scholarly journals Cellular senescence at the crossroads of inflammation and Alzheimer's disease

Author(s):  
Ana Guerrero ◽  
Bart De Strooper ◽  
I. Lorena Arancibia-Cárcamo
2013 ◽  
Vol 19 (9) ◽  
pp. 1727-1738 ◽  
Author(s):  
Tiziana Casoli ◽  
Marta Balietti ◽  
Belinda Giorgetti ◽  
Moreno Solazzi ◽  
Osvaldo Scarpino ◽  
...  

2016 ◽  
Vol 27 (8) ◽  
pp. 813-825 ◽  
Author(s):  
Siew Ying Wong ◽  
Bor Luen Tang

AbstractAlzheimer’s disease (AD) is the most prevalent cause of dementia in the aging population worldwide. SIRT1 deacetylation of histones and transcription factors impinge on multiple neuronal and non-neuronal targets, and modulates stress response, energy metabolism and cellular senescence/death pathways. Collectively, SIRT1 activity could potentially affect multiple aspects of hippocampal and cortical neuron function and survival, thus modifying disease onset and progression. In this review, the known and potential mechanisms of action of SIRT1 with regard to AD, and its potential as a therapeutic target, are discussed.


2013 ◽  
Vol 19 (9) ◽  
pp. 1727-1738
Author(s):  
Tiziana Casoli ◽  
Marta Balietti ◽  
Belinda Giorgetti ◽  
Moreno Solazzi ◽  
Osvaldo Scarpino ◽  
...  

2019 ◽  
Vol 3 (Supplement_1) ◽  
pp. S91-S92
Author(s):  
Angela Olson ◽  
Stacy A Hussong ◽  
Rakez Kayed ◽  
Veronica Galvan

Abstract Chronic sterile inflammation is a pathological feature of Alzheimer’s disease (AD) and other neurodegenerative diseases. The mechanisms that drive neuroinflammation and its impact on AD progression are still incompletely understood. The accumulation of molecular damage in somatic cells can trigger cellular senescence, an irreversible state of cell cycle arrest accompanied by the expression of proinflammatory mediators known collectively as the “senescence-associated secretory phenotype” (SASP). Misfolded tau forms pathogenic soluble aggregates that are released extracellularly and are transmitted trans-neuronally, promoting native tau phosphorylation and aggregation in target cells. We recently showed that, in addition to neurons, pathogenic soluble extracellular tau aggregates propagate to brain microvascular endothelial cells, where microtubule destabilization triggers senescence/SASP. Because astrocytes have a critical role in the regulation of both synaptic function and cerebral blood flow and are directly exposed to tau at its site of release at the tripartite synapse, we conducted studies to define whether soluble aggregated tau propagates to astrocytes, inducing astrocyte senescence/SASP and neuronal dysfunction/damage. Our studies indicate that tau can be propagated transcellularly to astrocytes, triggering cellular senescence/SASP. Our studies suggest that astrocyte senescence is detrimental to dendritic and synaptic structure and density, suggesting that pathogenic soluble tau-induced astrocyte senescence may contribute to synaptic dysfunction and loss in AD. Drugs that eliminate senescent cells are FDA-approved and antibody-based approaches to remove tau from brain are already in clinical trials. Our studies suggest that these interventions could be effective in the treatment of AD and other tauopathies.


Author(s):  
Angeles Tecalco–Cruz ◽  
Jesús Zepeda–Cervantes ◽  
Lilia López-Canovas ◽  
Josué O. Ramírez–Jarquín ◽  
José Pedraza-Chaverrí ◽  
...  

: Alzheimer's disease (AD) is characterized by progressive memory loss due to neurodegeneration that occurs mainly during aging. The accumulation of senescent cells has been related to aging. Furthermore, the expression of the variant ApoE ∈ 4 is a critical risk factor for AD. Some events that occur in senescence, such as the secretion of pro-inflammatory molecules, and metabolic and epigenetic changes, in addition to the detection of ApoE4, may accelerate the progression of AD. Here, we discuss the implications of cellular senescence and the presence of ApoE polymorphisms in AD. Molecular studies of these risk factors for AD may hence be pivotal to define new biomarkers and novel therapeutic strategies for this neurodegenerative pathology.


2021 ◽  
Vol 118 (37) ◽  
pp. e2011226118
Author(s):  
Yujun Hou ◽  
Yong Wei ◽  
Sofie Lautrup ◽  
Beimeng Yang ◽  
Yue Wang ◽  
...  

Alzheimer's disease (AD) is a progressive and fatal neurodegenerative disorder. Impaired neuronal bioenergetics and neuroinflammation are thought to play key roles in the progression of AD, but their interplay is not clear. Nicotinamide adenine dinucleotide (NAD+) is an important metabolite in all human cells in which it is pivotal for multiple processes including DNA repair and mitophagy, both of which are impaired in AD neurons. Here, we report that levels of NAD+ are reduced and markers of inflammation increased in the brains of APP/PS1 mutant transgenic mice with beta-amyloid pathology. Treatment of APP/PS1 mutant mice with the NAD+ precursor nicotinamide riboside (NR) for 5 mo increased brain NAD+ levels, reduced expression of proinflammatory cytokines, and decreased activation of microglia and astrocytes. NR treatment also reduced NLRP3 inflammasome expression, DNA damage, apoptosis, and cellular senescence in the AD mouse brains. Activation of cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) are associated with DNA damage and senescence. cGAS–STING elevation was observed in the AD mice and normalized by NR treatment. Cell culture experiments using microglia suggested that the beneficial effects of NR are, in part, through a cGAS–STING-dependent pathway. Levels of ectopic (cytoplasmic) DNA were increased in APP/PS1 mutant mice and human AD fibroblasts and down-regulated by NR. NR treatment induced mitophagy and improved cognitive and synaptic functions in APP/PS1 mutant mice. Our findings suggest a role for NAD+ depletion-mediated activation of cGAS–STING in neuroinflammation and cellular senescence in AD.


2019 ◽  
Author(s):  
Shoshik Amram ◽  
Tal Iram ◽  
Ekaterina Lazdon ◽  
Robert Vassar ◽  
Ittai Ben-Porath ◽  
...  

ABSTRACTAlterations in astrocyte function such as a pro-inflammatory phenotype are associated with Alzheimer’s disease (AD). We had shown impairments in the ability of aged astrocytes isolated from 5xFAD mice to clear and uptake amyloid-β (Aβ) as well as to support neuronal growth. Senescent cells accumulate with age and exhibit a senescence-associated secretory phenotype, which includes secretion of pro-inflammatory cytokines. In this study, we predicted that with age, astrocytes in 5xFAD mice would exhibit a cellular senescence phenotype that could promote neurodegeneration. We found an age-dependent increase in senescent astrocytes adjacent to Aβ plaques in 5xFAD mice. Inhibition of nuclear factor kappa-light-chain-enhancer of activated B cells reduced interelukin-6 secretion by senescent astrocytes and resulted in improved neuronal support. Moreover, senescent astrocytes exhibited an increase in the induction of the TGF-β1-SMAD2/3 pathway, and inhibition of this pathway resulted in a reduction of cellular senescence. We also discovered that soluble Aβ42 induced astrocyte senescence in young naïve mice in a SMAD2/3-dependent manner. Our results suggest an important role of astrocyte senescence in AD and its role in mediating the neurotoxicity properties of astrocytes in AD and related neurodegenerative diseases.


2015 ◽  
Vol 22 ◽  
pp. 1-8 ◽  
Author(s):  
Virginia Boccardi ◽  
Luca Pelini ◽  
Sara Ercolani ◽  
Carmelinda Ruggiero ◽  
Patrizia Mecocci

Cell Reports ◽  
2021 ◽  
Vol 36 (3) ◽  
pp. 109419
Author(s):  
Sagar Gaikwad ◽  
Nicha Puangmalai ◽  
Alice Bittar ◽  
Mauro Montalbano ◽  
Stephanie Garcia ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document