Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.221
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 186(20): 4260-4270, 2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37729908

RESUMEN

Recent Aß-immunotherapy trials have yielded the first clear evidence that removing aggregated Aß from the brains of symptomatic patients can slow the progression of Alzheimer's disease. The clinical benefit achieved in these trials has been modest, however, highlighting the need for both a deeper understanding of disease mechanisms and the importance of intervening early in the pathogenic cascade. An immunoprevention strategy for Alzheimer's disease is required that will integrate the findings from clinical trials with mechanistic insights from preclinical disease models to select promising antibodies, optimize the timing of intervention, identify early biomarkers, and mitigate potential side effects.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/prevención & control , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides , Anticuerpos/uso terapéutico , Inmunoterapia , Animales
2.
Cell ; 177(2): 256-271.e22, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30879788

RESUMEN

We previously reported that inducing gamma oscillations with a non-invasive light flicker (gamma entrainment using sensory stimulus or GENUS) impacted pathology in the visual cortex of Alzheimer's disease mouse models. Here, we designed auditory tone stimulation that drove gamma frequency neural activity in auditory cortex (AC) and hippocampal CA1. Seven days of auditory GENUS improved spatial and recognition memory and reduced amyloid in AC and hippocampus of 5XFAD mice. Changes in activation responses were evident in microglia, astrocytes, and vasculature. Auditory GENUS also reduced phosphorylated tau in the P301S tauopathy model. Furthermore, combined auditory and visual GENUS, but not either alone, produced microglial-clustering responses, and decreased amyloid in medial prefrontal cortex. Whole brain analysis using SHIELD revealed widespread reduction of amyloid plaques throughout neocortex after multi-sensory GENUS. Thus, GENUS can be achieved through multiple sensory modalities with wide-ranging effects across multiple brain areas to improve cognitive function.


Asunto(s)
Estimulación Acústica/métodos , Enfermedad de Alzheimer/terapia , Cognición/fisiología , Enfermedad de Alzheimer/patología , Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Percepción Auditiva/fisiología , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Ritmo Gamma/fisiología , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Placa Amiloide/metabolismo
3.
Cell ; 179(2): 312-339, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31564456

RESUMEN

Alzheimer disease (AD) is a heterogeneous disease with a complex pathobiology. The presence of extracellular ß-amyloid deposition as neuritic plaques and intracellular accumulation of hyperphosphorylated tau as neurofibrillary tangles remains the primary neuropathologic criteria for AD diagnosis. However, a number of recent fundamental discoveries highlight important pathological roles for other critical cellular and molecular processes. Despite this, no disease-modifying treatment currently exists, and numerous phase 3 clinical trials have failed to demonstrate benefits. Here, we review recent advances in our understanding of AD pathobiology and discuss current treatment strategies, highlighting recent clinical trials and opportunities for developing future disease-modifying therapies.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Placa Amiloide/metabolismo , Proteínas tau , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/terapia , Vacunas contra el Alzheimer/farmacología , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Animales , Apolipoproteínas E/antagonistas & inhibidores , Apolipoproteínas E/metabolismo , Ensayos Clínicos como Asunto , Humanos , Ratones , Proteínas tau/antagonistas & inhibidores , Proteínas tau/metabolismo
4.
Trends Immunol ; 45(10): 768-782, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39278789

RESUMEN

Recent discoveries of rare variants of human APOE may shed light on novel therapeutic strategies for Alzheimer's disease (AD). Here, we highlight the newly identified protective variant [APOE3 Christchurch (APOE3ch, R136S)] as an example. We summarize human AD and mouse amyloidosis and tauopathy studies from the past 5 years that have been associated with this R136S variant. We also propose a potential mechanism for how this point mutation might lead to protection against AD pathology, from the molecular level, to cells, to mouse models, and potentially, to humans. Lastly, we extend our discussion of the recent insights gained regarding different APOE variants to putative therapeutic approaches in AD.


Asunto(s)
Enfermedad de Alzheimer , Inmunidad Innata , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/terapia , Humanos , Animales , Apolipoproteínas E/genética , Ratones , Modelos Animales de Enfermedad
5.
PLoS Biol ; 21(3): e3002017, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36881554

RESUMEN

Alzheimer's disease (AD) is a heterogeneous disease with complex clinicopathological characteristics. To date, the role of m6A RNA methylation in monocyte-derived macrophages involved in the progression of AD is unknown. In our study, we found that methyltransferase-like 3 (METTL3) deficiency in monocyte-derived macrophages improved cognitive function in an amyloid beta (Aß)-induced AD mouse model. The mechanistic study showed that that METTL3 ablation attenuated the m6A modification in DNA methyltransferase 3A (Dnmt3a) mRNAs and consequently impaired YTH N6-methyladenosine RNA binding protein 1 (YTHDF1)-mediated translation of DNMT3A. We identified that DNMT3A bound to the promoter region of alpha-tubulin acetyltransferase 1 (Atat1) and maintained its expression. METTL3 depletion resulted in the down-regulation of ATAT1, reduced acetylation of α-tubulin and subsequently enhanced migration of monocyte-derived macrophages and Aß clearance, which led to the alleviated symptoms of AD. Collectively, our findings demonstrate that m6A methylation could be a promising target for the treatment of AD in the future.


Asunto(s)
Enfermedad de Alzheimer , Animales , Ratones , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides , Macrófagos , Acetilación , Metiltransferasas/genética
6.
Nucleic Acids Res ; 52(11): 6099-6113, 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38726879

RESUMEN

Divalent short-interfering RNA (siRNA) holds promise as a therapeutic approach allowing for the sequence-specific modulation of a target gene within the central nervous system (CNS). However, an siRNA modality capable of simultaneously modulating gene pairs would be invaluable for treating complex neurodegenerative disorders, where more than one pathway contributes to pathogenesis. Currently, the parameters and scaffold considerations for multi-targeting nucleic acid modalities in the CNS are undefined. Here, we propose a framework for designing unimolecular 'dual-targeting' divalent siRNAs capable of co-silencing two genes in the CNS. We systematically adjusted the original CNS-active divalent siRNA and identified that connecting two sense strands 3' and 5' through an intra-strand linker enabled a functional dual-targeting scaffold, greatly simplifying the synthetic process. Our findings demonstrate that the dual-targeting siRNA supports at least two months of maximal distribution and target silencing in the mouse CNS. The dual-targeting divalent siRNA is highly programmable, enabling simultaneous modulation of two different disease-relevant gene pairs (e.g. Huntington's disease: MSH3 and HTT; Alzheimer's disease: APOE and JAK1) with similar potency to a mixture of single-targeting divalent siRNAs against each gene. This work enhances the potential for CNS modulation of disease-related gene pairs using a unimolecular siRNA.


Asunto(s)
Sistema Nervioso Central , ARN Interferente Pequeño , Animales , Humanos , Ratones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/terapia , Apolipoproteínas E/genética , Sistema Nervioso Central/metabolismo , Silenciador del Gen , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/terapia , Ratones Endogámicos C57BL , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/química
7.
Proc Natl Acad Sci U S A ; 120(42): e2306714120, 2023 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-37816062

RESUMEN

Mutations in the Presenilin (PSEN1 and PSEN2) genes are the major cause of early-onset familial Alzheimer's disease (FAD). Presenilin (PS) is the catalytic subunit of the γ-secretase complex, which cleaves type I transmembrane proteins, such as Notch and the amyloid precursor protein (APP), and plays an evolutionarily conserved role in the protection of neuronal survival during aging. FAD PSEN1 mutations exhibit impaired γ-secretase activity in cell culture, in vitro, and knockin (KI) mouse brains, and the L435F mutation is the most severe in reducing γ-secretase activity and is located closest to the active site of γ-secretase. Here, we report that introduction of the codon-optimized wild-type human PSEN1 cDNA by adeno-associated virus 9 (AAV9) results in broadly distributed, sustained, low to moderate levels of human PS1 (hPS1) expression and rescues impaired γ-secretase activity in the cerebral cortex of Psen mutant mice either lacking PS or expressing the Psen1 L435F KI allele, as evaluated by endogenous γ-secretase substrates of APP and recombinant γ-secretase products of Notch intracellular domain and Aß peptides. Furthermore, introduction of hPS1 by AAV9 alleviates impairments of synaptic plasticity and learning and memory in Psen mutant mice. Importantly, AAV9 delivery of hPS1 ameliorates neurodegeneration in the cerebral cortex of aged Psen mutant mice, as shown by the reversal of age-dependent loss of cortical neurons and elevated microgliosis and astrogliosis. These results together show that moderate hPS1 expression by AAV9 is sufficient to rescue impaired γ-secretase activity, synaptic and memory deficits, and neurodegeneration caused by Psen mutations in mouse models.


Asunto(s)
Enfermedad de Alzheimer , Secretasas de la Proteína Precursora del Amiloide , Humanos , Ratones , Animales , Anciano , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/terapia , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Mutación , Trastornos de la Memoria/genética , Trastornos de la Memoria/terapia , Presenilina-2/genética , Péptidos beta-Amiloides/metabolismo
8.
J Neurosci ; 44(41)2024 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-39384409

RESUMEN

Alzheimer's disease (AD) and Alzheimer's disease-related dementias (ADRDs) are broad-impact multifactorial neurodegenerative diseases. Their complexity presents unique challenges for developing effective therapies. This review highlights research presented at the 2024 Society for Neuroscience meeting which emphasized the gut microbiome's role in AD pathogenesis by influencing brain function and neurodegeneration through the microbiota-gut-brain axis. This emerging evidence underscores the potential for targeting the gut microbiota to treat AD/ADRD.


Asunto(s)
Enfermedad de Alzheimer , Microbioma Gastrointestinal , Humanos , Enfermedad de Alzheimer/microbiología , Enfermedad de Alzheimer/prevención & control , Enfermedad de Alzheimer/terapia , Microbioma Gastrointestinal/fisiología , Animales , Eje Cerebro-Intestino/fisiología , Demencia/prevención & control , Demencia/microbiología , Encéfalo/microbiología
9.
Ann Neurol ; 95(4): 625-634, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38180638

RESUMEN

Alzheimer's disease (AD) is the most common neurodegenerative disorder and one of the leading causes of disability worldwide. The apolipoprotein E4 gene (APOE4) is the strongest genetic risk factor for AD. In 2023, the APOE4 National Institute on Aging/Alzheimer's Disease Sequencing Project working group came together to gather data and discuss the question of whether to reduce or increase APOE4 as a therapeutic intervention for AD. It was the unanimous consensus that cumulative data from multiple studies in humans and animal models support that lowering APOE4 should be a target for therapeutic approaches for APOE4 carriers. ANN NEUROL 2024;95:625-634.


Asunto(s)
Enfermedad de Alzheimer , Animales , Estados Unidos , Humanos , Enfermedad de Alzheimer/terapia , Enfermedad de Alzheimer/tratamiento farmacológico , Apolipoproteína E4/genética , Objetivos , National Institute on Aging (U.S.)
10.
Nat Rev Neurosci ; 21(1): 21-35, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31780819

RESUMEN

The global epidemic of Alzheimer disease (AD) is worsening, and no approved treatment can revert or arrest progression of this disease. AD pathology is characterized by the accumulation of amyloid-ß (Aß) plaques and tau neurofibrillary tangles in the brain. Genetic data, as well as autopsy and neuroimaging studies in patients with AD, indicate that Aß plaque deposition precedes cortical tau pathology. Because Aß accumulation has been considered the initial insult that drives both the accumulation of tau pathology and tau-mediated neurodegeneration in AD, the development of AD therapeutics has focused mostly on removing Aß from the brain. However, striking preclinical evidence from AD mouse models and patient-derived human induced pluripotent stem cell models indicates that tau pathology can progress independently of Aß accumulation and arises downstream of genetic risk factors for AD and aberrant metabolic pathways. This Review outlines novel insights from preclinical research that implicate apolipoprotein E, the endocytic system, cholesterol metabolism and microglial activation as Aß-independent regulators of tau pathology. These factors are discussed in the context of emerging findings from clinical pathology, functional neuroimaging and other approaches in humans. Finally, we discuss the implications of these new insights for current Aß-targeted strategies and highlight the emergence of novel therapeutic strategies that target processes upstream of both Aß and tau.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/terapia , Animales , Apolipoproteínas E/metabolismo , Colesterol/metabolismo , Endocitosis , Humanos , Microglía/metabolismo , Placa Amiloide/patología
11.
PLoS Biol ; 20(7): e3001694, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35862308

RESUMEN

Strong genetic evidence supports an imbalance between production and clearance of amyloid ß-protein (Aß) in people with Alzheimer disease (AD). Microglia that are potentially involved in alternative mechanisms are actually integral to the amyloid cascade. Fluid biomarkers and brain imaging place accumulation of Aß at the beginning of molecular and clinical changes in the disease. So why have clinical trials of anti-amyloid therapies not provided clear-cut benefits to patients with AD? Can anti-amyloid therapies robustly decrease Aß in the human brain, and if so, could this lowering be too little, too late? These central questions in research on AD are being urgently addressed.


Asunto(s)
Enfermedad de Alzheimer , Amiloidosis , Disfunción Cognitiva , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/terapia , Amiloide , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Disfunción Cognitiva/terapia , Humanos
12.
PLoS Comput Biol ; 20(1): e1011164, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38232116

RESUMEN

Transcranial direct current stimulation (tDCS) is a non-invasive brain stimulation technique with potential for counteracting disrupted brain network activity in Alzheimer's disease (AD) to improve cognition. However, the results of tDCS studies in AD have been variable due to different methodological choices such as electrode placement. To address this, a virtual brain network model of AD was used to explore tDCS optimization. We compared a large, representative set of virtual tDCS intervention setups, to identify the theoretically optimized tDCS electrode positions for restoring functional network features disrupted in AD. We simulated 20 tDCS setups using a computational dynamic network model of 78 neural masses coupled according to human structural topology. AD network damage was simulated using an activity-dependent degeneration algorithm. Current flow modeling was used to estimate tDCS-targeted cortical regions for different electrode positions, and excitability of the pyramidal neurons of the corresponding neural masses was modulated to simulate tDCS. Outcome measures were relative power spectral density (alpha bands, 8-10 Hz and 10-13 Hz), total spectral power, posterior alpha peak frequency, and connectivity measures phase lag index (PLI) and amplitude envelope correlation (AEC). Virtual tDCS performance varied, with optimized strategies improving all outcome measures, while others caused further deterioration. The best performing setup involved right parietal anodal stimulation, with a contralateral supraorbital cathode. A clear correlation between the network role of stimulated regions and tDCS success was not observed. This modeling-informed approach can guide and perhaps accelerate tDCS therapy development and enhance our understanding of tDCS effects. Follow-up studies will compare the general predictions to personalized virtual models and validate them with tDCS-magnetoencephalography (MEG) in a clinical AD patient cohort.


Asunto(s)
Enfermedad de Alzheimer , Estimulación Transcraneal de Corriente Directa , Humanos , Enfermedad de Alzheimer/terapia , Estimulación Transcraneal de Corriente Directa/métodos , Encéfalo/fisiología , Magnetoencefalografía , Redes Neurales de la Computación
15.
Cereb Cortex ; 34(3)2024 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-38517175

RESUMEN

Intermittent theta-burst stimulation (iTBS) is emerging as a noninvasive therapeutic strategy for Alzheimer's disease (AD). Recent advances highlighted a new accelerated iTBS (aiTBS) protocol, consisting of multiple sessions per day and higher overall pulse doses, in brain modulation. To examine the possibility of applying the aiTBS in treating AD patients, we enrolled 45 patients in AD at early clinical stages, and they were randomly assigned to either receive real or sham aiTBS. Neuropsychological scores were evaluated before and after treatment. Moreover, we detected cortical excitability and oscillatory activity changes in AD, by the single-pulse TMS in combination with EEG (TMS-EEG). Real stimulation showed markedly better performances in the group average of Auditory Verbal Learning Test scores compared to baseline. TMS-EEG revealed that aiTBS has reinforced this memory-related cortical mechanism by increasing cortical excitability and beta oscillatory activity underlying TMS target. We also found an enhancement of local natural frequency after aiTBS treatment. The novel findings implicated that high-dose aiTBS targeting left DLPFC is rapid-acting, safe, and tolerable in AD patients. Furthermore, TMS-related increase of specific neural oscillation elucidates the mechanisms of the AD cognitive impairment ameliorated by aiTBS.


Asunto(s)
Enfermedad de Alzheimer , Estimulación Magnética Transcraneal , Humanos , Estimulación Magnética Transcraneal/métodos , Enfermedad de Alzheimer/terapia , Corteza Prefrontal/fisiología , Encéfalo , Corteza Prefontal Dorsolateral
16.
Cereb Cortex ; 34(1)2024 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-38037857

RESUMEN

Repetitive transcranial magnetic stimulation (rTMS) and cognitive training for patients with Alzheimer's disease (AD) can change functional connectivity (FC) within gray matter (GM). However, the role of white matter (WM) and changes of GM-WM FC under these therapies are still unclear. To clarify this problem, we applied 40 Hz rTMS over angular gyrus (AG) concurrent with cognitive training to 15 mild-moderate AD patients and analyzed the resting-state functional magnetic resonance imaging before and after treatment. Through AG-based FC analysis, corona radiata and superior longitudinal fasciculus (SLF) were identified as activated WM tracts. Compared with the GM results with AG as seed, more GM regions were found with activated WM tracts as seeds. The averaged FC, fractional amplitude of low-frequency fluctuation (fALFF), and regional homogeneity (ReHo) of the above GM regions had stronger clinical correlations (r/P = 0.363/0.048 vs 0.299/0.108, 0.351/0.057 vs 0.267/0.153, 0.420/0.021 vs 0.408/0.025, for FC/fALFF/ReHo, respectively) and better classification performance to distinguish pre-/post-treatment groups (AUC = 0.91 vs 0.88, 0.65 vs 0.63, 0.87 vs 0.82, for FC/fALFF/ReHo, respectively). Our results indicated that rTMS concurrent with cognitive training could rewire brain network by enhancing GM-WM FC in AD, and corona radiata and SLF played an important role in this process.


Asunto(s)
Enfermedad de Alzheimer , Sustancia Blanca , Humanos , Sustancia Gris/patología , Sustancia Blanca/patología , Estimulación Magnética Transcraneal , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/terapia , Enfermedad de Alzheimer/patología , Entrenamiento Cognitivo , Imagen por Resonancia Magnética/métodos , Encéfalo
17.
Cereb Cortex ; 34(10)2024 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-39393920

RESUMEN

Neurovascular coupling plays an important role in the progression of Alzheimer's disease. However, it is unclear how ultrasound stimulation modulates neurovascular coupling in Alzheimer's disease. Here, we found that (i) transcranial ultrasound stimulation modulates the time domain and frequency domain characteristics of cerebral blood oxygen metabolism in Alzheimer's disease mice; (ii) transcranial ultrasound stimulation can significantly modulate the relative power of theta and gamma frequency of local field potential in Alzheimer's disease mice; and (iii) transcranial ultrasound stimulation can significantly modulate the neurovascular coupling in time domain and frequency domain induced by forepaw electrical stimulation in Alzheimer's disease mice. It provides a research basis for the clinical application of transcranial ultrasound stimulation in Alzheimer's disease patients.


Asunto(s)
Enfermedad de Alzheimer , Circulación Cerebrovascular , Modelos Animales de Enfermedad , Ratones Transgénicos , Acoplamiento Neurovascular , Animales , Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/terapia , Enfermedad de Alzheimer/metabolismo , Acoplamiento Neurovascular/fisiología , Circulación Cerebrovascular/fisiología , Ratones , Masculino , Ratones Endogámicos C57BL , Encéfalo/fisiopatología , Encéfalo/metabolismo , Estimulación Eléctrica/métodos
18.
Mol Ther ; 32(5): 1373-1386, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38504517

RESUMEN

Epidemiological studies show that individuals who carry the relatively uncommon APOE ε2 allele rarely develop Alzheimer disease, and if they do, they have a later age of onset, milder clinical course, and less severe neuropathological findings than people without this allele. The contrast is especially stark when compared with the major genetic risk factor for Alzheimer disease, APOE ε4, which has an age of onset several decades earlier, a more aggressive clinical course and more severe neuropathological findings, especially in terms of the amount of amyloid deposition. Here, we demonstrate that brain exposure to APOE ε2 via a gene therapy approach, which bathes the entire cortical mantle in the gene product after transduction of the ependyma, reduces Aß plaque deposition, neurodegenerative synaptic loss, and, remarkably, reduces microglial activation in an APP/PS1 mouse model despite continued expression of human APOE ε4. This result suggests a promising protective effect of exogenous APOE ε2 and reveals a cell nonautonomous effect of the protein on microglial activation, which we show is similar to plaque-associated microglia in the brain of Alzheimer disease patients who inherit APOE ε2. These data increase the potential that an APOE ε2 therapeutic could be effective in Alzheimer disease, even in individuals born with the risky ε4 allele.


Asunto(s)
Enfermedad de Alzheimer , Apolipoproteína E2 , Modelos Animales de Enfermedad , Terapia Genética , Ratones Transgénicos , Microglía , Placa Amiloide , Animales , Enfermedad de Alzheimer/terapia , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/etiología , Ratones , Terapia Genética/métodos , Humanos , Apolipoproteína E2/genética , Apolipoproteína E2/metabolismo , Placa Amiloide/metabolismo , Placa Amiloide/patología , Microglía/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Enfermedades Neuroinflamatorias/etiología , Enfermedades Neuroinflamatorias/terapia , Enfermedades Neuroinflamatorias/metabolismo , Péptidos beta-Amiloides/metabolismo , Biomarcadores
19.
Mol Cell Neurosci ; 128: 103917, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38244651

RESUMEN

Triggering receptor expressed on myeloid cells 2 (TREM2) is a membrane glycoprotein that plays a crucial role in the regulation of microglial survival, activation, phagocytosis, as well as in the maintenance of brain homeostasis and the inflammatory response to injury or neurodegeneration. This review provides a comprehensive overview of TREM2 structure and functions, highlighting the role of its variants in the development and progression of Alzheimer's disease (AD), a devastating neurodegenerative disease that affects millions of people worldwide. Additionally, the article discusses the potential of TREM2 as a therapeutic target in AD, analyzing the current state of research and future prospects. Given the significant challenges associated with the activation of TREM2, particularly due to its diverse isoforms and the delicate balance required to modulate the immune response without triggering hyperactivation, this review aims to enhance our understanding of TREM2 in AD and inspire further research into this promising yet challenging therapeutic target.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Humanos , Enfermedad de Alzheimer/terapia , Microglía/metabolismo , Encéfalo/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Péptidos beta-Amiloides , Receptores Inmunológicos/genética
20.
J Cell Mol Med ; 28(15): e18554, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39103747

RESUMEN

Alzheimer's disease (AD) is a progressive disease that causes an impairment of learning and memory. Despite the highly complex pathogenesis of AD, amyloid beta (Aß) deposition and neurofibrillary tangles (NFTs) formation are the main hallmarks of AD. Neuroinflammation also has a crucial role in the development of AD. As the central nervous system's innate immune cells, microglial cells are activated in AD and induce inflammation by producing pro-inflammatory mediators. However, microglial activation is not always deleterious. M2-activated microglial cells are considered anti-inflammatory cells, which develop neuroprotection. Various approaches are proposed for managing AD, yet no effective therapy is available for this disorder. Considering the potential protective role of M2 microglia in neurodegenerative disorders and the improvement of these disorders by preconditioning approaches, it can be suggested that preconditioning of microglial cells may be beneficial for managing AD progression. Therefore, this study review microglial preconditioning approaches for preventing and improving AD.


Asunto(s)
Enfermedad de Alzheimer , Microglía , Microglía/metabolismo , Microglía/patología , Enfermedad de Alzheimer/terapia , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/metabolismo , Humanos , Animales , Péptidos beta-Amiloides/metabolismo , Enfermedades Neuroinflamatorias/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA