Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 600
Filtrar
1.
Front Cell Neurosci ; 18: 1453038, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39355174

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disorder pathologically characterized by the deposition of amyloid beta (Aß) plaques and neurofibrillary tangles (NFTs) in the brain. The accumulation of these aggregated proteins causes memory and synaptic dysfunction, neuroinflammation, and oxidative stress. This research study is significant as it aims to assess the neuroprotective properties of vitamin E (VE) analog Trolox in an Aß1 - 42-induced AD mouse model. Aß1 - 42 5µL/5min/mouse was injected intracerebroventricularly (i.c.v.) into wild-type adult mice brain to induce AD-like neurotoxicity. For biochemical analysis, Western blotting and confocal microscopy were performed. Remarkably, intraperitoneal (i.p.) treatment of Trolox (30 mg/kg/mouse for 2 weeks) reduced the AD pathology by reducing the expression of Aß, phosphorylated tau (p-tau), and ß-site amyloid precursor protein cleaving enzyme1 (BACE1) in both cortex and hippocampus regions of mice brain. Furthermore, Trolox-treatment decreased neuroinflammation by inhibiting Toll-like receptor 4 (TLR4), phosphorylated nuclear factor-κB (pNF-κB) and interleukin-1ß (IL-1ß), and other inflammatory biomarkers of glial cells [ionized calcium-binding adaptor molecule 1 (Iba1) and glial fibrillary acidic protein (GFAP)]. Moreover, Trolox reduced oxidative stress by enhancing the expression of nuclear factor erythroid-related factor 2 (NRF2) and heme oxygenase 1 (HO1). Similarly, Trolox-induced synaptic markers, including synaptosomal associated protein 23 (SNAP23), synaptophysin (SYN), and post-synaptic density protein 95 (PSD-95), and memory functions in AD mice. Our findings could provide a useful and novel strategy for investigating new medications to treat AD-associated neurodegenerative diseases.

2.
Curr Alzheimer Res ; 2024 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-39279693

RESUMO

Alzheimer's disease (AD) is a devastating neurological disorder that affects synaptic transmission between neurons. Several theories and concepts have been postulated to explain its etiology and pathogenesis. The disease has no cure, and the drugs available to manage AD symptoms provide only modest benefits. It originates in the brain's entorhinal cortex (EC), with tau pathology that can proceed overt symptoms by decades and then spreads to other connected areas and networks to cause severe cognitive decline. Despite decades of research, the reason why the EC is the first region to be affected during AD pathophysiology remains unknown. The EC is well connected with surrounding areas to support the brain's structural and functional integrity, participating in navigation, working memory, memory consolidation, olfaction, and olfactory-auditory coordination. These actions require massive energy expenditure; thus, the EC is extremely vulnerable to severe hypometabolism and an energy crisis. Unfortunately, the crucial events/factors that make the EC vulnerable to pathological sequelae more than other brain regions have not been thoroughly explored. An in-depth analysis of available research on the role of the EC in AD could provide meaningful insights into the susceptibility of this region and its role in propagating AD. In this review article, we highlight how the functional complexities of the EC account for its vulnerability in AD.

3.
J Vet Sci ; 25(5): e60, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39231785

RESUMO

IMPORTANCE: Alzheimer's disease (AD) is the most common cause of dementia in the elderly with the incidence rising exponentially after the age of 65 years. Unfortunately, effective treatments are extremely limited and definite diagnosis can only be made at autopsy. This is in part due to our limited understanding of the complex pathophysiology, including the various genetic, environmental, and metabolic contributing factors. In an effort to better understand this complex disease, researchers have employed nonhuman primates as translational models. CASE PRESENTATION: This report aims to describe the AD-like neuropathology in the brain of a 37-year-old female baboon (Papio hamadryas), which at the time of her death made her the oldest hamadryas baboon at any member institution of the Association of Zoos and Aquariums. A diagnostic necropsy was performed, and the brain was evaluated for neurodegenerative disease. Frequent amyloid-ß deposits were identified, consistent with what has been described in other geriatric nonhuman primates. Phospho-tau pathology, including neurofibrillary tangles, a feature not well-described in other primate models, was also abundant. CONCLUSIONS AND RELEVANCE: Our results suggest that more detailed, prospective, longitudinal studies are warranted utilizing this particular species to see if they represent a viable model for human brain aging.


Assuntos
Doença de Alzheimer , Encéfalo , Doenças dos Macacos , Papio hamadryas , Animais , Feminino , Doença de Alzheimer/veterinária , Doença de Alzheimer/patologia , Encéfalo/patologia , Doenças dos Macacos/patologia , Envelhecimento/patologia
4.
J Alzheimers Dis ; 2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39302368

RESUMO

Background: Neuropathologic studies of brains from autopsy series show tau inclusions (pretangles, neuropils threads, neurofibrillary tangles) are detectable more than a decade before amyloid-ß (Aß) deposition in Alzheimer's disease (AD) and develop in a characteristic manner that forms the basis for AD staging. An alternative position views pathological tau without Aß deposition as a 'primary age-related tauopathy' (PART) rather than prodromal AD. Recently, an early focus of tau inclusions in the Ammon's horn second sector (CA2) with relative sparing of CA1 that occurs before tau inclusions develop in the entorhinal cortex (EC) was proposed as an additional feature of PART. Objective: To test the 'definite PART' hypothesis. Methods: We used AT8-immunohistochemistry in 100µm sections to examine the EC, transentorhinal cortex (TRE), and Ammon's horn in 325 brains with tau inclusions lacking Aß deposits (average age at death 66.7 years for females, 66.4 years for males). Results: 100% of cases displayed tau inclusions in the TRE. In 89% of cases, the CA1 tau rating was greater than or equal to that in CA2. In 25%, CA2 was devoid of tau inclusions. Only 4% displayed a higher tau score in CA2 than in the TRE, EC, and CA1. The perforant path also displayed early tau changes. APOE genotyping was available for 199/325 individuals. Of these, 44% had an ɛ4 allele that placed them at greater risk for developing later NFT stages and, therefore, clinical AD. Conclusions: Our new findings call into question the PART hypothesis and are consistent with the idea that our cases represent prodromal AD.

5.
Neurobiol Dis ; 201: 106656, 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39233131

RESUMO

Spleen tyrosine kinase (Syk), a non-receptor-type tyrosine kinase, has a wide range of physiological functions. A possible role of Syk in Alzheimer's disease (AD) has been proposed. We evaluated the localization of Syk in the brains of patients with AD and control participants. Human neuroblastoma M1C cells harboring wild-type tau (4R0N) were used with the tetracycline off (TetOff) induction system. In this model of neuronal tauopathy, the effects of the Syk inhibitors-BAY 61-3606 and R406-on tau phosphorylation and oligomerization were explored using several phosphorylated tau-specific antibodies and an oligomeric tau antibody, and the effects of these Syk inhibitors on autophagy were examined using western blot analyses. Moreover, the effects of the Syk inhibitor R406 were evaluated in vivo using wild-type mice. In AD brains, Syk and phosphorylated tau colocalized in the cytosol. In M1C cells, Syk protein (72 kDa) was detected using western blot analysis. Syk inhibitors decreased the expression levels of several tau phosphoepitopes including PHF-1, CP13, AT180, and AT270. Syk inhibitors also decreased the levels of caspase-cleaved tau (TauC3), a pathological tau form. Syk inhibitors increased inactivated glycogen synthase kinase 3ß expression and decreased active p38 mitogen-activated protein kinase expression and demethylated protein phosphatase 2 A levels, indicating that Syk inhibitors inactivate tau kinases and activate tau phosphatases. Syk inhibitors also activated autophagy, as indicated by increased LC3II and decreased p62 levels. In vivo, the Syk inhibitor R406 decreased phosphorylated tau levels in wild-type mice. These findings suggest that Syk inhibitors offer novel therapeutic strategies for tauopathies, including AD.

6.
Endocrinology ; 165(10)2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39276028

RESUMO

The identification of critical factors that can worsen the mechanisms contributing to the pathophysiology of Alzheimer disease is of paramount importance. Thyroid hormones (TH) fit this criterion. Epidemiological studies have identified an association between altered circulating TH levels and Alzheimer disease. The study of human and animal models indicates that TH can affect all the main cellular, molecular, and genetic mechanisms known as hallmarks of Alzheimer disease. This is true not only for the excessive production in the brain of protein aggregates leading to amyloid plaques and neurofibrillary tangles but also for the clearance of these molecules from the brain parenchyma via the blood-brain barrier and for the escalated process of neuroinflammation-and even for the effects of carrying Alzheimer-associated genetic variants. Suboptimal TH levels result in a greater accumulation of protein aggregates in the brain. The direct TH regulation of critical genes involved in amyloid beta production and clearance is remarkable, affecting the expression of multiple genes, including APP (related to amyloid beta production), APOE, LRP1, TREM2, AQP4, and ABCB1 (related to amyloid beta clearance). TH also affects microglia by increasing their migration and function and directly regulating the immunosuppressor gene CD73, impacting the immune response of these cells. Studies aiming to understand the mechanisms that could explain how changes in TH levels can contribute to the brain alterations seen in patients with Alzheimer disease are ongoing. These studies have potential implications for the management of patients with Alzheimer disease and ultimately can contribute to devising new interventions for these conditions.


Assuntos
Doença de Alzheimer , Hormônios Tireóideos , Humanos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/epidemiologia , Doença de Alzheimer/etiologia , Hormônios Tireóideos/metabolismo , Animais , Encéfalo/metabolismo , Peptídeos beta-Amiloides/metabolismo , Placa Amiloide/metabolismo
7.
Neurobiol Aging ; 144: 127-137, 2024 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-39326302

RESUMO

In Alzheimer's disease (AD), neuropsychiatric symptoms (NPS) correlate with tau deposition in the brain. Here, we investigated the association of PET-based Braak stages with NPS and assessed whether they predict annual changes in NPS. We evaluated 231 individuals in the aging and AD continuum. Participants were assigned a Braak stage at baseline and followed for 1.97 (s.d. 0.62) years. NPS were investigated using the Mild Behavioral Impairment Checklist (MBI-C) and the Neuropsychiatric Inventory Questionnaire severity (NPI-Q-S) and distress (NPI-Q-D) scales. Multiple linear regressions (MLR) assessed the association of Braak stages with baseline NPS and the annual change in NPS scores. At baseline, stages I-II, III-IV, and V-VI were associated with higher MBI-C, NPI-Q-S, and NPI-Q-D scores. Stages V-VI were associated with a significant annual increase in MBI-C scores. These findings suggest that tau accumulation may manifest clinically with an increase in NPS, which seems to be an early event in AD pathophysiology. Moreover, PET-based Braak staging appears to be a good predictor of NPS severity progression.

8.
Cells ; 13(18)2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39329695

RESUMO

Postencephalitic parkinsonism (PEP) is suggested to show a virus-induced pathology, which is different from classical idiopathic Parkinson's disease (PD) as there is no α-synuclein/Lewy body pathology. However, PEP shows a typical clinical representation of motor disturbances. In addition, compared to PD, there is no iron-induced pathology. The aim of this preliminary study was to compare PEP with PD regarding iron-induced pathology, using histochemistry methods on paraffin-embedded post-mortem brain tissue. In the PEP group, iron was not seen, except for one case with sparse perivascular depositions. Rather, PEP offers a pathology related to tau-protein/neurofibrillary tangles, with mild to moderate memory deficits only. It is assumed that this virus-induced pathology is due to immunological dysfunctions causing (neuro)inflammation-induced neuronal network disturbances as events that trigger clinical parkinsonism. The absence of iron deposits implies that PEP cannot be treated with iron chelators. The therapy with L-Dopa is also not an option, as L-Dopa only leads to an initial slight improvement in symptoms in isolated cases.


Assuntos
Doença de Parkinson Pós-Encefalítica , Humanos , Masculino , Feminino , Idoso , Doença de Parkinson Pós-Encefalítica/patologia , Pessoa de Meia-Idade , Ferro/metabolismo , Idoso de 80 Anos ou mais , Encéfalo/patologia , Proteínas tau/metabolismo , Transtornos Parkinsonianos/patologia , Transtornos Parkinsonianos/complicações
9.
Int J Mol Sci ; 25(15)2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-39125687

RESUMO

Human brain aging is characterized by the production and deposition of ß-amyloid (Aß) in the form of senile plaques and cerebral amyloid angiopathy and the intracellular accumulation of hyper-phosphorylated tau (Hp-tau) to form neurofibrillary tangles (NFTs) and dystrophic neurites of senile plaques. The process progresses for years and eventually manifests as cognitive impairment and dementia in a subgroup of aged individuals. Aß is produced and deposited first in the neocortex in most aged mammals, including humans; it is usually not accompanied by altered behavior and cognitive impairment. Hp-tau is less frequent than Aß pathology, and NFTs are rare in most mammals. In contrast, NFTs are familiar from middle age onward in humans; NFTs first appear in the paleocortex and selected brain stem nuclei. NFTs precede for decades or years Aß deposition and correlate with dementia in about 5% of individuals at the age of 65 and 25% at the age of 85. Based on these comparative data, (a) Aß deposition is the most common Alzheimer's disease neuropathological change (ADNC) in the brain of aged mammals; (b) Hp-tau is less common, and NFTs are rare in most aged mammals; however, NFTs are the principal cytoskeletal pathology in aged humans; (c) NFT in aged humans starts in selected nuclei of the brain stem and paleocortical brain regions progressing to the most parts of the neocortex and other regions of the telencephalon; (d) human brain aging is unique among mammalian species due to the early appearance and dramatic progression of NFTs from middle age onward, matching with cognitive impairment and dementia in advanced cases; (e) neither mammalian nor human brain aging supports the concept of the amyloid cascade hypothesis.


Assuntos
Envelhecimento , Doença de Alzheimer , Emaranhados Neurofibrilares , Proteínas tau , Doença de Alzheimer/patologia , Doença de Alzheimer/metabolismo , Animais , Humanos , Envelhecimento/patologia , Envelhecimento/metabolismo , Emaranhados Neurofibrilares/patologia , Emaranhados Neurofibrilares/metabolismo , Proteínas tau/metabolismo , Encéfalo/patologia , Encéfalo/metabolismo , Peptídeos beta-Amiloides/metabolismo , Mamíferos/metabolismo , Placa Amiloide/patologia , Placa Amiloide/metabolismo
10.
3 Biotech ; 14(8): 190, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39099620

RESUMO

The goal of this research was to study the effect of different doses of resveratrol (RS) and RS with donepezil (DPZ) on the deposition of amyloid beta (Aß) and neurofibrillary tangles (NFTs) in colchicine-induced Alzheimer's disease (AD) brain. The study included three months old male Albino Wistar rats and consisted of six animal groups: AD model (group 1), treatment groups, RS 10 mg/kg body weight (group 2), RS 20 mg/kg body weight (group 3), RS 10 mg/kg body weight along with DPZ 1 mg/kg body weight (group 6), prophylaxis groups, RS 10 mg/kg body weight (group 4) and RS 20 mg/kg body weight (group 5). In the treatment groups, RS was given for 7 consecutive days from the day of induction of AD, and in the prophylaxis groups, we started RS 7 days even before the induction of AD and continued for seven days after the induction. The number of Aßs and NFTs at the frontal region, cornu ammonis (CA) 1,2,3,4 and dentate gyrus regions of hippocampus were evaluated. The immunohistochemical analysis was performed by using mouse anti-ß-amyloid antibody for the Aß plaques and polyclonal rabbit anti-human tau for the tau-positive neurons. The present study observed the accumulation of Aß plaques and tau-positive neurons in the AD model. However, their numbers were significantly decreased in the treatment groups (p < 0.001). The best results were observed when RS 10 mg was given prophylactically (p < 0.01) and RS along with DPZ (p < 0.001), suggesting the neuroprotective effect of RS and its synergistic effect with the DPZ.

11.
Mol Neurobiol ; 2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-39102108

RESUMO

In recent years, significant advancements have been made in understanding Alzheimer's disease from both neurobiological and clinical perspectives. Exploring the complex systems underlying AD has unveiled insights that could potentially revolutionize therapeutic approaches. Recent investigations have highlighted intricate interactions among genetic, molecular, and environmental factors in AD. Optimism arises from neurobiological advancements and diverse treatment options, potentially slowing or halting disease progression. Amyloid-beta plaques and tau protein tangles crucially influence AD onset and progression. Emerging treatments involve diverse strategies, such as approaches targeting multiple pathways involved in AD pathogenesis, such as inflammation, oxidative stress, and synaptic dysfunction pathways. Clinical trials using humanized monoclonal antibodies, focusing on immunotherapies eliminating amyloid-beta, have shown promise. Nonpharmacological interventions such as light therapy, electrical stimulation, cognitive training, physical activity, and dietary changes have drawn attention for their potential to slow cognitive aging and enhance brain health. Precision medicine, which involves tailoring therapies to individual genetic and molecular profiles, has gained traction. Ongoing research and interdisciplinary collaboration are expected to yield more effective treatments.

12.
Adv Neurobiol ; 39: 345-382, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39190082

RESUMO

Besides its two defining misfolded proteinopathies-Aß plaques and tau neurofibrillary tangles-Alzheimer's disease (AD) is an exemplar of a neurodegenerative disease with prominent reactive astrogliosis, defined as the set of morphological, molecular, and functional changes that astrocytes suffer as the result of a toxic exposure. Reactive astrocytes can be observed in the vicinity of plaques and tangles, and the relationship between astrocytes and these AD neuropathological lesions is bidirectional so that each AD neuropathological hallmark causes specific changes in astrocytes, and astrocytes modulate the severity of each neuropathological feature in a specific manner. Here, we will review both how astrocytes change as a result of their chronic exposure to AD neuropathology and how those astrocytic changes impact each AD neuropathological feature. We will emphasize the repercussions that AD-associated reactive astrogliosis has for the astrocyte-neuron interaction and highlight areas of uncertainty and priorities for future research.


Assuntos
Doença de Alzheimer , Astrócitos , Neurônios , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Astrócitos/metabolismo , Astrócitos/patologia , Humanos , Neurônios/metabolismo , Neurônios/patologia , Gliose/metabolismo , Gliose/patologia , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Comunicação Celular/fisiologia , Proteínas tau/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais
13.
Cell Rep ; 43(8): 114574, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39096489

RESUMO

A prevailing hypothesis is that neurofibrillary tangles play a causal role in driving cognitive decline in Alzheimer's disease (AD) because tangles correlate anatomically with areas that undergo neuronal loss. We used two-photon longitudinal imaging to directly test this hypothesis and observed the fate of individual neurons in two mouse models. At any time point, neurons without tangles died at >3 times the rate as neurons with tangles. Additionally, prior to dying, they became >20% more distant from neighboring neurons across imaging sessions. Similar microstructural changes were evident in a population of non-tangle-bearing neurons in Alzheimer's donor tissues. Together, these data suggest that nonfibrillar tau puts neurons at high risk of death, and surprisingly, the presence of a tangle reduces this risk. Moreover, cortical microstructure changes appear to be a better predictor of imminent cell death than tangle status is and a promising tool for identifying dying neurons in Alzheimer's.


Assuntos
Doença de Alzheimer , Morte Celular , Emaranhados Neurofibrilares , Neurônios , Animais , Doença de Alzheimer/patologia , Emaranhados Neurofibrilares/patologia , Neurônios/patologia , Neurônios/metabolismo , Camundongos , Humanos , Proteínas tau/metabolismo , Modelos Animais de Doenças , Camundongos Transgênicos , Masculino , Feminino
14.
Eur J Med Chem ; 276: 116682, 2024 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-39053190

RESUMO

Timely diagnosis and therapy of Alzheimer's disease remains one of the greatest questions in medicinal chemistry of neurodegenerative disease. The lack of low-cost sensors capable of reliable detection of structural changes in AD-related proteins is the driving factor for the development of novel molecules with affinity for AD hallmarks. The development of cheap, safe diagnostic methods is a highly sought-after area of research. Optical fluorescent probes are of great interest due to their non-radioactivity, low cost, and ability of the real-time visualization of AD hallmarks. Boron dipyrromethene (BODIPY)-based fluorophore is one promising fluorescent unit for in vivo labeling due to its high photostability, easy modification, low toxicity, and cell-permeability. In recent years, many fluorescent BODIPY-based probes capable of Aß plaque, Aß soluble oligomers, neurofibrillary tangles (NFT) optical detection, as well as probes with copper ion chelating units and viscosity sensors have been developed. In this review, we summarized BODIPY derivatives as fluorescent sensors capable of detecting pathological features of Alzheimer's disease, published from 2009 to 2023, as well as their design strategies, optical properties, and in vitro and in vivo activities.


Assuntos
Doença de Alzheimer , Compostos de Boro , Corantes Fluorescentes , Doença de Alzheimer/diagnóstico , Compostos de Boro/química , Compostos de Boro/síntese química , Humanos , Corantes Fluorescentes/química , Corantes Fluorescentes/síntese química , Animais , Peptídeos beta-Amiloides/análise , Peptídeos beta-Amiloides/metabolismo , Estrutura Molecular
15.
Molecules ; 29(14)2024 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-39064887

RESUMO

Using a molecular modeling approach for Tau-binding sites, we modified our previously reported imaging agent, [125I]INFT, for the potential improvement of binding properties to Tau in an Alzheimer's disease (AD) brain. Two new derivatives, namely [125I]ISAS and [125I]NIPZ, were designed, where binding energies at site 1 of Tau were -7.4 and -6.0 kcal/mole, respectively, compared to [125I]INFT (-7.6 kcal/mole). The radiosynthesis of [125I]ISAS and [125I]NIPZ was carried out by using iodine-125 and purified chromatographically to achieve >90% purity. In vitro binding affinities (IC50) for Tau were as follows: INFT = 7.3 × 10-8 M; ISAS = 4.7 × 10-8 M; NIPZ > 10-6 M. The binding of [125I]ISAS to gray matter (GM) correlated with the presence of Tau in the AD brain, confirmed by anti-Tau immunohistochemistry. [125I]NIPZ did not bind to Tau, with similar levels of binding observed in GM and white matter (WM). Four radiotracers were compared and the rank order of binding to Tau was found to be [125I]IPPI > [125I]INFT > [125I]ISAS >>> [125I]NIPZ with GM/WM ratios of [125I]IPPI = 7.74 > [125I]INFT = 4.86 > [125I]ISAS = 3.62 >> [125I]NIPZ = 1.24. The predictive value of Chimera-AutoDock for structurally related compounds binding to the Tau binding sites (measured as binding energy) was good. A binding energy of less than -7 kcal/mole is necessary and less than -8 kcal/mole will be more suitable for developing imaging agents.


Assuntos
Doença de Alzheimer , Encéfalo , Radioisótopos do Iodo , Compostos Radiofarmacêuticos , Proteínas tau , Doença de Alzheimer/metabolismo , Doença de Alzheimer/diagnóstico por imagem , Proteínas tau/metabolismo , Proteínas tau/química , Humanos , Radioisótopos do Iodo/química , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/síntese química , Modelos Moleculares , Ligação Proteica , Sítios de Ligação , Masculino , Idoso , Autopsia , Feminino
16.
Curr Alzheimer Res ; 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38963100

RESUMO

Alzheimer's disease (AD) is the frequent form of dementia in the world. Despite over 100 years of research into the causes of AD, including amyloid and tau protein, the research has stalled and has not led to any conclusions. Moreover, numerous projects aimed at finding a cure for AD have also failed to achieve a breakthrough. Thus, the failure of anti-amyloid and anti-tau protein therapy to treat AD significantly influenced the way we began to think about the etiology of the disease. This situation prompted a group of researchers to focus on ischemic brain episodes, which, like AD, mostly present alterations in the hippocampus. In this context, it has been proposed that cerebral ischemic incidents may play a major role in promoting amyloid and tau protein in neurodegeneration in AD. In this review, we summarized the experimental and clinical research conducted over several years on the role of ischemic brain episodes in the development of AD. Studies have shown changes typical of AD in the course of brain neurodegeneration post-ischemia, i.e., progressive brain and hippocampal atrophy, increased amyloid production, and modification of tau protein. In the post-ischemic brain, the diffuse and senile amyloid plaques and the development of neurofibrillary tangles characteristic of AD were revealed. The above data evidently showed that after brain ischemia, there are modifications in protein folding, leading to massive neuronal death and damage to the neuronal network, which triggers dementia with the AD phenotype.

17.
Alzheimers Dement ; 20(7): 4803-4817, 2024 07.
Artigo em Inglês | MEDLINE | ID: mdl-38884346

RESUMO

INTRODUCTION: Tau aggregation into neurofibrillary tangles in Alzheimer's disease (AD) is a dynamic process involving changes in tau phosphorylation, isoform composition, and morphology. To facilitate studies of tangle maturity, we developed an image analysis pipeline to study antibody labeling signatures that can distinguish tangle maturity levels in AD brain tissue. METHODS: Using fluorescent immunohistochemistry, we co-labeled AD brain tissue with four antibodies that bind different tau epitopes. Mean fluorescence intensity of each antibody was measured, and spectral clustering was used to identify tangle immunophenotypes. RESULTS: Five distinct tangle populations were identified, and different tangle maturity immunophenotypes were identified with increasing Braak stage. Early tangle immunophenotypes were more prevalent in later affected regions and advanced immunophenotypes were associated with ghost morphology. DISCUSSION: Our findings indicate that tangle populations characterized by advanced tau immunophenotypes are associated with higher Braak stage and more mature morphology, providing a new framework for defining tangle maturity levels using tau antibody signatures. HIGHLIGHTS: Populations of neurofibrillary tangles exist in Alzheimer's disease. The immunophenotype of neurofibrillary tangle populations relates to their maturity. The most advanced immunophenotypes are associated with higher Braak stage. The most advanced immunophenotypes are associated with ghost morphology. The most immature immunophenotypes are associated with later affected regions.


Assuntos
Doença de Alzheimer , Encéfalo , Imunofenotipagem , Emaranhados Neurofibrilares , Proteínas tau , Doença de Alzheimer/patologia , Humanos , Emaranhados Neurofibrilares/patologia , Proteínas tau/metabolismo , Masculino , Encéfalo/patologia , Feminino , Idoso de 80 Anos ou mais , Idoso , Imuno-Histoquímica
18.
Acta Neuropathol ; 147(1): 101, 2024 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-38884806

RESUMO

Insoluble pathogenic proteins accumulate along blood vessels in conditions of cerebral amyloid angiopathy (CAA), exerting a toxic effect on vascular cells and impacting cerebral homeostasis. In this work, we provide new evidence from three-dimensional human brain histology that tau protein, the main component of neurofibrillary tangles, can similarly accumulate along brain vascular segments. We quantitatively assessed n = 6 Alzheimer's disease (AD), and n = 6 normal aging control brains and saw that tau-positive blood vessel segments were present in all AD cases. Tau-positive vessels are enriched for tau at levels higher than the surrounding tissue and appear to affect arterioles across cortical layers (I-V). Further, vessels isolated from these AD tissues were enriched for N-terminal tau and tau phosphorylated at T181 and T217. Importantly, tau-positive vessels are associated with local areas of increased tau neurofibrillary tangles. This suggests that accumulation of tau around blood vessels may reflect a local clearance failure. In sum, these data indicate that tau, like amyloid beta, accumulates along blood vessels and may exert a significant influence on vasculature in the setting of AD.


Assuntos
Doença de Alzheimer , Encéfalo , Emaranhados Neurofibrilares , Proteínas tau , Humanos , Doença de Alzheimer/patologia , Doença de Alzheimer/metabolismo , Proteínas tau/metabolismo , Emaranhados Neurofibrilares/patologia , Emaranhados Neurofibrilares/metabolismo , Encéfalo/patologia , Encéfalo/metabolismo , Feminino , Masculino , Idoso , Idoso de 80 Anos ou mais , Pessoa de Meia-Idade , Fosforilação
19.
J Neuropathol Exp Neurol ; 83(9): 752-762, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38812098

RESUMO

Neuropathological diagnosis of Alzheimer disease (AD) relies on semiquantitative analysis of phosphorylated tau-positive neurofibrillary tangles (NFTs) and neuritic plaques (NPs), without consideration of lesion heterogeneity in individual cases. We developed a deep learning workflow for automated annotation and segmentation of NPs and NFTs from AT8-immunostained whole slide images (WSIs) of AD brain sections. Fifteen WSIs of frontal cortex from 4 biobanks with varying tissue quality, staining intensity, and scanning formats were analyzed. We established an artificial intelligence (AI)-driven iterative procedure to improve the generation of expert-validated annotation datasets for NPs and NFTs thereby increasing annotation quality by >50%. This strategy yielded an expert-validated annotation database with 5013 NPs and 5143 NFTs. We next trained two U-Net convolutional neural networks for detection and segmentation of NPs or NFTs, achieving high accuracy and consistency (mean Dice similarity coefficient: NPs, 0.77; NFTs, 0.81). The workflow showed high generalization performance across different cases. This study serves as a proof-of-concept for the utilization of proprietary image analysis software (Visiopharm) in the automated deep learning segmentation of NPs and NFTs, demonstrating that AI can significantly improve the annotation quality of complex neuropathological features and enable the creation of highly precise models for identifying these markers in AD brain sections.


Assuntos
Doença de Alzheimer , Encéfalo , Aprendizado Profundo , Emaranhados Neurofibrilares , Placa Amiloide , Software , Doença de Alzheimer/patologia , Doença de Alzheimer/diagnóstico por imagem , Humanos , Emaranhados Neurofibrilares/patologia , Placa Amiloide/patologia , Encéfalo/patologia , Encéfalo/diagnóstico por imagem , Idoso , Feminino , Processamento de Imagem Assistida por Computador/métodos , Masculino
20.
Front Neurosci ; 18: 1373410, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38765673

RESUMO

Tauopathies are characterized by the abnormal buildup of tau protein, with early oligomeric forms associated with neurodegeneration and the later neurofibrillary tangles possibly conferring neuroprotection. The molecular mechanisms governing the formation of these tau species are unclear. Lately, there has been an increased focus on examining the interactions between tau and other proteins, along with their influence on the aggregation of tau. Our previous work revealed EFhd2's association with pathological tau in animal models and tauopathy brains. Herein, we examined the impact of EFhd2 on monomeric and filamentous tau in vitro. The results demonstrated that EFhd2 incubation with monomeric full length human tau (hTau40) formed amorphous aggregates, where both EFhd2 and hTau40 colocalized. Moreover, EFhd2 is entangled with arachidonic acid (ARA)-induced filamentous hTau40. Furthermore, EFhd2-induced aggregation with monomeric and filamentous hTau40 is EFhd2 concentration dependent. Using sandwich ELISA assays, we assessed the reactivity of TOC1 and Alz50-two conformation-specific tau antibodies-to EFhd2-hTau40 aggregates (in absence and presence of ARA). No TOC1 signal was detected in EFhd2 aggregates with monomeric hTau40 whereas EFhd2 aggregates with hTau in the presence of ARA showed a higher signal compared to hTau40 filaments. In contrast, EFhd2 aggregates with both monomeric and filamentous hTau40 reduced Alz50 reactivity. Taken together, our results illustrate for the first time that EFhd2, a tau-associated protein, interacts with monomeric and filamentous hTau40 to form large aggregates that are starkly different from tau oligomers and filaments. Given these findings and previous research, we hypothesize that EFhd2 may play a role in the formation of tau aggregates. Nevertheless, further in vivo studies are imperative to test this hypothesis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA