Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Brain ; 144(10): 3142-3158, 2021 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-34086871

RESUMO

Traumatic brain injury (TBI) leads to major brain anatomopathological damages underlined by neuroinflammation, oxidative stress and progressive neurodegeneration, ultimately leading to motor and cognitive deterioration. The multiple pathological events resulting from TBI can be addressed not by a single therapeutic approach, but rather by a synergistic biotherapy capable of activating a complementary set of signalling pathways and providing synergistic neuroprotective, anti-inflammatory, antioxidative, and neurorestorative activities. Human platelet lysate might fulfil these requirements as it is composed of a plethora of biomolecules readily accessible as a TBI biotherapy. In the present study, we tested the therapeutic potential of human platelet lysate using in vitro and in vivo models of TBI. We first prepared and characterized platelet lysate from clinical-grade human platelet concentrates. Platelets were pelletized, lysed by three freeze-thaw cycles, and centrifuged. The supernatant was purified by 56°C 30 min heat treatment and spun to obtain the heat-treated platelet pellet lysate that was characterized by ELISA and proteomic analyses. Two mouse models were used to investigate platelet lysate neuroprotective potential. The injury was induced by an in-house manual controlled scratching of the animals' cortex or by controlled cortical impact injury. The platelet lysate treatment was performed by topical application of 60 µl in the lesioned area, followed by daily 60 µl intranasal administration from Day 1 to 6 post-injury. Platelet lysate proteomics identified over 1000 proteins including growth factors, neurotrophins, and antioxidants. ELISA detected several neurotrophic and angiogenic factors at ∼1-50 ng/ml levels. We demonstrate, using two mouse models of TBI, that topical application and intranasal platelet lysate consistently improved mouse motor function in the beam and rotarod tests, mitigated cortical neuroinflammation, and oxidative stress in the injury area, as revealed by downregulation of pro-inflammatory genes and the reduction in reactive oxygen species levels. Moreover, platelet lysate treatment reduced the loss of cortical synaptic proteins. Unbiased proteomic analyses revealed that heat-treated platelet pellet lysate reversed several pathways promoted by both controlled cortical impact and cortical brain scratch and related to transport, postsynaptic density, mitochondria or lipid metabolism. The present data strongly support, for the first time, that human platelet lysate is a reliable and effective therapeutic source of neurorestorative factors. Therefore, brain administration of platelet lysate is a therapeutical strategy that deserves serious and urgent consideration for universal brain trauma treatment.


Assuntos
Terapia Biológica/métodos , Plaquetas/metabolismo , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/terapia , Administração Intranasal , Animais , Lesões Encefálicas Traumáticas/patologia , Linhagem Celular Tumoral , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
2.
FEBS J ; 288(5): 1457-1461, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32805742

RESUMO

In neurodegenerative diseases, a select set of neuron population displays early vulnerability and undergoes progressive degeneration. The heterogeneity of the cerebral cortex and the heterogeneity of patient populations diagnosed with the same disease offer many challenges for developing effective and long-term treatment options. Currently, patients who are considered to have a 'rare' disease are left with no hopes for cure, and many of the neurodegenerative diseases progress fast without any effective solutions. However, as our understanding of disease mechanisms evolve, we begin to realize that the boundaries between diseases are not as sharp as once believed. There are many patients who develop disease due to common underlying causes and mechanisms. As we move forward with drug discovery effort, it becomes obvious that we will have to shift our focus from finding a cure for a disease, to finding solutions to the disease-causing cellular mechanisms so that patients can be treated by mechanism-based strategies. This paradigm shift will lay the foundation for personalized medicine approaches for neurodegenerative disease patients and patients diagnosed with a rare disease.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Esclerose Lateral Amiotrófica/tratamento farmacológico , Drogas em Investigação/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Medicina de Precisão/tendências , Doenças Raras/tratamento farmacológico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Autofagia/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Reposicionamento de Medicamentos/métodos , Acessibilidade aos Serviços de Saúde/organização & administração , Humanos , Inflamação , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Doenças Raras/metabolismo , Doenças Raras/patologia , Resposta a Proteínas não Dobradas/efeitos dos fármacos
3.
J Prev Alzheimers Dis ; 6(4): 274-282, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31686100

RESUMO

BACKGROUND: There is a need to more fully characterize financial capacity losses in the preclinical and prodromal stages of Alzheimer's disease (AD) and their pathological substrates. OBJECTIVES: To test the association between financial skills and cortical ß-amyloid deposition in aging and subjects at risk for AD. DESIGN: Cross-sectional analyses of data from the Alzheimer's Disease Neuroimaging Initiative (ADNI-3) study conducted across 50 plus sites in the US and Canada. SETTING: Multicenter biomarker study. PARTICIPANTS: 243 subjects (144 cognitively normal, 79 mild cognitive impairment [MCI], 20 mild AD). MEASUREMENTS: 18F-Florbetapir brain PET scans to measure global cortical ß-amyloid deposition (SUVr) and the Financial Capacity Instrument Short Form (FCI-SF) to evaluate an individual's financial skills in monetary calculation, financial concepts, checkbook/register usage, and bank statement usage. There are five sub scores and a total score (range of 0-74) with higher scores indicating better financial skill. RESULTS: FCI-SF total score was significantly worse in MCI [Cohen's d= 0.9 (95%CI: 0.6-1.2)] and AD subjects [Cohen's d=3.1(CI: 2.5-3.7)] compared to normals. Domain scores and completion times also showed significant difference. Across all subjects, higher cortical ß-amyloid SUVr was significantly associated with worse FCI-SF total score after co-varying for age, education, and cognitive score [Cohen's f2=0.751(CI: 0.5-1.1)]. In cognitively normal subjects, after covarying for age, gender, and education, higher ß -amyloid PET SUVr was associated with longer task completion time [Cohen's f2=0.198(CI: 0.06-0.37)]. CONCLUSION: Using a multicenter study sample, we document that financial capacity is impaired in the prodromal and mild stages of AD and that such impairments are, in part, associated with the extent of cortical ß-amyloid deposition. In normal aging, ß-amyloid deposition is associated with slowing of financial tasks. These data confirm and extend prior research highlighting the utility of financial capacity assessments in at risk samples.


Assuntos
Envelhecimento/psicologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/diagnóstico por imagem , Disfunção Cognitiva/psicologia , Demência/psicologia , Administração Financeira , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/metabolismo , Compostos de Anilina , Encéfalo/metabolismo , Canadá , Estudos de Casos e Controles , Córtex Cerebral/diagnóstico por imagem , Córtex Cerebral/metabolismo , Disfunção Cognitiva/diagnóstico por imagem , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/fisiopatologia , Estudos Transversais , Demência/diagnóstico por imagem , Demência/metabolismo , Demência/fisiopatologia , Etilenoglicóis , Feminino , Humanos , Masculino , Tomografia por Emissão de Pósitrons , Sintomas Prodrômicos , Índice de Gravidade de Doença , Estados Unidos
4.
Acta Neuropathol Commun ; 7(1): 159, 2019 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-31640778

RESUMO

Tau aggregation is a hallmark feature in a subset of patients with frontotemporal dementia (FTD). Early and selective loss of von Economo neurons (VENs) and fork cells within the frontoinsular (FI) and anterior cingulate cortices (ACC) is observed in patients with sporadic behavioral variant FTD (bvFTD) due to frontotemporal lobar degeneration (FTLD), including FTLD with tau inclusions (FTLD-tau). Recently, we further showed that these specialized neurons show preferential aggregation of TDP-43 in FTLD-TDP. Whether VENs and fork cells are prone to tau accumulation in FTLD-tau remains unclear, and no previous studies of these neurons have focused on patients with pathogenic variants in the gene encoding microtubule-associated protein tau (FTLD-tau/MAPT). Here, we examined regional profiles of tau aggregation and neurodegeneration in 40 brain regions in 8 patients with FTLD-tau/MAPT and 7 with Pick's disease (PiD), a sporadic form of FTLD-tau that often presents with bvFTD. We further qualitatively assessed the cellular patterns of frontoinsular tau aggregation in FTLD-tau/MAPT using antibodies specific for tau hyperphosphorylation, acetylation, or conformational change. ACC and mid-insula were among the regions most affected by neurodegeneration and tau aggregation in FTLD-tau/MAPT and PiD. In these two forms of FTLD-tau, severity of regional neurodegeneration and tau protein aggregation were highly correlated across regions. In FTLD-tau/MAPT, VENs and fork cells showed disproportionate tau protein aggregation in patients with V337 M, A152T, and IVS10 + 16 variants, but not in patients with the P301L variant. As seen in FTLD-TDP, our data suggest that VENs and fork cells represent preferentially vulnerable neuron types in most, but not all of the MAPT variants we studied.


Assuntos
Córtex Cerebral/patologia , Degeneração Lobar Frontotemporal/patologia , Giro do Cíngulo/patologia , Neurônios/patologia , Proteínas tau/metabolismo , Idoso , Córtex Cerebral/metabolismo , Feminino , Degeneração Lobar Frontotemporal/metabolismo , Giro do Cíngulo/metabolismo , Humanos , Corpos de Inclusão/metabolismo , Corpos de Inclusão/patologia , Masculino , Pessoa de Meia-Idade , Neurônios/metabolismo , Doença de Pick/metabolismo , Doença de Pick/patologia
5.
Sci Rep ; 8(1): 14610, 2018 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-30279531

RESUMO

Whether hedonism or eudaimonia are two distinguishable forms of well-being is a topic of ongoing debate. To shed light on the relation between the two, large-scale available molecular genetic data were leveraged to gain more insight into the genetic architecture of the overlap between hedonic and eudaimonic well-being. Hence, we conducted the first genome-wide association studies (GWAS) of eudaimonic well-being (N = ~108 K) and linked it to a GWAS of hedonic well-being (N = ~222 K). We identified the first two genome-wide significant independent loci for eudaimonic well-being and six independent loci for hedonic well-being. Joint analyses revealed a moderate phenotypic correlation (r = 0.53) and a high genetic correlation (rg = 0.78) between eudaimonic and hedonic well-being. This indicates that the genetic etiology of hedonic and eudaimonic well-being is substantially shared, with divergent (environmental) factors contributing to their phenotypic divergence. Loci regulating expression showed significant enrichment in the brain cortex, brain cerebellum, frontal cortex, as well as the cerebellar hemisphere for eudaimonic well-being. No significant enrichment for hedonic well-being is observed, although brain tissues were top ranked. Genetic correlations patterns with a range of positive and negative related phenotypes were largely similar for hedonic -and eudaimonic well-being. Our results reveal a large overlap between the genes that influence hedonism and the genes that influence eudaimonia.


Assuntos
Loci Gênicos , Genoma Humano , Filosofia , Qualidade de Vida , Valor da Vida , Adulto , Idoso , Cerebelo/anatomia & histologia , Cerebelo/metabolismo , Córtex Cerebral/anatomia & histologia , Córtex Cerebral/metabolismo , Feminino , Lobo Frontal/anatomia & histologia , Lobo Frontal/metabolismo , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Estudos Prospectivos , Inquéritos e Questionários , Reino Unido
6.
Neurochem Int ; 121: 50-58, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30339985

RESUMO

Glutamate excitotoxicity plays a crucial role in the pathogenesis behind the development and progression of several neurodegenerative diseases. The study aimed to investigate the neuroprotective activity of Gallic acid (GA) against glutamate-induced neurotoxicity in primary rat cortex neurons (RCN). Treated the RCNs with GA 25 & 50 µg/ml for 2 h and later treated the cells with 100 µM glutamate (GLU) and incubated for 24 h at 37 °C. The results demonstrated that, the GA improved the antioxidant profile in the cortex neurons and inhibited the production of the proinflammatory cytokine. GA also maintained the Ca2+ homeostasis, IGF-1 expression, and protected the neurons from glutamate-induced neuronal toxicity. The neuroprotective activity of GA has further confirmed from the results of N-acetylaspartate and expression of microtubule-associated protein-2 expression. The reports suggest that, GA is significantly attenuated the glutamate-induced neurotoxicity and protected neurons from various chemical events that are involved in the pathogenesis of neurotoxicity.


Assuntos
Antioxidantes/farmacologia , Córtex Cerebral/efeitos dos fármacos , Ácido Gálico/farmacologia , Ácido Glutâmico/toxicidade , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/imunologia , Células Cultivadas , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Relação Dose-Resposta a Droga , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Ratos , Ratos Endogâmicos F344
7.
Neurotoxicology ; 65: 28-37, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29378300

RESUMO

Nerve agents (NAs) are potent organophosphorus (OP) compounds with applications in chemical warfare. OP compounds act by inhibiting acetylcholinesterase (AChE). Soman (O-pinacolyl methylphosphonofluoridate) is one of the most potent NAs. It is well known that small doses of NAs can be lethal, and that even non-lethal exposure leads to long-term mental debilitation/neurological damage. However, the neuropathology following exposure to sub-lethal nerve agents is not well understood. In this study, we examined changes in tissue oxygenation (pO2) in the cortex and hippocampus after a sub-lethal dose of soman [80-90 µg/kg; subcutaneous]. pO2 changes can provide information regarding oxygen delivery and utilization and may be indicative of a disruption in cerebral blood flow and/or metabolism. Changes in oxygenation were measured with chronically implanted oxygen sensors in awake and freely moving rats. Measurements were taken before, during, and after soman-induced convulsive seizures. Soman exposure resulted in an immediate increase in pO2 in the cortex, followed by an even greater increase that precedes the onset of soman-induced convulsive seizures. The rise in hippocampus pO2 was delayed relative to the cortex, although the general pattern of brain oxygenation between these two regions was similar. After convulsive seizures began, pO2 levels declined but usually remained hyperoxygenated. Following the decline in pO2, low frequency cycles of large amplitude changes were observed in both the cortex and hippocampus. This pattern is consistent with recurring seizures. Measuring real-time changes in brain pO2 provides new information on the physiological status of the brain following soman exposure. These results highlight that the measurement of brain oxygenation could provide a sensitive marker of nerve agent exposure and serve as a biomarker for treatment studies.


Assuntos
Córtex Cerebral/metabolismo , Hipocampo/metabolismo , Oxigênio/metabolismo , Soman/toxicidade , Animais , Implantes Experimentais , Masculino , Monitorização Ambulatorial , Ratos , Convulsões/induzido quimicamente , Convulsões/metabolismo , Fatores de Tempo
8.
J Neurosci Res ; 96(4): 487-500, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28463430

RESUMO

Traumatic brain injuries (TBIs) pose a massive burden of disease and continue to be a leading cause of morbidity and mortality throughout the world. A major obstacle in developing effective treatments is the lack of comprehensive understanding of the underlying mechanisms that mediate tissue damage and recovery after TBI. As such, our work aims to highlight the development of a novel experimental platform capable of fully characterizing the underlying pathobiology that unfolds after TBI. This platform encompasses an empirically optimized multiplex immunohistochemistry staining and imaging system customized to screen for a myriad of biomarkers required to comprehensively evaluate the extent of neuroinflammation, neural tissue damage, and repair in response to TBI. Herein, we demonstrate that our multiplex biomarker screening platform is capable of evaluating changes in both the topographical location and functional states of resident and infiltrating cell types that play a role in neuropathology after controlled cortical impact injury to the brain in male Sprague-Dawley rats. Our results demonstrate that our multiplex biomarker screening platform lays the groundwork for the comprehensive characterization of changes that occur within the brain after TBI. Such work may ultimately lead to the understanding of the governing pathobiology of TBI, thereby fostering the development of novel therapeutic interventions tailored to produce optimal tissue protection, repair, and/or regeneration with minimal side effects, and may ultimately find utility in a wide variety of other neurological injuries, diseases, and disorders that share components of TBI pathobiology.


Assuntos
Lesões Encefálicas Traumáticas/fisiopatologia , Imuno-Histoquímica/métodos , Neuroimagem/métodos , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Biomarcadores/metabolismo , Encéfalo/fisiopatologia , Lesões Encefálicas Traumáticas/metabolismo , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Masculino , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Ratos Sprague-Dawley , Doenças da Língua/metabolismo , Doenças da Língua/patologia
9.
Cereb Cortex ; 28(1): 131-144, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27913432

RESUMO

The human anterior cingulate and frontoinsular cortices are distinguished by 2 unique Layer 5 neuronal morphotypes, the von Economo neurons (VENs) and fork cells, whose biological identity remains mysterious. Insights could impact research on diverse neuropsychiatric diseases to which these cells have been linked. Here, we leveraged the Allen Brain Atlas to evaluate mRNA expression of 176 neurotransmitter-related genes and identified vesicular monoamine transporter 2 (VMAT2), gamma-aminobutyric acid (GABA) receptor subunit θ (GABRQ), and adrenoreceptor α-1A (ADRA1A) expression in human VENs, fork cells, and a minority of neighboring Layer 5 neurons. We confirmed these results using immunohistochemistry or in situ hybridization. VMAT2 and GABRQ expression was absent in mouse cerebral cortex. Although VMAT2 is known to package monoamines into synaptic vesicles, in VENs and fork cells its expression occurs in the absence of monoamine-synthesizing enzymes or reuptake transporters. Thus, VENs and fork cells may possess a novel, uncharacterized mode of cortical monoaminergic function that distinguishes them from most other mammalian Layer 5 neurons.


Assuntos
Monoaminas Biogênicas/metabolismo , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Adolescente , Adulto , Animais , Atlas como Assunto , Córtex Cerebral/crescimento & desenvolvimento , Criança , Expressão Gênica , Humanos , Lactente , Macaca mulatta , Camundongos , Pessoa de Meia-Idade , Proteínas da Membrana Plasmática de Transporte de Neurotransmissores/metabolismo , RNA Mensageiro/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Receptores de GABA-A/metabolismo , Especificidade da Espécie , Proteínas Vesiculares de Transporte de Monoamina/metabolismo
10.
Cereb Cortex ; 28(5): 1783-1793, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28407057

RESUMO

Thyroid hormones (THs, T4 and the transcriptionally active hormone T3) play an essential role in neurodevelopment; however, the mechanisms underlying T3 brain delivery during mice fetal development are not well known. This work has explored the sources of brain T3 during mice fetal development using biochemical, anatomical, and molecular approaches. The findings revealed that during late gestation, a large amount of fetal brain T4 is of maternal origin. Also, in the developing mouse brain, fetal T3 content is regulated through the conversion of T4 into T3 by type-2 deiodinase (D2) activity, which is present from earlier prenatal stages. Additionally, D2 activity was found to be essential to mediate expression of T3-dependent genes in the cerebral cortex, and also necessary to generate the transient cerebral cortex hyperthyroidism present in mice lacking the TH transporter Monocarboxylate transporter 8. Notably, the gene encoding for D2 (Dio2) was mainly expressed at the blood-cerebrospinal fluid barrier (BCSFB). Overall, these data signify that T4 deiodinated by D2 may be the only source of T3 during neocortical development. We therefore propose that D2 activity at the BCSFB converts the T4 transported across the choroid plexus into T3, thus supplying the brain with active hormone to maintain TH homeostasis.


Assuntos
Córtex Cerebral , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hormônios Tireóideos/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Peso Corporal/fisiologia , Córtex Cerebral/embriologia , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Feminino , Idade Gestacional , Iodeto Peroxidase/deficiência , Iodeto Peroxidase/genética , Isótopos de Iodo/metabolismo , Fígado/embriologia , Fígado/crescimento & desenvolvimento , Fígado/metabolismo , Proteínas de Membrana Transportadoras/deficiência , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transportadores de Ácidos Monocarboxílicos , Gravidez , RNA Mensageiro/metabolismo , Simportadores , Hormônios Tireóideos/farmacologia , Tirosina 3-Mono-Oxigenase/metabolismo , Iodotironina Desiodinase Tipo II
11.
JAMA Neurol ; 74(12): 1492-1501, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29114726

RESUMO

Importance: Visual assessment of amyloid positron emission tomographic (PET) images has been approved by regulatory authorities for clinical use. Several immunoassays have been developed to measure ß-amyloid (Aß) 42 in cerebrospinal fluid (CSF). The agreement between CSF Aß42 measures from different immunoassays and visual PET readings may influence the use of CSF biomarkers and/or amyloid PET assessment in clinical practice and trials. Objective: To determine the concordance between CSF Aß42 levels measured using 5 different immunoassays and visual amyloid PET analysis. Design, Setting, and Participants: The study included 262 patients with mild cognitive impairment or subjective cognitive decline from the Swedish BioFINDER (Biomarkers for Identifying Neurodegenerative Disorders Early and Reliably) cohort (recruited from September 1, 2010, through December 31, 2014) who had undergone flutemetamol F 18 ([18F]flutemetamol)-labeled PET. Levels of CSF Aß42 were analyzed using the classic INNOTEST and the newer modified INNOTEST, fully automated Lumipulse (FL), EUROIMMUN (EI), and Meso Scale Discovery (MSD) assays. Concentrations of CSF Aß were assessed using an antibody-independent mass spectrometry-based reference measurement procedure. Main Outcomes and Measures: The concordance of CSF Aß42 levels and Aß42:Aß40 and Aß42:tau ratios with visual [18F]flutemetamol PET status. Results: Of 262 participants (mean [SD] age, 70.9 [5.5] years), 108 were women (41.2%) and 154 were men (58.8%). The mass spectrometry-derived Aß42 values showed higher correlations with the modified Aß42-INNOTEST (r = 0.97), Aß42-FL (r = 0.93), Aß42-EI (r = 0.93), and Aß42-MSD (r = 0.95) assays compared with the classic Aß42-INNOTEST assay (r = 0.88; P ≤ .01). The signal in the classic Aß42-INNOTEST assay was partly quenched by recombinant Aß1-40 peptide. However, the classic Aß42-INNOTEST assay showed better concordance with visual [18F]flutemetamol PET status (area under the receiver operating characteristic curve [AUC], 0.92) compared with the newer assays (AUCs, 0.87-0.89; P ≤ .01). The accuracies of the newer assays improved significantly when Aß42:Aß40 (AUCs, 0.93-0.95; P ≤ .01), Aß42 to total tau (T-tau) (AUCs, 0.94; P ≤ .05), or Aß42 to phosphorylated tau (P-tau) (AUCs, 0.94-0.95; P ≤ .001) ratios were used. A combination of the Aß42:Aß40 ratio and T-tau or P-tau level did not improve the accuracy compared with the ratio alone. Conclusions and Relevance: Concentrations of CSF Aß42 derived from the new immunoassays (modified INNOTEST, FL, EI, and MSD) may correlate better with the antibody-independent mass spectrometry-based reference measurement procedure and may show improved agreement with visual [18F]flutemetamol PET assessment when using the Aß42:Aß40 or Aß42:tau ratios. These findings suggest the benefit of implementing the CSF Aß42:Aß40 or Aß42:tau ratios as a biomarker of amyloid deposition in clinical practice and trials.


Assuntos
Peptídeos beta-Amiloides/líquido cefalorraquidiano , Amiloide/metabolismo , Córtex Cerebral/diagnóstico por imagem , Disfunção Cognitiva/líquido cefalorraquidiano , Fragmentos de Peptídeos/líquido cefalorraquidiano , Proteínas tau/líquido cefalorraquidiano , Idoso , Compostos de Anilina , Área Sob a Curva , Benzotiazóis , Córtex Cerebral/metabolismo , Disfunção Cognitiva/diagnóstico por imagem , Disfunção Cognitiva/metabolismo , Estudos de Coortes , Feminino , Humanos , Imunoensaio , Masculino , Tomografia por Emissão de Pósitrons , Curva ROC , Compostos Radiofarmacêuticos , Suécia
12.
Am J Geriatr Psychiatry ; 25(4): 342-353, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28162919

RESUMO

OBJECTIVE: To compare regional nicotinic cholinergic receptor binding in older adults with Alzheimer disease (AD) and healthy older adults in vivo and to assess relationships between receptor binding and clinical symptoms. METHODS: Using cross-sectional positron emission tomography (PET) neuroimaging and structured clinical assessment, outpatients with mild to moderate AD (N = 24) and healthy older adults without cognitive complaints (C group; N = 22) were studied. PET imaging of α4ß2* nicotinic cholinergic receptor binding using 2-[18F]fluoro-3-(2(S)azetidinylmethoxy)pyridine (2FA) and clinical measures of global cognition, attention/processing speed, verbal memory, visuospatial memory, and neuropsychiatric symptoms were used. RESULTS: 2FA binding was lower in the AD group compared with the C group in the medial thalamus, medial temporal cortex, anterior cingulate, insula/opercula, inferior caudate, and brainstem (p < 0.05, corrected cluster), but binding was not associated with cognition. The C group had significant inverse correlations between 2FA binding in the thalamus (left: rs = -0.55, p = 0.008; right: rs = -0.50, p = 0.02; N = 22) and hippocampus (left: rs = -0.65, p = 0.001; right: rs = -0.55, p = 0.009; N = 22) and the Trails A score. The AD group had inverse correlation between 2FA binding in anterior cingulate (left: rs = -0.50, p = 0.01; right: rs = -0.50, p = 0.01; N = 24) and Neurobehavioral Rating Scale agitation/disinhibition factor score. CONCLUSION: Cholinergic receptor binding is reduced in specific brain regions in mild to moderate AD and is related to neuropsychiatric symptoms. Among healthy older adults, lower receptor binding may be associated with slower processing speed. Cholinergic receptor binding in vivo may reveal links to other key brain changes associated with aging and AD and may provide a potential molecular treatment target.


Assuntos
Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Tronco Encefálico/metabolismo , Córtex Cerebral/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Receptores Nicotínicos/metabolismo , Tálamo/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/fisiopatologia , Azetidinas , Tronco Encefálico/diagnóstico por imagem , Córtex Cerebral/diagnóstico por imagem , Estudos Transversais , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Piridinas , Tálamo/diagnóstico por imagem
13.
Neurobiol Aging ; 53: 20-26, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28199888

RESUMO

Accumulation of brain iron has been suggested as a biomarker of neurodegeneration. Increased iron has been seen in the cerebral cortex in postmortem studies of neurodegenerative diseases and healthy aging. Until recently, the diminutive thickness of the cortex and its relatively low iron content have hampered in vivo study of cortical iron accumulation. Using phase images of a T2*-weighted sequence at ultrahigh field strength (7 Tesla), we examined the iron content of 22 cortical regions in 70 healthy subjects aged 22-80 years. The cortex was automatically segmented and parcellated, and phase shift was analyzed using an in-house developed method. We found a significant increase in phase shift with age in 20 of 22 cortical regions, concurrent with current understanding of cortical iron accumulation. Our findings suggest that increased cortical iron content can be assessed in healthy aging in vivo. The high spatial resolution and sensitivity to iron of our method make it a potentially useful tool for studying cortical iron accumulation in healthy aging and neurodegenerative diseases.


Assuntos
Envelhecimento/metabolismo , Córtex Cerebral/metabolismo , Imagem de Difusão por Ressonância Magnética/métodos , Ferro/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/metabolismo , Córtex Cerebral/diagnóstico por imagem , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Doenças Neurodegenerativas/diagnóstico , Doenças Neurodegenerativas/diagnóstico por imagem , Sensibilidade e Especificidade , Adulto Jovem
14.
Chem Biol Interact ; 258: 79-88, 2016 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-27566243

RESUMO

BACKGROUND: The rapid economic development in the Arabian Gulf has resulted in lifestyle changes that have increased the prevalence of obesity and type 2 diabetes, with the greatest increases observed in Kuwait. Dyslipidemia and diabetes are risk factors for disruptions in cortical neurotransmitter homeostasis. This study investigated the effect of the antidiabetic medications canagliflozin (CAN) and metformin (MET) on the levels of cortical neurotransmitters in a diabetic rat model. MATERIALS AND METHODS: The rats were assigned to the control (C) group, the diabetic group that did not receive treatment (D) or the diabetic group treated with either CAN (10 mg/kg) or MET (100 mg/kg) for 2 or 4 weeks. Blood and urine glucose levels and cortical acetylcholinesterase (AChE) activity were assayed, and amino acid and monoamine levels were measured using HPLC. RESULTS: The diabetic group exhibited a significant increase in AChE activity and a decrease in monoamine and amino acid neurotransmitter levels. In the CAN group, AChE was significantly lower than that in the D and D + MET groups after 2 weeks of treatment. In addition, a significant increase in some cortical monoamines and amino acids was observed in the D + MET and D + CAN groups compared with the D group. Histopathological analysis revealed the presence of severe focal hemorrhage, neuronal degeneration, and cerebral blood vessel congestion, with gliosis in the cerebrum of rats in the D group. The CAN-treated group exhibited severe cerebral blood vessel congestion after 2 weeks of treatment and focal gliosis in the cerebrum after 4 weeks of treatment. Focal gliosis in the cerebrum of rats in the MET-treated group was observed after 2 and 4 weeks of treatment. CONCLUSIONS: We conclude that the effect of CAN and MET on neurotransmitters is potentially mediated by their antihyperglycemic and antihyperlipidemic effects. In addition, the effects of CAN on neurotransmitters might be associated with its receptor activity, and the effect of MET on neurotransmitters might be associated with cerebral metabolism.


Assuntos
Canagliflozina/farmacologia , Córtex Cerebral/metabolismo , Diabetes Mellitus Experimental/metabolismo , Metformina/farmacologia , Neurotransmissores/metabolismo , Acetilcolinesterase/metabolismo , Aminoácidos/metabolismo , Animais , Monoaminas Biogênicas/metabolismo , Glicemia/metabolismo , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/enzimologia , Córtex Cerebral/patologia , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/urina , Modelos Animais de Doenças , Glicosúria/sangue , Glicosúria/complicações , Glicosúria/tratamento farmacológico , Masculino , Ratos Wistar
15.
PLoS One ; 11(5): e0155303, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27175905

RESUMO

Cerebral metabolic alterations during cardiac arrest, cardiopulmonary resuscitation (CPR) and extracorporeal cardiopulmonary life support (ECLS) are poorly explored. Markers are needed for a more personalized resuscitation and post-resuscitation care. Aim of this study was to investigate early metabolic changes in the hippocampal CA1 region during ventricular fibrillation cardiac arrest (VF-CA) and ECLS versus conventional CPR. Male Sprague-Dawley rats (350g) underwent 8min untreated VF-CA followed by ECLS (n = 8; bloodflow 100ml/kg), mechanical CPR (n = 18; 200/min) until return of spontaneous circulation (ROSC). Shams (n = 2) were included. Glucose, glutamate and lactate/pyruvate ratio were compared between treatment groups and animals with and without ROSC. Ten animals (39%) achieved ROSC (ECLS 5/8 vs. CPR 5/18; OR 4,3;CI:0.7-25;p = 0.189). During VF-CA central nervous glucose decreased (0.32±0.1mmol/l to 0.04±0.01mmol/l; p<0.001) and showed a significant rise (0.53±0.1;p<0.001) after resuscitation. Lactate/pyruvate (L/P) ratio showed a 5fold increase (31 to 164; p<0.001; maximum 8min post ROSC). Glutamate showed a 3.5-fold increase to (2.06±1.5 to 7.12±5.1µmol/L; p<0.001) after CA. All parameters normalized after ROSC with no significant differences between ECLS and CPR. Metabolic changes during ischemia and resuscitation can be displayed by cerebral microdialysis in our VF-CA CPR and ECLS rat model. We found similar microdialysate concentrations and patterns of normalization in both resuscitation methods used. Institutional Protocol Number: GZ0064.11/3b/2011.


Assuntos
Reanimação Cardiopulmonar , Córtex Cerebral/irrigação sanguínea , Oxigenação por Membrana Extracorpórea , Parada Cardíaca/diagnóstico , Microdiálise , Perfusão , Animais , Biomarcadores , Pressão Sanguínea , Região CA1 Hipocampal/metabolismo , Córtex Cerebral/metabolismo , Modelos Animais de Doenças , Oxigenação por Membrana Extracorpórea/métodos , Glucose/metabolismo , Ácido Glutâmico/metabolismo , Parada Cardíaca/terapia , Hemodinâmica , Ácido Láctico/metabolismo , Masculino , Microdiálise/métodos , Oxigênio/sangue , Oxigênio/metabolismo , Ácido Pirúvico/metabolismo , Ratos
16.
J Neurochem ; 137(5): 806-19, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26811128

RESUMO

In this study, we used proton-localized spectroscopy ((1) H-MRS) for the acquisition of the neurochemical profile longitudinally in a novel rat model of human wild-type alpha-synuclein (α-syn) over-expression. Our goal was to find out if the increased α-syn load in this model could be linked to changes in metabolites in the frontal cortex. Animals injected with AAV vectors encoding for human α-syn formed the experimental group, whereas green fluorescent protein expressing animals were used as the vector-treated control group and a third group of uninjected animals were used as naïve controls. Data were acquired at 2, 4, and 8 month time points. Nineteen metabolites were quantified in the MR spectra using LCModel software. On the basis of 92 spectra, we evaluated any potential gender effect and found that lactate (Lac) levels were lower in males compared to females, while the opposite was observed for ascorbate (Asc). Next, we assessed the effect of age and found increased levels of GABA, Tau, and GPC+PCho. Finally, we analyzed the effect of treatment and found that Lac levels (p = 0.005) were specifically lower in the α-syn group compared to the green fluorescent protein and control groups. In addition, Asc levels (p = 0.05) were increased in the vector-injected groups, whereas glucose levels remained unchanged. This study indicates that the metabolic switch between glucose-lactate could be detectable in vivo and might be modulated by Asc. No concomitant changes were found in markers of neuronal integrity (e.g., N-acetylaspartate) consistent with the fact that α-syn over-expression in cortical neurons did not result in neurodegeneration in this model. We acquired the neurochemical profile longitudinally in a rat model of human wild-type alpha-synuclein (α-syn) over-expression in cortical neurons. We found that Lactate levels were reduced in the α-syn group compared to the control groups and Ascorbate levels were increased in the vector-injected groups. No changes were found in markers of neuronal integrity consistent with the fact that α-syn over-expression did not result in frank neurodegeneration.


Assuntos
Córtex Cerebral/metabolismo , Dependovirus , Espectroscopia de Ressonância Magnética/métodos , Neurônios/metabolismo , alfa-Sinucleína/biossíntese , Animais , Animais Recém-Nascidos , Córtex Cerebral/citologia , Feminino , Regulação da Expressão Gênica , Humanos , Hidrogênio , Estudos Longitudinais , Masculino , Gravidez , Ratos , Ratos Sprague-Dawley
17.
Brain ; 139(Pt 1): 193-203, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26490333

RESUMO

Non-invasive imaging of amyloid-ß in the brain, a hallmark of Alzheimer's disease, may support earlier and more accurate diagnosis of the disease. In this study, we assessed the novel single photon emission computed tomography tracer (123)I-ABC577 as a potential imaging biomarker for amyloid-ß in the brain. The radio-iodinated imidazopyridine derivative (123)I-ABC577 was designed as a candidate for a novel amyloid-ß imaging agent. The binding affinity of (123)I-ABC577 for amyloid-ß was evaluated by saturation binding assay and in vitro autoradiography using post-mortem Alzheimer's disease brain tissue. Biodistribution experiments using normal rats were performed to evaluate the biokinetics of (123)I-ABC577. Furthermore, to validate (123)I-ABC577 as a biomarker for Alzheimer's disease, we performed a clinical study to compare the brain uptake of (123)I-ABC577 in three patients with Alzheimer's disease and three healthy control subjects. (123)I-ABC577 binding was quantified by use of the standardized uptake value ratio, which was calculated for the cortex using the cerebellum as a reference region. Standardized uptake value ratio images were visually scored as positive or negative. As a result, (123)I-ABC577 showed high binding affinity for amyloid-ß and desirable pharmacokinetics in the preclinical studies. In the clinical study, (123)I-ABC577 was an effective marker for discriminating patients with Alzheimer's disease from healthy control subjects based on visual images or the ratio of cortical-to-cerebellar binding. In patients with Alzheimer's disease, (123)I-ABC577 demonstrated clear retention in cortical regions known to accumulate amyloid, such as the frontal cortex, temporal cortex, and posterior cingulate. In contrast, less, more diffuse, and non-specific uptake without localization to these key regions was observed in healthy controls. At 150 min after injection, the cortical standardized uptake value ratio increased by ∼ 60% in patients with Alzheimer's disease relative to healthy control subjects. Both healthy control subjects and patients with Alzheimer's disease showed minimal (123)I-ABC577 retention in the white matter. These observations indicate that (123)I-ABC577 may be a useful single photon emission computed tomography imaging maker to identify amyloid-ß in the human brain. The availability of an amyloid-ß tracer for single photon emission computed tomography might increase the accessibility of diagnostic imaging for Alzheimer's disease.


Assuntos
Doença de Alzheimer/diagnóstico , Peptídeos beta-Amiloides/metabolismo , Neuroimagem Funcional/métodos , Imidazóis/metabolismo , Imidazóis/farmacocinética , Piridinas/metabolismo , Piridinas/farmacocinética , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Animais , Biomarcadores/metabolismo , Estudos de Casos e Controles , Córtex Cerebral/metabolismo , Feminino , Humanos , Imidazóis/síntese química , Masculino , Piridinas/síntese química , Ratos , Distribuição Tecidual , Adulto Jovem
18.
Am J Alzheimers Dis Other Demen ; 31(4): 333-40, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26631686

RESUMO

OBJECTIVE: This study was designed to detect the brain glucose metabolism in rat models of Alzheimer's disease (AD) by the application of (18)F-2-fluoro-deoxy-d-glucose positron emission tomography ((18)F-FDG-PET) and to provide new insights for the early detection of AD. METHODS: Forty Wistar rats were randomly divided into 2 groups. Fifteen sham-operated rats were used as a control group. The remaining rats as a premodel group were intracerebroventricularly injected with ibotenic acid and were intraperitoneally injected with d-galactose, of which 15 rats were included as the experimental group. The above-mentioned 2 groups were assigned to Y-maze test and underwent (18)F-FDG-PET scanning. Positron emission tomography images were processed with SPM 2.0. RESULTS: The learning and memory skills were weakened in AD rats. Besides, the glucose metabolic activity of AD rats decreased in hippolampus, hypothalamus, insular cortex, piriform cortex, striatum, cingulate gyrus, stria terminalis, and parietal lobe and increased in olfactory bulb, cerebellum, midbrain, pontine, and retrosplenial cortex compared with the control group. Dorsal thalamus had shown both enhanced and reduced glucose metabolic activity. CONCLUSION: Our data indicate that the changed glucose metabolism in cerebral regions in (18)F-FDG-PET imaging could be an important predictor for early AD.


Assuntos
Doença de Alzheimer/diagnóstico , Modelos Animais de Doenças , Glucose/metabolismo , Animais , Córtex Cerebral/metabolismo , Feminino , Fluordesoxiglucose F18 , Masculino , Tomografia por Emissão de Pósitrons/métodos , Ratos , Ratos Wistar
19.
Lancet Neurol ; 14(8): 804-813, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26139022

RESUMO

BACKGROUND: The biomarker model of Alzheimer's disease postulates a dynamic sequence of amyloidosis, neurodegeneration, and cognitive decline as an individual progresses from preclinical Alzheimer's disease to dementia. Despite supportive evidence from cross-sectional studies, verification with long-term within-individual data is needed. METHODS: For this prospective cohort study, carriers of autosomal dominant Alzheimer's disease mutations (aged ≥21 years) were recruited from across the USA through referrals by physicians or from affected families. People with mutations in PSEN1, PSEN2, or APP were assessed at the University of Pittsburgh Alzheimer's Disease Research Center every 1-2 years, between March 23, 2003, and Aug 1, 2014. We measured global cerebral amyloid ß (Aß) load using (11)C-Pittsburgh Compound-B PET, posterior cortical metabolism with (18)F-fluorodeoxyglucose PET, hippocampal volume (age and sex corrected) with T1-weighted MRI, verbal memory with the ten-item Consortium to Establish a Registry for Alzheimer's Disease Word List Learning Delayed Recall Test, and general cognition with the Mini Mental State Examination. We estimated overall biomarker trajectories across estimated years from symptom onset using linear mixed models, and compared these estimates with cross-sectional data from cognitively normal control individuals (age 65-89 years) who were negative for amyloidosis, hypometabolism, and hippocampal atrophy. In the mutation carriers who had the longest follow-up, we examined the within-individual progression of amyloidosis, metabolism, hippocampal volume, and cognition to identify progressive within-individual changes (a significant change was defined as an increase or decrease of more than two Z scores standardised to controls). FINDINGS: 16 people with mutations in PSEN1, PSEN2, or APP, aged 28-56 years, completed between two and eight assessments (a total of 83 assessments) over 2-11 years. Significant differences in mutation carriers compared with controls (p<0·01) were detected in the following order: increased amyloidosis (7·5 years before expected onset), decreased metabolism (at time of expected onset), decreased hippocampal volume and verbal memory (7·5 years after expected onset), and decreased general cognition (10 years after expected onset). Among the seven participants with longest follow-up (seven or eight assessments spanning 6-11 years), three individuals had active amyloidosis without progressive neurodegeneration or cognitive decline, two amyloid-positive individuals showed progressive neurodegeneration and cognitive decline without further progressive amyloidosis, and two amyloid-positive individuals showed neither active amyloidosis nor progressive neurodegeneration or cognitive decline. INTERPRETATION: Our results support amyloidosis as the earliest component of the biomarker model in autosomal dominant Alzheimer's disease. Our within-individual examination suggests three sequential phases in the development of autosomal dominant Alzheimer's disease-active amyloidosis, a stable amyloid-positive period, and progressive neurodegeneration and cognitive decline-indicating that Aß accumulation is largely complete before progressive neurodegeneration and cognitive decline occur. These findings offer supportive evidence for efforts to target early Aß deposition for secondary prevention in individuals with autosomal dominant Alzheimer's disease. FUNDING: National Institutes of Health and Howard Hughes Medical Institute.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides/metabolismo , Amiloidose/metabolismo , Biomarcadores , Córtex Cerebral , Progressão da Doença , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Compostos de Anilina , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Feminino , Hipocampo/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Tiazóis
20.
Brain ; 138(Pt 4): 1059-69, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25688080

RESUMO

Perfusion is reduced in the cerebral neocortex in Alzheimer's disease. We have explored some of the mechanisms, by measurement of perfusion-sensitive and disease-related proteins in post-mortem tissue from Alzheimer's disease, vascular dementia and age-matched control brains. To distinguish physiological from pathological reduction in perfusion (i.e. reduction exceeding the decline in metabolic demand), we measured the concentration of vascular endothelial growth factor (VEGF), a protein induced under conditions of tissue hypoxia through the actions of hypoxia-inducible factors, and the myelin associated glycoprotein to proteolipid protein 1 (MAG:PLP1) ratio, which declines in chronically hypoperfused brain tissue. To evaluate possible mechanisms of hypoperfusion, we also measured the levels of amyloid-ß40, amyloid-ß42, von Willebrand factor (VWF; a measure of microvascular density) and the potent vasoconstrictor endothelin 1 (EDN1); we assayed the activity of angiotensin I converting enzyme (ACE), which catalyses the production of another potent vasoconstrictor, angiotensin II; and we scored the severity of arteriolosclerotic small vessel disease and cerebral amyloid angiopathy, and determined the Braak tangle stage. VEGF was markedly increased in frontal and parahippocampal cortex in Alzheimer's disease but only slightly and not significantly in vascular dementia. In frontal cortex the MAG:PLP1 ratio was significantly reduced in Alzheimer's disease and even more so in vascular dementia. VEGF but not MAG:PLP1 increased with Alzheimer's disease severity, as measured by Braak tangle stage, and correlated with amyloid-ß42 and amyloid-ß42: amyloid-ß40 but not amyloid-ß40. Although MAG:PLP1 tended to be lowest in cortex from patients with severe small vessel disease or cerebral amyloid angiopathy, neither VEGF nor MAG:PLP1 correlated significantly with the severity of structural vascular pathology (small vessel disease, cerebral amyloid angiopathy or VWF). However, MAG:PLP1 showed a significant negative correlation with the level of EDN1, which we previously showed to be elevated in the cerebral cortex Alzheimer's disease. These finding are in contrast with the previously demonstrated reduction in EDN1, and positive correlation with MAG:PLP1, in the hypoperfused white matter in Alzheimer's disease. The decline in MAG:PLP1 strongly suggests pathological hypoperfusion of the frontal cortex in Alzheimer's disease. Although severe small vessel disease or cerebral amyloid angiopathy may contribute in some cases, abnormal vascular contractility mediated by EDN1 is likely to be a more important overall contributor. Both amyloid-ß accumulation and hypoperfusion are likely to cause the upregulation of VEGF.


Assuntos
Doença de Alzheimer/metabolismo , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/metabolismo , Demência Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Autopsia , Córtex Cerebral/patologia , Demência Vascular/patologia , Feminino , Humanos , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA