Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42.779
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Annu Rev Immunol ; 42(1): 585-613, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38424470

RESUMO

Alzheimer disease (AD) is the most common neurodegenerative disease, and with no efficient curative treatment available, its medical, social, and economic burdens are expected to dramatically increase. AD is historically characterized by amyloid ß (Aß) plaques and tau neurofibrillary tangles, but over the last 25 years chronic immune activation has been identified as an important factor contributing to AD pathogenesis. In this article, we review recent and important advances in our understanding of the significance of immune activation in the development of AD. We describe how brain-resident macrophages, the microglia, are able to detect Aß species and be activated, as well as the consequences of activated microglia in AD pathogenesis. We discuss transcriptional changes of microglia in AD, their unique heterogeneity in humans, and emerging strategies to study human microglia. Finally, we expose, beyond Aß and microglia, the role of peripheral signals and different cell types in immune activation.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Microglia , Doença de Alzheimer/imunologia , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Humanos , Animais , Microglia/imunologia , Microglia/metabolismo , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/imunologia , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/patologia , Macrófagos/imunologia , Macrófagos/metabolismo
2.
Cell ; 187(2): 428-445.e20, 2024 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-38086389

RESUMO

A recent case report described an individual who was a homozygous carrier of the APOE3 Christchurch (APOE3ch) mutation and resistant to autosomal dominant Alzheimer's Disease (AD) caused by a PSEN1-E280A mutation. Whether APOE3ch contributed to the protective effect remains unclear. We generated a humanized APOE3ch knock-in mouse and crossed it to an amyloid-ß (Aß) plaque-depositing model. We injected AD-tau brain extract to investigate tau seeding and spreading in the presence or absence of amyloid. Similar to the case report, APOE3ch expression resulted in peripheral dyslipidemia and a marked reduction in plaque-associated tau pathology. Additionally, we observed decreased amyloid response and enhanced microglial response around plaques. We also demonstrate increased myeloid cell phagocytosis and degradation of tau aggregates linked to weaker APOE3ch binding to heparin sulfate proteoglycans. APOE3ch influences the microglial response to Aß plaques, which suppresses Aß-induced tau seeding and spreading. The results reveal new possibilities to target Aß-induced tauopathy.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Apolipoproteína E3 , Proteínas tau , Animais , Humanos , Camundongos , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Proteínas Amiloidogênicas/metabolismo , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Encéfalo/metabolismo , Modelos Animais de Doenças , Camundongos Transgênicos , Microglia/metabolismo , Placa Amiloide/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Relatos de Casos como Assunto
3.
Cell ; 186(22): 4757-4772, 2023 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-37848035

RESUMO

Alzheimer's disease (AD) research has entered a new era with the recent positive phase 3 clinical trials of the anti-Aß antibodies lecanemab and donanemab. Why did it take 30 years to achieve these successes? Developing potent therapies for reducing fibrillar amyloid was key, as was selection of patients at relatively early stages of disease. Biomarkers of the target pathologies, including amyloid and tau PET, and insights from past trials were also critical to the recent successes. Moving forward, the challenge will be to develop more efficacious therapies with greater efficiency. Novel trial designs, including combination therapies and umbrella and basket protocols, will accelerate clinical development. Better diversity and inclusivity of trial participants are needed, and blood-based biomarkers may help to improve access for medically underserved groups. Incentivizing innovation in both academia and industry through public-private partnerships, collaborative mechanisms, and the creation of new career paths will be critical to build momentum in these exciting times.


Assuntos
Doença de Alzheimer , Desenvolvimento de Medicamentos , Humanos , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides , Proteínas Amiloidogênicas , Anticorpos Monoclonais/uso terapêutico , Biomarcadores , Ensaios Clínicos como Assunto
4.
Cell ; 186(20): 4260-4270, 2023 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-37729908

RESUMO

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.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/imunologia , Doença de Alzheimer/prevenção & controle , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides , Anticorpos/uso terapêutico , Imunoterapia , Animais
5.
Cell ; 186(20): 4438-4453.e23, 2023 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-37774681

RESUMO

Cellular perturbations underlying Alzheimer's disease (AD) are primarily studied in human postmortem samples and model organisms. Here, we generated a single-nucleus atlas from a rare cohort of cortical biopsies from living individuals with varying degrees of AD pathology. We next performed a systematic cross-disease and cross-species integrative analysis to identify a set of cell states that are specific to early AD pathology. These changes-which we refer to as the early cortical amyloid response-were prominent in neurons, wherein we identified a transitional hyperactive state preceding the loss of excitatory neurons, which we confirmed by acute slice physiology on independent biopsy specimens. Microglia overexpressing neuroinflammatory-related processes also expanded as AD pathology increased. Finally, both oligodendrocytes and pyramidal neurons upregulated genes associated with ß-amyloid production and processing during this early hyperactive phase. Our integrative analysis provides an organizing framework for targeting circuit dysfunction, neuroinflammation, and amyloid production early in AD pathogenesis.


Assuntos
Doença de Alzheimer , Lobo Frontal , Microglia , Neurônios , Humanos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Amiloide , Peptídeos beta-Amiloides/metabolismo , Microglia/patologia , Neurônios/patologia , Células Piramidais , Biópsia , Lobo Frontal/patologia , Análise da Expressão Gênica de Célula Única , Núcleo Celular/metabolismo , Núcleo Celular/patologia
6.
Cell ; 185(22): 4135-4152.e22, 2022 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-36257314

RESUMO

Recent studies have begun to reveal critical roles for the brain's professional phagocytes, microglia, and their receptors in the control of neurotoxic amyloid beta (Aß) and myelin debris accumulation in neurodegenerative disease. However, the critical intracellular molecules that orchestrate neuroprotective functions of microglia remain poorly understood. In our studies, we find that targeted deletion of SYK in microglia leads to exacerbated Aß deposition, aggravated neuropathology, and cognitive defects in the 5xFAD mouse model of Alzheimer's disease (AD). Disruption of SYK signaling in this AD model was further shown to impede the development of disease-associated microglia (DAM), alter AKT/GSK3ß-signaling, and restrict Aß phagocytosis by microglia. Conversely, receptor-mediated activation of SYK limits Aß load. We also found that SYK critically regulates microglial phagocytosis and DAM acquisition in demyelinating disease. Collectively, these results broaden our understanding of the key innate immune signaling molecules that instruct beneficial microglial functions in response to neurotoxic material.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Animais , Camundongos , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides , Modelos Animais de Doenças , Camundongos Transgênicos , Microglia/patologia , Fagocitose
7.
Cell ; 185(26): 5040-5058.e19, 2022 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-36563667

RESUMO

Spatial molecular profiling of complex tissues is essential to investigate cellular function in physiological and pathological states. However, methods for molecular analysis of large biological specimens imaged in 3D are lacking. Here, we present DISCO-MS, a technology that combines whole-organ/whole-organism clearing and imaging, deep-learning-based image analysis, robotic tissue extraction, and ultra-high-sensitivity mass spectrometry. DISCO-MS yielded proteome data indistinguishable from uncleared samples in both rodent and human tissues. We used DISCO-MS to investigate microglia activation along axonal tracts after brain injury and characterized early- and late-stage individual amyloid-beta plaques in a mouse model of Alzheimer's disease. DISCO-bot robotic sample extraction enabled us to study the regional heterogeneity of immune cells in intact mouse bodies and aortic plaques in a complete human heart. DISCO-MS enables unbiased proteome analysis of preclinical and clinical tissues after unbiased imaging of entire specimens in 3D, identifying diagnostic and therapeutic opportunities for complex diseases. VIDEO ABSTRACT.


Assuntos
Doença de Alzheimer , Proteoma , Camundongos , Humanos , Animais , Proteoma/análise , Proteômica/métodos , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides , Espectrometria de Massas , Placa Amiloide
8.
Cell ; 185(22): 4153-4169.e19, 2022 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-36306735

RESUMO

Genetic studies have highlighted microglia as pivotal in orchestrating Alzheimer's disease (AD). Microglia that adhere to Aß plaques acquire a transcriptional signature, "disease-associated microglia" (DAM), which largely emanates from the TREM2-DAP12 receptor complex that transmits intracellular signals through the protein tyrosine kinase SYK. The human TREM2R47H variant associated with high AD risk fails to activate microglia via SYK. We found that SYK-deficient microglia cannot encase Aß plaques, accelerating brain pathology and behavioral deficits. SYK deficiency impaired the PI3K-AKT-GSK-3ß-mTOR pathway, incapacitating anabolic support required for attaining the DAM profile. However, SYK-deficient microglia proliferated and advanced to an Apoe-expressing prodromal stage of DAM; this pathway relied on the adapter DAP10, which also binds TREM2. Thus, microglial responses to Aß involve non-redundant SYK- and DAP10-pathways. Systemic administration of an antibody against CLEC7A, a receptor that directly activates SYK, rescued microglia activation in mice expressing the TREM2R47H allele, unveiling new options for AD immunotherapy.


Assuntos
Doença de Alzheimer , Microglia , Animais , Camundongos , Humanos , Microglia/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Peptídeos beta-Amiloides/metabolismo , Doença de Alzheimer/patologia , Placa Amiloide/metabolismo , Encéfalo/metabolismo , Modelos Animais de Doenças , Quinase Syk/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Receptores Imunológicos/metabolismo
9.
Nat Immunol ; 24(11): 1839-1853, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37749326

RESUMO

The APOE4 allele is the strongest genetic risk factor for late-onset Alzheimer's disease (AD). The contribution of microglial APOE4 to AD pathogenesis is unknown, although APOE has the most enriched gene expression in neurodegenerative microglia (MGnD). Here, we show in mice and humans a negative role of microglial APOE4 in the induction of the MGnD response to neurodegeneration. Deletion of microglial APOE4 restores the MGnD phenotype associated with neuroprotection in P301S tau transgenic mice and decreases pathology in APP/PS1 mice. MGnD-astrocyte cross-talk associated with ß-amyloid (Aß) plaque encapsulation and clearance are mediated via LGALS3 signaling following microglial APOE4 deletion. In the brains of AD donors carrying the APOE4 allele, we found a sex-dependent reciprocal induction of AD risk factors associated with suppression of MGnD genes in females, including LGALS3, compared to individuals homozygous for the APOE3 allele. Mechanistically, APOE4-mediated induction of ITGB8-transforming growth factor-ß (TGFß) signaling impairs the MGnD response via upregulation of microglial homeostatic checkpoints, including Inpp5d, in mice. Deletion of Inpp5d in microglia restores MGnD-astrocyte cross-talk and facilitates plaque clearance in APP/PS1 mice. We identify the microglial APOE4-ITGB8-TGFß pathway as a negative regulator of microglial response to AD pathology, and restoring the MGnD phenotype via blocking ITGB8-TGFß signaling provides a promising therapeutic intervention for AD.


Assuntos
Doença de Alzheimer , Feminino , Camundongos , Humanos , Animais , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Microglia/metabolismo , Galectina 3/genética , Galectina 3/metabolismo , Peptídeos beta-Amiloides/metabolismo , Camundongos Transgênicos , Modelos Animais de Doenças
10.
Cell ; 182(4): 976-991.e19, 2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32702314

RESUMO

Although complex inflammatory-like alterations are observed around the amyloid plaques of Alzheimer's disease (AD), little is known about the molecular changes and cellular interactions that characterize this response. We investigate here, in an AD mouse model, the transcriptional changes occurring in tissue domains in a 100-µm diameter around amyloid plaques using spatial transcriptomics. We demonstrate early alterations in a gene co-expression network enriched for myelin and oligodendrocyte genes (OLIGs), whereas a multicellular gene co-expression network of plaque-induced genes (PIGs) involving the complement system, oxidative stress, lysosomes, and inflammation is prominent in the later phase of the disease. We confirm the majority of the observed alterations at the cellular level using in situ sequencing on mouse and human brain sections. Genome-wide spatial transcriptomics analysis provides an unprecedented approach to untangle the dysregulated cellular network in the vicinity of pathogenic hallmarks of AD and other brain diseases.


Assuntos
Doença de Alzheimer/patologia , Análise de Sequência de DNA/métodos , Transcriptoma , Doença de Alzheimer/genética , Amiloide/metabolismo , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas do Sistema Complemento/genética , Proteínas do Sistema Complemento/metabolismo , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Humanos , Lisossomos/genética , Lisossomos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Bainha de Mielina/genética , Bainha de Mielina/metabolismo , Estresse Oxidativo/genética
11.
Cell ; 179(2): 312-339, 2019 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-31564456

RESUMO

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.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Placa Amiloide/metabolismo , Proteínas tau , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Vacinas contra Alzheimer/farmacologia , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/metabolismo , Animais , Apolipoproteínas E/antagonistas & inibidores , Apolipoproteínas E/metabolismo , Ensaios Clínicos como Assunto , Humanos , Camundongos , Proteínas tau/antagonistas & inibidores , Proteínas tau/metabolismo
12.
Cell ; 177(2): 256-271.e22, 2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30879788

RESUMO

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.


Assuntos
Estimulação Acústica/métodos , Doença de Alzheimer/terapia , Cognição/fisiologia , Doença de Alzheimer/patologia , Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Percepção Auditiva/fisiologia , Encéfalo/metabolismo , Modelos Animais de Doenças , Ritmo Gama/fisiologia , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Placa Amiloide/metabolismo
13.
Cell ; 178(3): 536-551.e14, 2019 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-31257024

RESUMO

The expression of some proteins in the autophagy pathway declines with age, which may impact neurodegeneration in diseases, including Alzheimer's Disease. We have identified a novel non-canonical function of several autophagy proteins in the conjugation of LC3 to Rab5+, clathrin+ endosomes containing ß-amyloid in a process of LC3-associated endocytosis (LANDO). We found that LANDO in microglia is a critical regulator of immune-mediated aggregate removal and microglial activation in a murine model of AD. Mice lacking LANDO but not canonical autophagy in the myeloid compartment or specifically in microglia have a robust increase in pro-inflammatory cytokine production in the hippocampus and increased levels of neurotoxic ß-amyloid. This inflammation and ß-amyloid deposition were associated with reactive microgliosis and tau hyperphosphorylation. LANDO-deficient AD mice displayed accelerated neurodegeneration, impaired neuronal signaling, and memory deficits. Our data support a protective role for LANDO in microglia in neurodegenerative pathologies resulting from ß-amyloid deposition.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Endocitose , Proteínas Associadas aos Microtúbulos/metabolismo , Doença de Alzheimer/metabolismo , Animais , Proteína 5 Relacionada à Autofagia/deficiência , Proteína 5 Relacionada à Autofagia/genética , Proteínas Relacionadas à Autofagia/deficiência , Proteínas Relacionadas à Autofagia/genética , Antígenos CD36/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Microglia/citologia , Microglia/metabolismo , Células RAW 264.7 , Receptores Imunológicos/metabolismo , Receptor 4 Toll-Like/metabolismo
14.
Nat Immunol ; 22(5): 586-594, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33859405

RESUMO

Two microglial TAM receptor tyrosine kinases, Axl and Mer, have been linked to Alzheimer's disease, but their roles in disease have not been tested experimentally. We find that in Alzheimer's disease and its mouse models, induced expression of Axl and Mer in amyloid plaque-associated microglia was coupled to induced plaque decoration by the TAM ligand Gas6 and its co-ligand phosphatidylserine. In the APP/PS1 mouse model of Alzheimer's disease, genetic ablation of Axl and Mer resulted in microglia that were unable to normally detect, respond to, organize or phagocytose amyloid-ß plaques. These major deficits notwithstanding, TAM-deficient APP/PS1 mice developed fewer dense-core plaques than APP/PS1 mice with normal microglia. Our findings reveal that the TAM system is an essential mediator of microglial recognition and engulfment of amyloid plaques and that TAM-driven microglial phagocytosis does not inhibit, but rather promotes, dense-core plaque development.


Assuntos
Doença de Alzheimer/imunologia , Microglia/patologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , c-Mer Tirosina Quinase/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/citologia , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Humanos , Microscopia Intravital , Masculino , Camundongos , Camundongos Knockout , Microglia/imunologia , Microscopia Confocal , Microscopia de Fluorescência por Excitação Multifotônica , Fagocitose/imunologia , Presenilina-1/genética , Proteínas Proto-Oncogênicas/genética , RNA-Seq , Receptores Proteína Tirosina Quinases/genética , Análise de Célula Única , c-Mer Tirosina Quinase/genética , Receptor Tirosina Quinase Axl
15.
Cell ; 169(7): 1249-1262.e13, 2017 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-28622510

RESUMO

Homeostasis of the gut microbiota critically influences host health and aging. Developing genetically engineered probiotics holds great promise as a new therapeutic paradigm to promote healthy aging. Here, through screening 3,983 Escherichia coli mutants, we discovered that 29 bacterial genes, when deleted, increase longevity in the host Caenorhabditis elegans. A dozen of these bacterial mutants also protect the host from age-related progression of tumor growth and amyloid-beta accumulation. Mechanistically, we discovered that five bacterial mutants promote longevity through increased secretion of the polysaccharide colanic acid (CA), which regulates mitochondrial dynamics and unfolded protein response (UPRmt) in the host. Purified CA polymers are sufficient to promote longevity via ATFS-1, the host UPRmt-responsive transcription factor. Furthermore, the mitochondrial changes and longevity effects induced by CA are conserved across different species. Together, our results identified molecular targets for developing pro-longevity microbes and a bacterial metabolite acting on host mitochondria to promote longevity.


Assuntos
Caenorhabditis elegans/microbiologia , Escherichia coli/genética , Longevidade , Envelhecimento/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Carga Bacteriana , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Escherichia coli/metabolismo , Deleção de Genes , Estudo de Associação Genômica Ampla , Dinâmica Mitocondrial , Modelos Animais , Polissacarídeos/metabolismo , Fatores de Transcrição/metabolismo , Resposta a Proteínas não Dobradas
16.
Physiol Rev ; 104(2): 533-587, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-37561137

RESUMO

Lung endothelia in the arteries, capillaries, and veins are heterogeneous in structure and function. Lung capillaries in particular represent a unique vascular niche, with a thin yet highly restrictive alveolar-capillary barrier that optimizes gas exchange. Capillary endothelium surveys the blood while simultaneously interpreting cues initiated within the alveolus and communicated via immediately adjacent type I and type II epithelial cells, fibroblasts, and pericytes. This cell-cell communication is necessary to coordinate the immune response to lower respiratory tract infection. Recent discoveries identify an important role for the microtubule-associated protein tau that is expressed in lung capillary endothelia in the host-pathogen interaction. This endothelial tau stabilizes microtubules necessary for barrier integrity, yet infection drives production of cytotoxic tau variants that are released into the airways and circulation, where they contribute to end-organ dysfunction. Similarly, beta-amyloid is produced during infection. Beta-amyloid has antimicrobial activity, but during infection it can acquire cytotoxic activity that is deleterious to the host. The production and function of these cytotoxic tau and amyloid variants are the subject of this review. Lung-derived cytotoxic tau and amyloid variants are a recently discovered mechanism of end-organ dysfunction, including neurocognitive dysfunction, during and in the aftermath of infection.


Assuntos
Pulmão , Insuficiência de Múltiplos Órgãos , Humanos , Insuficiência de Múltiplos Órgãos/metabolismo , Pulmão/metabolismo , Endotélio Vascular/metabolismo , Amiloide/química , Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo
17.
Physiol Rev ; 104(1): 103-197, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37843394

RESUMO

Alzheimer disease (AD) is associated with multiple etiologies and pathological mechanisms, among which oxidative stress (OS) appears as a major determinant. Intriguingly, OS arises in various pathways regulating brain functions, and it seems to link different hypotheses and mechanisms of AD neuropathology with high fidelity. The brain is particularly vulnerable to oxidative damage, mainly because of its unique lipid composition, resulting in an amplified cascade of redox reactions that target several cellular components/functions ultimately leading to neurodegeneration. The present review highlights the "OS hypothesis of AD," including amyloid beta-peptide-associated mechanisms, the role of lipid and protein oxidation unraveled by redox proteomics, and the antioxidant strategies that have been investigated to modulate the progression of AD. Collected studies from our groups and others have contributed to unraveling the close relationships between perturbation of redox homeostasis in the brain and AD neuropathology by elucidating redox-regulated events potentially involved in both the pathogenesis and progression of AD. However, the complexity of AD pathological mechanisms requires an in-depth understanding of several major intracellular pathways affecting redox homeostasis and relevant for brain functions. This understanding is crucial to developing pharmacological strategies targeting OS-mediated toxicity that may potentially contribute to slow AD progression as well as improve the quality of life of persons with this severe dementing disorder.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Qualidade de Vida , Estresse Oxidativo/fisiologia , Oxirredução , Lipídeos
18.
Immunity ; 55(12): 2236-2254, 2022 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-36351425

RESUMO

Alzheimer's disease (AD) is the most common neurodegenerative disease, with characteristic extracellular amyloid-ß (Aß) deposition and intracellular accumulation of hyperphosphorylated, aggregated tau. Several key regulators of innate immune pathways are genetic risk factors for AD. While these genetic risk factors as well as in vivo data point to key roles for microglia, emerging evidence also points to a role of the adaptive immune response in disease pathogenesis. We review the roles of innate and adaptive immunity, their niches, their communication, and their contributions to AD development and progression. We also summarize the cellular compositions and physiological functions of immune cells in the parenchyma, together with those in the brain border structures that form a dynamic disease-related immune niche. We propose that both innate and adaptive immune responses in brain parenchyma and border structures could serve as important therapeutic targets for treating both the pre-symptomatic and the symptomatic stages of AD.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Humanos , Doença de Alzheimer/genética , Doenças Neurodegenerativas/patologia , Imunidade Inata/fisiologia , Peptídeos beta-Amiloides/metabolismo , Imunidade Adaptativa , Microglia
19.
Immunity ; 55(5): 879-894.e6, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35443157

RESUMO

The principal signals that drive memory and cognitive impairment in Alzheimer's disease (AD) remain elusive. Here, we revealed brain-wide cellular reactions to type I interferon (IFN-I), an innate immune cytokine aberrantly elicited by amyloid ß plaques, and examined their role in cognition and neuropathology relevant to AD in a murine amyloidosis model. Using a fate-mapping reporter system to track cellular responses to IFN-I, we detected robust, Aß-pathology-dependent IFN-I activation in microglia and other cell types. Long-term blockade of IFN-I receptor (IFNAR) rescued both memory and synaptic deficits and resulted in reduced microgliosis, inflammation, and neuritic pathology. Microglia-specific Ifnar1 deletion attenuated the loss of post-synaptic terminals by selective engulfment, whereas neural Ifnar1 deletion restored pre-synaptic terminals and decreased plaque accumulation. Overall, IFN-I signaling represents a critical module within the neuroinflammatory network of AD and prompts concerted cellular states that are detrimental to memory and cognition.


Assuntos
Doença de Alzheimer , Interferon Tipo I , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Modelos Animais de Doenças , Imunidade Inata , Interferon Tipo I/metabolismo , Transtornos da Memória/metabolismo , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Placa Amiloide/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA