Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 May 2.
Published in final edited form as: Mech Ageing Dev. 2021 Oct 21;200:111589. doi: 10.1016/j.mad.2021.111589

A geroscience motivated approach to treat Alzheimer’s disease: Senolytics move to clinical trials

Mitzi M Gonzales a, Sudarshan Krishnamurthy b,c, Valentina Garbarino a, Ali S Daeihagh b, Gregory J Gillispie b, Gagan Deep d, Suzanne Craft b,e, Miranda E Orr b,e,f,*
PMCID: PMC9059898  NIHMSID: NIHMS1797561  PMID: 34687726

Abstract

The pathogenic processes driving Alzheimer’s disease (AD) are complex. An incomplete understanding of underlying disease mechanisms has presented insurmountable obstacles for developing effective disease-modifying therapies. Advanced chronological age is the greatest risk factor for developing AD. Intervening on biological aging may alter disease progression and represents a novel, complementary approach to current strategies. Toward this end, cellular senescence has emerged as a promising target. This complex stress response harbors damaged cells in a cell cycle arrested, apoptosis-resistant cell state. Senescent cells accumulate with age where they notoriously secrete molecules that contribute to chronic tissue dysfunction and disease. Thus, benefits of cell survival in a senescent fate are countered by their toxic secretome. The removal of senescent cells improves brain structure and function in rodent models at risk of developing AD, and in those with advanced Aβ and tau pathology. The present review describes the path to translating this promising treatment strategy to AD clinical trials. We review evidence for senescent cell accumulation in the human brain, considerations and strategies for senescence-targeting trials specific to AD, approaches to detect senescent brain cells in biofluids, and summarize the goals of the first senolytic trials for the treatment of AD (NCT04063124 and NCT04685590). This article is part of the Special Issue - Senolytics - Edited by Joao Passos and Diana Jurk.

Keywords: Cellular senescence, Alzheimer’s disease, Biology of aging, Neurodegeneration, Brain, Geroscience, Senolytics, Exosomes, Tauopathy, Clinical trials

1. Introduction

Cellular senescence is an elaborate stress response that involves initiating and maintaining a stable cell cycle arrest. It culminates as a change in cell fate often accompanied with a toxic proinflammatory secretory phenotype (Gorgoulis et al., 2019). Though initially defined as a finite replicative artifact of cells in culture (Hayflick and Moorhead, 1961), an appreciation for the role of senescence in driving aging and disease in vivo has emerged. Recent studies, including by our group (Musi et al., 2018), have demonstrated that postmitotic cells are vulnerable to senescence as well (von Zglinicki et al., 2021). Collectively the data indicate that mitotically competent cells and tissues, as well as those with limited regenerative potential, are vulnerable to the negative impacts of senescent cell accumulation. Specific to the brain, senescent cells contribute to poor brain aging and Alzheimer’s disease (for review, (DiBattista et al., 2020; Saez-Atienzar and Masliah, 2020). Senescence also plays a role in normal tissue repair and development, for recent reviews: (Rhinn et al., 2019; Wilkinson and Hardman, 2020). However, due to their notorious secretome, senescent cells become detrimental when they accumulate with advanced age. They secrete high levels of pro-inflammatory cytokines, chemokines, exosomes, and extracellular matrix-degrading proteins that negatively impact the extracellular environment and induce spread of senescence to non-senescent cells (Acosta et al., 2013; Coppe et al., 2010; Coppé et al., 2008; Kuilman and Peeper, 2009). This toxic secretome, referred to as senescence-associated secretory phenotype (SASP), contributes to inflammation, tissue degradation and ultimately chronic disease (He and Sharpless, 2017).

Several studies have chronicled senescent cell accumulation in the brains of rodents with aging (Jurk et al., 2012), tauopathy (Bussian et al., 2018; Musi et al., 2018), amyloid-β (Aβ) accumulation (Zhang et al., 2019) and other AD-associated risk factors (Table 1). Rodent models have the advantage of using senescent cell reporters (Bussian et al., 2018; Zhang et al., 2019), providing rapid access to fresh tissues for appropriately evaluating senescence-associated β-galactosidase (SA β-gal) (Jurk et al., 2012; Musi et al., 2018), and enabling longitudinal and cross-sectional studies of genetically identical individuals (Bussian et al., 2018; Jurk et al., 2012; Musi et al., 2018; Zhang et al., 2019), all of which are highly beneficial for identifying senescent cells in tissue. Studying senescent cells in the human brain presents obvious challenges. Identifying and tracking rare subpopulations of poorly defined cells in the ~100 billion brain cellular environment is not trivial. Archival tissue is not permissive to SA β-gal enzymatic assays (Debacq-Chainiaux et al., 2009) and its correlation to brain cell senescence is not clear given that some neuronal subpopulations express SA β-gal activity at young ages (Jurk et al., 2012; Musi et al., 2018). Similarly, lipofuscin, lysosomal biproducts indicative of senescence in some cell types, occurs even in young neurons in the human brain (Goyal, 1982). Moreover, the identity of the parent cell type and upstream signals may affect the post-senescence phenotype (Hernandez-Segura et al., 2017a). The resulting heterogeneity has presented challenges for identifying, defining, and studying senescent cells in vivo. While this is especially true for post-mitotic tissues such as the brain, much progress has been made in recent years, for review: (Gillispie et al., 2021; Sah et al., 2021).

Table 1.

Preclinical studies investigating cellular senescence in relation to AD risk factors and AD neuropathology.

Citation Aging/Neurodegenerative Disease Model Senotherapeutic Treatment and Dosing Strategy Senescent Cell Types Investigated Brief Summary of Outcomes
(Streit et al., 2009) Human postmortem AD brain tissue None Microglia Morphologically senescent-like, not activated, microglia were found near Aβ plagues and preceded tau tangles in human brains.
(Geng et al., 2010) Male Sprague-Dawley rats (6-, 18-, 24-month old) & in vitro hippocampal neurons None Neurons SA β-gal staining increased in the CA3, but not DG of the hippocampus with aging (51 % by 24 months)
(Bhat et al., 2012) Postmortem human brain & in vitro astrocytes None Astrocytes p16 positive astrocytes were increased with age and in AD brain tissue.
(Jurk et al., 2012) Male C57Bl/6 mice (4- & 32-month old) None Neurons Age and telomerase-related senescent phenotypes in neurons were mediated by the DNA damage response, slightly attenuated by caloric restriction, and fully rescued by the knockout of Cdkn1a.
(He et al., 2013) Male C57BL/6 and APP/PS1 (9-month-old) None Neural stem cells Aβ42 exposure increased senescent phenotypes in NSCs in the dentate gyrus of APP/PS1 mice.
(Al-Mashhadi et al., 2015) Postmortem human brains with or without white matter lesions None Oligodendrocytes Senescent phenotypes were identified in oligodendrocytes of aged brains, but were not associated with white matter lesions.
(Kang et al., 2015) Postmortem human brain tissue from young and aged adults without neuropathology None Astrocytes; neurons; oligodendrocytes Gata4, a SASP regulator, was highly associated with age and p16 expression in astrocytes, pyramidal neurons, and oligodendrocytes in humans.
(Turnquist et al., 2016) Postmortem brain tissues from AD and ALS patients None Astrocytes Greater senescent cell burden in AD and ALS tissue compared to non-diseased, age-matched and pediatric control tissue
(Bussian et al., 2018) Male and female PS19;ATTAC mice (weanling to 12-month-old) 2 mg/kg AP20187 twice weekly from weaning (long term) or 10 mg/kg for five consecutive days at 6 months of age (short-term) Astrocytes; microglia Clearance of senescent cells prevented gliosis, hyperphosphorylation of tau, and neuronal degeneration
50 mg/kg ABT263 for 5 consecutive days followed by 16 days of rest from weaning until 6 months
(Musi et al., 2018) Male and female rTg4510Mapt+/+, rTg4510Mapt0/0, rTg21221 Mapt+/+, 3xTgAD mice (2- to 23-month-old) 5 mg/kg dasatinib and 50 mg/kg quercetin for 5 consecutive days followed by 9 days rest for 6 treatment cycles Neurons Dasatinib + quercetin treatment reduced total NFT density, SASP gene expression, neuron loss, and ventricular enlargement.
Postmortem human brain: Alzheimer’s disease (AD) Progressive Supranuclear Palsy (PSP) None Neurons AD: Elevated senescent gene expression in NFT-bearing neurons compared to neurons without NFTs.
PSP: Elevated CDKN2D expression compared to age-matched controls that negatively correlated with cognition.
(Chow et al., 2019) Male and female C57BL/6 J mice (3- and 22–24 month old) +/− insulin resistance None Neurons Chronic insulin exposure led to senescence phenotype in neurons via dysregulation of hexokinase 2
(Moreno-Blas et al., 2019) Male Wistar rat brains (4- or 25-month-old) None Astrocytes; neurons Astrocytes and neurons showed a wide range of senescent phenotypes, which were attenuated or exacerbated in vitro via stimulation and inhibition of autophagy, respectively.
(Norton et al., 2019) Postmortem human brain without AD and with or without cerebral small vessel disease None Mural cells Senescent phenotypes were observed in mural cells of aged individuals, but were less prevalent in patients with cerebral small vessel disease.
(Ogrodnik et al., 2019) Male C57BL/6 INK-ATTAC mice (8-month-old) +/− high fat diet; INK-ATTAC; db/db mice 10 mg/kg AP20187 for 3 days every 2weeks for a total of 8–10 weeks Astrocytes; microglia Clearing obesity-induced senescent glial cells improved neurogenesis and alleviated anxiety-related behavior.
db/db mice 5 mg/kg dasatinib and 50 mg/kg quercetin for 5 days every 2 weeks for 8 weeks
(Ritzel et al., 2019) Male C57BL/6 mice (3- & 18-month-old) +/− TBI None Microglia Aging and TBI increased microglia senescent phenotypes including Bcl-2, p16INK4a, p21CIP1a, lipofuscin, and H2AX.
(Schwab et al., 2019) Postmortem human brains with or without a history of TBI None Astrocytes; ependymal cells; neurons; oligodendrocytes Brains from patients with a history of TBI showed extensive dysfunction, DNA damage, and other cellular senescence phenotypes in a variety of cell types.
(Stojiljkovic et al., 2019) Male C57BL/6J mice (3- & 24-month-old) None Microglia Various markers of microglial senescence distinct from microglial activation increased with age, although aged microglia in vivo may have been quiescent rather than senescent.
(Tominaga et al., 2019) Male C57BL/6 mice (10-weeks-old) +/− TBI None Astrocytes; microglia; neurons Cell cycle initiation increased for the first four days post-TBI, while markers of senescence were elevated on days 4, 7, and 14 with some cell type-specific differences in expression of senescence.
(Turnquist et al., 2019) Postmortem human brain tissue from patients that had received radiation and age-matched controls None Astrocytes Astrocyte senescence was the most prominent cell type in radiation exposed human tissues.
(Zhang et al., 2019) Male and female APP/PS1, WT mice and APP/PS1/ZsGreen mice (2.5- to 8-month-old) 12 mg/kg dasatinib and 50 mg/kg quercetin, once daily for 9 days (short-term) or 11 weeks (long-term) Oligodendrocyte progenitor cells Aβ induced senescence in oligodendrocyte progenitor cells and senolytic treatment reduced neuroinflammation, Aβ burden, and cognitive deficits.
( Arun et al., 2020 ) Male Sprague-Dawley rats (9–10-week-old to 1-year +/−TBI) None Did not investigate Single and repeated blast exposures increased markers of cellular senescence, particularly at 1 month, in several neuroanatomical structures.
(Bryant et al., 2020) Intact microvessels from postmortem human AD brain tissue None Endothelial cells Senescence-related genes were significantly upregulated in endothelial cells.
( Gao et al., 2020 ) Neural stem cell spheroids from 3-week-old and 23-month-old C57BL/6 mice 100 pg/mL Ribes meyeri anthocyanins extract Neural stem cells (NSCs) NSCs of aged mice showed increased senescence phenotypes and treatment with Ribes meyeri anthocyanins or Nar reduced senescence and improved cognition.
Female C57BL/6 mice (12-mo-old) 100 mg/kg Ribes meyeri anthocyanins extract for 2 months or 20 mg/kg Nar for 1 month
( Hu et al., 2021 ) Male SAMP8 mice (2-, 6-, 12-month-old) 1010 embryonic stem cell-derived extracellular vesicles (ESC-sEVs) twice weekly for 6 months NSCs ESC-sEV treatment alleviated senescence in hippocampal NSCs and improved age-related declines in cognition.
( Ogrodnik et al., 2021 ) Male INK-ATTAC and WT mice (4- & 24-month-old) 10 mg/kg AP20187 for 3 consecutive days once every two weeks for 8 weeks Microglia; oligodendrocyte progenitor cells Senolysis reduced p16INK4a expression, microglial activation, and SASP factors and improved cognitive function.
5 mg/kg dasatinib and 10 mg/kg quercetin for 3 consecutive days once every two weeks for 8 weeks
( Schwab et al., 2021 ) Male C57BL/6 mice (7–9-week-old) +/− repeated mild TBI None Did not investigate Following an initial DNA damage response at 24 h, senescent cell phenotypes were observed in the brain 7 days after traumatic brain injury.
( Yousefzadeh et al., 2021a ) Male and female mice with hematopoietic cell Ercc1 knockout (2- to 24-months old) None Did not investigate Whole-body immune senescence increased p16 expression in the brain, but showed no differences in p21 expression or age-associated lesions via H&E staining.

Abbreviations: amyloid-beta (Aβ); Alzheimer’s disease (AD); Amyotrophic lateral sclerosis (ALS); embryonic stem cell derived small embryonic vesicles (ESC-sEV); lipopolysaccharide (LPS); neural stem cell (NSC); neurofibrillary tangle (NFT); senescence-associated β-galactosidase (SA-βgal); senescence-associated heterochromatin foci (SAHF); senescence-associated secretory phenotype (SASP); traumatic brain injury (TBI).

Rodent models and postmortem human brain tissue have been invaluable in establishing the preclinical evidence necessary to begin translating senescence targeting strategies to the clinic (Tchkonia and Kirkland, 2018). Specifically, our group revealed an upregulation of genes and SASP factors in brain regions with tau-containing neurofibrillary tangles (NFTs) in four distinct transgenic mouse models of tau-pathogenesis and post-mortem human brain tissue (Musi et al., 2018). Transcriptomic comparisons between neurons with or without NFTs from postmortem human brains revealed a senescence signature in the NFT-bearing neurons. A causal role of NFT-mediated senescence was explored with pharmacological ablation of senescent cells in tau transgenic mice. The intermittent treatment with senolytic agents, dasatinib and quercetin (D + Q), selectively cleared senescent cells (Musi et al., 2018) which was independently reproduced in other models of brain aging (Ogrodnik et al., 2021) and pathogenesis relevant to AD (Ogrodnik et al., 2019; Zhang et al., 2019). Across studies, clearing senescent cells has been associated with diminished AD pathology (i.e., tau and Aβ deposition), reduced SASP and inflammation, decreased white matter hyperintensities (WMH), restored cerebral blood flow, and improved cognitive behavior (Bussian et al., 2018; Musi et al., 2018; Ogrodnik et al., 2019; Zhang et al., 2019). With these promising data, similar strategies are moving toward clinical testing in the context of amnestic mild cognitive impairment/early AD (NCT04063124 and NCT04685590). Overall, the present contribution aims to provide an accessible, updated summary of evidence that supports translating this therapeutic approach to older adults with amnestic MCI or early AD. Considerations unique to trials in AD, including potential challenges faced are also discussed.

2. Translating preclinical evidence to clinical trials

2.1. Evidence for senescent cell accumulation in the human brain with aging and neurodegeneration

Cell cycle activity in postmitotic brain cells in human AD was first reported, to our knowledge, in the late 1990s (Arendt et al., 1996; Luth et al., 2000; McShea et al., 1997).The elevated expression of cell cycle regulators, including senescence hallmark p16, was hypothesized to represent aberrant cell cycle activity leading to apoptosis in neurodegenerative diseases. Later work revealed that elevated p16 protected against, not induced, neuronal cell death (Park et al., 1998). While these early studies were investigating cell cycle activity, not senescence, several groups have since performed hypothesis-driven experiments focused on the role of cellular senescence in the brain (Table 1). Senescent cells indisputably accumulate in the brain, however the consequences of their accumulation and long-term survival in the context of human aging and disease remains poorly understood. In this section, we review evidence for human brain cellular senescence, and discuss whether the response represents a physiological benefit (i.e., alternative to cell death), or instead may contribute to chronic inflammation and degeneration as they are notoriously known.

2.1.1. Senescent cell accumulation associated with brain pathologies of advanced age

The privilege of growing old often comes with a cost of failing health. Common brain pathologies in non-demented older adults include vascular and white matter lesions (WML) (de Leeuw et al., 2001; Han et al., 2018) and cerebral small vessel disease (cSVD; alternatively termed arteriolosclerosis). In older adults, the prevalence of WML is >80 % (Longstreth et al., 1996); lesions associated with cSVD in older populations ranges from 8 % to 33 % for lacunes (Vermeer et al., 2007), 3 % to 35.7 % for cerebral microbleeds (Greenberg et al., 2009; Hilal et al., 2017; Poels et al., 2010), and 39 % to 96 % for WMHs, as reviewed in other studies (Prins and Scheltens, 2015). While vascular lesions and WML do not always co-occur, longitudinal magnetic resonance imaging (MRI) data suggest that small-vessel ischemic disease could contribute to white matter changes in advanced age (Erten-Lyons et al., 2013). Emerging evidence indicates cellular senescence may be involved with these pathologies, and in some cases, connect them.

Tissue-wide oxidative stress and DNA damage are more likely to be associated with WML in the presence of senescent cells (Al-Mashhadi et al., 2015). A neuropathological hallmark of cSVD is fibro-hyaline thickening of deep penetrating arteries resulting in elongated and tortuous vessels with narrowed lumina and thickened vessel walls. In rodent studies, this phenotype has been observed near NFT-bearing neurons (Bennett et al., 2018), cells which display features of cellular senescence (Musi et al., 2018). A follow-up study led by Bennett using postmortem human brain tissue from patients with AD found an association between cerebrovascular senescence and tau pathology (Bryant et al., 2020) (discussed below). While these data suggest that cellular senescence may contribute to cSVD in individuals with dementia, a separate study that used tissue from non-demented individuals found an inverse correlation between senescent myocytes and cSVD diagnosis. Specifically, Norton et al. investigated the prevalence of myocyte senescence in 60 adults, aged 80–96 years-old, with minimal AD pathology (i.e., Braak stage 0-II), and its association with neuropathological diagnosis of cSVD (Norton et al., 2019). Their study relied on histological analyses using antibodies to detect smooth-muscle myocytes, nuclear trimethylated Lys9 in histone-3 (H3K9me3) staining as a surrogate for senescence-associated heterochromatic foci (SAHF) and phosphorylated Ser139 on histone-2 (γH2AX) to detect DNA damage foci. The data indicated more abundant H3K9me3 and γH2AX positive myocytes in individuals without cSVD than those with a confirmed neuropathological diagnosis of cSVD. Total mural cell density (e.g., vascular smooth muscle cells and pericytes) was also higher in control cases, which is consistent with the depletion of vascular myocytes in cSVD (Pantoni, 2010). Since the loss of myocytes is closely associated with impaired cerebral blood flow and consequent ischemic lesions, it is tempting to speculate that retaining myocytes, even if senescent, may be preferred over their apoptosis. Similarly, given that inflammatory markers of the SASP were not evaluated in the aforementioned studies, and that not all senescent cells develop SASP (Coppe et al., 2011), the toxicity profile of senescent brain cells remains unknown. In this way, senescence may play a protective role for retaining brain vascular myocytes, especially if they do not adopt a toxic secretome. The intriguing inverse relationship between senescent myocytes and cSVD diagnosis warrants further consideration as senescent cell clearing therapies move to clinical trials.

2.1.2. Senescent cells and Alzheimer’s disease-associated neuropathologies

AD is the most common cause of dementia, and is responsible for 60–80 % of all known cases (2021a). The disease defining features include the accumulation of extracellular Aβ in the form of plaques and intracellular hyperphosphorylated tau forming NFTs (Jack et al., 2018). The pathogenic mechanisms connecting Aβ and tau protein accumulation with neurodegeneration and dementia, however, remain unclear. Many promising lines of investigation are underway, including a focus on the drivers of biological age, like cellular senescence. The formal investigation of senescence in the context of AD is a relatively recent field (DiBattista et al., 2020). However, as mentioned above, reports of elevated expression of cyclin dependent kinase (CDK) inhibitors p16 and p21, which are hallmarks of the senescence stress response, were found co-localized to NFT-bearing neurons in postmortem AD brain decades ago (Luth et al., 2000; McShea et al., 1997). Aberrant neuronal cell cycle re-entry was hypothesized to contribute to the slow death of neurons (Barrio-Alonso et al., 2018; Yang et al., 2001, 2003); however, recent data indicates this atypical neuronal phenotype resembles cellular senescence (Chow et al., 2019; Musi et al., 2018). The body of evidence now suggests that postmitotic cells are susceptible to becoming senescent. Neurons, for example, exist in quiescence. This reversable dormant cell state protects cells against stress but renders them vulnerable to enter senescence through lysosome-mediated mechanisms (Fujimaki et al., 2019). In this way it is tempting to speculate that neurons are primed for senescence given their quiescent state and susceptibility to age-associated lysosomal dysfunction (von Zglinicki et al., 2021). While it remains to be determined whether neurons slip directly into senescence, or abort cell-cycle re-entry induced apoptosis (Raina et al., 2003), brain cell senescence has emerged as a cell stress response to risk factors known to increase AD, including advanced age, Aβ, tau and traumatic brain injury (Table 1). Hypothesis-driven studies to investigate cellular senescence in postmortem human brains have revealed that multiple brain cell types may engage a senescence stress response (Bhat et al., 2012; Bryant et al., 2020; Musi et al., 2018; Zhang et al., 2019). Mechanistic experiments in mice have demonstrated that tau aggregation drives cellular senescence in neurons (Musi et al., 2018) and microglia (Bussian et al., 2018), whereas extracellular Aβ has a similar effect on oligodendrocyte precursor cells (Zhang et al., 2019). Similarly, in postmortem human AD brain, neurons (Musi et al., 2018), astrocytes (Bhat et al., 2012), oligodendrocyte precursor cells (Zhang et al., 2019), endothelial cells (Bryant et al., 2020) and microglia (Streit and Xue, 2014) display features of cellular senescence. The relative sensitivity of each of these cell types to senescence-targeted therapies, and outcomes of their removal remain to be determined in clinical trials.

2.1.3. Mixed neuropathology in AD

While neurodegenerative diseases have historically been characterized by disease-specific pathologies, recent postmortem analyses have firmly established the co-occurrence of pathologies in multiple dementia syndromes (Rabinovici et al., 2017). In AD, the most common Aβ plaque and NFT co-pathologies are Lewy bodies, TDP-43 and cerebrovascular lesions. Lewy bodies are primarily composed of α-synuclein and can co-exist with prototypical AD pathology (Rabinovici et al., 2017). Parkinson’s disease (PD), the second most common neurodegenerative disease following AD, is characterized by the presence of Lewy bodies, and is associated with progressive motor symptoms, static tremor, motor imbalance, and muscle rigidity (Zeng et al., 2018). PD brain tissue displays elevated levels of senescence markers p16INK4a, and SASP factors including IL-6, IL-1α, IL-8, and protease matrix metalloprotein-3 (MMP-3) (Chinta et al., 2018). Additionally, astrocytes in postmortem PD brain tissue were found to be lamin B1 deficient. Given that lamin B1 decline is induced by p53 activation or p16 expression and is closely associated with senescence (Freund et al., 2012), the reduction in lamin B1 in PD is suggestive of senescence (Chinta et al., 2018). The expression of nuclear lamin proteins in the brain is complex (Mahajani et al., 2017; Takamori et al., 2018; Young et al., 2014); we caution the need for careful consideration when attributing a senescence phenotype to lamin B1 expression level. Of note, senescent astrocytes have been reported in postmortem human AD, but neither lamin B1 nor Lewy body pathologies were evaluated (Bhat et al., 2012). Future studies on similarities or differences in the senescence profile in pure AD versus mixed pathologies (including others not discussed here, i.e., TDP-43) will be important for predicting outcomes in senescence-motivated therapies in future trials.

2.1.4. Partial AD pathology - pure tauopathies in the absence of Aβ

Tauopathies, including AD, are a group of neurodegenerative disorders pathologically defined by the deposition of tau. However, unlike mixed AD with multiple co-pathologies, pure tauopathies involve tau accumulation in the absence of Aβ. These diseases include progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), some frontotemporal dementias, and chronic traumatic encephalopathy among others (Orr et al., 2017). NFT-containing neurons from postmortem human AD brains exhibit a senescence-like phenotype (McShea et al., 1997; Musi et al., 2018). Moreover, tauopathy, even in the absence of Aβ, drives cellular senescence as evidenced by elevated CDKN2A in post-mortem human brain tissue with histopathologically confirmed PSP (Musi et al., 2018). To our knowledge, studies have not evaluated senescence in human brains with elevated Aβ in the absence of tau (i.e., cerebral amyloid angiopathy, CAA). These data will be critical to understanding whether patients with CAA may also benefit from therapies targeting cellular senescence, or whether the presence of tau pathology is a key feature in driving senescence in these diseases. Nevertheless, studies involving AD neuropathology [pure, mixed, or partial (i.e., tau)] all have reported a co-occurrence of senescent cells indicating a common stress response across many distinct neurodegenerative diseases.

2.2. Key considerations for translating senescence clearing therapies to AD trials

Prior AD trials have largely focused on restoring neurotransmitter levels (i.e., acetylcholine) or reducing AD protein accumulation (e.g. cerebral amyloid beta and tau). Despite strong rationale for these therapeutic targets, the trials have failed. Between 2002–2012 there was a 99.6 % failure rate of AD-drug trials (Cummings et al., 2014), and this has not significantly improved in the last decade (Rafii and Aisen, 2020). Targeting a fundamental cellular aging process, like cellular senescence, offers a fresh, distinct approach and is backed by compelling evidence. Advanced age is the strongest risk factor for AD, and data indicate a continuum of neurodegenerative changes from normal aging to dementia (Harada et al., 2013). These include brain atrophy, synaptic loss, and neuropathological protein accumulation. As the key processes of aging interact across molecular, cellular, and system levels, targeting one pathway (e.g., cellular senescence) may exert pleiotropic effects that are further beneficial for the treatment of AD (Mattson and Arumugam, 2018).

Removing senescent cells improves clinically relevant outcomes in AD mouse models Table 1, i.e., (Bussian et al., 2018; Musi et al., 2018; Zhang et al., 2019). Over 40 agents have been identified as ‘senolytic’, or capable of ablating senescent cells (Kirkland and Tchkonia, 2020; Zhu et al., 2015). A multitude of pharmacological agents have also been discovered that modulate broader key pathways relevant to senescence (Mongelli et al., 2020). Clinical trials using senolytics have been completed or are ongoing for several chronic diseases including renal insufficiency (NCT02848131), idiopathic pulmonary fibrosis (NCT0287498), and frailty (NCT03675724) (Hickson et al., 2019; Justice et al., 2019a; Kirkland and Tchkonia, 2020). Although still in early stages, clinical trials aimed at exploring cellular senescence as an innovative target for AD are imminent (NCT04063124 and NCT04685590). In the following sections, we review key critical considerations when translating promising senescence-modulating therapies to AD clinical trials that are unique to this study population, and distinct from trials focused on older adults with normal cognition.

2.2.1. General considerations for AD trials

2.2.1.1. Study partner.

Unique to AD trials, compared to other older adult study populations without dementia, is that all AD trials require two participants: the patient and a study partner. The study partner, typically the primary caregiver (e.g., spouse, adult child or other), is critical to a successful trial (Bernstein et al., 2021; Grill et al., 2013). They assist with informed consent, ensure protocol compliance, and serve as informants for cognitive, functional, and behavioral outcome measures.

2.2.1.2. Blinding and placebo effect.

Double-blinding and controlling for placebo effects are particularly critical to AD trials. Changes in cognition and biomarkers are often difficult to isolate from control groups due to limited statistical power, particularly in short trials.

2.2.1.3. Study duration.

Cognitive decline in placebo groups typically requires an 18-month period to observe (Ito et al., 2013), which is the basis for typical phase II and III AD trial duration (Green et al., 2009; Schneider et al., 2015; Schneider and Sano, 2009). Placebo lead-in of appropriate length can help identify placebo responders during the first couple weeks of the study, which could minimize trial failures (Ozawa et al., 2017). Target engagement for senolytics may be assessed with shorter trial durations but would require robust measures of senescent cell burden in the brain; such methodologies are described in a later section.

2.2.1.4. Medication considerations: interactions and stable dosing.

It is common for AD patients to suffer with co-occurring behavioral and psychological disturbance, which may be treated with antipsychotic and antidepressant medications often with more severe drug interaction profiles (Kettunen et al., 2019; Kuroda et al., 2019). Many medications, specifically commonly prescribed anti-depressants, are inhibitors or inducers of CYP450, and are often prescribed to AD patients who commonly have behavioral disturbance (30–90 %) (Garre-Olmo et al., 2010; Müller-Spahn, 2003) and depression (> 40 %) (Panza et al., 2010; Ryu et al., 2017). In considering the primary therapeutics prescribed for symptomologic maintenance in AD, there are a few specific considerations to make regarding co-administered medications. The anticholinesterase inhibitors donepezil, and galantamine (not true for rivastigmine) (Grossberg et al., 2000) are primarily metabolized by cytochrome P450 isoenzymes 34A (CYP3A4) and 2D6 (CYP2D6) (Farlow, 2003), and can interact with other anticholinesterase agents to produce synergistic effects. All three drugs have similar cholinergic effects that can increase risk of bradycardia, especially when paired with beta-blockers (2012; Grossberg et al., 2000; Tavassoli et al., 2007). Memantine, as a N-methyl-D-aspartate (NMDA) receptor antagonist, is unique amongst the other commonly prescribed medications for the symptomology of AD. Memantine inhibits glutamatergic signaling reducing excitotoxicity in AD that may lead to destruction of cholinergic neurons, and may even promote synaptic plasticity and preserve or enhance memory, but has not had long-term effectiveness in preventing disease progression (Paulison and Léos, 2010; Rogawski and Wenk, 2003). Memantine has an inhibitory effect on Cytochrome P450 (specifically of isoenzyme CYP2B6 and CYP2D6), and can also result in significant drug interactions (Micuda et al., 2004). Prior to enrolling a subject into an AD trial, participants should be maintained on a stable dose of any AD-specific medication for a minimum of 3 months. This will ensure that any cognitive changes, alternative study measures, or adverse events are due to the new investigational product (i.e., senolytic) rather than changes in prescription or dose of other medications (Tariot et al., 2004; Xu et al., 2021). Aside from the commonly prescribed medications to treat AD symptoms, patients are often also treated with a host of medications for co-occurring diseases and disorders (Chatterjee and Mudher, 2018; Janson et al., 2004; Santiago and Potashkin, 2021). Some of these medications have been suggested to have positive effects in AD (e.g., metformin (Koenig et al., 2017a) or antihypertensives (Group et al., 2019)) while others may have extensive side-effect profiles and potential for significant drug interactions (Ali et al., 2015; Ding et al., 2020; Pasqualetti et al., 2015). Therefore, especially in a population that may struggle with drug compliance and reporting of adverse events, physicians and research teams need to be aware of the increased risk of potential adverse drug interactions (Borah et al., 2010; Lima et al., 2016).

2.2.2. Determining target engagement

One existing barrier for cellular senescence trials is the lack of specific, reliable, accessible biomarkers to establish target engagement and monitor efficacy. Many ongoing efforts are focused on innovative approaches to identify senescent cells in human brain tissue and monitor target engagement in these trials. Newly emerging strategies aim to identify senescent-specific signatures by analyzing miRNAs, epigenetics (Horvath and Raj, 2018; Kritsilis et al., 2018) and transcriptomics (Hernandez-Segura et al., 2017b) that regulate gene expression and cellular functions integral to senescence (Selbach et al., 2008). Comparing transcriptomic networks across cultured fibroblasts, melanocytes, keratinocytes, and astrocytes exposed to various senescence-inducing stressors revealed cell type and stress type differences (Hernandez-Segura et al., 2017b). Only 4 % of senescence-associated genes were consistently differentially expressed across cell types; moreover, among 1311 differentially expressed genes in senescent fibroblasts, variances were noted across specific cellular stressors (replicative, oncogene-induced, and ionizing-induced). Results from this study highlight that senescence transcriptomes are unique to cell type and stressor (Hernandez-Segura et al., 2017b). Given that senescence signatures are expected to differ across tissues and cells, a concerted effort focused on characterizing senescent cells across the human body is greatly needed to advance clinical trials (Roy et al., 2020).

2.2.3. Biofluid markers of senescence to determine eligibility and monitor target engagement

In AD brain tissue, we find that 1–3 % of brain cells are senescent (Dehkordi et al., 2021). These results overlap with the prevalence of senescent cells reported in the blood from patients with idiopathic pulmonary fibrosis (IPF) (Justice et al., 2019b) and in adipose and skin (Hickson et al., 2019; Justice et al., 2018) to suggest a similar senescent cell burden across human tissues. Despite the limited number of senescent cells within a tissue, the diffuse paracrine transmission of the SASP induces broad changes in gene expression that may be detectable in biofluids distal from the site of origin. Successfully detecting their presence would then rely on senescence-specific molecules/phenotypes to target for analyses. Toward this end, much work has focused on the inflammatory response (Hickson et al., 2019; Schafer et al., 2020). While the specificity of these molecules to senescent brain cells remains unclear, this strategy was employed in a clinical trial of IPF where targeted gene expression analyses identified core SASP factors that were responsive to oral senolytic therapy (Justice et al., 2019b). The reduction in pro-inflammatory SASP factors corresponded to improvements in clinical outcomes. Notably, these core SASP factors are detectable in blood and/or CSF of patients with AD (Table 2) suggesting that SASPs may be useful to monitor senescence in AD as well. While the IPF study provides strong evidence for the utility of senolytic guided therapies in chronic conditions, many of the SASP factors measured co-occur with numerous inflammatory responses and highlights the need for continued biomarker discovery with higher specificity to senescence.

Table 2.

Core SASP in Human AD.

We also provide caution that the presence of SASP factors detected in the CSF may originate from peripheral sources. Though the brain is considered to be relatively immune-privileged, peripheral factors are able to cross the blood brain barrier through bidirectional transport systems, which may lead to senescence in the brain. This principle is demonstrated by findings of peripheral inflammation inducing transient alterations of mental status in the case of delirium (Lopez-Rodriguez et al., 2021; Munster et al., 2011), and even heterochronic parabiosis studies in mice (Villeda et al., 2011; Yousefzadeh et al., 2020). Mouse studies provide further evidence that senescent peripheral immune cells are able to increase markers of senescence in the central nervous system (Yousefzadeh et al., 2021b). Further, as blood brain barrier integrity declines with age, and to a greater extent with neurodegenerative disease (Farrall and Wardlaw, 2009; Montagne et al., 2015), considerations should be made to evaluate effects of target engagement of new therapeutics in the periphery versus the brain.

Exosomes are membrane-bound small extracellular vesicles (EVs) of endocytic origin with a size range of ~30–50 nm. These nanovesicles are secreted by cell types for intercellular communication, cellular homeostasis, stress response, and to improve cell survival (Kumar and Deep, 2020a, b; Misawa et al., 2020; Takahashi et al., 2017). They are being explored as potential biomarkers for the detection of neurodegenerative disease and senescence. Exosome biogenesis involves numerous steps, including a) the formation of clathrin-coated vesicles (CCV) from specific sites in the plasma membrane, b) fusion of CCV with the endosome, c) inward budding of the membrane of endosomes, and cargo (protein, nucleic acids, lipids, glycans, and metabolites, etc.) loading, leading to the formation of multi-vesicular endosomes (MVE) or multi-vesicular bodies (MVB) with intraluminal vesicles, e) MVE/MVB move along specific cytoskeleton track towards the plasma membrane, and lastly, f) MVE/MVB fuse with the plasma membrane and exosomes are released outside. There is a growing list of molecular regulators of exosome biogenesis and cargo loading, including the endosomal sorting complex required for transport (ESCRT), Rab GTPases, Alix, ceramides, tetraspanins, sphingomyelinases, HIF1α, syndecan, syntaxin, and syntenin etc (Colombo et al., 2013, 2014; Kowal et al., 2014; Kumar and Deep, 2020a, b; Peak et al., 2019; Ventimiglia and Alonso, 2016). Besides exosomes, cells secrete several other types of EVs, including microvesicles (~100 nm to >1000 nm in size), apoptotic bodies, and oncosomes (Jakhar and Crasta, 2019), underscoring the complexity and heterogeneity of EVs.

Exosomes have a lipid bilayer that protects their cargo from degradation, making these vesicles a useful tool for local and distant communication. These vesicles are found in all biofluids and exhibit certain common proteins, even in exosomes from different cellular origin. However, exosomes are also loaded with certain unique cargoes, including proteins, miRNAs, and metabolites, that could potentially relate to the cellular source of their origin and their molecular, physiologic, and metabolic state (de Jong et al., 2012; Eissa et al., 2016; Kumar and Deep, 2020a, b; Ramteke et al., 2015; Schlaepfer et al., 2015). Several studies have reported that exosomes released by cells under stressful physiological conditions or pathologic states have certain unique molecular features compared to those secreted under normal conditions (Chistiakov et al., 2016; de Jong et al., 2012; Hedlund et al., 2011; Kumar and Deep, 2020a, b; Panigrahi et al., 2018; Ramteke et al., 2015; Sheller et al., 2016). For example, under hypoxia (low oxygen condition), cells usually secrete higher amounts of exosomes loaded with relatively unique cargo and could transmit hypoxic phenotypes to recipient cells (Jung et al., 2017; Kumar and Deep, 2020a, b; Panigrahi et al., 2019; Ramteke et al., 2015; Zonneveld et al., 2019). Exosomes released under oxidative stress are loaded with certain unique proteins reflecting a pro-oxidative physiological state of the parent cells (de Jong et al., 2012; Eissa et al., 2016; Hedlund et al., 2011). Broadly, under certain conditions, the composition of exosomes could be a snapshot of the cell at the time of their biogenesis and secretion. Due to relative ease with which they can be obtained, less invasive access in various biofluids compared to tissue biopsies, and their unique cargo reflecting the pathophysiological state, exosomes have been extensively characterized for the diagnosis and prognosis of various pathologies including cancer, AD, and PD (Asai et al., 2015; Chistiakov et al., 2016; Eissa et al., 2016; Kalluri, 2016; Kumar and Deep, 2020a, b; Panigrahi and Deep, 2017; Soria et al., 2017; Winston et al., 2016).

Numerous studies have now established that exosomes secreted by various brain cells (e.g., neurons, astrocytes, and endothelial, etc) could be present in biofluids (blood, CSF, saliva, and urine), and could be useful to better understand molecular underpinnings of neurological disorders and neurodegenerative diseases including AD (Cha et al., 2019; Fiandaca et al., 2015; Goetzl, 2020; Goetzl et al., 2018, 2015; Goetzl et al., 2016a, b; Kapogiannis, 2020; Kapogiannis et al., 2015; Mustapic et al., 2019; Nogueras-Ortiz et al., 2020; Vella et al., 2016; Winston et al., 2016). Specific brain cell-derived exosomes could be isolated from blood plasma based upon their unique surface proteins and characterized for established AD biomarkers, and used to discover novel biomarkers. Fiandaca et al. reported that the level of established AD biomarkers (p-T181-tau, p-S396-tau, and Aβ42) in neuron-derived exosome (NDE), isolated from blood plasma, could predict the development of AD up to 10 years before the clinical onset (Fiandaca et al., 2015). Similarly, abnormal plasma NDE levels of p-Tau, Aβ42, neurogranin, and repressor element 1-silencing transcription factor precisely predicted the MCI conversion to AD (Winston et al., 2016). Goetzl et al. reported that astrocyte-derived exosomes (ADE) from AD patients exhibited higher levels of BACE-1 (β-site amyloid precursor protein-cleaving enzyme 1) and sAPPβ (soluble amyloid precursor protein β), and lower GDNF (glial-derived neurotrophic factor) compared to ADE from healthy controls. Few recent studies have also reported the usefulness of endothelial-derived exosomes (EDE) in better understanding cerebral vascular disease and cognitive function. For example, Abner et al. reported that high EDE levels of Aβ40, Aβ42, and p-Tau in patients with WMHs indicate the presence of small cerebral vascular disease in the pre-clinical stages of AD (Abner et al., 2020).

Senescent cells secrete not only inflammatory biomolecules but also release higher amounts of EVs with unique compositions, a phenomenon mainly regulated by p53 (Buratta et al., 2017; Carracedo et al., 2019; Jakhar and Crasta, 2019; Lehmann et al., 2008; Misawa et al., 2020; Saheera et al., 2020; Takahashi et al., 2017; Urbanelli et al., 2016). EVs could carry SASP factors and have been suggested to play a role as senescence effectors (Jakhar and Crasta, 2019). EV secretion by senescent cells is a defense mechanism to maintain cellular homeostasis (Misawa et al., 2020; Takahashi et al., 2017). EVs secreted by senescent cells could potentially contribute to detrimental effects (e.g., pro-tumorigenic effects, accelerate aging, and various age-associated pathologies) or beneficial outcomes (tissue regeneration and wound healing) (Misawa et al., 2020; Saheera et al., 2020; Takahashi et al., 2017; Urbanelli et al., 2016). Considering the importance of senescence in AD pathology, various brain-cell derived exosomes or EVs in the blood (or other biofluids) could offer important information regarding the senescent cell type. Furthermore, the expression of senescence-related biomarkers in brain cell-derived exosomes could potentially serve as novel and early diagnostic biomarkers for incipient AD.

Overall, the availability of easily accessible and less invasive exosomes in plasma has shown immense promise to better understand AD pathogenesis. Though, several challenges persist, such as lack of standard protocols for the collections and storage of biofluids as well as isolation and characterization of pure exosomes. For example, based upon currently available methods, it is impossible to isolate pure exosomes from biofluids and separate exosomes from other EVs of similar characteristics. Another key challenge is the specificity of exosomal surface proteins currently being used to isolate brain cell-derived exosomes from plasma. For example, L1CAM (L1 cell adhesion molecule) is widely used to isolate NDE from plasma; however, questions have been recently raised regarding the neuronal specificity of L1CAM and the presence of L1CAM on the NDE surface (Norman et al., 2021). It is also clear that more specific exosomal surface biomarkers are needed as currently used biomarkers do not provide any information on the brain region from which they originate. Isolation of exosomes from blood using surface markers unique to specific anatomical sites of the brain could enable a greater understanding of the biology of these sites (including senescence) and their relative contribution in AD. Though this research area is understudied and the role of exosomes secreted by senescent cells during AD pathogenesis is not well characterized, it holds great promise and warrants further investigation.

2.2.4. Relevant outcome measures for AD trials – considering pathogenesis and symptoms

For the purpose of research, AD is defined by the presence of Aβ plaques and tau-containing NFTs (Jack et al., 2018). While the mechanisms linking plaques and tangles to the behavioral and cognitive symptoms are not clear, strategies to reduce these protein aggregates are hypothesized to alter disease trajectory. Until recently, the FDA approved drugs for MCI and AD have only offered symptom management. For example, acetylcholinesterase inhibitors (donepezil, rivastigmine, galantamine) and a N-methyl-d-aspartate receptor antagonist (memantine) modulate cholinergic and glutamatergic signaling, respectively (Howard et al., 2012; Raina et al., 2008; Rogawski and Wenk, 2003). While they effectively modulate symptoms and behaviors, they are unable to reverse or delay the course of AD progression (Raina et al., 2008). However, in June 2021, the Aβ specific antibody, aducanumab was announced as the first FDA approved drug for altering AD disease trajectory (2021b; Sevigny et al., 2016). Aducanumab is unique compared to the historically utilized medications indicated for use in MCI or AD. Its purpose is to remove Aβ plaques and slow disease progression, whereas other FDA approved drugs modulate disease symptoms. Similar to aducanumab, the compounds described in Section 3 of this review, aim to target biological processes responsible for AD pathogenesis. Outcomes for senescence AD trials, therefore, need to consider the underlying pathogenesis (Aβ and tau), the downstream effects on cognition, and symptoms associated with disease progression, in addition to assessing markers of senescence and its consequences.

For AD clinical studies, there are several measures, when collected together, can contribute to a comprehensive summary of AD status. Imaging and biomarker protocols, behavioral assessments, neurological, physical, and functional assessments, as well as a harmonized cognitive/neuropsychological battery has been established by the National Alzheimer’s Coordinating Center (NACC) that promotes standardization of data collection among Alzheimer’s Disease Research Centers (ADRCs) (Weintraub et al., 2009). Similarly, the Alzheimer’s Disease Neuroimaging Initiative (ADNI) has worked hard to validate imaging biomarkers for consistent use in clinical studies (Veitch et al., 2019). Imaging data is commonly collected via MRI to assess morphologic and volumetric changes. PET tracer imaging has improved the utility of imaging data to help identify molecular brain changes in specific brain regions (Alexander et al., 2002; Frisoni et al., 2010; Johnson et al., 2005). Regarding biomarkers relevant to AD, collection of CSF via lumbar puncture often tests for Aβ (Aβ40 and Aβ42), phosphorylated-tau (p-Tau) and total-tau as markers of pathologic load, but tools are being developed to measure these and other (i.e. Aβ structure, Aβ42/Aβ43, Aβ42/APP669-711, p-Tau 181/217/231, flotillin) informative markers less invasively in blood (Suárez-Calvet et al., 2020; Zou et al., 2020). With the identification and improved measuring techniques for AD biomarkers in plasma and CSF, including specificity for tau isoforms, these new tools should be incorporated into newly proposed AD clinical trials. Similar biomarker and imaging strategies to reliably identify senescent cells in vivo would significantly benefit senescence trials. Until these advances are made, levels of senescent cells in the blood or biopsied tissues, and composite measures of SASP factors remain the gold standard for senescence trials (Hickson et al., 2019; Justice et al., 2019b).

3. Clinical trials in AD that directly or indirectly target cellular senescence

3.1. Senolytic compounds

The first senolytic compounds were identified using a hypothesis-driven bioinformatics approach (Zhu et al., 2015). Senescent cells are resistant to apoptosis, which is governed through the upregulation of senescent cell anti-apoptotic pathways (SCAPs). Compounds were subsequently identified that disrupted the SCAPs, inducing death of senescent cells while leaving healthy cells unaffected. Forty-six potential senolytic agents were discovered through this process. To advance translational efforts, the majority of research has focused on agents with known safety profiles and limited off-target effects (Kirkland and Tchkonia, 2020). The best characterized senolytic agents are dasatinib, a tyrosine kinase inhibitor approved for use in humans for cancer treatment, and quercetin, a naturally occurring plant flavonoid. The agents have a synergistic effect, making their combination more potent for senescent cell clearance (Zhu et al., 2015). As senescent cells do not divide and accumulate over a period of weeks, they can be administered using an intermittent approach, which further serves to reduce the risk of side effects (Kirkland and Tchkonia, 2020). In preclinical trials, the combination of dasatinib and quercetin (D + Q) have been found to alleviate numerous chronic medical conditions including vascular stiffness, osteoporosis, frailty, and hepatic stenosis (Kirkland and Tchkonia, 2020; Ogrodnik et al., 2017; Roos et al., 2016; Tchkonia and Kirkland, 2018). The first clinical trial of D + Q was conducted as an open-labeled study in adults with IPF (Justice et al., 2019a). The treatment was generally well-tolerated with no trial discontinuation due to side effects. Although IPF is a fatally progressive disease, participants demonstrated improvements in physical functioning after three weeks of intermittent treatment. A subsequent open label trial of D + Q conducted in adults with diabetic kidney disease demonstrated effective senescent cell clearance in adipose tissue after only 11 days of intermittent treatment (Hickson et al., 2019).

Initial proof-of-concept data for clearing senescent cells as a therapeutic treatment for AD was conducted by our lab using tau transgenic mice (Musi et al., 2018) and has since been replicated by independent teams (Bussian et al., 2018; Zhang et al., 2019). In our study, oral D + Q were intermittently administered to tau transgenic mice with late-stage pathology (approximated to a 70-year-old human with advanced AD) (Musi et al., 2018). The treatment effectively reduced cellular senescence and associated SASP. The 35 % reduction in NFTs was accompanied by enhanced neuron density, decreased ventricular enlargement, diminished tau accumulation, and restoration of aberrant cerebral blood flow. A subsequent preclinical study validated the findings, reporting that intermittently administered D + Q cleared senescent cells in the central nervous system, reduced Aβ plaques, attenuated neuroinflammation, and enhanced cognition (Zhang et al., 2019).

The strong mechanistic support for cellular senescence as a novel target for AD in preclinical trials, coupled with the encouraging safety data from human studies for other conditions, provided the framework for translational research in neurodegenerative disease. The first clinical trial of D + Q for early-stage AD has completed enrollment (Gonzales et al., 2021). The primary aim of the open-label pilot study was to examine the central nervous system penetrance of D and Q in a small sample of older adults with early-stage AD (NCT04063124). In addition, two placebo-controlled trials of D + Q for neurodegenerative disease are underway (NCT04685590 and NCT04785300). One of the trials in development is a multi-site, double blind, randomized, placebo-controlled study of senolytic therapy in older adults with amnestic mild cognitive impairment (MCI) or early-stage dementia (Clinical Dementia Rating Scale (CDR) Global 0.5–1) due to AD (elevated CSF total tau/Aβ42 ratio). The treatment regimen will consist of 12-weeks of intermittently administered oral D + Q. Participants will continue to be monitored across a 9-month follow-up period to evaluate changes in senescence markers and disease trajectory. Across the study duration, safety will be closely monitored with assessment of adverse events, vital signs, physical examination, ECG, and laboratory parameters (biochemistry, hematology, liver, and renal function). Baseline, post-treatment (12 weeks), and end of study (48 weeks) changes will be examined across indices of blood and CSF senescence and AD markers, tau PET retention, cognition, functional status, brain MRI, and physical functioning. The results will shed insight on efficacy, safety, and feasibility of senolytic treatment for AD and will be used to evaluate the decision to pursue a larger trial.

3.2. Senomorphic compounds

While senolytics ablate senescent cells, senomorphics attenuate senescence without affecting the total number of senescent cells (Mongelli et al., 2020). Several senomorphics exert their effects by modulating the SASP and may extend organism lifespan (Kirkland and Tchkonia, 2020). Similar to senolytics, clinical trials of senomorphic agents for neurodegenerative disease are presently underway. The sections below highlight a few examples of senomorphics and select clinical trials targeting neurodegenerative disease. As these agents have pleiotropic effects on pathways relevant to aging (Kirkland and Tchkonia, 2020; Mongelli et al., 2020), assessment of their potential impact directly attributable to senomorphic properties will require thorough investigation.

3.2.1. Rapamycin

The mechanistic target of rapamycin (mTOR) is a serine-kinase that is integral to cellular metabolism and its dysregulation propagates multi-system degradation, potentially contributing to age-related chronic diseases (Stanfel et al., 2009). mTOR is a member of the phosphatidylinositol-3 kinases (PI3K)-related kinase (PIKK) family that is present in at least two protein complexes, mTOR complex 1 (mTORC1) and mTORC2 (Saxton and Sabatini, 2017). MTORC1 has been directly implicated in propagating the SASP through upregulation of cytokine IL1A (Laberge et al., 2015). mTORC1 is also hypothesized to contribute to the metabolic dysregulation of senescent cells by inhibiting autophagy of dysfunctional mitochondria (Carroll et al., 2017).

Rapamycin, an FDA-approved immune modulator, effectively suppresses the mTOR pathway and extends lifespan in model organisms (Arriola Apelo and Lamming, 2016). Rapamycin dampens the SASP through inhibition of IL1α, which in turn reduces the transcription of inflammatory genes regulated by NF-κB (Laberge et al., 2015; Mongelli et al., 2020). In preclinical trials, rapamycin has been shown to ameliorate several age-related conditions including neurodegenerative disease. In mouse models of AD, oral rapamycin administration attenuated cerebral mTOR signaling, and reduced pro-inflammatory cytokine levels in the central nervous system (Van Skike et al., 2018). Rapamycin has also been shown to reduce the accumulation of Aβ and tau, enhance cerebral blood flow, and improve cognition in preclinical studies (Caccamo et al., 2010; Lin et al., 2013; Tang et al., 2013). A double blind, randomized, placebo-controlled Phase 2 clinical trial of rapamycin for MCI and early-stage dementia (CDR Global 0.5–1) due to AD (amyloid PET positivity) is presently underway (NCT04629495). Enrolled participants will be randomized 1:1 to rapamycin (1 mg/day) or placebo for 12-months followed by an extended 6-month observation period. Safety will be evaluated across the study duration with assessment of adverse events, vital signs, and laboratory parameters. Baseline, post-treatment (12 months) and end of study (18 months) assessments include brain MRI, FDG PET, cognitive evaluation, questionnaires, and blood and CSF markers of AD and neuroinflammation. A multicenter, phase 2 randomized, double-blind, placebo-controlled clinical trial of rapamycin for ALS is also underway (NCT03359538) (Mandrioli et al., 2018). The primary aim of the investigation is to evaluate changes in regulatory T lymphocytes (Treg), which have been previously associated with ALS disease progression (Beers et al., 2011). Secondary outcomes include broader assessments of changes in immune parameters, inflammatory markers, safety, and clinical outcomes.

3.2.2. Nicotinamide riboside

Sirtuins are a class of histone deacetylases, which play a critical role in the modulation of chromatin structure and enable gene transcription (North and Verdin, 2004; Toiber et al., 2011). They also increase anti-oxidant production and have been shown to extend lifespan in preclinical studies (Kaeberlein et al., 1999). Sirtuins attenuate cellular senescence by preventing age-related telomere attrition, maintaining chromatin integrity, and promoting DNA repair (Lee et al., 2019). Sirtuin activity requires nicotinamide adenine dinucleotide (NAD+), a coenzyme crucial for cellular metabolism (Mouchiroud et al., 2013). Nicotinamide riboside is an NAD + precursor that has been shown to increase lifespan in animal studies (Cantó et al., 2012). Nicotinamide riboside further contributes to axonal integrity and prevents neuronal death in model organisms (Schöndorf et al., 2018; Vaur et al., 2017). We recently completed a double-blinded RCT pilot study of nicotinamide riboside for individuals with MCI. The 10-week study increased blood NAD, was well-tolerated, and showed evidence of altered cerebral blood flow in disease relevant brain regions (Orr et al., 2020). Additional trials examining the efficacy of nicotinamide riboside for subjective cognitive decline, MCI, and AD are underway (NCT04078178, NCT04430517).

3.2.3. Metformin

Metformin is an FDA-approved medication for diabetes, which modulates several key pathways underlying aging (Barzilai et al., 2016). These include reducing IGF-1 and mTOR signaling, attenuating reactive oxygen species production, and decreasing cellular senescence (Barzilai et al., 2016; Batandier et al., 2006; Kickstein et al., 2010; Liu et al., 2011; Moiseeva et al., 2013). Metformin has been shown to reduce cellular senescence by stimulating autophagy and inhibiting the NF-κB pathway (Deschênes-Simard et al., 2019; Moiseeva et al., 2013). In model organisms, metformin effectively extends lifespan and healthspan (Chen et al., 2017; Martin-Montalvo et al., 2013). Observational studies have further highlighted the agent’s potential cognitive benefits. Within the Singapore Longitudinal Aging Study, metformin use was associated with a 51 % reduction in risk for cognitive impairment over and above adjustment for vascular risk factors (Ng et al., 2014). A small placebo-controlled clinical trial of individuals with major depression and type 2 diabetes reported that metformin enhanced cognition and reduced depressive symptoms relative to placebo (Guo et al., 2014). However, metformin has also been associated with increased risk of AD in population studies (Imfeld et al., 2012). A large-scale, multi-site NIH-funded initiative is underway to examine the potential of metformin for attenuating age-related diseases including cognitive decline as part of the Target Aging with Metformin (TAME) trial (Barzilai, 2017). Additional clinical trials of metformin for preventing or halting cognitive decline in healthy older adults and individuals with MCI and dementia have been conducted or are presently ongoing (NCT00620191, NCT01965756, NCT03757910, NCT04098666) with initial data supporting treatment-related improvements in cognition (Koenig et al., 2017b; Luchsinger et al., 2016). In a 16-week randomized crossover pilot trial of metformin in individuals with MCI or dementia due to AD, it was found to be safe, tolerable, and detected in the CSF of participants, indicating penetration of the blood brain barrier (Koenig et al., 2017b). Although no changes in AD biomarkers (including Aβ42, total tau, or p-Tau) were detected in the CSF, significant improvements in executive functioning and trends suggesting improvements in attention, learning, and memory were seen (Koenig et al., 2017b). While initial analyses revealed no significant differences in cerebral blood flow with metformin, post-hoc analyses revealed an improvement in orbitofrontal cerebral blood flow in the intervention group compared to placebo (Koenig et al., 2017b). Though the crossover trial did not include a washout period, included a small sample size (n = 20), and was too short to account for placebo responders, it is promising and warrants further investigation.

4. Concluding Remarks

Clinical trials focused on removing senescent cells and their toxicity in the context of AD are in early stages. These pioneering trials face daunting failure rates that have plagued the AD field and are tasked with overcoming challenges associated with determining efficacy/target engagement for a complex senescence phenotype in a notoriously difficult tissue. We also acknowledge that senescent cells contribute to both healthy physiology and disease. The early safety trials will be responsible for identifying potential negative effects of senescent cell removal. The intermittent dosing regimen planned for these trials was designed to optimally minimize potential drug side effects, reduce senescent cells and their detrimental SASP effects while preserving important function. Nevertheless, data from postmortem human brains and mechanistic studies in AD mouse models overwhelmingly support this next step. On the side of optimism, we note that many of the past failed studies focused on clinical outcomes relevant to cognition. With new, improved, sensitive imaging and biomarker assays, trials are now better positioned for success. They are able to recruit subjects with earlier disease stages, assess more sensitive outcomes that reflect the underlying biology, all while tracking changes in important cognitive outcomes (Rafii and Aisen, 2020).

We also note a potential need for interventions that target multiple pathways; senolytic trials innately test this approach. They target a hallmark driver of biological aging, and may have potential to broadly impact cellular and molecular processes across tissues. Indeed, the hallmarks of aging interact across molecular, cellular, and system levels - targeting one (i.e., senescence) may exert pleiotropic effects that may be potentially beneficial for the treatment of AD. Moreover, combining senolytics (or other agents targeting mechanisms underlying the biology of aging) with the newly approved aducanumab, or other promising protein-clearing therapies (i.e., tau reducing approaches) may be considered in future trials. Such a combination strategy could potentially clear existing pathologies and unhealthy cells to improve tissue health and function. In summary, the upcoming senolytic trials are backed by strong evidence and will provide a first glimpse into the efficacy of targeting an aging hallmark to treat AD. Regardless of outcome, these studies are critically important and will lay the groundwork for future geroscience trials in AD.

Acknowledgements

This work was made possible by grants through the Alzheimer’s Drug Discovery Foundation, GC-201908-2019443; the Alzheimer’s Association Part of the Cloud Gate Partnership, PTCG-20-695184; and the Coordinating Center for Claude D. Pepper Older Americans Independence Centers, U24AG059624. Dr. Garbarino is supported by T32AG021890. Drs. Gonzales and Orr also thank the Institute on Methods and Protocols for Advancement of Clinical Trials in ADRD.

References

  1. Abe K, Chiba Y, Hattori S, Yoshimi A, Asami T, Katsuse O, Suda A, Hishimoto A, Alzheimer’s Disease Neuroimaging, I, 2020. Influence of plasma matrix metalloproteinase levels on longitudinal changes in Alzheimer’s disease (AD) biomarkers and cognitive function in patients with mild cognitive impairment due to AD registered in the Alzheimer’s Disease Neuroimaging Initiative database. J. Neurol. Sci 416, 116989. [DOI] [PubMed] [Google Scholar]
  2. Abner EL, Elahi FM, Jicha GA, Mustapic M, Al-Janabi O, Kramer JH, Kapogiannis D, Goetzl EJ, 2020. Endothelial-derived plasma exosome proteins in Alzheimer’s disease angiopathy. FASEB J. 34, 5967–5974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Acosta JC, Banito A, Wuestefeld T, Georgilis A, Janich P, Morton JP, Athineos D, Kang TW, Lasitschka F, Andrulis M, Pascual G, Morris KJ, Khan S, Jin H, Dharmalingam G, Snijders AP, Carroll T, Capper D, Pritchard C, Inman GJ, Longerich T, Sansom OJ, Benitah SA, Zender L, Gil J, 2013. A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nat. Cell Biol 15, 978–990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Alexander GE, Chen K, Pietrini P, Rapoport SI, Reiman EM, 2002. Longitudinal PET evaluation of cerebral metabolic decline in dementia: a potential outcome measure in alzheimer’s disease treatment studies. Am. J. Psychiatry 159, 738–745. [DOI] [PubMed] [Google Scholar]
  5. Ali TB, Schleret TR, Reilly BM, Chen WY, Abagyan R, 2015. Adverse effects of cholinesterase inhibitors in dementia, according to the pharmacovigilance databases of the United-States and Canada. PLoS One 10, e0144337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Al-Mashhadi S, Simpson JE, Heath PR, Dickman M, Forster G, Matthews FE, Brayne C, Ince PG, Wharton SB, Medical Research Council Cognitive, F, Ageing, S., 2015. Oxidative glial cell damage associated with white matter lesions in the aging human brain. Brain Pathol. 25, 565–574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Anon, 2012. In: Eisai Co L (Ed.), ARICEPT (Donepezil Hydrochloride) Tablets. Woodcliff Lake, NJ, 07677. [Google Scholar]
  8. Anon, 2021a. 2021 Alzheimer’s disease facts and figures. Alzheimer’s Dement. 17, 327–406. [DOI] [PubMed] [Google Scholar]
  9. Anon, 2021b. In: Inc B (Ed.), ADUHELM (aducanumab-avwa) Injection for Intravenous Use. Cambridge, MA: 02142. [Google Scholar]
  10. Arendt T, Rodel L, Gartner U, Holzer M, 1996. Expression of the cyclin-dependent kinase inhibitor p16 in Alzheimer’s disease. Neuroreport 7, 3047–3049. [DOI] [PubMed] [Google Scholar]
  11. Arriola Apelo SI, Lamming DW, 2016. Rapamycin: an InhibiTOR of aging emerges from the soil of Easter Island. J. Gerontol. A Biol. Sci. Med. Sci 71, 841–849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Arun P, Rossetti F, Wilder DM, Sajja S, Van Albert SA, Wang Y, Gist ID, Long JB, 2020. Blast exposure leads to accelerated cellular senescence in the rat brain. Front. Neurol 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Asai H, Ikezu S, Tsunoda S, Medalla M, Luebke J, Haydar T, Wolozin B, Butovsky O, Kugler S, Ikezu T, 2015. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat. Neurosci 18, 1584–1593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Barrio-Alonso E, Hernandez-Vivanco A, Walton CC, Perea G, Frade JM, 2018. Cell cycle reentry triggers hyperploidization and synaptic dysfunction followed by delayed cell death in differentiated cortical neurons. Sci. Rep 8, 14316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Barzilai N, 2017. Targeting aging with metformin (TAME). Innov. Aging 743. [Google Scholar]
  16. Barzilai N, Crandall JP, Kritchevsky SB, Espeland MA, 2016. Metformin as a tool to target aging. Cell Metab. 23, 1060–1065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Batandier C, Guigas B, Detaille D, El-Mir MY, Fontaine E, Rigoulet M, Leverve XM, 2006. The ROS production induced by a reverse-electron flux at respiratory-chain complex 1 is hampered by metformin. J. Bioenerg. Biomembr 38, 33–42. [DOI] [PubMed] [Google Scholar]
  18. Beers DR, Henkel JS, Zhao W, Wang J, Huang A, Wen S, Liao B, Appel SH, 2011. Endogenous regulatory T lymphocytes ameliorate amyotrophic lateral sclerosis in mice and correlate with disease progression in patients with amyotrophic lateral sclerosis. Brain 134, 1293–1314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Bennett RE, Robbins AB, Hu M, Cao X, Betensky RA, Clark T, Das S, Hyman BT, 2018. Tau induces blood vessel abnormalities and angiogenesis-related gene expression in P301L transgenic mice and human Alzheimer’s disease. Proc. Natl. Acad. Sci. U. S. A 115, E1289–E1298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Bernstein OM, Grill JD, Gillen DL, 2021. Recruitment and retention of participant and study partner dyads in two multinational Alzheimer’s disease registration trials. Alzheimer’s Res. Ther 13, 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Bhat R, Crowe EP, Bitto A, Moh M, Katsetos CD, Garcia FU, Johnson FB, Trojanowski JQ, Sell C, Torres C, 2012. Astrocyte senescence as a component of Alzheimer’s disease. PLoS One 7, e45069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Blasko I, Lederer W, Oberbauer H, Walch T, Kemmler G, Hinterhuber H, Marksteiner J, Humpel C, 2006. Measurement of thirteen biological markers in CSF of patients with Alzheimer’s disease and other dementias. Dement. Geriatr. Cogn. Disord 21, 9–15. [DOI] [PubMed] [Google Scholar]
  23. Blum-Degen D, Muller T, Kuhn W, Gerlach M, Przuntek H, Riederer P, 1995. Interleukin-1 beta and interleukin-6 are elevated in the cerebrospinal fluid of Alzheimer’s and de novo Parkinson’s disease patients. Neurosci. Lett 202, 17–20. [DOI] [PubMed] [Google Scholar]
  24. Borah B, Sacco P, Zarotsky V, 2010. Predictors of adherence among Alzheimer’s disease patients receiving oral therapy. Curr. Med. Res. Opin 26, 1957–1965. [DOI] [PubMed] [Google Scholar]
  25. Brosseron F, Krauthausen M, Kummer M, Heneka MT, 2014. Body fluid cytokine levels in mild cognitive impairment and Alzheimer’s disease: a comparative overview. Mol. Neurobiol 50, 534–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Bryant AG, Hu M, Carlyle BC, Arnold SE, Frosch MP, Das S, Hyman BT, Bennett RE, 2020. Cerebrovascular senescence is associated with tau pathology in Alzheimer’s disease. Front. Neurol 11, 575953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Buratta S, Urbanelli L, Sagini K, Giovagnoli S, Caponi S, Fioretto D, Mitro N, Caruso D, Emiliani C, 2017. Extracellular vesicles released by fibroblasts undergoing H-Ras induced senescence show changes in lipid profile. PLoS One 12, e0188840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Burgos K, Malenica I, Metpally R, Courtright A, Rakela B, Beach T, Shill H, Adler C, Sabbagh M, Villa S, 2014. Profiles of extracellular miRNA in cerebrospinal fluid and serum from patients with Alzheimer’s and Parkinson’s diseases correlate with disease status and features of pathology. PLoS One 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Bussian TJ, Aziz A, Meyer CF, Swenson BL, van Deursen JM, Baker DJ, 2018. Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature 562, 578–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Caccamo A, Majumder S, Richardson A, Strong R, Oddo S, 2010. Molecular interplay between mammalian target of rapamycin (mTOR), amyloid-beta, and Tau: effects on cognitive impairments. J. Biol. Chem 285, 13107–13120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Cantó C, Houtkooper RH, Pirinen E, Youn DY, Oosterveer MH, Cen Y, Fernandez-Marcos PJ, Yamamoto H, Andreux PA, Cettour-Rose P, Gademann K, Rinsch C, Schoonjans K, Sauve AA, Auwerx J, 2012. The NAD(+) precursor nicotinamide riboside enhances oxidative metabolism and protects against high-fat diet-induced obesity. Cell Metab 15, 838–847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Carracedo J, Alique M, Ramirez-Carracedo R, Bodega G, Ramirez R, 2019. Endothelial extracellular vesicles produced by senescent cells: pathophysiological role in the cardiovascular disease associated with all types of diabetes mellitus. Curr. Vasc. Pharmacol 17, 447–454. [DOI] [PubMed] [Google Scholar]
  33. Carroll B, Nelson G, Rabanal-Ruiz Y, Kucheryavenko O, Dunhill-Turner NA, Chesterman CC, Zahari Q, Zhang T, Conduit SE, Mitchell CA, Maddocks ODK, Lovat P, von Zglinicki T, Korolchuk VI, 2017. Persistent mTORC1 signaling in cell senescence results from defects in amino acid and growth factor sensing. J. Cell Biol 216, 1949–1957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Cha DJ, Mengel D, Mustapic M, Liu W, Selkoe DJ, Kapogiannis D, Galasko D, Rissman RA, Bennett DA, Walsh DM, 2019. miR-212 and miR-132 are downregulated in neurally derived plasma exosomes of Alzheimer’s patients. Front. Neurosci 13, 1208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Chatterjee S, Mudher A, 2018. Alzheimer’s disease and type 2 diabetes: a critical assessment of the shared pathological traits. Front. Neurosci 12, 383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Chen J, Ou Y, Li Y, Hu S, Shao L-W, Liu Y, 2017. Metformin extends C. elegans lifespan through lysosomal pathway. Elife 6, e31268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Chinta SJ, Woods G, Demaria M, Rane A, Zou Y, McQuade A, Rajagopalan S, Limbad C, Madden DT, Campisi J, Andersen JK, 2018. Cellular senescence is induced by the environmental neurotoxin paraquat and contributes to neuropathology linked to parkinson’s disease. Cell Rep 22, 930–940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Chistiakov DA, Orekhov AN, Bobryshev YV, 2016. Cardiac extracellular vesicles in normal and infarcted heart. Int. J. Mol. Sci 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Chow HM, Shi M, Cheng A, Gao Y, Chen G, Song X, So RWL, Zhang J, Herrup K, 2019. Age-related hyperinsulinemia leads to insulin resistance in neurons and cell-cycle-induced senescence. Nat. Neurosci 22, 1806–1819. [DOI] [PubMed] [Google Scholar]
  40. Colombo M, Moita C, van Niel G, Kowal J, Vigneron J, Benaroch P, Manel N, Moita LF, Thery C, Raposo G, 2013. Analysis of ESCRT functions in exosome biogenesis, composition and secretion highlights the heterogeneity of extracellular vesicles. J. Cell. Sci 126, 5553–5565. [DOI] [PubMed] [Google Scholar]
  41. Colombo M, Raposo G, Thery C, 2014. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol 30, 255–289. [DOI] [PubMed] [Google Scholar]
  42. Coppé JP, Patil C, Rodier F, Sun Y, Muñoz DP, Goldstein J, Nelson PS, Desprez PY, Campisi J, 2008. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol 6, 2853–2868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Coppe JP, Desprez PY, Krtolica A, Campisi J, 2010. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu. Rev. Pathol 5, 99–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Coppe JP, Rodier F, Patil CK, Freund A, Desprez PY, Campisi J, 2011. Tumor suppressor and aging biomarker p16(INK4a) induces cellular senescence without the associated inflammatory secretory phenotype. J. Biol. Chem 286, 36396–36403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Cummings JL, Morstorf T, Zhong K, 2014. Alzheimer’s disease drug-development pipeline: few candidates, frequent failures. Alzheimer’s Res. Ther 6, 37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. de Jong OG, Verhaar MC, Chen Y, Vader P, Gremmels H, Posthuma G, Schiffelers RM, Gucek M, van Balkom BW, 2012. Cellular stress conditions are reflected in the protein and RNA content of endothelial cell-derived exosomes. J. Extracell. Vesicles 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. de Leeuw FE, de Groot JC, Achten E, Oudkerk M, Ramos LM, Heijboer R, Hofman A, Jolles J, van Gijn J, Breteler MM, 2001. Prevalence of cerebral white matter lesions in elderly people: a population based magnetic resonance imaging study. The Rotterdam Scan Study. J. Neurol. Neurosurg. Psychiatry 70, 9–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Debacq-Chainiaux F, Erusalimsky JD, Campisi J, Toussaint O, 2009. Protocols to detect senescence-associated beta-galactosidase (SA-betagal) activity, a biomarker of senescent cells in culture and in vivo. Nat. Protoc 4, 1798–1806. [DOI] [PubMed] [Google Scholar]
  49. Dehkordi SK, Walker J, Sah E, Bennett E, Atrian F, Frost B, Woost B, Bennett RE, Orr TC, Zhou Y, Andhey PS, Colonna M, Sudmant PH, Xu P, Wang M, Zhang B, Zare H, Orr ME, 2021. Profiling senescent cells in human brains reveals neurons with CDKN2D/p19 and tau neuropathology. Nat. Aging (Accepted) [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Deschênes-Simard X, Parisotto M, Rowell MC, Le Calvé B, Igelmann S, Moineau-Vallée K, Saint-Germain E, Kalegari P, Bourdeau V, Kottakis F, 2019. Circumventing senescence is associated with stem cell properties and metformin sensitivity. Aging Cell 18, e12889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. DiBattista AM, Sierra F, Masliah E, 2020. NIA workshop on senescence in brain aging and Alzheimer’s disease and its related dementias. Geroscience 42, 389–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Ding J, Davis-Plourde KL, Sedaghat S, Tully PJ, Wang W, Phillips C, Pase MP, Himali JJ, Gwen Windham B, Griswold M, Gottesman R, Mosley TH, White L, Guðnason V, Debette S, Beiser AS, Seshadri S, Ikram MA, Meirelles O, Tzourio C, Launer LJ, 2020. Antihypertensive medications and risk for incident dementia and Alzheimer’s disease: a meta-analysis of individual participant data from prospective cohort studies. Lancet Neurol 19, 61–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Eissa S, Matboli M, Aboushahba R, Bekhet MM, Soliman Y, 2016. Urinary exosomal microRNA panel unravels novel biomarkers for diagnosis of type 2 diabetic kidney disease. J. Diabetes Compl 30, 1585–1592. [DOI] [PubMed] [Google Scholar]
  54. Erten-Lyons D, Woltjer R, Kaye J, Mattek N, Dodge HH, Green S, Tran H, Howieson DB, Wild K, Silbert LC, 2013. Neuropathologic basis of white matter hyperintensity accumulation with advanced age. Neurology 81, 977–983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Farlow MR, 2003. Clinical pharmacokinetics of galantamine. Clin. Pharmacokinet 42, 1383–1392. [DOI] [PubMed] [Google Scholar]
  56. Farrall AJ, Wardlaw JM, 2009. Blood-brain barrier: ageing and microvascular disease–systematic review and meta-analysis. Neurobiol. Aging 30, 337–352. [DOI] [PubMed] [Google Scholar]
  57. Fiandaca MS, Kapogiannis D, Mapstone M, Boxer A, Eitan E, Schwartz JB, Abner EL, Petersen RC, Federoff HJ, Miller BL, Goetzl EJ, 2015. Identification of preclinical Alzheimer’s disease by a profile of pathogenic proteins in neurally derived blood exosomes: a case-control study. Alzheimer’s Dement. 11, 600–607 e601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Freund A, Laberge RM, Demaria M, Campisi J, 2012. Lamin B1 loss is a senescence-associated biomarker. Mol. Biol. Cell 23, 2066–2075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Frisoni GB, Fox NC, Jack CR Jr., Scheltens P, Thompson PM, 2010. The clinical use of structural MRI in Alzheimer disease. Nat. Rev. Neurol 6, 67–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Fujimaki K, Li R, Chen H, Della Croce K, Zhang HH, Xing J, Bai F, Yao G, 2019. Graded regulation of cellular quiescence depth between proliferation and senescence by a lysosomal dimmer switch. Proc. Natl. Acad. Sci. U. S. A 116, 22624–22634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Gao J, Wu Y, He D, Zhu X, Li H, Liu H, Liu H, 2020. Anti-aging effects of Ribes meyeri anthocyanins on neural stem cells and aging mice. Aging 12, 17738–17753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Garre-Olmo J, López-Pousa S, Vilalta-Franch J, de Gracia Blanco M, Vilarrasa AB, 2010. Grouping and trajectories of neuropsychiatric symptoms in patients with Alzheimer’s disease. Part II: two-year patient trajectories. J. Alzheimer’s Dis 22, 1169–1180. [DOI] [PubMed] [Google Scholar]
  63. Geng Y-Q, Guan J-T, Xu X-H, Fu Y-C, 2010. Senescence-associated beta-galactosidase activity expression in aging hippocampal neurons. Biochem. Biophys. Res. Commun 396, 866–869. [DOI] [PubMed] [Google Scholar]
  64. Gillispie GJ, Sah E, Krishnamurthy S, Ahmidouch MY, Zhang B, Orr ME, 2021. Evidence of the cellular senescence stress response in mitotically active brain cells—implications for cancer and neurodegeneration. Life 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Goetzl EJ, 2020. Advancing medicine for Alzheimer’s disease: a plasma neural exosome platform. FASEB J 34 (10), 13079–13084. [DOI] [PubMed] [Google Scholar]
  66. Goetzl EJ, Boxer A, Schwartz JB, Abner EL, Petersen RC, Miller BL, Kapogiannis D, 2015. Altered lysosomal proteins in neural-derived plasma exosomes in preclinical Alzheimer disease. Neurology 85, 40–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Goetzl EJ, Kapogiannis D, Schwartz JB, Lobach IV, Goetzl L, Abner EL, Jicha GA, Karydas AM, Boxer A, Miller BL, 2016a. Decreased synaptic proteins in neuronal exosomes of frontotemporal dementia and Alzheimer’s disease. FASEB J. 30 (12), 4141–4148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Goetzl EJ, Mustapic M, Kapogiannis D, Eitan E, Lobach IV, Goetzl L, Schwartz JB, Miller BL, 2016b. Cargo proteins of plasma astrocyte-derived exosomes in Alzheimer’s disease. FASEB J 30, 3853–3859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Goetzl EJ, Abner EL, Jicha GA, Kapogiannis D, Schwartz JB, 2018. Declining levels of functionally specialized synaptic proteins in plasma neuronal exosomes with progression of Alzheimer’s disease. FASEB J 32, 888–893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Gonzales MM, Garbarino VR, Marques Zilli E, Petersen RC, Kirkland JL, Tchkonia T, Musi N, Seshadri S, Craft S, Orr ME, 2021. Senolytic therapy to modulate the progression of Alzheimer’s disease (SToMP-AD): a pilot clinical trial. J. Prev. Alzheimer’s Dis. (Accepted) [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O, Bishop C, Campisi J, Collado M, Evangelou K, Ferbeyre G, Gil J, Hara E, Krizhanovsky V, Jurk D, Maier AB, Narita M, Niedernhofer L, Passos JF, Robbins PD, Schmitt CA, Sedivy J, Vougas K, von Zglinicki T, Zhou D, Serrano M, Demaria M, 2019. Cellular senescence: defining a path forward. Cell 179, 813–827. [DOI] [PubMed] [Google Scholar]
  72. Goyal VK, 1982. Lipofuscin pigment accumulation in human brain during aging. Exp. Gerontol 17, 481–487. [DOI] [PubMed] [Google Scholar]
  73. Green RC, Schneider LS, Amato DA, Beelen AP, Wilcock G, Swabb EA, Zavitz KH, 2009. Effect of tarenflurbil on cognitive decline and activities of daily living in patients with mild Alzheimer disease: a randomized controlled trial. JAMA 302, 2557–2564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Greenberg SM, Vernooij MW, Cordonnier C, Viswanathan A, Al-Shahi Salman R, Warach S, Launer LJ, Van Buchem MA, Breteler MM, Microbleed Study G, 2009. Cerebral microbleeds: a guide to detection and interpretation. Lancet Neurol. 8, 165–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Grill JD, Raman R, Ernstrom K, Aisen P, Karlawish J, 2013. Effect of study partner on the conduct of Alzheimer disease clinical trials. Neurology 80, 282–288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Grossberg GT, Stahelin HB, Messina JC, Anand R, Veach J, 2000. Lack of adverse pharmacodynamic drug interactions with rivastigmine and twenty-two classes of medications. Int. J. Geriatr. Psychiatry 15, 242–247. [DOI] [PubMed] [Google Scholar]
  77. Group, S.M.I.f.t.S.R, Williamson JD, Pajewski NM, Auchus AP, Bryan RN, Chelune G, Cheung AK, Cleveland ML, Coker LH, Crowe MG, Cushman WC, Cutler JA, Davatzikos C, Desiderio L, Erus G, Fine LJ, Gaussoin SA, Harris D, Hsieh MK, Johnson KC, Kimmel PL, Tamura MK, Launer LJ, Lerner AJ, Lewis CE, Martindale-Adams J, Moy CS, Nasrallah IM, Nichols LO, Oparil S, Ogrocki PK, Rahman M, Rapp SR, Reboussin DM, Rocco MV, Sachs BC, Sink KM, Still CH, Supiano MA, Snyder JK, Wadley VG, Walker J, Weiner DE, Whelton PK, Wilson VM, Woolard N, Wright JT Jr., Wright CB, 2019. Effect of intensive vs standard blood pressure control on probable dementia: a randomized clinical trial. JAMA 321, 553–561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Guo M, Mi J, Jiang QM, Xu JM, Tang YY, Tian G, Wang B, 2014. Metformin may produce antidepressant effects through improvement of cognitive function among depressed patients with diabetes mellitus. Clin. Exp. Pharmacol. Physiol 41, 650–656. [DOI] [PubMed] [Google Scholar]
  79. Han F, Zhai FF, Wang Q, Zhou LX, Ni J, Yao M, Li ML, Zhang SY, Cui LY, Jin ZY, Zhu YC, 2018. Prevalence and risk factors of cerebral small vessel disease in a chinese population-based sample. J. Stroke 20, 239–246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Hanneken A, Frautschy S, Galasko D, Baird A, 1995. A fibroblast growth factor binding protein in human cerebral spinal fluid. Neuroreport 6, 886–888. [DOI] [PubMed] [Google Scholar]
  81. Harada CN, Natelson Love MC, Triebel KL, 2013. Normal cognitive aging. Clin. Geriatr. Med 29, 737–752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Hayflick L, Moorhead PS, 1961. The serial cultivation of human diploid cell strains. Exp. Cell Res 25, 585–621. [DOI] [PubMed] [Google Scholar]
  83. He S, Sharpless NE, 2017. Senescence in health and disease. Cell 169, 1000–1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. He N, Jin WL, Lok KH, Wang Y, Yin M, Wang ZJ, 2013. Amyloid-β1–42 oligomer accelerates senescence in adult hippocampal neural stem/progenitor cells via formylpeptide receptor 2. Cell Death Dis. 4, e924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Hedlund M, Nagaeva O, Kargl D, Baranov V, Mincheva-Nilsson L, 2011. Thermal- and oxidative stress causes enhanced release of NKG2D ligand-bearing immunosuppressive exosomes in leukemia/lymphoma T and B cells. PLoS One 6, e16899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Hernandez-Segura A, de Jong TV, Melov S, Guryev V, Campisi J, Demaria M, 2017a. Unmasking transcriptional heterogeneity in senescent cells. Curr. Biol 27, 2652–2660 e2654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Hernandez-Segura A, de Jong TV, Melov S, Guryev V, Campisi J, Demaria M, 2017b. Unmasking transcriptional heterogeneity in senescent cells. Curr. Biol 27, 2652–2660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Hickson LJ, Langhi Prata LGP, Bobart SA, Evans TK, Giorgadze N, Hashmi SK, Herrmann SM, Jensen MD, Jia Q, Jordan KL, Kellogg TA, Khosla S, Koerber DM, Lagnado AB, Lawson DK, LeBrasseur NK, Lerman LO, McDonald KM, McKenzie TJ, Passos JF, Pignolo RJ, Pirtskhalava T, Saadiq IM, Schaefer KK, Textor SC, Victorelli SG, Volkman TL, Xue A, Wentworth MA, Wissler Gerdes EO, Zhu Y, Tchkonia T, Kirkland JL, 2019. Senolytics decrease senescent cells in humans: preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease. EBioMedicine 47, 446–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Hilal S, Mok V, Youn YC, Wong A, Ikram MK, Chen CL, 2017. Prevalence, risk factors and consequences of cerebral small vessel diseases: data from three Asian countries. J. Neurol. Neurosurg. Psychiatry 88, 669–674. [DOI] [PubMed] [Google Scholar]
  90. Horvath S, Raj K, 2018. DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nat. Rev. Genet 19, 371. [DOI] [PubMed] [Google Scholar]
  91. Howard R, McShane R, Lindesay J, Ritchie C, Baldwin A, Barber R, Burns A, Dening T, Findlay D, Holmes C, Hughes A, Jacoby R, Jones R, Jones R, McKeith I, Macharouthu A, O’Brien J, Passmore P, Sheehan B, Juszczak E, Katona C, Hills R, Knapp M, Ballard C, Brown R, Banerjee S, Onions C, Griffin M, Adams J, Gray R, Johnson T, Bentham P, Phillips P, 2012. Donepezil and memantine for moderate-to-severe Alzheimer’s disease. N. Engl. J. Med 366, 893–903. [DOI] [PubMed] [Google Scholar]
  92. Hu G, Xia Y, Chen B, Zhang J, Gong L, Chen Y, Li Q, Wang Y, Deng Z, 2021. ESC-sEVs rejuvenate aging hippocampal NSCs by transferring SMADs to regulate the MYT1-Egln3-Sirt1 axis. Mol. Ther 29, 103–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Imfeld P, Bodmer M, Jick SS, Meier CR, 2012. Metformin, other antidiabetic drugs, and risk of Alzheimer’s disease: a population-based case–control study. J. Am. Geriatr. Soc 60, 916–921. [DOI] [PubMed] [Google Scholar]
  94. Italiani P, Puxeddu I, Napoletano S, Scala E, Melillo D, Manocchio S, Angiolillo A, Migliorini P, Boraschi D, Vitale E, Di Costanzo A, 2018. Circulating levels of IL-1 family cytokines and receptors in Alzheimer’s disease: new markers of disease progression? J. Neuroinflammation 15, 342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Ito K, Corrigan B, Romero K, Anziano R, Neville J, Stephenson D, Lalonde R, 2013. Understanding placebo responses in Alzheimer’s disease clinical trials from the literature meta-data and CAMD database. J. Alzheimer’s Dis 37, 173–183. [DOI] [PubMed] [Google Scholar]
  96. Jack CR Jr., Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, Holtzman DM, Jagust W, Jessen F, Karlawish J, Liu E, Molinuevo JL, Montine T, Phelps C, Rankin KP, Rowe CC, Scheltens P, Siemers E, Snyder HM, Sperling R, Contributors, 2018. NIA-AA Research Framework: toward a biological definition of Alzheimer’s disease. Alzheimer’s Dement 14, 535–562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Jakhar R, Crasta K, 2019. Exosomes as emerging pro-tumorigenic mediators of the senescence-associated secretory phenotype. Int. J. Mol. Sci 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Janson J, Laedtke T, Parisi JE, O’Brien P, Petersen RC, Butler PC, 2004. Increased risk of type 2 diabetes in Alzheimer disease. Diabetes 53, 474–481. [DOI] [PubMed] [Google Scholar]
  99. Johnson NA, Jahng GH, Weiner MW, Miller BL, Chui HC, Jagust WJ, Gorno-Tempini ML, Schuff N, 2005. Pattern of cerebral hypoperfusion in Alzheimer disease and mild cognitive impairment measured with arterial spin-labeling MR imaging: initial experience. Radiology 234, 851–859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Jung KO, Youn H, Lee CH, Kang KW, Chung JK, 2017. Visualization of exosome-mediated miR-210 transfer from hypoxic tumor cells. Oncotarget 8, 9899–9910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Jurk D, Wang C, Miwa S, Maddick M, Korolchuk V, Tsolou A, Gonos ES, Thrasivoulou C, Saffrey MJ, Cameron K, von Zglinicki T, 2012. Postmitotic neurons develop a p21-dependent senescence-like phenotype driven by a DNA damage response. Aging Cell 11, 996–1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Justice JN, Gregory H, Tchkonia T, LeBrasseur NK, Kirkland JL, Kritchevsky SB, Nicklas BJ, 2018. Cellular senescence biomarker p16INK4a+ cell burden in thigh adipose is associated with poor physical function in older women. J. Gerontol. A Biol. Sci. Med. Sci 73, 939–945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Justice JN, Nambiar AM, Tchkonia T, LeBrasseur NK, Pascual R, Hashmi SK, Prata L, Masternak MM, Kritchevsky SB, Musi N, 2019a. Senolytics in idiopathic pulmonary fibrosis: results from a first-in-human, open-label, pilot study. EBioMedicine 40, 554–563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Justice JN, Nambiar AM, Tchkonia T, LeBrasseur NK, Pascual R, Hashmi SK, Prata L, Masternak MM, Kritchevsky SB, Musi N, Kirkland JL, 2019b. Senolytics in idiopathic pulmonary fibrosis: results from a first-in-human, open-label, pilot study. EBioMedicine 40, 554–563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Kaeberlein M, McVey M, Guarente L, 1999. The SIR2/3/4 complex and SIR2 alone promote longevity in Saccharomyces cerevisiae by two different mechanisms. Genes Dev. 13, 2570–2580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Kalluri R, 2016. The biology and function of exosomes in cancer. J. Clin. Invest 126, 1208–1215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Kang C, Xu Q, Martin TD, Li MZ, Demaria M, Aron L, Lu T, Yankner BA, Campisi J, Elledge SJ, 2015. The DNA damage response induces inflammation and senescence by inhibiting autophagy of GATA4. Science 349, aaa5612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Kapogiannis D, 2020. Exosome biomarkers revolutionize preclinical diagnosis of neurodegenerative diseases and assessment of treatment responses in clinical trials. Adv. Exp. Med. Biol 1195, 149. [DOI] [PubMed] [Google Scholar]
  109. Kapogiannis D, Boxer A, Schwartz JB, Abner EL, Biragyn A, Masharani U, Frassetto L, Petersen RC, Miller BL, Goetzl EJ, 2015. Dysfunctionally phosphorylated type 1 insulin receptor substrate in neural-derived blood exosomes of preclinical Alzheimer’s disease. FASEB J. 29, 589–596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Kettunen R, Taipale H, Tolppanen A-M, Tanskanen A, Tiihonen J, Hartikainen S, Koponen M, 2019. Duration of new antidepressant use and factors associated with discontinuation among community-dwelling persons with Alzheimer’s disease. Eur. J. Clin. Pharmacol 75, 417–425. [DOI] [PubMed] [Google Scholar]
  111. Kickstein E, Krauss S, Thornhill P, Rutschow D, Zeller R, Sharkey J, Williamson R, Fuchs M, Köhler A, Glossmann H, Schneider R, Sutherland C, Schweiger S, 2010. Biguanide metformin acts on tau phosphorylation via mTOR/protein phosphatase 2A (PP2A) signaling. Proc. Natl. Acad. Sci. U. S. A 107, 21830–21835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Kiko T, Nakagawa K, Tsuduki T, Furukawa K, Arai H, Miyazawa T, 2014. MicroRNAs in plasma and cerebrospinal fluid as potential markers for Alzheimer’s disease. J. Alzheimer’s Dis 39, 253–259. [DOI] [PubMed] [Google Scholar]
  113. Kirkland JL, Tchkonia T, 2020. Senolytic drugs: from discovery to translation. J. Intern. Med 288, 518–536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Koenig AM, Mechanic-Hamilton D, Xie SX, Combs MF, Cappola AR, Xie L, Detre JA, Wolk DA, Arnold SE, 2017a. Effects of the insulin sensitizer metformin in Alzheimer disease: pilot data from a randomized placebo-controlled crossover study. Alzheimer Dis. Assoc. Disord 31, 107–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Koenig AM, Mechanic-Hamilton D, Xie SX, Combs MF, Cappola AR, Xie L, Detre JA, Wolk DA, Arnold SE, 2017b. Effects of the insulin sensitizer metformin in Alzheimer’s disease: pilot data from a randomized placebo-controlled crossover study. Alzheimer Dis. Assoc. Disord 31, 107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Kowal J, Tkach M, Thery C, 2014. Biogenesis and secretion of exosomes. Curr. Opin. Cell Biol 29, 116–125. [DOI] [PubMed] [Google Scholar]
  117. Kritsilis M, V Rizou S, Koutsoudaki PN, Evangelou K, Gorgoulis VG, Papadopoulos D, 2018. Ageing, cellular senescence and neurodegenerative disease. Int. J. Mol. Sci 19, 2937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Kuilman T, Peeper DS, 2009. Senescence-messaging secretome: SMS-ing cellular stress. Nat. Rev. Cancer 9, 81–94. [DOI] [PubMed] [Google Scholar]
  119. Kumar A, Deep G, 2020a. Exosomes in hypoxia-induced remodeling of the tumor microenvironment. Cancer Lett 488, 1–8. [DOI] [PubMed] [Google Scholar]
  120. Kumar A, Deep G, 2020b. Hypoxia in tumor microenvironment regulates exosome biogenesis: molecular mechanisms and translational opportunities. Cancer Lett. 479 (Jun 1), 23–30. [DOI] [PubMed] [Google Scholar]
  121. Kumar P, Dezso Z, MacKenzie C, Oestreicher J, Agoulnik S, Byrne M, Bernier F, Yanagimachi M, Aoshima K, Oda Y, 2013. Circulating miRNA biomarkers for Alzheimer’s disease. PLoS One 8, e69807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Kuroda N, Hamada S, Sakata N, Jeon B, Iijima K, Yoshie S, Ishizaki T, Jin X, Watanabe T, Tamiya N, 2019. Antipsychotic use and related factors among people with dementia aged 75 years or older in Japan: a comprehensive population-based estimation using medical and long-term care data. Int. J. Geriatr. Psychiatry 34, 472–479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Laberge RM, Sun Y, Orjalo AV, Patil CK, Freund A, Zhou L, Curran SC, Davalos AR, Wilson-Edell KA, Liu S, Limbad C, Demaria M, Li P, Hubbard GB, Ikeno Y, Javors M, Desprez PY, Benz CC, Kapahi P, Nelson PS, Campisi J, 2015. MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation. Nat. Cell Biol 17, 1049–1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Lai KSP, Liu CS, Rau A, Lanctot KL, Kohler CA, Pakosh M, Carvalho AF, Herrmann N, 2017. Peripheral inflammatory markers in Alzheimer’s disease: a systematic review and meta-analysis of 175 studies. J. Neurol. Neurosurg. Psychiatry 88, 876–882. [DOI] [PubMed] [Google Scholar]
  125. Lee SH, Lee JH, Lee HY, Min KJ, 2019. Sirtuin signaling in cellular senescence and aging. BMB Rep. 52, 24–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Lehmann BD, Paine MS, Brooks AM, McCubrey JA, Renegar RH, Wang R, Terrian DM, 2008. Senescence-associated exosome release from human prostate cancer cells. Cancer Res 68, 7864–7871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Lima S, Gago M, Garrett C, Pereira MG, 2016. Medication adherence in Alzheimer’s disease: the mediator role of mindfulness. Arch. Gerontol. Geriatr 67, 92–97. [DOI] [PubMed] [Google Scholar]
  128. Lin A-L, Zheng W, Halloran JJ, Burbank RR, Hussong SA, Hart MJ, Javors M, Shih Y-YI, Muir E, Solano Fonseca R, Strong R, Richardson AG, Lechleiter JD, Fox PT, Galvan V, 2013. Chronic rapamycin restores brain vascular integrity and function through NO synthase activation and improves memory in symptomatic mice modeling Alzheimer’s disease. J. Cereb. Blood Flow Metab 33, 1412–1421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Liu B, Fan Z, Edgerton SM, Yang X, Lind SE, Thor AD, 2011. Potent anti-proliferative effects of metformin on trastuzumab-resistant breast cancer cells via inhibition of erbB2/IGF-1 receptor interactions. Cell Cycle 10, 2959–2966. [DOI] [PubMed] [Google Scholar]
  130. Longstreth WT Jr., Manolio TA, Arnold A, Burke GL, Bryan N, Jungreis CA, Enright PL, O’Leary D, Fried L, 1996. Clinical correlates of white matter findings on cranial magnetic resonance imaging of 3301 elderly people. The Cardiovascular Health Study. Stroke 27, 1274–1282. [DOI] [PubMed] [Google Scholar]
  131. Lopez-Rodriguez AB, Hennessy E, Murray CL, Nazmi A, Delaney HJ, Healy D, Fagan SG, Rooney M, Stewart E, Lewis A, de Barra N, Scarry P, Riggs-Miller L, Boche D, Cunningham MO, Cunningham C, 2021. Acute systemic inflammation exacerbates neuroinflammation in Alzheimer’s disease: IL-1beta drives amplified responses in primed astrocytes and neuronal network dysfunction. Alzheimer’s Dement. (Jun 3) 10.1002/alz.12341. Online ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Lorenzl S, Albers DS, Relkin N, Ngyuen T, Hilgenberg SL, Chirichigno J, Cudkowicz ME, Beal MF, 2003. Increased plasma levels of matrix metalloproteinase-9 in patients with Alzheimer’s disease. Neurochem. Int 43, 191–196. [DOI] [PubMed] [Google Scholar]
  133. Lorenzl S, Buerger K, Hampel H, Beal MF, 2008. Profiles of matrix metalloproteinases and their inhibitors in plasma of patients with dementia. Int. Psychogeriatr 20, 67–76. [DOI] [PubMed] [Google Scholar]
  134. Luchsinger JA, Perez T, Chang H, Mehta P, Steffener J, Pradabhan G, Ichise M, Manly J, Devanand DP, Bagiella E, 2016. Metformin in amnestic mild cognitive impairment: results of a pilot randomized placebo controlled clinical trial. J. Alzheimer’s Dis 51, 501–514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Luth HJ, Holzer M, Gertz HJ, Arendt T, 2000. Aberrant expression of nNOS in pyramidal neurons in Alzheimer’s disease is highly co-localized with p21ras and p16INK4a. Brain Res. 852, 45–55. [DOI] [PubMed] [Google Scholar]
  136. Mahajani S, Giacomini C, Marinaro F, De Pietri Tonelli D, Contestabile A, Gasparini L, 2017. Lamin B1 levels modulate differentiation into neurons during embryonic corticogenesis. Sci. Rep 7, 4897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Mandrioli J, D’Amico R, Zucchi E, Gessani A, Fini N, Fasano A, Caponnetto C, Chiò A, Dalla Bella E, Lunetta C, Mazzini L, Marinou K, Sorarù G, de Biasi S, Lo Tartaro D, Pinti M, Cossarizza A, 2018. Rapamycin treatment for amyotrophic lateral sclerosis: protocol for a phase II randomized, double-blind, placebo-controlled, multicenter, clinical trial (RAP-ALS trial). Medicine (Baltimore) 97, e11119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Martin-Montalvo A, Mercken EM, Mitchell SJ, Palacios HH, Mote PL, Scheibye-Knudsen M, Gomes AP, Ward TM, Minor RK, Blouin M-J, 2013. Metformin improves healthspan and lifespan in mice. Nat. Commun 4, 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Mattson MP, Arumugam TV, 2018. Hallmarks of brain aging: adaptive and pathological modification by metabolic states. Cell Metab. 27, 1176–1199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. McShea A, Harris PL, Webster KR, Wahl AF, Smith MA, 1997. Abnormal expression of the cell cycle regulators P16 and CDK4 in Alzheimer’s disease. Am. J. Pathol 150, 1933–1939. [PMC free article] [PubMed] [Google Scholar]
  141. Micuda S, Mundlova L, Anzenbacherova E, Anzenbacher P, Chladek J, Fuksa L, Martinkova J, 2004. Inhibitory effects of memantine on human cytochrome P450 activities: prediction of in vivo drug interactions. Eur. J. Clin. Pharmacol 60, 583–589. [DOI] [PubMed] [Google Scholar]
  142. Misawa T, Tanaka Y, Okada R, Takahashi A, 2020. Biology of extracellular vesicles secreted from senescent cells as senescence-associated secretory phenotype factors. Geriatr. Gerontol. Int 20, 539–546. [DOI] [PubMed] [Google Scholar]
  143. Moiseeva O, Descĥenes-Simard X, St-Germain E, Igelmann S, Huot G, Cadar AE, Bourdeau V, Pollak MN, Ferbeyre G, 2013. Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-κB activation. Aging Cell 12, 489–498. [DOI] [PubMed] [Google Scholar]
  144. Mongelli A, Atlante S, Barbi V, Bachetti T, Martelli F, Farsetti A, Gaetano C, 2020. Treating senescence like Cancer: novel perspectives in senotherapy of chronic diseases. Int. J. Mol. Sci 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Montagne A, Barnes SR, Sweeney MD, Halliday MR, Sagare AP, Zhao Z, Toga AW, Jacobs RE, Liu CY, Amezcua L, Harrington MG, Chui HC, Law M, Zlokovic BV, 2015. Blood-brain barrier breakdown in the aging human hippocampus. Neuron 85, 296–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Moreno-Blas D, Gorostieta-Salas E, Pommer-Alba A, Muciño-Hernández G, Gerónimo-Olvera C, Maciel-Barón LA, Konigsberg M, Massieu L, Castro-Obregón S, 2019. Cortical neurons develop a senescence-like phenotype promoted by dysfunctional autophagy. Aging 11, 6175–6198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Mouchiroud L, Houtkooper RH, Moullan N, Katsyuba E, Ryu D, Cantó C, Mottis A, Jo YS, Viswanathan M, Schoonjans K, Guarente L, Auwerx J, 2013. The NAD(+)/sirtuin pathway modulates longevity through activation of mitochondrial UPR and FOXO signaling. Cell 154, 430–441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Müller-Spahn F, 2003. Behavioral disturbances in dementia. Dialogues Clin. Neurosci 5, 49–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Munster BC, Aronica E, Zwinderman AH, Eikelenboom P, Cunningham C, Rooij SE, 2011. Neuroinflammation in delirium: a postmortem case-control study. Rejuvenation Res. 14, 615–622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Musi N, Valentine JM, Sickora KR, Baeuerle E, Thompson CS, Shen Q, Orr ME, 2018. Tau protein aggregation is associated with cellular senescence in the brain. Aging Cell 17, e12840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Mustapic M, Tran J, Craft S, Kapogiannis D, 2019. Extracellular vesicle biomarkers track cognitive changes following intranasal insulin in Alzheimer’s disease. J. Alzheimer’s Dis 69, 489–498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Ng TP, Feng L, Yap KB, Lee TS, Tan CH, Winblad B, 2014. Long-term metformin usage and cognitive function among older adults with diabetes. J. Alzheimer’s Dis 41, 61–68. [DOI] [PubMed] [Google Scholar]
  153. Nogueras-Ortiz CJ, Mahairaki V, Delgado-Peraza F, Das D, Avgerinos K, Eren E, Hentschel M, Goetzl EJ, Mattson MP, Kapogiannis D, 2020. Astrocyte- and neuron-derived extracellular vesicles from Alzheimer’s disease patients effect complement-mediated neurotoxicity. Cells 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Norman M, Ter-Ovanesyan D, Trieu W, Lazarovits R, Kowal EJK, Lee JH, Chen-Plotkin AS, Regev A, Church GM, Walt DR, 2021. L1CAM is not associated with extracellular vesicles in human cerebrospinal fluid or plasma. Nat. Methods 18, 631–634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. North BJ, Verdin E, 2004. Sirtuins: Sir2-related NAD-dependent protein deacetylases. Genome Biol. 5, 224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Norton EJ, Bridges LR, Kenyon LC, Esiri MM, Bennett DC, Hainsworth AH, 2019. Cell senescence and cerebral small vessel disease in the brains of people aged 80 years and older. J. Neuropathol. Exp. Neurol 78, 1066–1072. [DOI] [PubMed] [Google Scholar]
  157. Ogrodnik M, Miwa S, Tchkonia T, Tiniakos D, Wilson CL, Lahat A, Day CP, Burt A, Palmer A, Anstee QM, 2017. Cellular senescence drives age-dependent hepatic steatosis. Nat. Commun 8, 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Ogrodnik M, Zhu Y, Langhi LGP, Tchkonia T, Kruger P, Fielder E, Victorelli S, Ruswhandi RA, Giorgadze N, Pirtskhalava T, Podgorni O, Enikolopov G, Johnson KO, Xu M, Inman C, Palmer AK, Schafer M, Weigl M, Ikeno Y, Burns TC, Passos JF, von Zglinicki T, Kirkland JL, Jurk D, 2019. Obesity-induced cellular senescence drives anxiety and impairs neurogenesis. Cell Metab 29, 1061–1077 e1068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Ogrodnik M, Evans SA, Fielder E, Victorelli S, Kruger P, Salmonowicz H, Weigand BM, Patel AD, Pirtskhalava T, Inman CL, Johnson KO, Dickinson SL, Rocha A, Schafer MJ, Zhu Y, Allison DB, von Zglinicki T, LeBrasseur NK, Tchkonia T, Neretti N, Passos JF, Kirkland JL, Jurk D, 2021. Whole-body senescent cell clearance alleviates age-related brain inflammation and cognitive impairment in mice. Aging Cell 20, e13296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Orr ME, Sullivan AC, Frost B, 2017. A brief overview of tauopathy: causes, consequences, and therapeutic strategies. Trends Pharmacol. Sci 38, 637–648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Orr ME, Kotkowski E, Bair-Kelps D, Romo T, Espinoza S, Musi N, Powers B, Center SACDP, 2020. Results from a pilot study: the effects of nicotinamide riboside on mild cognitive impairment. Alzheimer’s Dement. e044746. [Google Scholar]
  162. Ozawa C, Roberts R, Yoshida K, Suzuki T, Lebowitz B, Reeves S, Howard R, Abe T, Mimura M, Uchida H, 2017. Placebo effects in the treatment of noncognitive symptoms of Alzheimer’s disease: analysis of the CATIE-AD data. J. Clin. Psychiatry 78, e1204–e1210. [DOI] [PubMed] [Google Scholar]
  163. Panigrahi GK, Deep G, 2017. Exosomes-based biomarker discovery for diagnosis and prognosis of prostate cancer. Front. Biosci (Landmark; Ed) 22, 1682–1696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Panigrahi GK, Praharaj PP, Peak TC, Long J, Singh R, Rhim JS, Elmageed ZYA, Deep G, 2018. Hypoxia-induced exosome secretion promotes survival of African-American and Caucasian prostate cancer cells. Sci. Rep 8, 3853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Panigrahi GK, Praharaj PP, Kittaka H, Mridha AR, Black OM, Singh R, Mercer R, van Bokhoven A, Torkko KC, Agarwal C, Agarwal R, Abd Elmageed ZY, Yadav H, Mishra SK, Deep G, 2019. Exosome proteomic analyses identify inflammatory phenotype and novel biomarkers in African American prostate cancer patients. Cancer Med 8, 1110–1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Pantoni L, 2010. Cerebral small vessel disease: from pathogenesis and clinical characteristics to therapeutic challenges. Lancet Neurol. 9, 689–701. [DOI] [PubMed] [Google Scholar]
  167. Panza F, Frisardi V, Capurso C, D’Introno A, Colacicco AM, Imbimbo BP, Santamato A, Vendemiale G, Seripa D, Pilotto A, Capurso A, Solfrizzi V, 2010. Late-life depression, mild cognitive impairment, and dementia: possible continuum? Am. J. Geriatr. Psychiatry 18, 98–116. [DOI] [PubMed] [Google Scholar]
  168. Park DS, Morris EJ, Padmanabhan J, Shelanski ML, Geller HM, Greene LA, 1998. Cyclin-dependent kinases participate in death of neurons evoked by DNA-damaging agents. J. Cell Biol 143, 457–467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Pasqualetti G, Tognini S, Calsolaro V, Polini A, Monzani F, 2015. Potential drug-drug interactions in Alzheimer patients with behavioral symptoms. Clin. Interv. Aging 10, 1457–1466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Paulison B, Léos CL, 2010. Potential cardiotoxic reaction involving rivastigmine and beta-blockers: a case report and review of the literature. Cardiovasc. Toxicol 10, 306–310. [DOI] [PubMed] [Google Scholar]
  171. Peak TC, Panigrahi GK, Praharaj PP, Su Y, Shi L, Chyr J, Rivera-Chavez J, Flores-Bocanegra L, Singh R, Vander Griend DJ, Oberlies NH, Kerr BA, Hemal A, Bitting RL, Deep G, 2019. Syntaxin 6-mediated exosome secretion regulates enzalutamide resistance in prostate cancer. Mol. Carcinog 59 (1), 62–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Poels MM, Vernooij MW, Ikram MA, Hofman A, Krestin GP, van der Lugt A, Breteler MM, 2010. Prevalence and risk factors of cerebral microbleeds: an update of the Rotterdam scan study. Stroke 41, S103–106. [DOI] [PubMed] [Google Scholar]
  173. Prins ND, Scheltens P, 2015. White matter hyperintensities, cognitive impairment and dementia: an update. Nat. Rev. Neurol 11, 157–165. [DOI] [PubMed] [Google Scholar]
  174. Rabinovici GD, Carrillo MC, Forman M, DeSanti S, Miller DS, Kozauer N, Petersen RC, Randolph C, Knopman DS, Smith EE, Isaac M, Mattsson N, Bain LJ, Hendrix JA, Sims JR, 2017. Multiple comorbid neuropathologies in the setting of Alzheimer’s disease neuropathology and implications for drug development. Alzheimers Dement. (N Y) 3, 83–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Rafii MS, Aisen PS, 2020. The search for Alzheimer disease therapeutics - same targets, better trials? Nat. Rev. Neurol 16, 597–598. [DOI] [PubMed] [Google Scholar]
  176. Raina AK, Zhu X, Shimohama S, Perry G, Smith MA, 2003. Tipping the apoptotic balance in Alzheimer’s disease: the abortosis concept. Cell Biochem. Biophys 39, 249–255. [DOI] [PubMed] [Google Scholar]
  177. Raina P, Santaguida P, Ismaila A, Patterson C, Cowan D, Levine M, Booker L, Oremus M, 2008. Effectiveness of cholinesterase inhibitors and memantine for treating dementia: evidence review for a clinical practice guideline. Ann. Intern. Med 148, 379–397. [DOI] [PubMed] [Google Scholar]
  178. Ramteke A, Ting H, Agarwal C, Mateen S, Somasagara R, Hussain A, Graner M, Frederick B, Agarwal R, Deep G, 2015. Exosomes secreted under hypoxia enhance invasiveness and stemness of prostate cancer cells by targeting adherens junction molecules. Mol. Carcinog 54, 554–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Rhinn M, Ritschka B, Keyes WM, 2019. Cellular senescence in development, regeneration and disease. Development 146. [DOI] [PubMed] [Google Scholar]
  180. Ritzel RM, Doran SJ, Glaser EP, Meadows VE, Faden AI, Stoica BA, Loane DJ, 2019. Old age increases microglial senescence, exacerbates secondary neuroinflammation, and worsens neurological outcomes after acute traumatic brain injury in mice. Neurobiol. Aging 77, 194–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Rogawski MA, Wenk GL, 2003. The neuropharmacological basis for the use of memantine in the treatment of Alzheimer’s disease. CNS Drug Rev. 9, 275–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Roos CM, Zhang B, Palmer AK, Ogrodnik MB, Pirtskhalava T, Thalji NM, Hagler M, Jurk D, Smith LA, Casaclang-Verzosa G, 2016. Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging Cell 15, 973–977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Roy AL, Sierra F, Howcroft K, Singer DS, Sharpless N, Hodes RJ, Wilder EL, Anderson JM, 2020. A blueprint for characterizing senescence. Cell 183, 1143–1146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Ryu SH, Jung HY, Lee KJ, Moon SW, Lee DW, Hong N, Kee BS, Kim DH, Han C, Lee CU, 2017. Incidence and course of depression in patients with Alzheimer’s disease. Psychiatry Investig. 14, 271–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Saez-Atienzar S, Masliah E, 2020. Cellular senescence and Alzheimer disease: the egg and the chicken scenario. Nat. Rev. Neurosci 21 (Aug (8)), 433–444. [DOI] [PubMed] [Google Scholar]
  186. Sah E, Krishnamurthy S, Gillispie GJ, Milligan C, Orr ME, 2021. The cellular senescence stress response in post-mitotic brain cells – cell survival at the expense of tissue degeneration. Life 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Saheera S, Potnuri AG, Krishnamurthy P, 2020. Nano-vesicle (Mis)communication in senescence-related pathologies. Cells 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Santiago JA, Potashkin JA, 2021. The impact of disease comorbidities in alzheimer’s disease. Front. Aging Neurosci 13, 631770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Saxton RA, Sabatini DM, 2017. mTOR signaling in growth, metabolism, and disease. Cell 168, 960–976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Schafer MJ, Zhang X, Kumar A, Atkinson EJ, Zhu Y, Jachim S, Mazula DL, Brown AK, Berning M, Aversa Z, Kotajarvi B, Bruce CJ, Greason KL, Suri RM, Tracy RP, Cummings SR, White TA, LeBrasseur NK, 2020. The senescence-associated secretome as an indicator of age and medical risk. JCI Insight 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. Schlaepfer IR, Nambiar DK, Ramteke A, Kumar R, Dhar D, Agarwal C, Bergman B, Graner M, Maroni P, Singh RP, Agarwal R, Deep G, 2015. Hypoxia induces triglycerides accumulation in prostate cancer cells and extracellular vesicles supporting growth and invasiveness following reoxygenation. Oncotarget 6, 22836–22856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Schneider LS, Sano M, 2009. Current Alzheimer’s disease clinical trials: methods and placebo outcomes. Alzheimer’s Dement. 5, 388–397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Schneider LS, Kennedy RE, Wang G, Cutter GR, 2015. Differences in Alzheimer disease clinical trial outcomes based on age of the participants. Neurology 84, 1121–1127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Schöndorf DC, Ivanyuk D, Baden P, Sanchez-Martinez A, De Cicco S, Yu C, Giunta I, Schwarz LK, Di Napoli G, Panagiotakopoulou V, 2018. The NAD+ precursor nicotinamide riboside rescues mitochondrial defects and neuronal loss in iPSC and fly models of Parkinson’s disease. Cell Rep. 23, 2976–2988. [DOI] [PubMed] [Google Scholar]
  195. Schwab N, Grenier K, Hazrati L-N, 2019. DNA repair deficiency and senescence in concussed professional athletes involved in contact sports. Acta Neuropathol. Commun 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Schwab N, Ju Y, Hazrati L-N, 2021. Early onset senescence and cognitive impairment in a murine model of repeated mTBI. Acta Neuropathol. Commun 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Selbach M, Schwanhäusser B, Thierfelder N, Fang Z, Khanin R, Rajewsky N, 2008. Widespread changes in protein synthesis induced by microRNAs. Nature 455, 58–63. [DOI] [PubMed] [Google Scholar]
  198. Sevigny J, Chiao P, Bussière T, Weinreb PH, Williams L, Maier M, Dunstan R, Salloway S, Chen T, Ling Y, O’Gorman J, Qian F, Arastu M, Li M, Chollate S, Brennan MS, Quintero-Monzon O, Scannevin RH, Arnold HM, Engber T, Rhodes K, Ferrero J, Hang Y, Mikulskis A, Grimm J, Hock C, Nitsch RM, Sandrock A, 2016. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature 537, 50–56. [DOI] [PubMed] [Google Scholar]
  199. Sheller S, Papaconstantinou J, Urrabaz-Garza R, Richardson L, Saade G, Salomon C, Menon R, 2016. Amnion-epithelial-cell-derived exosomes demonstrate physiologic state of cell under oxidative stress. PLoS One 11, e0157614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Soria FN, Pampliega O, Bourdenx M, Meissner WG, Bezard E, Dehay B, 2017. Exosomes, an unmasked culprit in neurodegenerative diseases. Front. Neurosci 11, 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. Stanfel MN, Shamieh LS, Kaeberlein M, Kennedy BK, 2009. The TOR pathway comes of age. Biochim. Biophys. Acta (BBA)-Gen. Subj. 1790, 1067–1074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Stojiljkovic MR, Ain Q, Bondeva T, Heller R, Schmeer C, Witte OW, 2019. Phenotypic and functional differences between senescent and aged murine microglia. Neurobiol. Aging 74, 56–69. [DOI] [PubMed] [Google Scholar]
  203. Streit WJ, Xue QS, 2014. Human CNS immune senescence and neurodegeneration. Curr. Opin. Immunol 29, 93–96. [DOI] [PubMed] [Google Scholar]
  204. Streit WJ, Braak H, Xue Q-S, Bechmann I, 2009. Dystrophic (senescent) rather than activated microglial cells are associated with tau pathology and likely precede neurodegeneration in Alzheimer’s disease. Acta Neuropathol. 118, 475–485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Suárez-Calvet M, Karikari TK, Ashton NJ, Lantero Rodríguez J, Milà-Alomà M, Gispert JD, Salvadó G, Minguillon C, Fauria K, Shekari M, Grau-Rivera O, Arenaza-Urquijo EM, Sala-Vila A, Sánchez-Benavides G, González-de-Echávarri JM, Kollmorgen G, Stoops E, Vanmechelen E, Zetterberg H, Blennow K, Molinuevo JL, 2020. Novel tau biomarkers phosphorylated at T181, T217 or T231 rise in the initial stages of the preclinical Alzheimer’s continuum when only subtle changes in Aβ pathology are detected. EMBO Mol. Med 12, e12921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Swardfager W, Lanctot K, Rothenburg L, Wong A, Cappell J, Herrmann N, 2010. A meta-analysis of cytokines in Alzheimer’s disease. Biol. Psychiatry 68, 930–941. [DOI] [PubMed] [Google Scholar]
  207. Taipa R, das Neves SP, Sousa AL, Fernandes J, Pinto C, Correia AP, Santos E, Pinto PS, Carneiro P, Costa P, Santos D, Alonso I, Palha J, Marques F, Cavaco S, Sousa N, 2019. Proinflammatory and anti-inflammatory cytokines in the CSF of patients with Alzheimer’s disease and their correlation with cognitive decline. Neurobiol. Aging 76, 125–132. [DOI] [PubMed] [Google Scholar]
  208. Takahashi A, Okada R, Nagao K, Kawamata Y, Hanyu A, Yoshimoto S, Takasugi M, Watanabe S, Kanemaki MT, Obuse C, Hara E, 2017. Exosomes maintain cellular homeostasis by excreting harmful DNA from cells. Nat. Commun 8, 15287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Takamori Y, Hirahara Y, Wakabayashi T, Mori T, Koike T, Kataoka Y, Tamura Y, Kurebayashi S, Kurokawa K, Yamada H, 2018. Differential expression of nuclear lamin subtypes in the neural cells of the adult rat cerebral cortex. IBRO Rep. 5, 99–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  210. Tan L, Yu JT, Liu QY, Tan MS, Zhang W, Hu N, Wang YL, Sun L, Jiang T, Tan L, 2014. Circulating miR-125b as a biomarker of Alzheimer’s disease. J. Neurol. Sci 336, 52–56. [DOI] [PubMed] [Google Scholar]
  211. Tang Z, Bereczki E, Zhang H, Wang S, Li C, Ji X, Branca RM, Lehtiö J, Guan Z, Filipcik P, Xu S, Winblad B, Pei J-J, 2013. Mammalian target of rapamycin (mTor) mediates tau protein dyshomeostasis: implication for Alzheimer disease. J. Biol. Chem 288, 15556–15570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Tariot PN, Farlow MR, Grossberg GT, Graham SM, McDonald S, Gergel I, 2004. Memantine treatment in patients with moderate to severe Alzheimer disease already receiving donepezil: a randomized controlled trial. JAMA 291, 317–324. [DOI] [PubMed] [Google Scholar]
  213. Tavassoli N, Sommet A, Lapeyre-Mestre M, Bagheri H, Montrastruc JL, 2007. Drug interactions with cholinesterase inhibitors: an analysis of the French pharmacovigilance database and a comparison of two national drug formularies (Vidal, British National Formulary). Drug Saf. 30, 1063–1071. [DOI] [PubMed] [Google Scholar]
  214. Tchkonia T, Kirkland JL, 2018. Aging, cell senescence, and chronic disease: emerging therapeutic strategies. JAMA 320, 1319–1320. [DOI] [PubMed] [Google Scholar]
  215. Toiber D, Sebastian C, Mostoslavsky R, 2011. Characterization of nuclear sirtuins: molecular mechanisms and physiological relevance. Handb. Exp. Pharmacol 206, 189–224. [DOI] [PubMed] [Google Scholar]
  216. Tominaga T, Shimada R, Okada Y, Kawamata T, Kibayashi K, 2019. Senescence-associated-β-galactosidase staining following traumatic brain injury in the mouse cerebrum. PLoS One 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Tsai CL, Ukropec J, Ukropcova B, Pai MC, 2018. An acute bout of aerobic or strength exercise specifically modifies circulating exerkine levels and neurocognitive functions in elderly individuals with mild cognitive impairment. Neuroimage Clin. 17, 272–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Turnquist C, Horikawa I, Foran E, Major EO, Vojtesek B, Lane DP, Lu X, Harris BT, Harris CC, 2016. p53 isoforms regulate astrocyte-mediated neuroprotection and neurodegeneration. Cell Death Differ. 23, 1515–1528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Turnquist C, Beck JA, Horikawa I, Obiorah IE, Von Muhlinen N, Vojtesek B, Lane DP, Grunseich C, Chahine JJ, Ames HM, Smart DD, Harris BT, Harris CC, 2019. Radiation-induced astrocyte senescence is rescued by Δ133p53. Neuro-Oncology 21, 474–485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  220. Urbanelli L, Buratta S, Sagini K, Tancini B, Emiliani C, 2016. Extracellular vesicles as new players in cellular senescence. Int. J. Mol. Sci 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  221. Van Skike CE, Jahrling JB, Olson AB, Sayre NL, Hussong SA, Ungvari Z, Lechleiter JD, Galvan V, 2018. Inhibition of mTOR protects the blood-brain barrier in models of Alzheimer’s disease and vascular cognitive impairment. Am. J. Physiol. Heart Circ. Physiol 314, H693–H703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  222. Vaur P, Brugg B, Mericskay M, Li Z, Schmidt MS, Vivien D, Orset C, Jacotot E, Brenner C, Duplus E, 2017. Nicotinamide riboside, a form of vitamin B3, protects against excitotoxicity-induced axonal degeneration. FASEB J. 31, 5440–5452. [DOI] [PubMed] [Google Scholar]
  223. Veitch DP, Weiner MW, Aisen PS, Beckett LA, Cairns NJ, Green RC, Harvey D, Jack CR Jr., Jagust W, Morris JC, Petersen RC, Saykin AJ, Shaw LM, Toga AW, Trojanowski JQ, 2019. Understanding disease progression and improving Alzheimer’s disease clinical trials: recent highlights from the Alzheimer’s Disease neuroimaging Initiative. Alzheimer’s Dement. 15, 106–152. [DOI] [PubMed] [Google Scholar]
  224. Vella LJ, Hill AF, Cheng L, 2016. Focus on extracellular vesicles: exosomes and their role in protein trafficking and biomarker potential in Alzheimer’s and Parkinson’s disease. Int. J. Mol. Sci 17, 173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  225. Ventimiglia LN, Alonso MA, 2016. Biogenesis and function of t cell-derived exosomes. Front. Cell Dev. Biol 4, 84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  226. Vermeer SE, Longstreth WT Jr., Koudstaal PJ, 2007. Silent brain infarcts: a systematic review. Lancet Neurol. 6, 611–619. [DOI] [PubMed] [Google Scholar]
  227. Villeda SA, Luo J, Mosher KI, Zou B, Britschgi M, Bieri G, Stan TM, Fainberg N, Ding Z, Eggel A, Lucin KM, Czirr E, Park JS, Couillard-Despres S, Aigner L, Li G, Peskind ER, Kaye JA, Quinn JF, Galasko DR, Xie XS, Rando TA, Wyss-Coray T, 2011. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature 477, 90–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  228. von Zglinicki T, Wan T, Miwa S, 2021. Senescence in post-mitotic cells: a driver of aging? Antioxid. Redox Signal 34, 308–323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  229. Weintraub S, Salmon D, Mercaldo N, Ferris S, Graff-Radford NR, Chui H, Cummings J, DeCarli C, Foster NL, Galasko D, Peskind E, Dietrich W, Beekly DL, Kukull WA, Morris JC, 2009. The Alzheimer’s Disease Centers’ Uniform Data Set (UDS): the neuropsychologic test battery. Alzheimer Dis. Assoc. Disord 23, 91–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. Wilkinson HN, Hardman MJ, 2020. Senescence in wound repair: emerging strategies to target chronic healing wounds. Front. Cell Dev. Biol 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  231. Winston CN, Goetzl EJ, Akers JC, Carter BS, Rockenstein EM, Galasko D, Masliah E, Rissman RA, 2016. Prediction of conversion from mild cognitive impairment to dementia with neuronally derived blood exosome protein profile. Alzheimers Dement. (Amst) 3, 63–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Xu H, Garcia-Ptacek S, Jönsson L, Wimo A, Nordström P, Eriksdotter M, 2021. Long-term effects of cholinesterase inhibitors on cognitive decline and mortality. Neurology 96, e2220–e2230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Yang Y, Geldmacher DS, Herrup K, 2001. DNA replication precedes neuronal cell death in Alzheimer’s disease. J. Neurosci 21, 2661–2668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  234. Yang Y, Mufson EJ, Herrup K, 2003. Neuronal cell death is preceded by cell cycle events at all stages of Alzheimer’s disease. J. Neurosci 23, 2557–2563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Young SG, Jung HJ, Lee JM, Fong LG, 2014. Nuclear lamins and neurobiology. Mol. Cell. Biol 34, 2776–2785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Yousefzadeh MJ, Wilkinson JE, Hughes B, Gadela N, Ladiges WC, Vo N, Niedernhofer LJ, Huffman DM, Robbins PD, 2020. Heterochronic parabiosis regulates the extent of cellular senescence in multiple tissues. Geroscience 42, 951–961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  237. Yousefzadeh MJ, Flores RR, Zhu Y, Schmiechen ZC, Brooks RW, Trussoni CE, Cui Y, Angelini L, Lee K-A, McGowan SJ, Burrack AL, Wang D, Dong Q, Lu A, Sano T, O’Kelly RD, McGuckian CA, Kato JI, Bank MP, Wade EA, Pillai SPS, Klug J, Ladiges WC, Burd CE, Lewis SE, LaRusso NF, Vo NV, Wang Y, Kelley EE, Huard J, Stromnes IM, Robbins PD, Niedernhofer LJ, 2021a. An aged immune system drives senescence and ageing of solid organs. Nature 594, 100–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  238. Yousefzadeh MJ, Flores RR, Zhu Y, Schmiechen ZC, Brooks RW, Trussoni CE, Cui Y, Angelini L, Lee KA, McGowan SJ, Burrack AL, Wang D, Dong Q, Lu A, Sano T, O’Kelly RD, McGuckian CA, Kato JI, Bank MP, Wade EA, Pillai SPS, Klug J, Ladiges WC, Burd CE, Lewis SE, LaRusso NF, Vo NV, Wang Y, Kelley EE, Huard J, Stromnes IM, Robbins PD, Niedernhofer LJ, 2021b. An aged immune system drives senescence and ageing of solid organs. Nature 594, 100–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Zeng XS, Geng WS, Jia JJ, Chen L, Zhang PP, 2018. Cellular and molecular basis of neurodegeneration in Parkinson disease. Front. Aging Neurosci 10, 109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  240. Zhang P, Kishimoto Y, Grammatikakis I, Gottimukkala K, Cutler RG, Zhang S, Abdelmohsen K, Bohr VA, Sen JM, Gorospe M, 2019. Senolytic therapy alleviates Aβ-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimer’s disease model. Nat. Neurosci 22, 719–728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Zhu YI, Tchkonia T, Pirtskhalava T, Gower AC, Ding H, Giorgadze N, Palmer AK, Ikeno Y, Hubbard GB, Lenburg M, 2015. The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell 14, 644–658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Zonneveld MI, Keulers TGH, Rouschop KMA, 2019. Extracellular vesicles as transmitters of hypoxia tolerance in solid cancers. Cancers (Basel) 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  243. Zou K, Abdullah M, Michikawa M, 2020. Current biomarkers for Alzheimer’s disease: from CSF to blood. J. Pers. Med 10. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES