Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Pharmacol Res ; 187: 106631, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36586644

RESUMO

According to the ß-amyloid (Aß) hypothesis of Alzheimer's disease (AD), brain Aß accumulation is the primary cascade event leading to cognitive deficit and dementia. Numerous anti-Aß drugs either inhibiting production or aggregation of Aß or stimulating its clearance have failed to show clinical benefit in large scale AD trials, with ß- and γ-secretase inhibitors consistently worsening cognitive and clinical decline. In June 2021, the FDA approved aducanumab, an anti-Aß monoclonal antibody for early AD based on its ability to reduce brain amyloid plaques, while two other amyloid-clearing antibodies (lecanemab and donanemab) have recently produced encouraging cognitive and clinical results. We reviewed AD trials using PubMed, meeting abstracts and ClinicalTrials.gov and evaluated the effects of such drugs on cerebrospinal fluid (CSF) Aß levels, correlating them with cognitive effects. We found that ß-secretase and γ-secretase inhibitors produce detrimental cognitive effects by significantly reducing CSF Aß levels. We speculate that monoclonal antibodies targeting Aß protofibrils, fibrils or plaques may improve cognitive performance in early AD by increasing soluble Aß levels through Aß aggregate disassembly and/or stabilization of existing Aß monomers.These findings suggest that the real culprit in AD may be decreased levels of soluble monomeric Aß due to sequestration into brain Aß aggregates and plaques.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/tratamento farmacológico , Secretases da Proteína Precursora do Amiloide , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Peptídeos beta-Amiloides , Cognição
2.
J Lipid Res ; 62: 100014, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33518512

RESUMO

Apolipoproteins C-I, C-II, and C-III interact with ApoE to regulate lipoprotein metabolism and contribute to Alzheimer's disease pathophysiology. In plasma, apoC-I and C-II exist as truncated isoforms, while apoC-III exhibits multiple glycoforms. This study aimed to 1) delineate apoC-I, C-II, and C-III isoform profiles in cerebrospinal fluid (CSF) and plasma in a cohort of nondemented older individuals (n = 61), and 2) examine the effect of APOE4 on these isoforms and their correlation with CSF Aß42, a surrogate of brain amyloid accumulation. The isoforms of the apoCs were immunoaffinity enriched and measured with MALDI-TOF mass spectrometry, revealing a significantly higher percentage of truncated apoC-I and apoC-II in CSF compared with matched plasma, with positive correlation between CSF and plasma. A greater percentage of monosialylated and disialylated apoC-III isoforms was detected in CSF, accompanied by a lower percentage of the two nonsialylated apoC-III isoforms, with significant linear correlations between CSF and plasma. Furthermore, a greater percentage of truncated apoC-I in CSF and apoC-II in plasma and CSF was observed in individuals carrying at least one APOE Ɛ4 allele. Increased apoC-I and apoC-II truncations were associated with lower CSF Aß42. Finally, monosialylated apoC-III was lower, and disialylated apoC-III greater in the CSF of Ɛ4 carriers. Together, these results reveal distinct patterns of the apoCs isoforms in CSF, implying CSF-specific apoCs processing. These patterns were accentuated in APOE Ɛ4 allele carriers, suggesting an association between APOE4 genotype and Alzheimer's disease pathology with apoCs processing and function in the brain.


Assuntos
Doença de Alzheimer
3.
Brain ; 143(3): 976-992, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-32091109

RESUMO

Research into hippocampal self-regulation abilities may help determine the clinical significance of hippocampal hyperactivity throughout the pathophysiological continuum of Alzheimer's disease. In this study, we aimed to identify the effects of amyloid-ß peptide 42 (amyloid-ß42) and phosphorylated tau on the patterns of functional connectomics involved in hippocampal downregulation. We identified 48 cognitively unimpaired participants (22 with elevated CSF amyloid-ß peptide 42 levels, 15 with elevated CSF phosphorylated tau levels, mean age of 62.705 ± 4.628 years), from the population-based 'Alzheimer's and Families' study, with baseline MRI, CSF biomarkers, APOE genotyping and neuropsychological evaluation. We developed a closed-loop, real-time functional MRI neurofeedback task with virtual reality and tailored it for training downregulation of hippocampal subfield cornu ammonis 1 (CA1). Neurofeedback performance score, cognitive reserve score, hippocampal volume, number of apolipoprotein ε4 alleles and sex were controlled for as confounds in all cross-sectional analyses. First, using voxel-wise multiple regression analysis and controlling for CSF biomarkers, we identified the effect of healthy ageing on eigenvector centrality, a measure of each voxel's overall influence based on iterative whole-brain connectomics, during hippocampal CA1 downregulation. Then, controlling for age, we identified the effects of abnormal CSF amyloid-ß42 and phosphorylated tau levels on eigenvector centrality during hippocampal CA1 downregulation. Across subjects, our main findings during hippocampal downregulation were: (i) in the absence of abnormal biomarkers, age correlated with eigenvector centrality negatively in the insula and midcingulate cortex, and positively in the inferior temporal gyrus; (ii) abnormal CSF amyloid-ß42 (<1098) correlated negatively with eigenvector centrality in the anterior cingulate cortex and primary motor cortex; and (iii) abnormal CSF phosphorylated tau levels (>19.2) correlated with eigenvector centrality positively in the ventral striatum, anterior cingulate and somatosensory cortex, and negatively in the precuneus and orbitofrontal cortex. During resting state functional MRI, similar eigenvector centrality patterns in the cingulate had previously been associated to CSF biomarkers in mild cognitive impairment and dementia patients. Using the developed closed-loop paradigm, we observed such patterns, which are characteristic of advanced disease stages, during a much earlier presymptomatic phase. In the absence of CSF biomarkers, our non-invasive, interactive, adaptive and gamified neuroimaging procedure may provide important information for clinical prognosis and monitoring of therapeutic efficacy. We have released the developed paradigm and analysis pipeline as open-source software to facilitate replication studies.


Assuntos
Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/diagnóstico por imagem , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Região CA1 Hipocampal/metabolismo , Neurorretroalimentação/métodos , Fragmentos de Peptídeos/líquido cefalorraquidiano , Proteínas tau/líquido cefalorraquidiano , Fatores Etários , Idoso , Doença de Alzheimer/complicações , Apolipoproteína E4/genética , Biomarcadores/líquido cefalorraquidiano , Estudos de Casos e Controles , Disfunção Cognitiva/complicações , Disfunção Cognitiva/metabolismo , Conectoma , Estudos Transversais , Regulação para Baixo , Feminino , Genótipo , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Neuroimagem , Testes Neuropsicológicos , Fosforilação , Software , Realidade Virtual
4.
Neurodegener Dis ; 17(6): 242-250, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28787714

RESUMO

BACKGROUND: Available drugs for the global Alzheimer disease (AD) epidemic only treat the symptoms without modifying disease progression. Accumulating evidence supports amyloid-ß42 (Aß42)as the key triggering agent in AD, making it the ideal target for disease-modifying therapies. Preclinical studies provided extensive support for passive Aß42 immunotherapy, leading to human clinical trials with different antibodies. OBJECTIVE: Examine the status of clinical trials for passive immunotherapy against Aß42. METHODS: We performed a thorough literature review of passive Aß42 immunotherapy. RESULTS: Ten anti-Aß42 antibodies targeting lineal or conformational epitopes have been tested in clinical trials. Antibody engineering and appropriate dosing have overcome undesired side effects, leading to increased safety profiles. Unfortunately, few trials have shown cognitive protection, leading to legitimate questions about the utility of Aß42 as an AD target. There is still hope that solanezumab, aducanumab, and other ongoing trials will identify antibodies, patient subpopulations, and administration protocols, with consistent clinical benefits. CONCLUSIONS: Despite the overall disappointing results, there is still hope that Aß immunotherapy in presymptomatic patients will prevent neuronal loss and provide significant clinical benefits that can be applied to larger populations as preventive therapies. Advances with other targets may soon provide additional therapeutic options for AD with increased efficacy.


Assuntos
Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/imunologia , Anticorpos/uso terapêutico , Imunoterapia/métodos , Doença de Alzheimer/imunologia , Humanos
5.
Biochim Biophys Acta ; 1852(4): 633-43, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25241263

RESUMO

Sporadic inclusion-body myositis (s-IBM) is the most common degenerative muscle disease in which aging appears to be a key risk factor. In this review we focus on several cellular molecular mechanisms responsible for multiprotein aggregation and accumulations within s-IBM muscle fibers, and their possible consequences. Those include mechanisms leading to: a) accumulation in the form of aggregates within the muscle fibers, of several proteins, including amyloid-ß42 and its oligomers, and phosphorylated tau in the form of paired helical filaments, and we consider their putative detrimental influence; and b) protein misfolding and aggregation, including evidence of abnormal myoproteostasis, such as increased protein transcription, inadequate protein disposal, and abnormal posttranslational modifications of proteins. Pathogenic importance of our recently demonstrated abnormal mitophagy is also discussed. The intriguing phenotypic similarities between s-IBM muscle fibers and the brains of Alzheimer and Parkinson's disease patients, the two most common neurodegenerative diseases associated with aging, are also discussed. This article is part of a Special Issue entitled: Neuromuscular Diseases: Pathology and Molecular Pathogenesis.


Assuntos
Envelhecimento , Encéfalo , Fibras Musculares Esqueléticas , Miosite de Corpos de Inclusão , Agregação Patológica de Proteínas , Deficiências na Proteostase , Envelhecimento/metabolismo , Envelhecimento/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Humanos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Miosite de Corpos de Inclusão/metabolismo , Miosite de Corpos de Inclusão/patologia , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Fragmentos de Peptídeos , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/patologia , Biossíntese de Proteínas , Processamento de Proteína Pós-Traducional , Deficiências na Proteostase/metabolismo , Deficiências na Proteostase/mortalidade
6.
Ann Nucl Med ; 38(2): 120-130, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37921921

RESUMO

OBJECTIVE: 2-[18F]fluoro-2-deoxy-D-glucose positron emission tomography ([18F]-FDG-PET) is a imaging modality that has been used to measure of glucose metabolism in the brain in Alzheimer's disease (AD). Clinically, decreased glucose uptake has been reported in the brain of AD, although the precise underlying mechanisms have not yet been elucidated. To elucidate the mechanisms of decreased [18F]-FDG uptake in the AD by PET, [18F]-FDG uptake in the brain of aged model mouse of AD was investigated using a dynamic autoradiography technique "bioradiography". A X-ray phase-contrast imaging (X-PCI) and a histopathological evaluation were also investigated to elucidate the mechanisms underlying the relationships between decreased [18F]-FDG uptake and the pathological changes in the brain of AD mouse. METHODS: In this study, AD model mouse (5XFAD, APP+/PS1+) were used. [18F]-FDG-bioradiography was conducted in fresh slices of brain tissue under the condition of resting (slices immersed in 5 mM K+ solution) and metabolically active (in 50 mM K+ solution). Amyloid ß42 (Aß42) deposition in the brain of AD mouse was confirmed by X-PCI. In addition, the positive cells of phosphated tau protein (P-tau) and deposition of Aß42 were also examined by immunohistochemical staining. RESULTS: No significant differences were observed between the two groups in the resting condition. In the activate condition of the brain, [18F]-FDG uptake was significantly decreased in AD mice compared to WT mice. In X-PCI showed Aß deposition in the AD mouse, but not in the WT. The AD mouse also showed increased P-tau, accumulation of Aß42, increase in neuronal apoptosis, and decrease in the number of neurons than that of the WT mouse. CONCLUSION: Neuronal damage, and induction of neuronal apoptosis, decreased [18F]-FDG uptake, increased Aß accumulation and P-tau induced neurofibrillary degeneration are observed in AD mouse. In clinical diagnosis, reduction of [18F]-FDG uptake by PET is one of the means of diagnosing the onset of AD. Our results suggest that decreased uptake of [18F]-FDG in the brains of AD may be associated with neuronal dysfunction and cell death in the brain.


Assuntos
Doença de Alzheimer , Intervenção Coronária Percutânea , Camundongos , Animais , Doença de Alzheimer/metabolismo , Fluordesoxiglucose F18/metabolismo , Autorradiografia , Encéfalo/metabolismo , Amiloide/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Peptídeos beta-Amiloides/metabolismo
7.
Antioxidants (Basel) ; 13(8)2024 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-39199177

RESUMO

Phytoene is a colourless carotenoid widely available from dietary sources and a precursor for the synthesis of other carotenoids. Although present at high concentrations across different tissues, phytoene is largely viewed as not having physiological activity. Here, we utilize the model organism C. elegans to show that phytoene is bioactive and has anti-ageing properties. Supplementation with phytoene protects against oxidative damage and amyloid-ß42 proteotoxicity (a major pathology of Alzheimer's disease), and extends lifespan. We also examine extracts from two microalgae, Chlorella sorokiniana and Dunaliella bardawil. We show that the extracts contain high levels of phytoene, and find that these phytoene-rich extracts have protective effects similar to pure phytoene. Our findings show that phytoene is a bioactive molecule with positive effects on ageing and longevity. Our work also suggests that phytoene-rich microalgae extracts can utilized to produce foods or supplements that promote healthy ageing and prevent the development of chronic age-related diseases.

8.
Curr Alzheimer Res ; 2024 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-39041277

RESUMO

OBJECT: The study aims to determine whether multimorbidity status is associated with cerebrospinal fluid (CSF) biomarkers for neurodegenerative disorders. METHODS: A total of 827 patients were enrolled from the Parkinson's Progression Markers Initiative (PPMI) database, including 638 patients with early-stage Parkinson's disease (PD) and 189 healthy controls (HCs). Multimorbidity status was evaluated based on the count of long-term conditions (LTCs) and the multimorbidity pattern. Using linear regression models, cross-sectional and longitudinal analyses were conducted to assess the associations of multimorbidity status with CSF biomarkers for neurodegenerative disorders, including α-synuclein (αSyn), amyloid-ß42 (Aß42), total tau (t-tau), phosphorylated tau (p-tau), glial fibrillary acidic protein (GFAP), and neurofilament light chain protein (NfL). RESULTS: At baseline, the CSF t-tau (p = 0.010), p-tau (p = 0.034), and NfL (p = 0.049) levels showed significant differences across the three categories of LTC counts. In the longitudinal analysis, the presence of LTCs was associated with lower Aß42 (ß < -0.001, p = 0.020), and higher t-tau (ß = 0.007, p = 0.026), GFAP (ß = 0.013, p = 0.022) and NfL (ß = 0.020, p = 0.012); Participants with tumor/musculoskeletal/mental disorders showed higher CSF levels of t-tau (ß = 0.016, p = 0.011) and p-tau (ß = 0.032, p = 0.044) than those without multimorbidity. CONCLUSION: Multimorbidity, especially severe multimorbidity and the pattern of mental/musculoskeletal/ tumor disorders, was associated with CSF biomarkers for neurodegenerative disorders in early-stage PD patients, suggesting that multimorbidity might play a crucial role in aggravating neuronal damage in neurodegenerative diseases.

9.
J Leukoc Biol ; 2024 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-39243388

RESUMO

Dysfunctional phagocytic clearance of ß-amyloid (Aß) in microglia and peripheral macrophages/monocytes has been implicated in Alzheimer's disease (AD), but the mechanisms underlying this dysfunction are not yet well understood. In this study, we examined the role of glia maturation factor-γ (GMFG), an actin-disassembly protein that is highly expressed in immune cells, in macrophage Aß phagocytosis and in regulating scavenger receptor AI (SR-AI), a cell-surface receptor that has previously been implicated in Aß clearance. GMFG knockdown increased phagocytosis of Aß42 in BMDMs and RAW264.7 murine macrophages, while GMFG overexpression reduced Aß42 uptake in these cells. Blocking with anti-SR-AI antibodies inhibited Aß42 uptake in GMFG-knockdown cells, establishing a role for SR-AI in Aß42 phagocytosis. GMFG knockdown increased SR-AI protein expression under both basal conditions and in response to Aß42 treatment via both the transcriptional and post-transcriptional level in RAW264.7 macrophages. GMFG knockdown modulated Aß42-induced K48-linked and K63-polyubiquitination of SR-AI, the phosphorylation of SR-AI and JNK, suggesting that GMFG plays a role for intracellular signaling in the SR-AI-mediated uptake of Aß. Further, GMFG-knockdown cells displayed increased levels of the transcriptional factor MafB, and silencing of MafB in these cells reduced their SR-AI expression. Finally, GMFG was found to interact with the nuclear pore complex component RanBP2, and silencing of RanBP2 in GMFG-knockdown cells reduced their SR-AI expression. Collectively, these data support the role of GMFG as a novel regulator of SR-AI in macrophage Aß phagocytosis, and may provide insight into therapeutic approaches to potentially slow or prevent the progression of AD.

10.
JACC Heart Fail ; 11(1): 93-102, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36372727

RESUMO

BACKGROUND: Amyloid-ß (Aß) may be related to cardiac function. However, there are limited data on the association of plasma Aß with cardiac function and risk of heart failure (HF) in the general population. OBJECTIVES: This study sought to determine the associations of plasma amyloid-ß40 (Aß40) and amyloid-ß42 (Aß42) with echocardiographic measurements of cardiac dysfunction and with incident HF in the general population. METHODS: The study included 4,156 participants of the population-based Rotterdam Study (mean age: 71.4 years; 57.1% women), who had plasma Aß samples collected between 2002 and 2005 and had no established dementia and HF at baseline. Multivariable linear regression models were used to explore the cross-sectional association of plasma Aß with echocardiographic measures. Participants were followed up until December 2016. Cox proportional hazards models were used to assess the association of Aß levels with incident HF. Models were adjusted for cardiovascular risk factors. RESULTS: A per 1-SD increase in log-transformed plasma Aß40 was associated with a 0.39% (95% CI: -0.68 to -0.10) lower left ventricular ejection fraction and a 0.70 g/m2 (95% CI: 0.06-1.34) larger left ventricular mass indexed by body surface area. Aß42 was not significantly associated with echocardiographic measures cross-sectionally. During follow-up (median: 10.2 years), 472 incident HF cases were identified. A per 1-SD increase in log-transformed Aß40 was associated with a 32% greater risk of HF (HR: 1.32; 95% CI: 1.15-1.51), and the association was significant in men, but not in women. Higher plasma Aß42 levels were associated with an increased risk of HF (HR: 1.12; 95% CI: 1.02-1.24), although the association was attenuated after further adjustment for concomitant Aß40 (HR: 1.03; 95% CI: 0.92-1.16). CONCLUSIONS: Higher levels of Aß40 were associated with worse cardiac function and higher risk of new onset HF in the general population, in particular among men.


Assuntos
Insuficiência Cardíaca , Masculino , Humanos , Feminino , Idoso , Volume Sistólico , Estudos Transversais , Insuficiência Cardíaca/epidemiologia , Função Ventricular Esquerda , Peptídeos beta-Amiloides
11.
ACS Appl Mater Interfaces ; 15(23): 27789-27800, 2023 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-37261999

RESUMO

Alzheimer's disease is associated with the aggregation of the misfolded neuronal peptide, amyloid-ß42 (Aß42). Evidence has suggested that several reasons are responsible for the toxicity caused by the aggregation of Aß42, including the conformational restriction of Aß42. In this study, one of the toxic conformers of Aß42, which contains a Glu-to-Pro substitution (E22P-Aß42), was explored using atomic force microscopy and molecular docking to study the aggregation dynamics. We proposed a systematic model of fibril formation to better understand the molecular basis of conformational transitions in the Aß42 species. Our results demonstrated the formation of amorphous aggregates in E22P-Aß42 that are stem-based, network-like structures, while the formation of mature fibrils occurred in the less toxic conformer of Aß42, E22-Aß42, that are sphere-like flexible structures. A comparison was made between the biophysical properties of E22P-Aß42 and E22-Aß42 that revealed that E22P-Aß42 had greater stiffness, dihedral angle, number of ß sheets involved, and elasticity, compared with E22-Aß42. These findings will have considerable implications toward our understanding of the structural basis of the toxicity caused by conformational diversity in Aß42 species.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Humanos , Peptídeos beta-Amiloides/toxicidade , Peptídeos beta-Amiloides/química , Simulação de Acoplamento Molecular , Microscopia de Força Atômica , Amiloide , Proteínas Amiloidogênicas , Fragmentos de Peptídeos/química
12.
J Parkinsons Dis ; 12(2): 537-544, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34864690

RESUMO

BACKGROUND: Several biomarkers have been evaluated in Parkinson's disease (PD); cerebrospinal fluid (CSF) levels of lactate may reflect cerebral metabolism function and CSF amyloid-ß42 (Aß42), total tau (t-tau) and phosphorylated tau (p-tau) concentrations may detect an underlying neurodegenerative process. OBJECTIVE: CSF levels of lactate, Aß42, t-tau, and p-tau were measured in patients with mild to moderate PD. CSF levels of dopamine (DA) and its metabolite 3,4-Dihydroxyphenylacetic acid (DOPAC) were also assessed, exploring their relations with the other CSF biomarkers. METHODS: 101 drug-naive PD patients and 60 controls were included. Participants underwent clinical assessments and CSF biomarker analysis. Patients were divided into subgroups according to their Hoehn & Yahr stage (PD-1, PD-2, PD-3). RESULTS: PD patients showed higher lactate levels (M = 1.91; p = 0.03) and lower Aß42 (M = 595; p < 0.001) and DA levels (M = 0.32; p = 0.04) than controls (Mlactate = 1.72; MAß42 = 837; MDA = 0.50), while no significant differences were found in t-tau, p-tau and DOPAC concentrations. Considering the subgroup analysis, PD-3 group had higher lactate (M = 2.12) and t-tau levels (M = 333) than both PD-1 (Mlactate = 1.75, p = 0.006; Mt - tau = 176, p = 0.008) and PD-2 groups (Mlactate = 1.91, p = 0.01; Mt - tau = 176, p = 0.03), as well as the controls (Mlactate = 1.72, p = 0.04; Mt - tau = 205, p = 0.04). PD-2 group showed higher lactate levels than PD-1 group (p = 0.04) and controls (p = 0.03). Finally, CSF lactate levels negatively correlated with DA (r = -0.42) and positively with t-tau CSF levels (r = 0.33). CONCLUSION: This CSF-based study shows that lactate levels in PD correlated with both clinical disease progression and neurodegeneration biomarkers, such as tau proteins and DA. Further studies should explore the clinical potential of measuring CSF biomarkers for better understanding the role of brain energy metabolism in PD, for research and therapeutic options.


Assuntos
Doença de Parkinson , Ácido 3,4-Di-Hidroxifenilacético , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Biomarcadores/líquido cefalorraquidiano , Dopamina , Glucose , Humanos , Lactatos , Fragmentos de Peptídeos/líquido cefalorraquidiano , Receptor de Morte Celular Programada 1
13.
J Alzheimers Dis ; 75(3): 827-843, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32333592

RESUMO

BACKGROUND: Numerous studies have reported on cerebrospinal fluid (CSF) and blood biomarkers of Alzheimer's disease (AD); however, to date, none has compared biomarker patterns across the early-onset subtypes, i.e., early onset sporadic AD (EOsAD) and autosomal dominant AD (ADAD), qualitatively and quantitatively. OBJECTIVE: To compare the fluid biomarker patterns in early-onset subtypes of AD; EOsAD and ADAD. METHODS: Six scientific databases were searched for peer-reviewed research publications. The total number of individuals used in all the meta-analysis were 2,427, comprised of 1,337 patients and 1,090 controls. RESULTS: In the subset of EOsAD cases without APP, PSEN1/PSEN2 mutations, CSF Aß42 and tau levels were higher when compared to the EOsAD group as a whole. Prevalence of the APOEɛ4 allele was more elevated in EOsAD relative to controls, and not significantly elevated in ADAD cases. CONCLUSION: Established CSF biomarkers confirmed quantitative differences between variants of EOAD. EOsAD is enriched with APOEɛ4, but the level is not higher than generally reported in late-onset AD. The results prompt further exploration of the etiopathogenesis of EOsAD, which accounts for ∼4-10% of all AD cases, but the reasons for the early onset remain poorly understood.


Assuntos
Doença de Alzheimer , Apolipoproteínas E/sangue , Apolipoproteínas E/líquido cefalorraquidiano , Doença de Alzheimer/sangue , Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/sangue , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Biomarcadores/sangue , Biomarcadores/líquido cefalorraquidiano , Humanos , Proteínas tau/sangue , Proteínas tau/líquido cefalorraquidiano
14.
Biol Open ; 8(2)2019 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-30670376

RESUMO

Ionizing radiation is widely used in medicine and is valuable in both the diagnosis and treatment of many diseases. However, its health effects are ambiguous. Here, we report that low-dose ionizing radiation has beneficial effects in human amyloid-ß42 (Aß42)-expressing Drosophila Alzheimer's disease (AD) models. Ionizing radiation at a dose of 0.05 Gy suppressed AD-like phenotypes, including developmental defects and locomotive dysfunction, but did not alter the decreased survival rates and longevity of Aß42-expressing flies. The same dose of γ-irradiation reduced Aß42-induced cell death in Drosophila AD models through downregulation of head involution defective (hid), which encodes a protein that activates caspases. However, 4 Gy of γ-irradiation increased Aß42-induced cell death without modulating pro-apoptotic genes grim, reaper and hid The AKT signaling pathway, which was suppressed in Drosophila AD models, was activated by either 0.05 or 4 Gy γ-irradiation. Interestingly, p38 mitogen-activated protein-kinase (MAPK) activity was inhibited by exposure to 0.05 Gy γ-irradiation but enhanced by exposure to 4 Gy in Aß42-expressing flies. In addition, overexpression of phosphatase and tensin homolog (PTEN), a negative regulator of the AKT signaling pathway, or a null mutant of AKT strongly suppressed the beneficial effects of low-dose ionizing radiation in Aß42-expressing flies. These results indicate that low-dose ionizing radiation suppresses Aß42-induced cell death through regulation of the AKT and p38 MAPK signaling pathways, suggesting that low-dose ionizing radiation has hormetic effects on the pathogenesis of Aß42-associated AD.

15.
Curr Alzheimer Res ; 16(11): 1050-1054, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31660827

RESUMO

INTRODUCTION: We recently showed that Amyloid Beta (Aß)40 accumulates in erythrocytes and possibly causes cell damage as evidenced by an increased number of assumed injured low-density (kg/L) erythrocytes. Furthermore, we have suggested a separation technique to isolate and concentrate such damaged red blood cells for subsequent analysis. OBJECTIVES: We isolated high- and low-density erythrocytes and investigated the accumulation patterns of the Aß peptides (Aß40, Aß42, and Aß43) in Alzheimer (AD), mild cognitive impairment (MCI), and Subjective Cognitive Impairment (SCI). METHODS: Whole blood was fractionated through a density gradient, resulting in two concentrated highand presumed injured low-density erythrocyte fractions. After cell lysis, intracellular Aß40, Aß42, and Aß43 were quantified by ELISA. RESULTS: In both high- and low-density erythrocytes, Aß40 displayed the lowest concentration in MCI, while it was equal and higher in AD and SCI. Aß40 was detected at a 10-fold higher level than Aß42, and in injured low-density erythrocytes, the lowest quantity of Aß42 was found in AD and MCI. Aß40 exhibited a 100-fold greater amount than Aß43, and lighter erythrocytes of MCI subjects displayed less intracellular Aß43 than SCI. CONCLUSION: Red blood cell accumulation patterns of Aß40, Aß42, and Aß43 differ significantly between AD, MCI, and SCI. The data must be verified through larger clinical trials. It is, however, tenable that Aß peptide distributions in erythrocyte subpopulations have the potential to be used for diagnostic purposes.


Assuntos
Doença de Alzheimer/sangue , Peptídeos beta-Amiloides/metabolismo , Disfunção Cognitiva/sangue , Eritrócitos/metabolismo , Eritrócitos/patologia , Idoso , Biomarcadores/sangue , Feminino , Humanos , Masculino , Isoformas de Proteínas/sangue
16.
ACS Chem Neurosci ; 10(6): 2830-2842, 2019 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-30917651

RESUMO

The presence of partially structured helices in natively unfolded amyloid-ß42 (Aß42) and α-synuclein (αS) has been shown to accelerate fibrillation in the onset of Alzheimer's and Parkinson's disease, respectively. At the other extreme, folded stable helical conformers have also been reported to resist amyloid formation. Recent studies indicate that amyloidogenic aggregation can be impeded using small molecules that stabilize the α-helical monomers and switch off the neurotoxic pathway. We predict a common intrapeptide route to stabilization based on the plasticity of helical conformations of Aß42 and αS as assessed through extensive atomistic molecular dynamics (MD) computer simulations (∼36 µs) across ten distinct protein force field and water model combinations. Computed free energies and interaction maps (not obtainable from experiments alone) show that flexible terminal groups (N-terminus of Aß42 and C-terminus of αS) show a tendency to stabilize folded helical conformations in both peptides via primary hydrophobic interactions with central hydrophobic domains, and secondary salt bridges with other domains. These interactions confer aggregation resistance by decreasing the population of partially structured helices and are absent in control simulations of complete unfolding. Computed helical stability is also significantly reduced in terminal-deleted variants. The models suggest new strategies to tackle neurodegeneration by rationally re-engineering terminal groups to optimize their predicted ability to deactivate helical monomers.


Assuntos
Peptídeos beta-Amiloides/química , alfa-Sinucleína/química , Simulação de Dinâmica Molecular , Conformação Proteica em alfa-Hélice , Dobramento de Proteína , Estabilidade Proteica
17.
J Alzheimers Dis ; 67(3): 875-891, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30689565

RESUMO

We previously discovered microRNAs (miRNAs) in cerebrospinal fluid (CSF) that differentiate Alzheimer's disease (AD) patients from Controls. Here we examined the performance of 37 candidate AD miRNA biomarkers in a new and independent cohort of CSF from 47 AD patients and 71 Controls on custom TaqMan arrays. We employed a consensus ranking approach to provide an overall priority score for each miRNA, then used multimarker models to assess the relative contributions of the top-ranking miRNAs to differentiate AD from Controls. We assessed classification performance of the top-ranking miRNAs when combined with apolipoprotein E4 (APOE4) genotype status or CSF amyloid-ß42 (Aß42):total tau (T-tau) measures. We also assessed whether miRNAs that ranked higher as AD markers correlate with Mini-Mental State Examination (MMSE) scores. We show that of 37 miRNAs brought forth from the discovery study, 26 miRNAs remained viable as candidate biomarkers for AD in the validation study. We found that combinations of 6-7 miRNAs work better to identify AD than subsets of fewer miRNAs. Of 26 miRNAs that contribute most to the multimarker models, 14 have higher potential than the others to predict AD. Addition of these 14 miRNAs to APOE4 status or CSF Aß42:T-tau measures significantly improved classification performance for AD. We further show that individual miRNAs that ranked higher as AD markers correlate more strongly with changes in MMSE scores. Our studies validate that a set of CSF miRNAs serve as biomarkers for AD, and support their advancement toward development as biomarkers in the clinical setting.


Assuntos
Doença de Alzheimer/líquido cefalorraquidiano , MicroRNAs/líquido cefalorraquidiano , Idoso , Doença de Alzheimer/diagnóstico , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Apolipoproteína E4/genética , Biomarcadores/líquido cefalorraquidiano , Estudos de Casos e Controles , Feminino , Genótipo , Humanos , Masculino , Fragmentos de Peptídeos/líquido cefalorraquidiano , Reação em Cadeia da Polimerase em Tempo Real , Reprodutibilidade dos Testes , Proteínas tau/líquido cefalorraquidiano
18.
Prion ; 12(5-6): 315-319, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30394185

RESUMO

Although colocalization of amyloid ß (Aß) with prion protein (PrP) in the kuru plaque has previously been observed in the brain of prion diseases patients, the participating Aß species has not been identified. Here, we present an immunohistochemical assessment of the brain and spinal cord of a 69-year-old Japanese female patient with Gerstmann-Sträussler-Scheinker disease with a P105L mutation on the PRNP gene (GSS-P105L). Immunohistochemical assessment of serial brain sections was performed using anti-PrP and -Aß antibodies in the hippocampus, frontal and occipital lobes. She died 69 years after a 21-year clinical course. Immunohistochemistorical examination revealed that ~50% of the kuru plaques in the cerebrum were colocalized with Aß, and Aß42 was predominantly observed to be colocalized with PrP-plaques. The Aß deposition patterns were unique, and distinct from diffuse plaques observed in the normal aging brain or Alzheimer's disease brain. The spinal cord exhibited degeneration in the lateral corticospinal tract, posterior horn, and fasciculus gracilis. We have demonstrated for the first time that Aß42, rather than Aß40, is the main Aß component associated with PrP-plaques, and also the degeneration of the fasciculus gracilis in the spinal cord in GSS-P105L, which could be associated with specific clinical features of GSS-P105L.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Encéfalo/patologia , Doença de Gerstmann-Straussler-Scheinker/metabolismo , Mutação de Sentido Incorreto/genética , Fragmentos de Peptídeos/metabolismo , Proteínas Priônicas/metabolismo , Idoso , Substituição de Aminoácidos/genética , Peptídeos beta-Amiloides/genética , Evolução Fatal , Feminino , Humanos , Imuno-Histoquímica , Fragmentos de Peptídeos/genética , Placa Amiloide/patologia , Proteínas Priônicas/genética , Medula Espinal/patologia
19.
Anim Cells Syst (Seoul) ; 21(3): 160-168, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-30460065

RESUMO

Drosophila melanogaster expressing amyloid-ß42 (Aß42) transgenes have been used as models to study Alzheimer's disease. Various Aß42 transgenes with different structures induce different phenotypes, which make it difficult to compare data among studies which use different transgenic lines. In this study, we compared the phenotypes of four frequently used Aß42 transgenic lines, UAS-Aß422X , UAS-Aß42BL33770 , UAS-Aß4211C39 , and UAS-Aß42H29.3 . Among the four transgenic lines, only UAS-Aß422X has two copies of the upstream activation sequence-amyloid-ß42 (UAS-Aß42) transgene, while remaining three have one copy. UAS-Aß42BL33770 has the 3' untranslated region of Drosophila α-tubulin, while the others have that of SV40. UAS-Aß4211C39 and UAS-Aß42H29.3 have the rat pre-proenkephalin signal peptide, while UAS-Aß422X and UAS-Aß42BL33770 have that of the fly argos protein. When the transgenes were expressed ectopically in the developing eyes of the flies, UAS-Aß422X transgene resulted in a strongly reduced and rough eye phenotype, while UAS-Aß42BL33770 only showed a strong rough eye phenotype; UAS-Aß42H29.3 and UAS-Aß4211C39 had mild rough eyes. The levels of cell death and reactive oxygen species (ROS) in the eye imaginal discs were consistently the highest in UAS-Aß422X , followed by UAS-Aß42BL33770 , UAS-Aß4211C39 , and UAS-Aß42H29.3 . Surprisingly, the reduction in survival during the development of these lines did not correlate with cell death or ROS levels. The flies which expressed UAS-Aß4211C39 or UAS-Aß42H29.3 experienced greatly reduced survival rates, although low levels of ROS or cell death were detected. Collectively, our results demonstrated that different Drosophila AD models show different phenotypic severity, and suggested that different transgenes may have different modes of cytotoxicity. Abbreviations: Aß42: amyloid-ß42; AD: Alzheimer's disease; UAS: upstream activation sequence.

20.
Dis Model Mech ; 9(3): 295-306, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26659252

RESUMO

Expression of the Down syndrome critical region 1 (DSCR1) protein, an inhibitor of the Ca(2+)-dependent phosphatase calcineurin, is elevated in the brains of individuals with Down syndrome (DS) or Alzheimer's disease (AD). Although increased levels of DSCR1 were often observed to be deleterious to neuronal health, its beneficial effects against AD neuropathology have also been reported, and the roles of DSCR1 on the pathogenesis of AD remain controversial. Here, we investigated the role of sarah (sra; also known as nebula), a Drosophila DSCR1 ortholog, in amyloid-ß42 (Aß42)-induced neurological phenotypes in Drosophila. We detected sra expression in the mushroom bodies of the fly brain, which are a center for learning and memory in flies. Moreover, similar to humans with AD, Aß42-expressing flies showed increased Sra levels in the brain, demonstrating that the expression pattern of DSCR1 with regard to AD pathogenesis is conserved in Drosophila. Interestingly, overexpression of sra using the UAS-GAL4 system exacerbated the rough-eye phenotype, decreased survival rates and increased neuronal cell death in Aß42-expressing flies, without modulating Aß42 expression. Moreover, neuronal overexpression of sra in combination with Aß42 dramatically reduced both locomotor activity and the adult lifespan of flies, whereas flies with overexpression of sra alone showed normal climbing ability, albeit with a slightly reduced lifespan. Similarly, treatment with chemical inhibitors of calcineurin, such as FK506 and cyclosporin A, or knockdown of calcineurin expression by RNA interference (RNAi), exacerbated the Aß42-induced rough-eye phenotype. Furthermore, sra-overexpressing flies displayed significantly decreased mitochondrial DNA and ATP levels, as well as increased susceptibility to oxidative stress compared to that of control flies. Taken together, our results demonstrating that sra overexpression augments Aß42 cytotoxicity in Drosophila suggest that DSCR1 upregulation or calcineurin downregulation in the brain might exacerbate Aß42-associated neuropathogenesis in AD or DS.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Inibidores de Calcineurina/metabolismo , Modelos Animais de Doenças , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fragmentos de Peptídeos/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas de Ligação ao Cálcio , Morte Celular , Peróxido de Hidrogênio/metabolismo , Larva/metabolismo , Longevidade , Mitocôndrias/metabolismo , Corpos Pedunculados/metabolismo , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Neuroglia/metabolismo , Neurônios/metabolismo , Óxido Nítrico/metabolismo , Estresse Oxidativo , Fenótipo , Células Fotorreceptoras de Invertebrados/metabolismo , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA