism , Animals , Carrier Proteins/genetics , Female , Gene Expression , Humans , Male , Mediodorsal Thalamic Nucleus/metabolism , Mediodorsal Thalamic Nucleus/physiopathology , Mice , Middle Aged , Neurons/metabolism , RNA, Messenger/metabolism , Schizophrenia/genetics , Schizophrenia/metabolism , Thalamus/physiopathology
6.
Mol Psychiatry ; 21(8): 1099-111, 2016 08.
Article in English | MEDLINE | ID: mdl-26552589

ABSTRACT

Identification and characterization of molecular mechanisms that connect genetic risk factors to initiation and evolution of disease pathophysiology represent major goals and opportunities for improving therapeutic and diagnostic outcomes in Alzheimer's disease (AD). Integrative genomic analysis of the human AD brain transcriptome holds potential for revealing novel mechanisms of dysfunction that underlie the onset and/or progression of the disease. We performed an integrative genomic analysis of brain tissue-derived transcriptomes measured from two lines of mice expressing distinct mutant AD-related proteins. The first line expresses oligomerogenic mutant APP(E693Q) inside neurons, leading to the accumulation of amyloid beta (Aß) oligomers and behavioral impairment, but never develops parenchymal fibrillar amyloid deposits. The second line expresses APP(KM670/671NL)/PSEN1(Δexon9) in neurons and accumulates fibrillar Aß amyloid and amyloid plaques accompanied by neuritic dystrophy and behavioral impairment. We performed RNA sequencing analyses of the dentate gyrus and entorhinal cortex from each line and from wild-type mice. We then performed an integrative genomic analysis to identify dysregulated molecules and pathways, comparing transgenic mice with wild-type controls as well as to each other. We also compared these results with datasets derived from human AD brain. Differential gene and exon expression analysis revealed pervasive alterations in APP/Aß metabolism, epigenetic control of neurogenesis, cytoskeletal organization and extracellular matrix (ECM) regulation. Comparative molecular analysis converged on FMR1 (Fragile X Mental Retardation 1), an important negative regulator of APP translation and oligomerogenesis in the post-synaptic space. Integration of these transcriptomic results with human postmortem AD gene networks, differential expression and differential splicing signatures identified significant similarities in pathway dysregulation, including ECM regulation and neurogenesis, as well as strong overlap with AD-associated co-expression network structures. The strong overlap in molecular systems features supports the relevance of these findings from the AD mouse models to human AD.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Presenilin-1/genetics , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Fibrillar Collagens , Fragile X Mental Retardation Protein/metabolism , Humans , Mice , Mice, Transgenic , Mutation , Neurogenesis , Neurons/metabolism , Plaque, Amyloid/pathology , Risk Factors , Transcriptome/genetics
7.
Transl Psychiatry ; 5: e612, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26241350

ABSTRACT

Inhibitory neurotransmission is primarily mediated by γ-aminobutyric acid (GABA) activating synaptic GABA type A receptors (GABA(A)R). In schizophrenia, presynaptic GABAergic signaling deficits are among the most replicated findings; however, postsynaptic GABAergic deficits are less well characterized. Our lab has previously demonstrated that although there is no difference in total protein expression of the α1-6, ß1-3 or γ2 GABA(A)R subunits in the superior temporal gyrus (STG) in schizophrenia, the α1, ß1 and ß2 GABA(A)R subunits are abnormally N-glycosylated. N-glycosylation is a posttranslational modification that has important functional roles in protein folding, multimer assembly and forward trafficking. To investigate the impact that altered N-glycosylation has on the assembly and trafficking of GABA(A)Rs in schizophrenia, this study used western blot analysis to measure the expression of α1, α2, ß1, ß2 and γ2 GABA(A)R subunits in subcellular fractions enriched for endoplasmic reticulum (ER) and synapses (SYN) from STG of schizophrenia (N = 16) and comparison (N = 14) subjects and found evidence of abnormal localization of the ß1 and ß2 GABA(A)R subunits and subunit isoforms in schizophrenia. The ß2 subunit is expressed as three isoforms at 52 kDa (ß2(52 kDa)), 50 kDa (ß2(50 kDa)) and 48 kDa (ß2(48 kDa)). In the ER, we found increased total ß2 GABA(A)R subunit (ß2(ALL)) expression driven by increased ß2(50 kDa), a decreased ratio of ß(248 kDa):ß2(ALL) and an increased ratio of ß2(50 kDa):ß2(48 kDa). Decreased ratios of ß1:ß2(ALL) and ß1:ß2(50 kDa) in both the ER and SYN fractions and an increased ratio of ß2(52 kDa):ß(248 kDa) at the synapse were also identified in schizophrenia. Taken together, these findings provide evidence that alterations of N-glycosylation may contribute to GABAergic signaling deficits in schizophrenia by disrupting the assembly and trafficking of GABA(A)Rs.


Subject(s)
Brain/metabolism , Receptors, GABA-A/metabolism , Schizophrenia/metabolism , Aged , Blotting, Western , Brain/physiopathology , Case-Control Studies , Endoplasmic Reticulum/chemistry , Female , Glycosylation , Humans , Male , Receptors, GABA-A/analysis , Receptors, GABA-A/physiology , Receptors, GABA-B/metabolism , Receptors, GABA-B/physiology , Schizophrenia/physiopathology , Subcellular Fractions/chemistry , Synapses/chemistry , Temporal Lobe/chemistry
8.
Transl Psychiatry ; 5: e579, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26057049

ABSTRACT

Dysregulation of the glutamate transporters EAAT1 and EAAT2 and their isoforms have been implicated in schizophrenia. EAAT1 and EAAT2 expression has been studied in different brain regions but the prevalence of astrocytic glutamate transporter expression masks the more subtle changes in excitatory amino acid transporters (EAATs) isoforms in neurons in the cortex. Using laser capture microdissection, pyramidal neurons were cut from the anterior cingulate cortex of postmortem schizophrenia (n = 20) and control (n = 20) subjects. The messenger RNA (mRNA) levels of EAAT1, EAAT2 and the splice variants EAAT1 exon9skipping, EAAT2 exon9skipping and EAAT2b were analyzed by real time PCR (RT-PCR) in an enriched population of neurons. Region-level expression of these transcripts was measured in postmortem schizophrenia (n = 25) and controls (n = 25). The relationship between selected EAAT polymorphisms and EAAT splice variant expression was also explored. Anterior cingulate cortex pyramidal cell expression of EAAT2b mRNA was increased (P < 0.001; 67%) in schizophrenia subjects compared with controls. There was no significant change in other EAAT variants. EAAT2 exon9skipping mRNA was increased (P < 0.05; 38%) at region level in the anterior cingulate cortex with no significant change in other EAAT variants at region level. EAAT2 single-nucleotide polymorphisms were significantly associated with changes in EAAT2 isoform expression. Haloperidol decanoate-treated animals, acting as controls for possible antipsychotic effects, did not have significantly altered neuronal EAAT2b mRNA levels. The novel finding that EAAT2b levels are increased in populations of anterior cingulate cortex pyramidal cells further demonstrates a role for neuronal glutamate transporter splice variant expression in schizophrenia.


Subject(s)
Excitatory Amino Acid Transporter 1/genetics , Glutamate Plasma Membrane Transport Proteins/genetics , Gyrus Cinguli/metabolism , Protein Isoforms/genetics , Pyramidal Cells/metabolism , RNA, Messenger/metabolism , Schizophrenia/genetics , Aged , Aged, 80 and over , Animals , Antipsychotic Agents/pharmacology , Brain/drug effects , Brain/metabolism , Case-Control Studies , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2/drug effects , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 2/metabolism , Female , Glutamate Plasma Membrane Transport Proteins/metabolism , Gyrus Cinguli/drug effects , Haloperidol/pharmacology , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Protein Isoforms/metabolism , Pyramidal Cells/drug effects , Rats , Real-Time Polymerase Chain Reaction , Schizophrenia/metabolism
9.
Schizophr Res ; 164(1-3): 92-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25702973

ABSTRACT

The number of Genome Wide Association Studies (GWAS) of schizophrenia is rapidly growing. However, the small effect of individual genes limits the number of reliably implicated genes, which are too few and too diverse to perform reliable pathway analysis; hence the biological roles of the genes implicated in schizophrenia are unclear. To overcome these limitations we combine GWAS with genome-wide expression data from human post-mortem brain samples of schizophrenia patients and controls, taking these steps: 1) Identify 36 GWAS-based genes which are expressed in our dataset. 2) Find a cluster of 19 genes with highly correlated expression. We show that this correlation pattern is robust and statistically significant. 3) GO-enrichment analysis of these 19 genes reveals significant enrichment of ion channels and calcium-related processes. This finding (based on analyzing a small number of coherently expressed genes) is validated and enhanced in two ways: First, the emergence of calcium channels and calcium signaling is corroborated by identifying proteins that interact with those encoded by the cluster of 19. Second, extend the 19 cluster genes into 1028 genes, whose expression is highly correlated with the cluster's average profile. When GO-enrichment analysis is performed on this extended set, many schizophrenia related pathways appear, with calcium-related processes enriched with high statistical significance. Our results give further, expression-based validation to GWAS results, support a central role of calcium-signaling in the pathogenesis of schizophrenia, and point to additional pathways potentially related to the disease.


Subject(s)
Calcium Signaling/genetics , Gene Expression/genetics , Schizophrenia/genetics , Schizophrenia/physiopathology , Aged , Aged, 80 and over , Female , Genetic Association Studies , Humans , Male , Middle Aged , Statistics as Topic
10.
Glia ; 62(11): 1856-77, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25056210

ABSTRACT

Historically, the human brain has been conceptually segregated from the periphery and further dichotomized into gray matter (GM) and white matter (WM) based on the whitish appearance of the exceptionally high lipid content of the myelin sheaths encasing neuronal axons. These simplistic dichotomies were unfortunately extended to conceptually segregate neurons from glia, cognition from behavior, and have been codified in the separation of clinical and scientific fields into medicine, psychiatry, neurology, pathology, etc. The discrete classifications have helped obscure the importance of continual dynamic communication between all brain cell types (neurons, astrocytes, microglia, oligodendrocytes, and precursor (NG2) cells) as well as between brain and periphery through multiple signaling systems. The signaling systems range from neurotransmitters to insulin, angiotensin, and multiple kinases such a glycogen synthase kinase 3 (GSK-3) that together help integrate metabolism, inflammation, and myelination processes and orchestrate the development, plasticity, maintenance, and repair that continually optimize function of neural networks. A more comprehensive, evolution-based, systems biology approach that integrates brain, body, and environmental interactions may ultimately prove more fruitful in elucidating the complexities of human brain function. The historic focus on neurons/GM is rebalanced herein by highlighting the importance of a systems-level understanding of the interdependent age-related shifts in both central and peripheral homeostatic mechanisms that can lead to remarkably prevalent and devastating neuropsychiatric diseases. Herein we highlight the role of glia, especially the most recently evolved oligodendrocytes and the myelin they produce, in achieving and maintaining optimal brain function. The human brain undergoes exceptionally protracted and pervasive myelination (even throughout its GM) and can thus achieve and maintain the rapid conduction and synchronous timing of neural networks on which optimal function depends. The continuum of increasing myelin vulnerability resulting from the human brain's protracted myelination underlies underappreciated communalities between different disease phenotypes ranging from developmental ones such as schizophrenia (SZ) and bipolar disorder (BD) to degenerative ones such as Alzheimer's disease (AD). These shared vulnerabilities also expose significant yet underexplored opportunities for novel treatment and prevention approaches that have the potential to considerably reduce the tremendous burden of neuropsychiatric disease.


Subject(s)
Brain , Mental Disorders/pathology , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Aging , Brain/metabolism , Brain/pathology , Brain/physiopathology , Humans , Neurotransmitter Agents/metabolism
11.
Transl Psychiatry ; 2: e137, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22781170

ABSTRACT

Glutamatergic signaling through N-methyl-D-aspartate receptors (NMDARs) is required for synaptic plasticity. Disruptions in glutamatergic signaling are proposed to contribute to the behavioral and cognitive deficits observed in schizophrenia (SZ). One possible source of compromised glutamatergic function in SZ is decreased surface expression of GluN2B-containing NMDARs. STEP(61) is a brain-enriched protein tyrosine phosphatase that dephosphorylates a regulatory tyrosine on GluN2B, thereby promoting its internalization. Here, we report that STEP(61) levels are significantly higher in the postmortem anterior cingulate cortex and dorsolateral prefrontal cortex of SZ patients, as well as in mice treated with the psychotomimetics MK-801 and phencyclidine (PCP). Accumulation of STEP(61) after MK-801 treatment is due to a disruption in the ubiquitin proteasome system that normally degrades STEP(61). STEP knockout mice are less sensitive to both the locomotor and cognitive effects of acute and chronic administration of PCP, supporting the functional relevance of increased STEP(61) levels in SZ. In addition, chronic treatment of mice with both typical and atypical antipsychotic medications results in a protein kinase A-mediated phosphorylation and inactivation of STEP(61) and, consequently, increased surface expression of GluN1/GluN2B receptors. Taken together, our findings suggest that STEP(61) accumulation may contribute to the pathophysiology of SZ. Moreover, we show a mechanistic link between neuroleptic treatment, STEP(61) inactivation and increased surface expression of NMDARs, consistent with the glutamate hypothesis of SZ.


Subject(s)
Antipsychotic Agents/pharmacology , Gyrus Cinguli/metabolism , Phosphorylation/drug effects , Prefrontal Cortex/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Schizophrenia/metabolism , Analysis of Variance , Animals , Antipsychotic Agents/therapeutic use , Dizocilpine Maleate/pharmacology , Gyrus Cinguli/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phencyclidine/pharmacology , Prefrontal Cortex/drug effects , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Schizophrenia/drug therapy , Schizophrenia/etiology
12.
Curr Alzheimer Res ; 8(3): 303-12, 2011 May.
Article in English | MEDLINE | ID: mdl-21244352

ABSTRACT

UNLABELLED: Elevated serum total cholesterol (TC) has been considered a risk factor for Alzheimer's disease (AD), but conflicting results have confused understanding of the relationships of serum lipids to the presence of AD in the elderly. METHODS: To clarify these issues, we evaluated correlations of admission TC, low-density (LDL) and high-density (HDL)cholesterol directly with the densities of Alzheimer hallmarks--neuritic plaques (NP) and neurofibrillary tangles (NFT)--in nursing home residents (n=281). RESULTS: Significant positive associations of TC and LDL with NP densities were found in both the neocortex (TC: r=0.151, p=0.013 and LDL: r=0.190, p=0.005) and the hippocampal/entorhinal (allocortical)region (TC: r=0.182, p=0.002 and LDL: r=0.203, p=0.003). Associations of HDL with NP were less strong but also significant.In contrast, after adjustment for confounders, no correlations of NFT with any lipid were significant.When subjects with any non-AD neuropathology (largely vascular) were excluded, the TC-plaque and LDL-plaque associations for the remaining "Pure AD" subgroup were consistently stronger than for the full sample. The TC- and LDL-plaque correlations were also stronger for the subgroup of 87 subjects with an APOE ε4 allele. CONCLUSIONS: The findings indicate that serum TC and LDL levels clearly relate to densities of NP, but not to densities of NFT. The stronger associations found in the subgroup that excluded all subjects with non-AD neuropathology suggest that cerebrovascular involvement does not explain these lipid-plaque relationships. Since the associations of TC/LDL with NP were particularly stronger in ε4 carriers, varying prevalence of this allele may explain some discrepancies among prior studies.


Subject(s)
Alzheimer Disease/blood , Alzheimer Disease/pathology , Apolipoprotein E4/genetics , Cholesterol/blood , Neurofibrillary Tangles/pathology , Plaque, Amyloid/pathology , Aged, 80 and over , Alzheimer Disease/genetics , Cholesterol, LDL/blood , Female , Humans , Male
13.
Eur J Neurol ; 18(3): 410-24, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20695885

ABSTRACT

BACKGROUND: Alzheimer's disease and Schizophrenia are two common diseases of the brain with significant differences in neuropathology, etiology and symptoms. This dissimilarity in the two diseases makes a comparison of the two ideal for detecting molecular substrates that are common to brain disorders in general. METHODS: In this study, we compared gene expression profiles across multiple brain areas, taken postmortem from patients with well-characterized Alzheimer's disease and Schizophrenia, and from cognitively normal control group with no neuro- or psychopathology. RESULTS: Although the totality of gene expression changes in the two diseases is dissimilar, a subset of genes appears to play a role in both diseases in specific brain regions. We find at Brodmann area 22, the superior temporal gyrus, a statistically significant number of genes with apparently disregulated expression in both diseases. Furthermore, we found genes that differentiate the two diseases from the control across multiple brain regions, and note that these genes were usually down-regulated. Brodmann area 8, part of the superior frontal cortex, is relatively abundant with them. CONCLUSION: We show overwhelming statistical evidence for Alzheimer's and Schizophrenia sharing a specific molecular background at the superior temporal gyrus. We suggest that impairment of the regulation of autophagy pathway is shared, in BA 22, by the two diseases.


Subject(s)
Alzheimer Disease/genetics , Gene Expression Profiling , Schizophrenia/genetics , Temporal Lobe , Aged , Aged, 80 and over , Female , Humans , Male , Oligonucleotide Array Sequence Analysis
14.
Schizophr Res ; 119(1-3): 198-209, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20347576

ABSTRACT

Dysregulated glutamate neurotransmission has been implicated in the pathophysiology of schizophrenia. In particular, hypofunction of the NMDA glutamate receptor has been proposed to play an important role in mediating cognitive deficits in patients. The two NMDA receptor subunits, NR2A and NR2B, are distinctly regulated during development and are associated with different intracellular pathways and functions, which suggest that these receptors play separate roles in the control of higher cognitive functions such as learning and memory. Trafficking of the NR2B subunit-containing receptor is regulated by a microtubule-associated trafficking complex consisting of the KIF17, APBA1, CASK, and mLin7 proteins. Several studies have demonstrated an integrated functional regulation of this trafficking complex with NR2B receptor subunit expression, which in turn has been linked to higher cognitive functions. In the present work, we investigated whether expression of this NR2B-associated trafficking complex might be abnormal in schizophrenia. We analyzed the expression of KIF17, APBA1, CASK, mLin7A and mLin7C in postmortem brain from patients with schizophrenia a comparison group. Analysis of transcripts for all of these proteins revealed particularly prominent expression in cortical layer III and layer IV, which overlapped with NR2B but not NR2A transcripts. We found altered expression of transcripts for the CASK, ABPA1, and mLin7 molecules and the CASK, mLin7 proteins, suggesting that NR2B-containing NMDA receptor transport could be selectively compromised in schizophrenia, and that these changes likely involve altered NR2B function in a subset of cortical neurons.


Subject(s)
Frontal Lobe/pathology , Prefrontal Cortex/pathology , Receptors, N-Methyl-D-Aspartate/genetics , Schizophrenia/genetics , Aged , Aged, 80 and over , Animals , Antipsychotic Agents/pharmacology , Cadherins/genetics , Carrier Proteins/genetics , Cerebral Cortex/pathology , Female , Gene Expression/drug effects , Gene Expression/genetics , Guanylate Kinases/genetics , Haloperidol/pharmacology , Humans , Injections, Intramuscular , Kinesins/genetics , Male , Middle Aged , Nerve Tissue Proteins/genetics , Neurons/pathology , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Schizophrenia/pathology , Transcription, Genetic/genetics
15.
Neurology ; 72(20): 1720-6, 2009 May 19.
Article in English | MEDLINE | ID: mdl-19228583

ABSTRACT

OBJECTIVE: To test the hypothesis that use of antihypertensive medication is associated with lower Alzheimer disease (AD) neuropathology. METHODS: This was a postmortem study of 291 brains limited to those with normal neuropathology or with uncomplicated AD neuropathology (i.e., without other dementia-associated neuropathology) in persons with or without hypertension (HTN) who were and were not treated with antihypertensive medications. Neuritic plaque (NP) and neurofibrillary tangle (NFT) densities, quantified in selected brain regions according to the Consortium to Establish a Registry for Alzheimer's Disease (CERAD) neuropathologic criteria, with additional cortical NP counts, yielded 24 neuropathologic regional measures or summaries. Medicated hypertension (HTN-med; n = 77), nonmedicated HTN (HTN-nomed; n = 42), and non-HTN (no-HTN; n = 172) groups were compared by analyses of variance. RESULTS: The HTN-med group had significantly less neuropathology than the no-HTN group. The no-HTN group averaged over 50% higher mean NP and NFT ratings, and double the mean NP count, of the HTN-med group. The HTN-nomed group had significantly more neuropathology than the HTN-med group, but not significantly less than the no-HTN group. CONCLUSIONS: There was substantially less Alzheimer disease (AD) neuropathology in the medicated hypertension group than the nonhypertensive group, which may reflect a salutary effect of antihypertensive medication against AD-associated neuropathology.


Subject(s)
Alzheimer Disease/pathology , Antihypertensive Agents/therapeutic use , Dementia/pathology , Hypertension , Aged , Aged, 80 and over , Blood Pressure/physiology , Brain/anatomy & histology , Brain/pathology , Female , Humans , Hypertension/drug therapy , Hypertension/physiopathology , Neurofibrillary Tangles/pathology , Plaque, Amyloid/pathology , Risk Factors
16.
Neurology ; 71(10): 750-7, 2008 Sep 02.
Article in English | MEDLINE | ID: mdl-18765651

ABSTRACT

OBJECTIVE: To examine the association between treatment for diabetes and Alzheimer disease (AD) neuropathology. METHODS: This postmortem study matched 124 subjects with diabetes to 124 without diabetes from the Mount Sinai School of Medicine Brain Bank, on age (mean = 81.2 + 9.3), sex (57.3% F), and severity of dementia (Clinical Dementia Rating [CDR] 2.4 + 1.7). Densities of neuritic plaques (NPs) and of neurofibrillary tangles (NFTs) were assessed in several neocortical regions and in the hippocampus, entorhinal cortex, and amygdala. Diabetic subjects were classified according to their recorded lifetime antidiabetic medications: none (n = 29), insulin only (n = 49), diabetes medications other than insulin only (n = 28), or concomitant use of both insulin and any oral antidiabetic medications (n = 18). For each dependent variable, analysis of covariance controlling for age at death, sex, and CDR distinguished among the nondiabetic patients and four diabetic subgroups. RESULTS: There were differences among the five groups for NP ratings in the entorhinal cortex (p = 0.003), amygdala (p = 0.009), and overall NP (p = 0.014) as well as counts of NPs in all regions examined (p values ranging from 0.009 to 0.04). NP ratings in the hippocampus (p = 0.057) and the combined neocortical measure (p = 0.052) approached significance. In each analysis, the concomitant medication group had significantly fewer NPs (approximately 20%) than any of the other groups, which were relatively similar. No significant NFT differences were found. CONCLUSION: The results of this study suggest that the combination of insulin with other diabetes medication is associated with substantially lower neuritic plaque density consistent with the effects of both on the neurobiology of insulin.


Subject(s)
Brain/pathology , Diabetes Mellitus/drug therapy , Diabetes Mellitus/pathology , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Neurofibrillary Tangles/pathology , Plaque, Amyloid/pathology , Aged , Aged, 80 and over , Analysis of Variance , Brain/drug effects , Case-Control Studies , Drug Therapy, Combination , Female , Humans , Male , Neurofibrillary Tangles/drug effects , Plaque, Amyloid/drug effects , Postmortem Changes , Psychiatric Status Rating Scales , Severity of Illness Index
17.
Mol Psychiatry ; 13(11): 1001-10, 2008 Nov.
Article in English | MEDLINE | ID: mdl-17848916

ABSTRACT

Suicide is a major public health problem with approximately 1 million victims each year worldwide. Up to 90% of adults who commit suicide have at least one psychiatric diagnosis such as major depression, bipolar disorder (BPD), schizophrenia (SZ), substance abuse or dependence. A question that has remained unanswered is whether the biological substrates of suicide are distinct from those of the psychiatric disorders in which it occurs. The serotonin 2C receptor (5-HT 2C R) has been implicated in depression and suicide. We, therefore, compared the frequencies of its mRNA editing variants in postmortem prefrontal cortical specimens from subjects who committed suicide or who died from other causes. All suicides occurred in the context of either SZ or BPD. The non-suicide cases included subjects with either SZ or BPD as well as subjects with no psychiatric diagnosis. We identified 5-HT 2CR mRNA editing variations that were associated with suicide but not with the comorbid psychiatric diagnoses, and were not influenced by demographic characteristics (age and sex) and alcohol or drug use. These variations consisted of a significant increase in the pool of mRNA variants (ACD and ABCD) that encode one of the most prevalent and highly edited isoforms of 5-HT 2C R, that is, VSV (Val156-Ser158-Val160). Because the VSV isoform of 5-HT 2C R exhibits low functional activity, an increase in its expression frequency may significantly influence the serotonergic regulation of the brain. Thus, at least in patients with SZ or BPD, overexpression of the VSV isoform in the prefrontal cortex may represent an additional risk factor for suicidal behavior.


Subject(s)
RNA Editing/genetics , RNA, Messenger/metabolism , Receptor, Serotonin, 5-HT2C/genetics , Risk Factors , Suicide/psychology , Adult , Bipolar Disorder/genetics , Bipolar Disorder/pathology , Bipolar Disorder/psychology , Female , Gene Expression , Humans , Male , Middle Aged , Models, Molecular , Prefrontal Cortex/metabolism , Receptor, Serotonin, 5-HT2C/metabolism , Schizophrenia/genetics , Schizophrenia/pathology , Schizophrenic Psychology , Serine/genetics , Valine/genetics
18.
Neurobiol Aging ; 28(11): 1700-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-16914231

ABSTRACT

The opioid peptides dynorphins may be involved in pathogenesis of Alzheimer disease (AD) by inducing neurodegeneration or cognitive impairment. To test this hypothesis, the dynorphin system was analyzed in postmortem samples from AD and control subjects, and subjects with Parkinson or cerebro-vascular diseases for comparison. Dynorphin A, dynorphin B and related neuropeptide nociceptin were determined in the Brodmann area 7 by radioimmunoassay. The precursor protein prodynorphin, processing convertase PC2 and the neuroendocrine pro7B2 and 7B2 proteins required for PC2 maturation were analyzed by Western blot. AD subjects displayed robustly elevated levels of dynorphin A and no differences in dynorphin B and nociceptin compared to controls. Subjects with Parkinson or cerebro-vascular diseases did not differ from controls with respect to any of the three peptides. PC2 levels were also increased, whereas, those of prodynorphin and pro7B2/7B2 were not changed in AD. Dynorphin A levels correlated with the neuritic plaque density. These results along with the known non-opioid ability of dynorphin A to induce neurodegeneration suggest a role for this neuropeptide in AD neuropathology.


Subject(s)
Alzheimer Disease/metabolism , Dynorphins/biosynthesis , Endorphins/biosynthesis , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Brain/metabolism , Brain/pathology , Dynorphins/genetics , Endorphins/genetics , Female , Humans , Male , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Opioid Peptides/biosynthesis , Opioid Peptides/genetics , Up-Regulation/physiology , Nociceptin
19.
Neurobiol Aging ; 28(1): 1-7, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16343696

ABSTRACT

The degree to which neurofibrillary tangles (NFT), the hallmark lesions of Alzheimer disease (AD), contribute to the development of the cognitive symptoms of AD has been debated. NFTs are comprised of abnormally phosphorylated and conformationally altered tau proteins. Conformational changes in tau have been proposed to be among the earliest neurobiological changes in AD. This study examined whether conformational changes detected by antibodies MC1 and TG3 represent early abnormalities in the disease process by assessing their presence at different stages of dementia in multiple brain regions. Postmortem specimens from several neocortical regions were examined for conformational changes in tau by ELISA in subjects [n=81] who died at different stages of cognitive impairment. Concentrations of conformationally altered tau increased with increasing dementia severity and the levels of MC1 immunoreactivity increased in the frontal cortex of mildly demented subjects before the appearance of NFT bearing neurons, suggesting that conformational alterations in tau occur early in the course of AD and its cognitive symptoms and may precede histologically identified NFTs.


Subject(s)
Aging/metabolism , Aging/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Brain Chemistry , Brain/pathology , tau Proteins/chemistry , Aged , Aged, 80 and over , Disease Progression , Female , Humans , In Vitro Techniques , Male , Middle Aged , tau Proteins/classification
20.
Neurology ; 67(1): 69-75, 2006 Jul 11.
Article in English | MEDLINE | ID: mdl-16832080

ABSTRACT

OBJECTIVE: To determine whether changes in brain biometals in Alzheimer disease (AD) and in normal brain tissue are tandemly associated with amyloid beta-peptide (Abeta) burden and dementia severity. METHODS: The authors measured zinc, copper, iron, manganese, and aluminum and Abeta levels in postmortem neocortical tissue from patients with AD (n = 10), normal age-matched control subjects (n = 14), patients with schizophrenia (n = 26), and patients with schizophrenia with amyloid (n = 8). Severity of cognitive impairment was assessed with the Clinical Dementia Rating Scale (CDR). RESULTS: There was a significant, more than twofold, increase of tissue zinc in the AD-affected cortex compared with the other groups. Zinc levels increased with tissue amyloid levels. Zinc levels were significantly elevated in the most severely demented cases (CDR 4 to 5) and in cases that had an amyloid burden greater than 8 plaques/mm(2). Levels of other metals did not differ between groups. CONCLUSIONS: Brain zinc accumulation is a prominent feature of advanced Alzheimer disease (AD) and is biochemically linked to brain amyloid beta-peptide accumulation and dementia severity in AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Cerebral Cortex/metabolism , Zinc/metabolism , Age Factors , Aged , Aged, 80 and over , Alzheimer Disease/physiopathology , Amyloid/metabolism , Amyloid beta-Peptides/metabolism , Analysis of Variance , Case-Control Studies , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Immunohistochemistry/methods , Male , Metals, Heavy/metabolism , Neurofibrillary Tangles/metabolism , Postmortem Changes , Schizophrenia/metabolism , Schizophrenia/pathology , Statistics as Topic
SELECTION OF CITATIONS
SEARCH DETAIL
...