Next Article in Journal
The Connectivity of the Resting Brain in Primary Open-Angle Glaucoma: A Systematic Review
Previous Article in Journal
Pulmonary and Renal Predictors of Mortality in ANCA-Associated Vasculitis: A Regional Experience from Türkiye
Previous Article in Special Issue
Connexins and Aging-Associated Respiratory Disorders: The Role in Intercellular Communications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Cellular Senescence in Parkinson’s Disease: Potential for Disease-Modification Through Senotherapy

by
David J. Rademacher
1,*,
Jacob E. Exline
2,3 and
Eileen M. Foecking
3,4,5,6
1
Department of Microbiology and Immunology and Core Microscopy Facility, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
2
Neuroscience Graduate Program, Loyola University Chicago, Maywood, IL 60153, USA
3
Department of Research and Developmental Services, Edward Hines Jr. VA Hospital, Hines, IL 60141, USA
4
The Burn and Shock Trauma Research Institute, Loyola University Chicago Medical Center, Maywood, IL 60153, USA
5
Department of Molecular Pharmacology and Neuroscience, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
6
Department of Otolaryngology, Head and Neck Surgery, Loyola University Chicago Medical Center, Maywood, IL 60153, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2025, 13(6), 1400; https://doi.org/10.3390/biomedicines13061400 (registering DOI)
Submission received: 1 April 2025 / Revised: 29 May 2025 / Accepted: 30 May 2025 / Published: 7 June 2025

Abstract

:
Parkinson’s disease (PD) is an aging-related neurodegenerative disease characterized by a progressive loss of dopamine (DA)-secreting neurons in the substantia nigra. Most of the currently available treatments attempt to alleviate the disease symptoms by increasing DA transmission in the brain and are associated with unpleasant side effects. Since there are no treatments that modify the course of PD or regenerate DA neurons, identifying therapeutic strategies that slow, stop, or reverse cell death in PD is of critical importance. Here, factors that confer vulnerability of substantia nigra DA neurons to cell death and the primary mechanisms of PD pathogenesis, including cellular senescence, a cellular stress response that elicits a stable cell cycle arrest in mitotic cells and profound phenotypic changes including the implementation of a pro-inflammatory secretome, are reviewed. Additionally, a discussion of the characteristics, mechanisms, and markers of cellular senescence and the development of approaches to target senescent cells, referred to as senotherapeutics, is included. Although the senotherapeutics curcumin, fisetin, GSK-650394, and astragaloside IV had disease-modifying effects in in vitro and in vivo models of PD, the potential long-term side effects of these compounds remain unclear. It remains to be elucidated whether their beneficial effects will translate to non-human primate models and/or human PD patients. The enhanced selectivity, safety, and/or selectivity of next generation senotherapeutic strategies including senolytic peptides, senoreverters, proteolysis-targeting chimeras, pro-drugs, immunotherapy, and nanoparticles will also be reviewed. Although these next generation senotherapeutics may have advantages, none have been tried in models of PD.

1. Introduction

Parkinson’s disease (PD) is the most common neurodegenerative movement disorder and affects 1–2% of individuals over the age of 65 making it the second most common neurodegenerative disease in the world after Alzheimer’s disease [1]. The worldwide prevalence of PD is over seven million with one million cases existing in the United States alone [2]. The burden of PD on society will rise significantly as the population ages. PD is characterized by its pathological changes including the presence of Lewy bodies (LBs) (Figure 1), eosinophilic inclusions in the cytoplasm of cell bodies, and thread-like Lewy neurites (LNs) in the cellular processes of dopamine (DA)-secreting neurons in the substantia nigra (SN) [3] leading to a progressive loss of these neurons. PD patients also have a significant decrease in DA in the striatum [3]. Classical parkinsonian motor symptoms (i.e., bradykinesia, tremor, and rigidity) and a variety of non-motor symptoms (e.g., depression, constipation, pain, gastrointestinal dysfunction, Parkinson disease psychosis, and sleep problems) that worsen over time are part of the progressive PD symptomology [4,5].
Pharmacological and nonpharmacological treatment options are available for PD [6]. Nonpharmacological options include deep brain stimulation, transplantation of pluripotent stem cells or DA neurons into the brain, gene therapy, exercise, improved nutrition, and palliative care [6]. Pharmacological options include L-3,4-hydroxyphenyl-ethylamine (L-dopa), monoamine oxidase inhibitors, catechol-O-methyltransferase inhibitors, anti-cholinergic drugs, and anti-diabetic drugs. Most of the currently available treatments target the symptoms of the disease and act by increasing dopaminergic (DAergic) transmission. Due to its remarkable effectiveness in reducing PD motor symptoms like tremor, bradykinesia, and rigidity, the DA precursor L-dopa is the gold standard for treating PD [7]. L-dopa is more efficacious and cost effective than other anti-parkinsonian medications, such as DA agonists [8,9]. Life expectancy is less for PD patients than for the general population, and L-dopa is the only anti-parkinsonian medication that increases the life expectancy of PD patients [10,11]. As the disease progresses, patients experience a decreased response to L-dopa or a wearing-off, necessitating higher doses of L-dopa over time to manage the PD motor symptoms [12]. Within 5 years of starting L-dopa treatment, 50% of PD patients experience L-dopa-related complications, fluctuating efficacy, and L-dopa-induced dyskinesia (LID) [13], and 80% of patients experience these side effects after 10 years [14]. LIDs are typically managed by reducing the dose of L-dopa or DA agonist. New approaches to manage LIDs are promising, such as the use of compounds that have a non-DAergic target [15]. The most prevalent kind of kinesia is peak dose dyskinesia [16]. Early morning off, also known as early morning akinesia, occurs in the morning before the first dose of L-dopa [17]. Biphasic dyskinesia can happen 10 to 15 min after L-dopa is administered and is caused by fluctuating plasma concentrations of the drug. LIDs are more likely to happen if SNpc DA neuron degeneration is severe and early [18]. Because they stimulate peripheral DA receptors, PD medications that increase DAergic transmission—like L-dopa and DA agonists—are also linked to a higher risk of side effects. Nausea, vomiting, and hypotension are among the side effects. Hallucinations and psychosis are among the more likely adverse reactions brought on by the stimulation of DA receptors in the central nervous system [19,20]. Of patients with PD, 15–20% have orthostatic hypotension [21], which gets worse when drugs that increase DAergic transmission are taken. L-dopa generally has a poor effect on non-motor symptoms, and anti-parkinsonian drugs may make them worse. The response of the typical motor issues of PD, such as dysarthria, postural instability, gait dysfunction, falls, and freezing, to drugs that enhance DA transmission is limited [22,23,24]. The quality of life for PD patients tends to gradually deteriorate as the disease relentlessly advances, even in cases that had an initial positive response to L-dopa or other DAergic medications. Therefore, a treatment approach that slows, halts, or reverses cell death in PD is critically needed. Indeed, the failure of all clinical studies of drugs to alter the progression of PD or regenerate DA neurons is one of the main concerns in PD research [25].

2. Relationship Between PD and Aging

The most important risk factor for PD is aging [26,27,28]. A meta-analysis of worldwide data revealed that PD prevalence increases directly with age. The prevalence of PD was found to be 41 per 100,000, 107 per 100,000, 428 per 100,000, 1087 per 100,000, and 1903 per 100,000 in those aged 40–49 years, 50–59 years, 60–69 years, 70–79 years, and 80 years of age or older, respectively [29]. A progressive loss of motor abilities and neural degeneration in the brain, akin to that seen in PD, are hallmarks of aging. However, aging does not result in all of the clinical signs of PD. In a study of 2500 aged individuals, 18.6% exhibited global parkinsonism [30]. A post-mortem examination of 744 of these aged individuals, who did not have PD, revealed the following: (a) ~one-third had mild or more severe loss of SN neurons; (b) ~17% had LBs; and (c) 10% had both neuron loss in the SN and LBs [30]. Notably, many of the cellular processes involved in aging, such as inflammation [31,32], mitochondrial dysfunction [33,34], oxidative stress [35,36], loss of protein homeostasis [3,37,38], a decrease in ubiquitin-proteasome system (UPS) function [39,40,41,42], dysregulation of autophagy [43,44,45], and cellular senescence [46,47], are also involved in PD pathogenesis.

3. Vulnerability of SN DA Neurons to Cell Death

The gradual loss of DA neurons in the substantia nigra pars compacta (SNpc), which innervate the basal ganglia, cause the classical parkinsonian motor symptoms [48]. Once about half of the SNpc DA neurons are gone, the motor symptoms start to show [49]. Since DA neurons in the ventral tegmental area (VTA) show significantly less degeneration, the neuronal loss is specific to the SNpc [50]. The axon of SNpc DA neurons is long, highly branched, and contains a large number of neurotransmitter release sites [51], which allows individual SNpc DA neurons to broadcast a DA signal over a wide area. Consequently, a significant number of neurons in the striatum are strongly influenced by SNpc DA neurons. Axonal arbors of DA neurons in the striatum are also an order of magnitude larger than those of other DA neuron classes, such as those found in the VTA and other brain regions [52]. Large axonal arbors and a high degree of arborization are features of the extensive axonal network of these neurons. As a result, more energy is required for synaptic transmission, membrane potential maintenance, and action potential propagation (Figure 1) [53,54]. Adenosine triphosphate (ATP)-dependent calcium (Ca2+) influx through L-type Cav1.3 Ca2+ channels, which is required to sustain the autonomous pacemaker firing activity of SNpc DA neurons, also comes with a high energy cost [55]. Interestingly, Ca2+ buffering in these neurons is poor (Figure 1) [56], rendering these neurons vulnerable to Ca2+ toxicity. The SNpc DA neurons have a limited ability to enhance oxidative phosphorylation in response to elevated energy demands [57]. These characteristics make the SNpc DA neurons vulnerable to even mild energy stress. Lastly, the SNpc lacks oxidative stress-protective mechanisms and is highly susceptible to oxidative stress (Figure 1) [58]. There is an inherent propensity for DA and its metabolites to generate reactive oxygen species (ROS), which ultimately kills neurons [59]. Increased levels of iron and copper, essential cofactors for biosynthetic enzymes involved in catecholamine synthesis, are found in the SNpc. The oxidation-reduction cycle of iron can generate free radicals and toxic metabolites, such as hydrogen peroxide [58], that, ultimately, contribute to the progressive degeneration of the DA neurons. SNpc DA neurons have a relative lack of neuroprotective factors such as the DA vesicle transport protein, vesicular monoamine transporter (VMAT) [60], and several trophic and growth factors (Figure 1) [61,62,63,64]. SNpc DA neurons also exhibit increased expression of vulnerability-related factors, such as D2 DA autoreceptors [65], G-protein inward rectifying potassium-2 channels [66], lactotransferrin [67], and the dopamine transporter (DAT) [68]. Thus, the combination of reduced neuroprotective factors with increased vulnerability-related factors makes SNpc DA neurons more vulnerable to death than other neurons.

4. Pathogenesis of PD

Although the exact cause of PD is still unknown, ~15% of patients worldwide report having a family history of the disease. They are categorized as familial PD patients, while the remaining 85% are considered sporadic PD patients [69]. In both familial and sporadic PD, a number of genetic risk factors have been found to interact with aging and environmental factors. Based on the extensive study of familial and sporadic PD, the emerging picture of PD pathogenesis includes oxidative stress, diminished anti-oxidant defenses, mitochondrial dysfunction, protein misfolding, autophagic-lysosomal pathway (ALP) dysfunction, impaired UPS function, impaired Ca2+ handling, inflammation, and cellular senescence [70].

4.1. Oxidative Stress

Oxidative stress mechanisms have been repeatedly demonstrated to play a role in the degeneration of SNpc DA neurons in PD [71]. When the production of ROS exceeds the ability of endogenous anti-oxidant enzymes and molecular chaperones to remove them, oxidative stress is the result. ROS buildup can cause oxidative damage to proteins, lipids, DNA, and RNA, which can impair neuronal structure and function [72]. It can also directly or indirectly trigger a number of cell death pathways, such as cytoplasmic cell death, autophagic cell death, and apoptosis [73,74]. Post-mortem brain tissue from patients with familial and sporadic PD has been found to contain oxidized proteins, lipids, and DNA [75,76]. In patients with early-stage PD, increased oxidative stress was observed prior to significant neuron loss [77], suggesting a causal link between the production of ROS and SNpc DA neuron death. Several environmental toxins and pesticides demonstrate preferential cytotoxicity to SNpc DA neurons [78]. These toxins freely pass through lipid membranes and build up in mitochondria after being consumed or inhaled [79]. Accumulation of mitochondrial toxins increases the production of superoxide and hinders the redox activity of mitochondrial complex I. Almost all genetic mutations associated with PD are linked to increased ROS production and reduced mitochondrial complex I activity.
Before striatal DA loss occurs, the accumulation of wild-type or mutant α-synuclein (α-syn) [80,81] in the mitochondria of DA neurons decreases mitochondrial complex I activity and increases ROS production [82]. Therefore, α-syn is thought to contribute to the death of DA neurons in human PD. Oxidative metabolism of DA by SNpc DA neurons and the surrounding glia also produce elevated levels of ROS [83]. Additionally, interactions with labile iron and other metals may cause DA to auto-oxidize, producing ROS [84]. There is a preferential build-up of labile iron in the aging SNpc, which enhances pro-oxidant interactions between iron and DA [84]. Post-mortem SNpc iron levels of PD patients are twice as high as those of age-matched controls [85,86]. DAT and VMAT play key roles in the defense mechanisms against iron-DA-induced ROS production. After being extracted from the synapse by DAT and VMAT, free DA is packaged into synaptic vesicles, where it is shielded from oxidation [87]. Due to DAT and VMAT dysregulation, DA neurons in the PD SNpc produce more DA while simultaneously reducing synaptic DA clearance and repackaging into vesicles [88,89]. VMAT and DAT changes are linked to abnormal α-syn post-translational modification or mutation, which can hinder VMAT-mediated DA packaging into synaptic vesicles [90] and disrupt cell surface regulation of DAT expression [91]. These modifications work together to increase free cytoplasmic DA, which in turn enhances the production of ROS and other neurotoxic redox species.

4.2. Diminished Anti-Oxidant Defenses

Aging-related decreases in anti-oxidant levels occur in the healthy SNpc, but severe and pervasive anti-oxidant deficiencies occur in the PD SNpc, further exacerbating the production of ROS linked to the disease (Figure 1). In the SNpc of PD patients, a significant reduction in glutathione protein and activity has been observed [92,93] suggesting a diminished ability to clear hydrogen peroxide. Notably, the degree of glutathione loss in the SNpc is correlated with the severity of the disease [92]. Dysfunction of superoxide dismutase 1 may reduce superoxide clearance in the PD SNpc [94]. Deficits in glutathione [95] and superoxide dismutase 1 [96] also occur in incidental Lewy body disease, a condition believed to be preclinical PD [97]. Thus, decreases in glutathione and superoxide dismutase 1 likely take place before neuronal loss in early-stage PD and could be a causal factor of the disease.
Astrocytes and microglia express larger quantities and higher levels of anti-oxidant genes compared to neurons [98]. Therefore, the anti-oxidant deficiencies observed in the SNpc are likely to reflect glial rather than neuronal reductions in anti-oxidant production. Two well-established characteristics of the PD SNpc, substantial microglial activation [99] and reactive gliosis [100], are linked to PD-linked mutations in both sporadic and familial PD. Hyperactivation and reactive gliosis of both microglia and astrocytes reduce anti-oxidant production in SNpc DA neurons [101].

4.3. Mitochondrial Dysfunction

Mitochondrial dysfunction is a major factor in the degeneration of SNpc DA neurons in PD. The toxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), which was discovered in 1976 by a chemistry student who attempted to synthesize synthetic heroin, but instead synthesized MPTP, eliminates SNpc DA neurons. Consumption of MPTP produces severe PD-like symptoms when administered to humans, non-human primates, and mice [102]. MPTP is converted to its toxic metabolite, the 1-methyl-4-phenylpyridinium ion (MPP+) in astrocytes, and is taken up by DA neurons via the DAT [102]. MPP+ is an inhibitor of complex I of the electron transport chain in mitochondria, and, as a result, a rapid decrease in ATP occurs in the striatum and SNpc [102]. Although the primary mechanism of MPTP-induced neurotoxicity is an ATP crisis, other mechanisms of MPTP-induced cell death include oxidative stress, upregulation of components of the mitochondrial apoptotic cascade, excitotoxicity, inhibition of the UPS, and neuroinflammation [102].
Blood levels of mitochondrial DNA copies and mitochondrial biogenesis gene expression are lower in PD patients [47]. The SNpc of PD patients contains mitochondrial DNA deletions, mutations, and rearrangements [103]. These mutations disrupt the mitochondria respiratory chain, resulting in a complex I deficiency [104]. As mitochondrial DNA mutations accumulate, SNpc DA neurons experience increased oxidative stress and changes in mitochondrial respiration. Additionally, a 30–40% decrease in activity and immunoreactivity for mitochondrial complex I has been observed in the SNpc of PD patients [105,106]. This specific mitochondrial complex I deficit does not affect other components of the mitochondria respiratory chain and has not been observed in other brain areas or other neurodegenerative diseases [106]. As mitochondria have been implicated in apoptotic cell death [107], it is possible that the specific mitochondrial complex I deficit contributes to PD by making neurons more prone to apoptosis.
Mutations in the α-syn gene, SNCA [108,109,110,111], increase mitochondrial fragmentation and cell death [112,113]. In PD models, α-syn accumulation leads to mitochondria dysfunction. In particular, deficiencies in the motility, function, and dynamics of mitochondria result from the overexpression of an A53T mutation [114]. In PD patients, α-syn accumulates in mitochondria in the striatum, impairs mitochondrial complex I function, and increases oxidative stress [115]. In PD patients, mitochondrial dysfunction in SNpc DA neurons promotes α-syn aggregation [116]. Mutations in the parkin gene, PRKN, result in a loss of parkin activity, protein accumulation, mitochondrial accumulation, low mitochondrial respiration, and the death of DA neurons [117]. Parkin is recruited to dysfunctional mitochondria and promotes autophagic degradation, a process impaired by pathogenic mutations in PRKN and PTEN-induced putative kinase (PINK1), another PD-associated gene [118]. Increased oxidative stress, neurodegeneration, and impaired ATP synthesis are all caused by PINK1 mutations [117,119]. The scavenging of ROS in mitochondria is disrupted by mutations in deglycase 1 (DJ-1), which makes SNpc DA neurons more susceptible to oxidative stress [120]. Mutations in LRRK2 affect mitochondrial fission and increase the susceptibility to mitochondrial toxins [121]. Lastly, GBA1 mutations cause membrane potential changes, diminished complex I function, and mitochondrial fragmentation [122]. Readers interested in a more comprehensive review of the multi-faceted role of mitochondria dysfunction in the pathogenesis of PD should read published reviews on the topic [123,124,125].

4.4. Protein Misfolding

The discovery of α-syn aggregates in LBs and LNs, as well as the discovery that SNCA mutations accelerated the pathological aggregation of α-syn and caused familial PD, strongly suggest that α-syn plays a role in the pathogenesis of PD [126]. In healthy neurons, α-syn is in a state of equilibrium between being soluble and membrane-bound [127]. A monomeric form of α-syn is found in axon terminals. It is thought to regulate neurotransmitter release by regulating the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex [128,129]. Post-translationally modified or misfolded α-syn monomers aggregate into intermediate oligomeric structures that subsequently assemble into insoluble fibrillar aggregates [130], forming LBs and LNs, the main pathological hallmarks of PD [131]. These proteinaceous aggregates cause neuronal dysfunction and death by interfering with synaptic transmission, mitochondrial activity, and proteasomal degradation. According to the prion hypothesis of PD, like prion proteins, α-syn pathology spreads in the PD brain through the transmission of misfolded α-syn from diseased neurons, astrocytes, and glia to healthy neurons, astrocytes, and glia. The transmitted α-syn acts as a seed or template to induce the conversion of soluble, monomeric α-syn to insoluble, fibrillar aggregates in the recipient cell (Figure 1). The α-syn aggregates activate microglia and astrocytes. The neurodegenerative processes in PD are facilitated by the release of ROS, pro-inflammatory cytokines, and pro-inflammatory chemokines by activated astrocytes and microglia [126]. The finding of more than 200 different oligomeric proteins [132] in LBs suggests that the pathological misfolding and aggregations of proteins other than α-syn play a role in the pathogenesis of PD.

4.5. Dysfunction of the ALP

The turnover of protein aggregates through lysosomal degradation is attributed to the ALP, one of the main degradative pathways. In macroautophagy, phagophore formation enables the engulfment of cytosolic proteins and organelles. A vesicular compartment called an autophagosome is created when the phagophore with the engulfed organelles and cytosolic proteins closes. When autophagosomes fuse with lysosomes, the engulfed contents are exposed to the lysosome’s hydrolytic enzymes, causing degradation [133,134].
α-Syn is degraded by autophagic mechanisms [135]. Several PD risk genes have been identified in patient genome-wide association studies that are critical for autophagy and lysosomal function [136], and more than 40 genes associated with PD are also involved with the ALP [45]. Vacuolar protein sorting-associated protein 35, which is encoded by VPS35, participates in endosomal-lysosomal trafficking linked to autophagy. A rare form of autosomal-dominant PD has been linked to mutations in VPS35 [45]. The most common genetic cause of PD is a result of mutations in LRRK2. The most common pathogenic mutation of LRRK2 causes a substantial build-up of intracellular α-syn by increasing LRRK2 activity and compromising autophagy [137]. Glucosylcerimidase beta 1 (GBA1) encodes for β-glucocerebrosidase (GCase), a lysosomal acidic hydrolase involved in glycosphingolipid catabolism. Heterozygous GBA1 mutation carriers have a 5- to 6-fold increased risk of developing PD [138]. Reduced GCase activity has been observed in the SNpc of PD patients [139] and has been reported to be more evident at the earlier PD stages [140]. Lastly, secretory autophagy is an unconventional secretory mechanism that is promoted by proteins typically linked to ALP degradation [141]. Interestingly, proteins associated with ALP degradation have been observed in LBs [142], and an autophagic secretion pathway mediates the secretion of α-syn [143].

4.6. Impaired UPS Function

The main function of the UPS is to eliminate mutant, damaged, and misfolded intracellular proteins [144,145]. For degradation to occur, unwanted proteins are conjugated to a chain of ubiquitin molecules, which acts as a degradation signal to the proteasome [144]. Polyubiquitin-protein conjugates and nonubiquitinated proteins are degraded by 26S and 20S proteasomes [145]. If the UPS does not appropriately clear unwanted proteins, they accumulate and aggregate to form insoluble inclusion bodies, which can disrupt cellular homeostasis and cause apoptotic cell death [146].
A role for the UPS in the pathogenesis of Parkinson’s disease is suggested by multiple lines of evidence. Proteasome subunits and ubiquitin co-localize in LBs [147]. Aggregated α-syn directly binds to and inhibits the proteasome [148]. Deletions, truncations, and point mutations in PRKN, the gene that codes for parkin, cause autosomal recessive juvenile parkinsonism [149], characterized by cell death preferentially in the SNpc [150]. In patients with this disorder, parkin, an E3 ubiquitin protein ligase, is either absent or has low levels in the brain [151,152]. Mutations in PRKN lead to an accumulation of parkin-ubiquitinated proteins due to the failure of the proteasome to degrade and clear substrate proteins. In addition, a missense mutation in ubiquitin C-terminal hydrolase L1 (UCH-L1) has also been found in a family with PD [153]. UCH-L1 belongs to a family of deubiquitinating enzymes, is only expressed in neurons, and has been observed in LBs [154,155]. Reduced enzyme activity, decreased availability of free monomeric ubiquitin required for labeling undesirable proteins, and impaired protein clearance are the results of UCH-L1 mutations. Structural and functional deficits in the 26/20S proteasomal complex have been observed solely within SNpc DA neurons of PD patients [41], and the three 20S proteasomal enzymatic activities have been found to be inhibited by ~40% [41,156].

4.7. Impaired Ca2+ Handling

DA neurons in the SNpc are self-contained pacemakers because they can produce action potentials slowly without synaptic input [157] by adjusting intracellular Ca2+ concentration through the release of Ca2+ from the endoplasmic reticulum, the opening of plasma membrane Ca2+ channels, and the lack of robust intracellular Ca2+ buffering [158]. Consequently, Ca2+ concentrations within mitochondria rise, stimulating oxidative phosphorylation and ATP production while increasing ROS [159]. Ca2+ promotes α-syn aggregation [160], which is boosted by oxidative stress [161] or DA [162]. Furthermore, α-syn-mediated toxicity is promoted by calcineurin, a Ca2+-dependent protein phosphatase [163]. Increased cytosolic Ca2+ hinders misfolded protein turnover and lysosomal motility [164]. Notably, there is an inverse relationship between the expression of the Ca2+ binding protein, calbindin, and the risk of neurodegeneration in PD patients [165], underscoring the importance of Ca2+ buffering to the survival of SNpc DA neurons. Hypertension treatment with dihydropyridine inhibitors, which are negative allostatic modulators of Cav1 Ca2+ channels [166], has been associated with an ~30% lower chance of receiving a PD diagnosis [167].

4.8. Inflammation

Inflammatory processes in the periphery and the central nervous system play a role in PD pathogenesis. A systematic review and meta-analysis demonstrated significantly increased levels of interleukin-6 (IL-6), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), monocyte chemoattractant protein-1 (MCP-1), and C-reactive protein (CRP) in both the blood and cerebral spinal fluid (CSF), as well as increased levels of interleukin-4 (IL-4), interferon-γ (IFN-γ), soluble tumor necrosis factor receptor 1 (STNFR1), and fractalkine only in blood samples of PD patients [168]. According to this clinical evidence, PD may be linked to a specific peripheral inflammatory response in addition to central inflammation. Toll-like receptor (TLR) overexpression can cause innate inflammatory processes to become overactive and contribute to the loss of neurons in PD. Circulating monocytes and the SNpc of PD patients have been found to have elevated levels of toll-like receptor 4 (TLR4) [169,170]. Immunoreactivity for TLR4 and a pathogenic form of α-syn was observed in LBs in SNpc DA neurons of PD patients [170]. PD is characterized by a shift toward inflammatory monocytes [171] and a dysfunction in the T lymphocyte compartment. Fewer regulatory T cells are present to suppress effector T cells, leading to an increase in effector T cell subsets with neurotoxic potential and fewer regulatory T cells [172,173]. Effector T lymphocytes isolated from patients with PD increased pro-inflammatory cytokine production when exposed to T cell activators [174]. Additionally, a number of studies show an association between PD susceptibility and polymorphisms in the human leukocyte antigen region or enrichment for genes of the adaptive immune system, including cytokine-mediated pathways and the regulation of leukocyte/lymphocyte activity [175,176].
Glial cells might play a significant role in the pathogenesis of neuronal death in PD. In the areas of the SNpc that have undergone the most neurodegeneration, activated microglia have been found [177]. Aggregates of pathogenic α-syn in microglial cell bodies and processes co-localized with TLR4 have been observed in PD patients [170]. Glial cell activation also increases the production and release of cytokines, ROS, and nitric oxide (NO), all of which can cause neurodegeneration [178]. The cytokines IL-1β, INF-γ, and TNF-α are increased in the SNpc of PD patients by 7.5- to 15-fold [179,180,181]. TNF-α receptors have been found in DA neurons [181]. Activation of TNF-α receptors leads to nuclear translocation of nuclear factor kappa-B (NFκ-B) and apoptotic cell death [182]. NFκ-B nuclear translocation has been found to increase 70-fold in the SNpc of PD patients [182].
Inflammatory processes in PD perpetuate disease progression through their role in the development and progression of blood-brain barrier (BBB) alterations. The inner portion of the BBB is made up of endothelial cells joined by tight junction proteins. Endothelial cells contract as a result of chronic inflammation, creating spaces between them that allow blood-borne molecules and cells to enter the brain and intensify inflammation (Figure 1) [183]. Activation of endothelial cells by pro-inflammatory mediators stimulates the expression of cell adhesion molecules, such as intercellular adhesion molecule-1 (ICAM-1), promoting the invasion of peripheral immune cells [184]. Accordingly, CD8+ and CD4+ T cells were observed in the SNpc of patients with PD [185]. Overexpression of ICAM-1 has been observed in the SNpc of PD patients and MPTP-treated non-human primates [180,186]. TNF-α and other pro-inflammatory cytokines, including IL-6 and IL-1β, can degrade tight junction proteins and produce a reactive microglial phenotype [187]. Chronic inflammation causes reactive microglia to phagocytose astrocytic end-feet, which compromises the function of the BBB [188].

4.9. Cellular Senescence

A complex stress response known as cellular senescence causes the cell cycle to stabilize at the G1/S-Phase or G2/M-Phase checkpoints and triggers profound phenotypic changes, including chromatin remodeling, metabolic reprogramming, and the implementation of a bioactive secretome, referred to as the senescence-associated secretory phenotype (SASP) [189]. The SASP is comprised of hundreds of factors, including pro-inflammatory cytokines, chemokines, growth factors, and proteases [190,191], with the specific combination of secreted factors thought to depend on the cell type and senescence inducer. In wound healing, the SASP mediates the activation and recruitment of the adaptive and innate immune system to eliminate cellular debris and senescent cells [192,193]. Notably, autocrine transmission of the SASP can reinforce the SASP phenotype, and paracrine transmission of the SASP can induce senescence in neighboring cells (Figure 1) [194]. Senescence is a powerful anti-tumorigenic response to oncogenic or mitogenic stimuli [195,196,197,198,199,200,201], though specific SASP factors have been shown to promote tumorigenesis [202,203]. Tissue remodeling during embryonic development and wound healing is promoted by cellular senescence and generally occurs in the absence of DNA damage [204,205]. Numerous disease states and aging have also been linked to cellular senescence [206]. The various mechanisms by which different stressors induce cellular senescence vary in a cell type- and tissue-dependent manner, making the characterization of senescent cells and the identification of a single, highly specific senescence-associated marker quite difficult.
A great deal of data support a role for cellular senescence in the pathogenesis of PD. The expression of cell cycle genes is upregulated in PD. p16INK4A mRNA was elevated in PD brain samples [207]. Increased levels of senescence-associated β-galactosidase (SA-β-gal) have been observed in the CSF [208] and brain tissue [207] from PD patients. In addition, SASP-related factors are upregulated in PD. The SASP factors, IL-1β and IL-6, are elevated in the CSF [209] and the striatum of PD patients [180]. Overexpression of IL-6 causes neurodegeneration in vivo [210]. IL-6 levels are positively correlated with the severity of motor symptoms in PD patients [211]. Elevated TNFα levels have been observed in the striatum [212] and the CSF from PD patients [209]. The SASP factor matrix metalloproteinase-3 was co-localized with α-syn in LBs in PD patients [213]. Interestingly, Bae and colleagues observed that the cell-to-cell transfer of α-syn was enhanced in senescent neurons [214].
Senescence in neurons, astrocytes, and microglia contribute to PD [215]. SNpc DA neuron degeneration was alleviated upon senescence cell elimination [207]. Compared to healthy individuals, higher numbers of senescent astrocytes have been observed in the SN of PD patients, and the PD-associated herbicide, paraquat, induces senescence in human astrocytes [207]. Senescent astrocytes exhibit increased secretion of SASP factors and dysfunctional mitochondria leading to elevated levels of ROS [216,217], which can perpetuate cellular senescence and damage neighboring cells. A change in the level of matrix metalloproteinases is a feature of the SASP [190]. MMP3 was upregulated in senescent astrocytes [218]. Matrix metalloproteinase-3 activates microglial cells [219] and causes leakage of the BBB [220], which has been observed in the PD brain post-mortem [221]. Microglial activation occurs in the SN [222], where they are more than 4 times more abundant than other brain regions [223]. Microglial activation results in the loss of SNpc DA neurons [224]. The senescent changes in microglia cause inflammaging, a chronic, low-grade inflammation that contributes to the progression of PD [225]. SASP mediators from senescent cells are thought to underlie inflammaging [226]. Importantly, studies indicate that the senescence of SN DA neurons precedes their loss [227,228], and it has been proposed that the SASP of senescent SNpc DA neurons in PD patients triggers an immune response that leads to their elimination [229]. Senescent cells are resistant to apoptosis and accumulate with age [230,231].

5. Mechanisms and Markers of Cellular Senescence

The diverse and highly context-dependent nature of cellular senescence has prevented the identification of a universal senescence biomarker [232,233,234]. Therefore, multiple senescence-associated characteristics must be evaluated when determining senescence levels within a given tissue. Replicative senescence or replicative exhaustion was first observed by Hayflick and Moorhead and refers to the cessation of the cell cycle because of repeated mitosis [233]. Repeated cell division and DNA replication result in the progressive shortening of telomeres and the eventual loss of the telomere t-loops which cap and protect the ends of DNA strands [235]. Telomerase prevents telomere attrition under normal circumstances; however, the activity of this enzyme is diminished with age and disease [236]. The cell recognizes the loss of t-loop capping as double-strand DNA breaks and triggers the DNA damage response, which can initiate cellular senescence [237]. Replicative senescence is distinct from the other forms of DNA damage response-induced senescence such as bleomycin-caused oxidative DNA damage-induced senescence [234]. Yet, no differences in conventional senescence-associated markers were detected between the two types of senescence. However, the 18S, 5.8S, and 28S ribosome promoter regions in the DNA were hypermethylated in the case of replicative senescence but absent in the bleomycin-caused oxidative DNA damage-induced senescence. This hypermethylation of ribosomal promoter regions resulted in the reduced expression of the corresponding ribosomal RNA in replicative senescence alone [234,237] and highlights the intricacies of cellular senescence.
ROS act as regulators of various cellular processes under homeostatic conditions such as metabolic regulation; however, exposure and aberrant production of ROS cause dysfunction in a multitude of cellular processes, many of which can be senescence-initiating factors including ROS itself [238]. Interestingly, senescent cells further increase ROS production [239]. Increased ROS production can lead to irreparable DNA damage, mitochondrial and metabolic dysfunction, and protein misfolding, all of which are characteristics and inducers of cellular senescence [240]. Aside from developmental-, nutrient deprivation-, and some forms of tissue repair-induced senescence, DNA damage is a common underlying feature of senescent cells [240,241,242]. Non-telomere attrition-associated DNA damage occurs in oxidative stress-, oncogene-, irradiation-, and chemically-induced senescence [243]. The distinguishing factors between the types of senescence are the mechanisms by which DNA damage was accrued and the type of DNA damage detected by the cell. Furthermore, cellular senescence can result from both nuclear and mitochondrial DNA damage [244].
The tumor suppressor protein p53 is widely recognized as “the guardian of the genome” and indirectly halts cell cycle progression or induces apoptosis in response to DNA damage [245]. Cellular senescence has been linked to two positive p53 regulatory mechanisms. Mouse double minute 2 (MDM2) negatively regulates p53 under homeostatic conditions by promoting p53 proteasomal degradation to ensure cell cycle continuation [245]. This negative regulatory mechanism is circumvented when the serine/threonine kinase, ataxia telangiectasia and Rad-3-related (ATR), phosphorylates p53 in rapid response to DNA damage [246]. The degradation of p53 can also be avoided by p14ARF (p19ARF in mice)-mediated inhibition of MDM2 in the context of chronic or irreparable DNA damage [247]. p53 can then act as a transcription factor and/or protein binding partner to ultimately activate numerous genes and proteins that result in cell cycle arrest and DNA damage repair [248].
Consequentially, cellular senescence can arise when DNA damage is chronic or beyond repair [248]. Two senescence-associated genes, CDKN1A and CDKN2A, encode the cyclin-dependent kinase inhibitors that directly prevent cell cycle progression, known as p21WAF1/CIP1 and p16INK4A, respectively [249]. The direct binding of p53 to the promoter region of the CDKN1A gene results in the upregulation of p21WAF1/CIP1 [250,251]. This mechanism is known as the p53-p21WAF1/CIP1 pathway and is generally associated with the induction of senescence when DNA damage accrues [252]. The p16INK4A pathway is a p53-independent mechanism of cellular senescence [252]. p16INK4A is encoded by CDKN2A, which also encodes p14ARF through an alternate reading frame [253]. The maintenance of cellular senescence has been associated with the p16INK4A pathway in the context of chronic or irreparable DNA damage but can also be activated by ROS sensing mechanisms [254]. The p21WAF1/CIP1 and p16INK4A proteins prevent cell cycle progression by inhibiting the association of cyclins with their respective cyclin-dependent kinases, preventing the phosphorylation of the Retinoblastoma (Rb) protein [255]. Hypophosphorylation of Rb results in the sequestration of E2F transcription factor 1, ultimately preventing the transcription of genes necessary for DNA replication and cell cycle progression [240]. Increases in p53, p21WAF1/CIP1, and p16INK4A are, consequentially, the most commonly used identifiers when evaluating senescence.
Substantial cross-activation between the p53-p21WAF1/CIP1 and p16INK4A pathways exists despite the ability of these two pathways to induce senescence independently. For instance, p53 can stabilize Lamin A/C to promote the degradation of Polycomb repressor complex 1 components, resulting in the derepression of CDKN2A and a subsequent increase in p16INK4A [256]. p21WAF1/CIP1 has been shown to induce p16INK4A expression by promoting SP1 transcription factor association with the p16INK4A promoter region [193]. Conversely, p16INK4A can stabilize p21WAF1/CIP1 mRNA and prevent MDM2-mediated p53 degradation, leading to subsequent increases in both proteins [257,258]. The complex interactions between the senescence-related markers make it difficult to identify a single, definable senescence marker.
Nicotinamide adenosine dinucleotide-dependent deacetylase sirtuin-1 (SIRT1) regulates a myriad of cellular processes including cellular senescence, metabolism, aging, and inflammation, through its ability to deacetylate a diverse range of target proteins [259,260,261]. Several mechanisms contribute to the downregulation of SIRT1 during senescence. Conversely, overexpression of SIRT1 can prevent various forms of senescence [262,263,264]. SIRT1 is further associated with the regulation of senescence genes through its inherent histone deacetylase activity [265]. Senescence studies have shown that SIRT1 regulates the SASP by directly repressing SASP gene promoter sequences and indirectly by deacetylating p53 at lysine 382 to limit the SASP-promoting p53 transcriptional activity [263,266]. p53 binding to the SIRT1 promoter lowers SIRT1 levels in response to DNA damage and results in the derepression of SASP genes [263,267]. Increased lysosomal activity further decreases SIRT1 protein levels through selective autophagy [264]. The implications of SIRT1 overexpression ameliorating senescence and aging along with the observed decreases in gene and protein expression in senescence make SIRT1 a prominent marker of senescence.
Lamin B1 is also degraded through selective autophagy in senescent cells [264]. Under homeostatic conditions, Lamin B1 functions as a nuclear structural protein that assists in maintaining the epigenetic configuration of DNA and, therefore, indirectly regulates gene expression by controlling gene accessibility [268]. The maintenance of heterochromatin by Lamin B1 helps to suppress senescence-associated gene transcription [269,270]. Senescence-associated degradation of Lamin B1 results in the rearrangement of genetic material and the loosening of heterochromatic DNA [270]. The transition of Lamin B1-bound loci to euchromatin allows the transcription of senescence-associated genes [270]. These epigenetic rearrangements also contribute to characteristic senescence-associated heterochromatic foci (SAHF) [271]. Observing decreases in Lamin B1 and the observation of SAHF, therefore, serves as a proxy for senescence-associated epigenetic alterations. The aforementioned increased lysosomal activity of senescent cells, specifically at pH 6, can be observed through SA β-gal assays and is among the most used senescent cell markers [230].
Markers of senescence are not limited to those mentioned above and include various senescence-associated cell surface proteins, microribonucleic acid expression, and the phosphorylation state of specific proteins. However, a major limitation in senescence and aging research is that many of these senescence-associated alterations occur in other cellular processes and require the detection of multiple markers to discriminate cellular senescence from other phenomena. Recent advances in techniques such as matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) and spatial transcriptomics offer an opportunity for investigators to extensively characterize the transcriptional and proteomic differences in senescence between tissues and cell types with single-cell resolution. However, the use of such techniques to study senescence is beyond the scope of this review, and more comprehensive reviews of recent advances and the capabilities of MALDI-MSI and single-cell multi-omics are discussed by Ngai and colleagues and Sun and colleagues, respectively [272,273].

6. Role of Cellular Senescence in Aging

Cellular senescence is thought to be the underlying mechanism behind organismal aging; however, when compared to healthy cells, the percentage of senescent cells in a particular tissue is negligible [274,275]. In young, healthy mice, transplanting a small number of senescent cells (1 × 106, ~0.01–0.03 percent of the recipient’s total cells) resulted in cellular senescence in host cells and persistent physical dysfunction [46]. In older mice, transplanting fewer senescent cells (0.5 × 106, 0.007% of the total cells in the recipient) resulted in cellular senescence in host cells, decreased lifespan, and chronic physical dysfunction [46]. If senescent cell transplantation results in physical dysfunction and shortens lifespan, then senescent cell removal should lengthen lifespan and/or postpone aging-related tissue dysfunction. To address this question, researchers designed a novel transgene, INK-ATTAC, for the inducible elimination of p16INK4A-positive cells [206]. In the BubR1 progeroid mouse background, INK-ATTAC removed p16INK4A-positive cells upon drug treatment. Using this mouse model, p16INK4A was demonstrated to play a role in the development of aging-related pathologies in the eye, skeletal muscle, and adipose tissue because the onset of aging-related pathological phenotypes was postponed by lifelong deletion of p16INK4A-expressing cells. INK-ATTAC’s late-life clearance of p16INK4A-expressing cells slowed the development of aging-related diseases that were already well-established [206]. These results imply that cellular senescence contributes to aging-related phenotypes and that senescent cell removal can prolong life by halting or postponing aging-related tissue dysfunction. Though senescent cells are sparse, they have an appreciable impact on surrounding tissue. Senescent cells generate a chronic pro-inflammatory environment and increased ROS, promoting an environment conducive to many of the previously discussed pathogenic factors of PD such as oxidative stress, macromolecular damage, and misfolded proteins [276,277,278,279]. Aging is the single most critical risk factor for PD [26,27,28]; therefore, it must be considered that cellular senescence may play a role in PD pathogenesis.

7. Effects of Senotherapeutics in PD Models

The clearance of senescent cells by genetic methods has led to a rush in the discovery and development of pharmacological approaches to target senescent cells without using a transgene, known as senotherapeutics. Whereas senolytics selectively kill senescent cells, senomorphics spare senescent cells yet attenuate the pathological SASP. Notably, depending on the cell type, compounds can be senolytic and/or senomorphic.
The purpose of first generation senolytic compounds was to interfere with senescent cells’ pro-survival pathways. Dasatinib functions as a receptor tyrosine kinase (RTK) antagonist, which promotes survival [280]. PF-04691502 inhibits the pro-survival phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway [281]. Both quercetin and fisetin block NFκB and the PI3K/Akt pathway [280]. Geldanamycin, tanespinmycin, and 17-dimethylaminoethylamino (17-DMAG) are examples of heat shock protein 90 (HSP90) inhibitors [282] that produce a senolytic effect by blocking pro-survival pathways and disrupting intracellular protein homeostasis [283]. Senescent cells that are the target of other first generation senolytics have higher levels of the pro-survival B-cell lymphoma-2 (Bcl-2) protein family, which includes Bcl-2, B-cell lymphoma-extra large (Bcl-xl), and B-cell lymphoma-w (Bcl-w) [284]. Specifically, digoxin, ouabain, and epigallocatechin gallate inhibit Bcl-2. ABT-263 (Navitoclax), ABT-737, and PZ15227 inhibit Bcl-w. A1331852, UBX1325, and A1155463 inhibit BCL-xl [285,286,287]. Senescent cells exhibit a higher concentration of protons in their plasma membrane as this membrane is more depolarized than non-senescent cells. Thus, inhibitors of sodium/potassium adenosine triphosphatase (Na+/K+ ATPase), such as digoxin and digitoxin, have a senolytic effect [288]. Despite the fact that numerous senolytics and compounds have been developed with a wide range of subcellular targets (Figure 2), few have been tested in in vitro or in vivo PD models.
The primary strategy for first generation senomorphics is the targeting of pathways related to SASP expression, such as p38 mitogen-activated protein kinase (p38-MAPK), PI3K/Akt, mechanistic target of rapamycin (mTOR), and Janus kinase/signal transducer and activation of transcription (JAK/STAT) pathways and transcription factors [289]. Everlimus and rapamycin inhibit mTOR, which suppresses the SASP [290,291,292]. Metformin, apigenin, kaempferol, and the glucocorticoids cortisol and corticosterone have strong senomorphic activity through NF-κB inhibition [293,294,295]. Ruxolitinib, a selective JAK1/2 inhibitor, reduced the expression of SASP in senescent cells [296]. In addition, blocking and neutralizing antibodies against the SASP factors interleukin-1 (IL-1) (anakinra, canakinumab, and riloncept), IL-6 (tocilizumab and siltuximab), and tumor necrosis factor (TNF) (etanercept and infliximab) have senomorphic effects [297]. Inhibitors of the p38 pathway, such as SB203580, have been shown to inhibit the SASP [298]. Pharmacological inhibition of cyclic GMP-AMP synthase (cGAS) via administration of RU.521 diminished senescence [299]. The selective NOD-like receptor domain-containing protein 3 (NLRP3) inhibitor, MCC950, ameliorated senescence [300]. Finally, the compounds FK866 and mitoquinone (mitoQ) have a senomorphic effect by blocking the production of ROS within mitochondria [301]. Despite the fact that numerous senomorphic compounds have been developed with a wide range of subcellular targets (Figure 3), few have been tested in in vitro or in vivo PD models.

7.1. Effects of Curcumin in PD Models

The main biologically active component of turmeric, curcumin (diferuloylmethane), has therapeutic effects via its anti-oxidant and anti-inflammatory properties [302]. Without having any harmful effects on healthy cells, curcumin had cytotoxic effects on cancer cells [303]. Curcumin selectively induces apoptosis in senescent but not proliferating cells [304] and disrupts several cellular functions such as proliferation, cell survival, and apoptosis [305]. Epidemiological data point to a decline in the prevalence of PD in India [306], where ~200 mg of curcumin is ingested daily [307]. The advantages of curcumin as a potential disease-modifying therapeutic for treating PD include low cost, safety, and the ability to cross the BBB. Curcumin has been tested in in vitro and in vivo models of PD (Table 1). Curcumin disrupted preformed α-syn aggregates, enhanced the solubility of α-syn aggregates in catecholaminergic SH-SY5Y cells, and dose-dependently inhibited α-syn aggregation [308]. In a lipopolysaccharide (LPS) model of PD, an intranigral injection of LPS in rats activated astrocytes, a significant inflammatory response was triggered, and intracellular α-syn aggregates were formed. Systemic administration of curcumin once daily for 21 days prevented the activation of astrocytes caused by LPS, the upregulation of NFκB, the pro-inflammatory cytokines TNF-α, IL-1β, and IL-1α, inducible nitric oxide synthase (iNOS), and regulating molecules of the intrinsic apoptotic pathway such as Bax, Bcl-2, caspase-3, and caspase-9. Additionally, curcumin treatment inhibited α-syn aggregation in DA neurons and enhanced the glutathione system [309]. Curcumin pretreatment preserved DA content in the striatum and protected SN DA neurons in rats in the 6-hydroxydopamine (6-OHDA) model of PD [310]. In MPP+-treated catecholaminergic SH-SY5Y cells, curcumin-loaded nanoparticles (NPs) restored tyrosine hydroxylase (TH) to control levels, removed α-syn aggregates, reversed MPP+-induced morphological alterations, and safeguarded against cell death induced by MPP+. In MPTP-treated mice, curcumin-loaded NPs reversed MPTP-induced motor deficits and restored SN TH and striatal DA to control levels [311]. These findings imply that NPs loaded with curcumin may be a disease-modifying treatment for PD.

7.2. Effects of Fisetin in PD Models

Apples, persimmon, grapes, onions, cucumbers, strawberries, and many other fruits and vegetables contain the flavonoid fisetin (3,3′,4′,7-tetrahydroxyflavone) [312]. Fisetin, even when administered at high doses, has not been reported to have adverse effects [313]. Fisetin has been shown to be neuroprotective by reducing tumorigenesis, ROS production, and inflammation [314,315,316]. Fisetin can specifically trigger the apoptosis of senescent cells without altering proliferating human umbilical vein endothelial cells [317]. Fisetin decreased senescence markers in human adipose tissue explants, senescent mouse fibroblasts, progeroid mice, and aged, wild-type mice. Furthermore, fisetin extended the lifespan of wild-type mice, even when treatment began in old age [318]. Fisetin has been evaluated in both in vitro and in vivo PD models (Table 1). The 6-OHDA-mediated death of catecholaminergic SH-SY5Y cells, elevation of oxidative stress-related genes, and the activation of caspase-3 and caspase-9 were all inhibited by fisetin pretreatment. Fisetin attenuated a 6-OHDA-induced increase in the ratio of the expression of the pro-apoptotic Bax protein to the anti-apoptotic Bcl-2 protein in these cells. The fisetin-mediated blockade of 6-OHDA-induced cell death occurred through the activation of the PI3K/Akt pathway, which inhibits pro-apoptotic proteins, such as caspase-3 and caspase-9 [319]. Fisetin protected against MPTP-induced cell death of rat PC12 cells, blocked MPTP-induced increases in NO and α-syn expression, suppressed pro-apoptotic signaling, and attenuated MPTP-induced increased expression of NFκB as well as the pro-inflammatory cytokines, TNF-α and IL-1β [320]. The effects of fisetin have been examined in two rodent mouse models [321,322]. Once daily oral administration of fisetin protected against MPTP-induced decreases in striatal DA and TH immunoreactivity in mice [321]. Once daily oral administration of fisetin attenuated MPTP-induced motor deficits and apoptosis of SN DA neurons in mice [323]. Fisetin was given orally once daily in the rotenone model of PD 10 days prior to the onset of rotenone administration and continued until the end of rotenone administration. Fisetin attenuated rotenone-induced motor behavior deficits, decreases in striatal DA and TH immunoreactivity, deficits in midbrain mitochondrial function, and increases in oxidative stress markers [322].

7.3. Effects of a Serum/Glucocorticoid-Related Kinase 1 Inhibitor in PD Models

Osmotic and isotonic cell shrinkage, cytokines, growth factors, and steroid and peptide hormones all influence the serine/threonine kinase known as serum/glucocorticoid-related kinase 1 (SGK1) [324]. In PD patients and the MPTP model of PD, SGK1 inducers like cytokines and glucocorticoids are upregulated [325,326,327]. All regions of the brain express SGK1, and DA neurons in the ventral midbrain (VM) were found to express it. It is present in astrocytes but is preferentially expressed in neurons. Interestingly, SGK1 mRNA was upregulated in the SN for at least 14 days after a single, intra-striatal injection of 6-OHDA [328], and MPTP-induced SN DA neuron death began at the same time as an MPTP-induced upregulation of sgk1 [329].
The effects of pharmacological inhibition of SGK1 in multiple in vitro and in vivo models of PD have been examined (Table 1). Treatment of VM glial cultures with the specific SGK1 inhibitor, GSK-650394, downregulated pro-oxidant and SASP genes. GSK-650394 treatment decreased the toxin-induced rise in mitochondrial ROS and fall in mitochondrial membrane potential in VM glial cultures. The levels of pro-senescence proteins, SASP proteins, and senescence markers returned to normal when VM glial cultures were exposed to toxins while GSK-650394 was present. In addition, pharmacological inhibition of SGK1 prevented α-syn aggregation in co-cultures of midbrain DA neurons overexpressing α-syn and VM glial cells treated with α-syn preformed fibrils; this effect was due to an effect on glial cells. To determine if pharmacological inhibition of SGK1 could rescue a PD phenotype in vivo, mice received once-daily MPTP injections for 5 days. One week after these injections, mice received once-daily injections of GSK-650394. Treatment with GSK-650394 alleviated the MPTP-induced motor deficits, preserved DA fibers in the striatum, and attenuated SN DA neuron loss [330]. In an in vivo PD model produced by α-syn overexpression and the intra-nigral injection of α-syn preformed fibrils, treatment with GSK-650394 alleviated the progression of motor deficits, decreased α-syn pathology, and reduced the degeneration of DA fibers and SN DA neurons [330]. SGK1 inhibitors may, therefore, be able to change the course of PD.
Table 1. Effects of senotherapeutics in cell culture and animal models of PD. Abbreviations: 6-OHDA, 6-hydroxydopamine; SN, substantia nigra; DA, dopamine; PI3K, phosphoinositide 3-kinase; Akt, protein kinase B; α-syn, α-synuclein; TH, tyrosine hydroxylase; mDA, midbrain dopamine; SGK1, serum/glucocorticoid-related kinase 1; NFκB, nuclear factor kappa-B; iNOS, inducible nitric oxide synthase; MPP+, 1-methyl-4-phenylpyridinium ion; MPTP, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; NPs, nanoparticles; NO, nitric oxide; LPS, lipopolysaccharide.
Table 1. Effects of senotherapeutics in cell culture and animal models of PD. Abbreviations: 6-OHDA, 6-hydroxydopamine; SN, substantia nigra; DA, dopamine; PI3K, phosphoinositide 3-kinase; Akt, protein kinase B; α-syn, α-synuclein; TH, tyrosine hydroxylase; mDA, midbrain dopamine; SGK1, serum/glucocorticoid-related kinase 1; NFκB, nuclear factor kappa-B; iNOS, inducible nitric oxide synthase; MPP+, 1-methyl-4-phenylpyridinium ion; MPTP, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; NPs, nanoparticles; NO, nitric oxide; LPS, lipopolysaccharide.
SerotherapeuticMain Finding(s)Citation
CurcuminCurcumin inhibited α-syn aggregation and increased α-syn solubility in SH-SY5Y cells.[308]
CurcuminCurcumin prevented LPS-induced astrocyte activation and upregulation of NFκB, pro-inflammatory cytokines, iNOS, and regulating molecules of the intrinsic apoptotic pathway in rats. Curcumin improved the glutathione system and prevented iron deposition and α-syn aggregation in DA neurons in rats.[309]
CurcuminCurcumin protected against 6-OHDA-induced death of SN DA neurons and decreased striatal DA content in rats.[310]
CurcuminCurcumin-loaded NPs restored TH to control levels, removed α-syn aggregates, reversed morphological alterations, and protected against cell death in MPP+-treated SH-SY5Y cells. Curcumin-loaded NPs reversed motor deficits and restored striatal DA levels and SN TH immunoreactivity in MPTP-treated mice.[311]
FisetinFisetin protected against 6-OHDA-induced elevation of oxidative stress-related genes, activation of caspase-3 and caspase-9, and cell death by activating PI3K-Akt signaling in SH-SY5Y cells.[319]
FisetinFisetin protected against MPTP-induced cell death, blocked MPTP-induced increases in NO and α-syn expression, attenuated MPTP-induced increased expression of pro-inflammatory cytokines, and suppressed pro-apoptotic signaling in rat PC12 cells.[320]
FisetinFisetin protected against MPTP-induced decreases in striatal DA and TH immunoreactivity in mice.[321]
FisetinFisetin attenuated rotenone-induced motor behavior deficits, decreases in striatal DA and TH immunoreactivity, deficits in midbrain mitochondrial function, and increases in oxidative stress markers in rats.[322]
FisetinFisetin prevented MPTP-induced motor deficits and protected SN DA neurons from apoptosis in mice.[323]
GSK-650394Pharmacological inhibition of glial cell SGK1 in glia-mDA neuron co-cultures attenuated mDA neuron death and neurite degeneration. Glial cell SGK1 inhibition alleviated MPTP- and α-syn overexpression-induced behavioral deficits, SN mDA neuron loss, and striatal TH immunoreactivity in mice.[330]
Astragaloside IVAstragaloside IV prevented the induction of senescence in astrocytes in vitro. Astragaloside IV prevented MPTP-induced motor deficits, SNpc DA neuron loss, and accumulation of senescent astrocytes in the SNpc in mice.[331]

7.4. Effects of Astragaloside IV in PD Models

Astragaloside IV (AS-IV) is one of the main active ingredients of the Chinese medicinal herb Astragalus membranaceus. AS-IV exerts anti-tumor, anti-inflammation, and anti-oxidative stress activities [332,333,334]. Although the effect of AS-IV on cellular senescence was not examined, Chan and colleagues [335] discovered that, in primary nigral cell cultures, pretreatment with AS-IV protected against 6-OHDA-mediated DA neuron loss and degeneration of neurites. Interestingly, treatment of nigral cell cultures with AS-IV alone increased the length and branching of axons of DA neurons [335]. AS-IV prevented both LPS- and MPP+-mediated induction of senescence in astrocytes in culture (Table 1). Moreover, AS-IV prevented MPTP-mediated loss of DA neurons in the SNpc, motor deficits, and the accumulation of senescent astrocytes in the SNpc. AS-IV promoted mitophagy, which reduced the accumulation of damaged mitochondria and the generation of mitochondrial ROS, thereby preventing the induction of senescence in astrocytes (Table 1) [331]. Thus, AS-IV has potential as a disease-modifying therapeutic for PD.

8. Challenges Associated with Senolytics and Senomorphics

Senolytic and senomorphic compounds are attractive as therapeutics as they have the potential to treat many aging-related conditions at once and have demonstrated efficacy in extending healthspan and lifespan in animal models. However, their use has drawbacks. Senescent cells are heterogeneous in their transcriptional, metabolic, and SASP profiles between species, cell types, and individual cells, and the specific targeting of pathways shared between senescent and non-senescent cells can result in off-target effects. As mentioned previously, senescent cells play an important role in tissue renewal, wound healing, and cancer prevention. The potential long-term side effects of senolytics and senomorphics remain unclear as, to date, these compounds have been administered to mice for a short duration, ranging from 3 to 6 months [336]. Studies in which senolytics and senomorphics are administered for an extended time course are needed to better understand the potential toxicity of the drugs. Studies that employ the long-term administration of senolytics are needed to understand the potential negative consequences of long-term removal of senescent cells. Notably, as senescent cells accumulate slowly, senolytics are often dosed intermittently, using a hit-and-run strategy, which likely lessens the side effects that would otherwise occur if the compounds were administered continuously [337]. In contrast to senolytics, senomorphics may be beneficial when taken regularly for an extended period. However, some side effects have been reported for some senomorphics. For example, the senomorphic rapamycin has beneficial effects in animal models, but it also induces thrombocytopenia, metabolic dysregulation, hyperlipidemia, and impairs wound healing [291]. Importantly, many senotherapeutic molecules have only been examined at one or a few concentrations.

9. Next Generation Senotherapeutics

Efforts are underway to develop next generation senotherapeutic strategies, often with better selectivity, safety, and/or efficacy. Next generation therapeutics include senolytic peptides, senoreverters, proteolysis-targeting chimeras (PROTACs), pro-drugs, immunotherapy, and NPs. Although these next generation senotherapeutics may have advantages, none have been tried in in vitro and in vivo models of PD.

9.1. Senolytic Peptides

The protein forkhead box subclass O protein 4 (FOXO4) was highly expressed in senescent cells but not in healthy cells. The D-retro inverse (DRI) isoform of FOXO4, the FOX-DRI peptide, is designed to perturb the interaction between FOXO4 and p53. Disrupting FOXO4-p53 binding causes p53 to be excluded from the nucleus, and consequently, mitochondrial-induced apoptosis of senescent cells occurs (Figure 2) [338].

9.2. Senoreverters

Although cellular senescence has been viewed as an irreversible cell fate, studies suggest that senescence, in specific cell types, is a dynamic process that can be reverted to allow senescent cells to re-enter the cell cycle [254]. For example, a six-factor gene cocktail reversed the cellular senescence of senescent and centenarian fibroblasts into pluripotent stem cells that were indistinguishable from human embryonic stem cells. Incredibly, the senescent and centenarian-derived pluripotent stem cells were redifferentiated into fully rejuvenated cells [339]. Thus, senoreverters and/or cell rejuvenation may be a novel approach for the treatment of PD.

9.3. PROTACs

PROTACs are an innovative technology to induce the degradation of a specific protein of interest [340]. PROTACs, which hijack the UPS system, are comprised of a ligand that binds to a target protein of interest, an E3 ligase recruiting ligand, and a flexible linker between the two ligands. Thus, a PROTAC can form a stable complex with a protein of interest and E3 ligase, resulting in the subsequent ubiquitination and proteasomal degradation of the protein of interest. PROTACs have several advantages, such as increased potency, higher selectivity, prolonged activity, and reduced toxicity, making them an attractive strategy for developing senotherapeutics [341].

9.4. Pro-Drugs

One common characteristic shared by senescent cells is the elevated activity of the lysosomal SA-β-gal, which hydrolyses the bond formed between a galactose and its organic moiety. According to this novel approach, pro-drugs with a cleavable galactose moiety attached to a cytotoxic compound can be processed specifically in senescent cells after cellular uptake, releasing the cytotoxic drug and resulting in the selective killing of senescent cells. Several galactose-based pro-drugs, such as SKK1, prodrug A, and Nav-Gal, have been developed and selectively kill senescent cells while sparing non-senescent normal and proliferating cells [342,343,344].

9.5. Immunotherapy

Immunotherapy is based on stimulating the ability of immune cells to target senescent cells. Immunotherapies take advantage of proteins upregulated on the cell surface of senescent cells. One immunotherapy approach used antibodies to guide immune cells toward cytotoxic clearance of senescent cells. Dipeptidyl peptidase 4 (DDP4) is a protein preferentially upregulated on the cell membrane of senescent cells. An anti-DDP4 antibody labeled DDP4-expressing senescent cells, which were subsequently cleared by immune cells [345]. Another immunotherapy approach involves using antibody-drug conjugates, which are monoclonal antibodies attached to cytotoxic drugs. In this approach, the antibody recognizes and binds to an extracellular epitope preferentially expressed on the cell surface of the senescent cells. After internalization, the drug is cleaved from the antibody, releasing the toxin, which causes the death of the senescent cell [346]. Finally, neutralizing antibodies can target specific SASP components (e.g., IL-1β, IL-6, and IL-8) or their cell surface receptors (Figure 3) for senescence suppression.

9.6. NPs

NPs have been used to increase therapeutic efficacy, reduce off-target effects, and deliver cytotoxic or therapeutic cargo to specific cells. For example, to increase the bioavailability of the senotherapeutic curcumin, polysorbate 80-modified ceresome (PS80 CPC) NPs were fabricated and loaded with curcumin. Compared to the systemic administration of free curcumin, the systemic administration of PS80 CPC NPs substantially increased the circulation lifetime and amount of curcumin that reached the brain. The circulation lifetime and amount of curcumin that reached the brain were more than 8 times and 100 times greater, respectively, for curcumin-loaded PS80 CPC NPs than for free curcumin [311]. Additionally, SA-β-gal has been used to produce NPs for preferential delivery into senescent cells. The senolytic, ABT-263 (Navitoclax), was loaded into NPs to generate senolytic NPs. After cellular uptake, fusion with lysosomes, and hydrolysis of the galacto-oligosaccharide coat by SA-β-gal, these NPs released their cytotoxic cargo to selectively kill senescent cells while sparing normal, healthy cells [347]. Advantages to the use of exosomes (EXs), membrane-enclosed NPs released by cells into the extracellular space, as carriers of therapeutic agents into the diseased brain, include their ability to readily cross the BBB, the potential for targeted delivery of therapeutic cargo over a long distance, and immune resistance. In addition, a wide variety of cargo can be loaded into EXs. Loading therapeutic agents into EXs often results in greater stability, increased bioavailability, protection from degradation, and reduced immunogenicity. Genetic modification of EX-producing cells and/or EXs and chemical modification of EXs have emerged as powerful approaches for the targeted delivery of therapeutics to neurons and/or glia. Thus, EXs hold great promise as carriers of disease-modifying therapeutics for treating PD [126].

10. Conclusions

PD is an aging-related progressive neurodegenerative disease that affects 1–2% of the population over the age of 65. One of the biggest concerns in PD research is the failure of all clinical studies of administering drugs to modify the course of PD or regenerate DA neurons. Indeed, there is an urgent need for disease-modifying therapeutics (specifically, those that slow, stop, or reverse cell death) in PD. PD pathogenesis includes oxidative stress, diminished anti-oxidant defenses, mitochondrial dysfunction, protein misfolding, dysfunction of the ALP, impaired UPS function, impaired Ca2+ handling, inflammation, and cellular senescence (Figure 1). Cellular senescence plays an important role in aging and aging-related disorders such as PD, and studies suggest that the removal of senescent cells or the modulation of the SASP can extend healthspan and lifespan and may modify the course of PD. Although a host of senolytics and senomorphic compounds have been developed with a wide range of subcellular targets (see Figure 2 and Figure 3), few have been tested in in vitro or in vivo PD models (Table 1). Although the senotherapeutics curcumin, fisetin, GSK-650394, and AS-IV have impressive disease-modifying effects in culture and animal models of PD, their efficacy has not been determined in higher-order animal models of PD, such as non-human primate models, and scientists do not yet know whether their beneficial effects will translate to human PD patients. Studies are needed to determine whether existing senotherapeutics have side effects when administered long-term, and there is a need for clinical trials for PD with senotherapeutics that have shown promise in PD models (Table 1). Although next generation senotherapeutic strategies are very promising and offer distinct advantages compared to first generation senotherapeutics, studies that examine the disease-modifying potential of senolytic peptides, senoreverters, PROTACs, pro-drugs, immunotherapy, and NPs in cell culture and animal models of PD are needed.

Author Contributions

Conceptualization, D.J.R.; writing-review and editing, D.J.R., J.E.E. and E.M.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Lebouvier, T.; Chaumette, T.; Paillusson, S.; Duyckaerts, C.; Bruley des Varannes, S.; Neunlist, M.; Derkinderen, P. The second brain and Parkinson’s disease. Eur. J. Neurosci. 2009, 30, 735–741. [Google Scholar] [CrossRef] [PubMed]
  2. Marras, C.; Beck, J.C.; Bower, J.H.; Roberts, E.; Ritz, B.; Ross, G.W.; Abbott, R.D.; Savica, R.; Van Den Eeden, S.K.; Willis, A.W.; et al. Prevalence of Parkinson’s disease across North America. NPJ Park. Dis. 2018, 4, 21. [Google Scholar] [CrossRef] [PubMed]
  3. Spillantini, M.G.; Schmidt, M.L.; Lee, V.M.; Trojanowski, J.Q.; Jakes, R.; Goedert, M. Alpha-synuclein in Lewy bodies. Nature 1997, 388, 839–840. [Google Scholar] [CrossRef]
  4. Kalia, L.V.; Lang, A.E. Parkinson’s disease. Lancet 2015, 386, 896–912. [Google Scholar] [CrossRef]
  5. Chaudhuri, K.R.; Healy, D.G.; Schapira, A.H.; National Institute for Clinical Excellence. Non-motor symptoms of Parkinson’s disease: Diagnosis and management. Lancet Neurol. 2006, 5, 235–245. [Google Scholar] [CrossRef] [PubMed]
  6. Muleiro Alvarez, M.; Cano-Herrera, G.; Osorio Martinez, M.F.; Vega Gonzales-Portillo, J.; Monroy, G.R.; Murguiondo Perez, R.; Torres-Rios, J.A.; van Tienhoven, X.A.; Garibaldi Bernot, E.M.; Esparza Salazar, F.; et al. A Comprehensive Approach to Parkinson’s Disease: Addressing Its Molecular, Clinical, and Therapeutic Aspects. Int. J. Mol. Sci. 2024, 25, 7183. [Google Scholar] [CrossRef]
  7. Cilia, R.; Cereda, E.; Akpalu, A.; Sarfo, F.S.; Cham, M.; Laryea, R.; Obese, V.; Oppon, K.; Del Sorbo, F.; Bonvegna, S.; et al. Natural history of motor symptoms in Parkinson’s disease and the long-duration response to levodopa. Brain 2020, 143, 2490–2501. [Google Scholar] [CrossRef]
  8. Rascol, O.; Brooks, D.J.; Brunt, E.R.; Korczyn, A.D.; Poewe, W.H.; Stocchi, F. Ropinirole in the treatment of early Parkinson’s disease: A 6-month interim report of a 5-year levodopa-controlled study. 056 Study Group. Mov. Disord. 1998, 13, 39–45. [Google Scholar] [CrossRef]
  9. Parkinson Study Group. Pramipexole vs levodopa as initial treatment for Parkinson disease: A randomized controlled trial. Parkinson Study Group. JAMA 2000, 284, 1931–1938. [Google Scholar] [CrossRef]
  10. Rascol, O. L-dopa-induced peak-dose dyskinesias in patients with Parkinson’s disease: A clinical pharmacologic approach. Mov. Disord. 1999, 14 (Suppl. 1), 19–32. [Google Scholar]
  11. Bezard, E.; Brotchie, J.M.; Gross, C.E. Pathophysiology of levodopa-induced dyskinesia: Potential for new therapies. Nat. Rev. Neurosci. 2001, 2, 577–588. [Google Scholar] [CrossRef] [PubMed]
  12. Armstrong, M.J.; Okun, M.S. Diagnosis and Treatment of Parkinson Disease: A Review. JAMA 2020, 323, 548–560. [Google Scholar] [CrossRef]
  13. Lang, A.E.; Lozano, A.M. Parkinson’s disease. Second of two parts. N. Engl. J. Med. 1998, 339, 1130–1143. [Google Scholar] [CrossRef] [PubMed]
  14. Manson, A.; Stirpe, P.; Schrag, A. Levodopa-induced-dyskinesias clinical features, incidence, risk factors, management and impact on quality of life. J. Park. Dis. 2012, 2, 189–198. [Google Scholar] [CrossRef] [PubMed]
  15. Cenci, M.A.; Skovgard, K.; Odin, P. Non-dopaminergic approaches to the treatment of motor complications in Parkinson’s disease. Neuropharmacology 2022, 210, 109027. [Google Scholar] [CrossRef]
  16. Muller, T. Pharmacokinetic considerations for the use of levodopa in the treatment of Parkinson disease: Focus on levodopa/carbidopa/entacapone for treatment of levodopa-associated motor complications. Clin. Neuropharmacol. 2013, 36, 84–91. [Google Scholar] [CrossRef]
  17. Kataoka, H.; Sugie, K. Early-morning OFF in Parkinson’s disease: A systematic literature review and current therapeutics. Clin. Neurol. Neurosurg. 2024, 245, 108493. [Google Scholar] [CrossRef]
  18. Smith, Y.; Wichmann, T.; Factor, S.A.; DeLong, M.R. Parkinson’s disease therapeutics: New developments and challenges since the introduction of levodopa. Neuropsychopharmacology 2012, 37, 213–246. [Google Scholar] [CrossRef]
  19. Bodagh, I.Y.; Robertson, D.R. A risk-benefit assessment of drugs used in the management of Parkinson’s disease. Drug Saf. 1994, 11, 94–103. [Google Scholar] [CrossRef]
  20. Factor, S.A.; Molho, E.S.; Podskalny, G.D.; Brown, D. Parkinson’s disease: Drug-induced psychiatric states. Adv. Neurol. 1995, 65, 115–138. [Google Scholar]
  21. Senard, J.M.; Rai, S.; Lapeyre-Mestre, M.; Brefel, C.; Rascol, O.; Rascol, A.; Montastruc, J.L. Prevalence of orthostatic hypotension in Parkinson’s disease. J. Neurol. Neurosurg. Psychiatry 1997, 63, 584–589. [Google Scholar] [CrossRef]
  22. Gentil, M.; Pollak, P.; Perret, J. [Parkinsonian dysarthria]. Rev. Neurol. 1995, 151, 105–112. [Google Scholar] [PubMed]
  23. Bloem, B.R.; van Vugt, J.P.; Beckley, D.J. Postural instability and falls in Parkinson’s disease. Adv. Neurol. 2001, 87, 209–223. [Google Scholar]
  24. Giladi, N.; Kao, R.; Fahn, S. Freezing phenomenon in patients with parkinsonian syndromes. Mov. Disord. 1997, 12, 302–305. [Google Scholar] [CrossRef] [PubMed]
  25. Athauda, D.; Foltynie, T. The ongoing pursuit of neuroprotective therapies in Parkinson disease. Nat. Rev. Neurol. 2015, 11, 25–40. [Google Scholar] [CrossRef] [PubMed]
  26. Tanner, C.M.; Goldman, S.M. Epidemiology of Parkinson’s disease. Neurol. Clin. 1996, 14, 317–335. [Google Scholar] [CrossRef]
  27. Bennett, D.A.; Beckett, L.A.; Murray, A.M.; Shannon, K.M.; Goetz, C.G.; Pilgrim, D.M.; Evans, D.A. Prevalence of parkinsonian signs and associated mortality in a community population of older people. N. Engl. J. Med. 1996, 334, 71–76. [Google Scholar] [CrossRef]
  28. Collier, T.J.; Kanaan, N.M.; Kordower, J.H. Ageing as a primary risk factor for Parkinson’s disease: Evidence from studies of non-human primates. Nat. Rev. Neurosci. 2011, 12, 359–366. [Google Scholar] [CrossRef]
  29. Pringsheim, T.; Jette, N.; Frolkis, A.; Steeves, T.D. The prevalence of Parkinson’s disease: A systematic review and meta-analysis. Mov. Disord. 2014, 29, 1583–1590. [Google Scholar] [CrossRef]
  30. Buchman, A.S.; Shulman, J.M.; Nag, S.; Leurgans, S.E.; Arnold, S.E.; Morris, M.C.; Schneider, J.A.; Bennett, D.A. Nigral pathology and parkinsonian signs in elders without Parkinson disease. Ann. Neurol. 2012, 71, 258–266. [Google Scholar] [CrossRef]
  31. Ownby, R.L. Neuroinflammation and cognitive aging. Curr. Psychiatry Rep. 2010, 12, 39–45. [Google Scholar] [CrossRef]
  32. Zhu, B.; Park, J.M.; Coffey, S.R.; Russo, A.; Hsu, I.U.; Wang, J.; Su, C.; Chang, R.; Lam, T.T.; Gopal, P.P.; et al. Single-cell transcriptomic and proteomic analysis of Parkinson’s disease brains. Sci. Transl. Med. 2024, 16, eabo1997. [Google Scholar] [CrossRef] [PubMed]
  33. Grimm, A.; Eckert, A. Brain aging and neurodegeneration: From a mitochondrial point of view. J. Neurochem. 2017, 143, 418–431. [Google Scholar] [CrossRef]
  34. Sherer, T.B.; Betarbet, R.; Greenamyre, J.T. Environment, mitochondria, and Parkinson’s disease. Neuroscientist 2002, 8, 192–197. [Google Scholar] [CrossRef] [PubMed]
  35. Harman, D. Aging: A theory based on free radical and radiation chemistry. J. Gerontol. 1956, 11, 298–300. [Google Scholar] [CrossRef]
  36. Zhang, Y.; Dawson, V.L.; Dawson, T.M. Oxidative stress and genetics in the pathogenesis of Parkinson’s disease. Neurobiol. Dis. 2000, 7, 240–250. [Google Scholar] [CrossRef] [PubMed]
  37. Powers, E.T.; Morimoto, R.I.; Dillin, A.; Kelly, J.W.; Balch, W.E. Biological and chemical approaches to diseases of proteostasis deficiency. Annu. Rev. Biochem. 2009, 78, 959–991. [Google Scholar] [CrossRef]
  38. Walther, D.M.; Kasturi, P.; Zheng, M.; Pinkert, S.; Vecchi, G.; Ciryam, P.; Morimoto, R.I.; Dobson, C.M.; Vendruscolo, M.; Mann, M.; et al. Widespread Proteome Remodeling and Aggregation in Aging C. elegans. Cell 2015, 161, 919–932. [Google Scholar] [CrossRef]
  39. Nago, N.; Murata, S.; Tanaka, K.; Tanahashi, N. Changes in brain proteasome dynamics associated with aging. Genes. Cells 2024, 29, 438–445. [Google Scholar] [CrossRef]
  40. Zeng, B.Y.; Medhurst, A.D.; Jackson, M.; Rose, S.; Jenner, P. Proteasomal activity in brain differs between species and brain regions and changes with age. Mech. Ageing Dev. 2005, 126, 760–766. [Google Scholar] [CrossRef]
  41. McNaught, K.S.; Belizaire, R.; Isacson, O.; Jenner, P.; Olanow, C.W. Altered proteasomal function in sporadic Parkinson’s disease. Exp. Neurol. 2003, 179, 38–46. [Google Scholar] [CrossRef] [PubMed]
  42. Chung, K.K.; Dawson, V.L.; Dawson, T.M. The role of the ubiquitin-proteasomal pathway in Parkinson’s disease and other neurodegenerative disorders. Trends Neurosci. 2001, 24, S7–S14. [Google Scholar] [CrossRef]
  43. Lipinski, M.M.; Zheng, B.; Lu, T.; Yan, Z.; Py, B.F.; Ng, A.; Xavier, R.J.; Li, C.; Yankner, B.A.; Scherzer, C.R.; et al. Genome-wide analysis reveals mechanisms modulating autophagy in normal brain aging and in Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2010, 107, 14164–14169. [Google Scholar] [CrossRef]
  44. Raz, Y.; Guerrero-Ros, I.; Maier, A.; Slagboom, P.E.; Atzmon, G.; Barzilai, N.; Macian, F. Activation-Induced Autophagy Is Preserved in CD4+ T-Cells in Familial Longevity. J. Gerontol. A Biol. Sci. Med. Sci. 2017, 72, 1201–1206. [Google Scholar] [CrossRef] [PubMed]
  45. Gan-Or, Z.; Dion, P.A.; Rouleau, G.A. Genetic perspective on the role of the autophagy-lysosome pathway in Parkinson disease. Autophagy 2015, 11, 1443–1457. [Google Scholar] [CrossRef]
  46. Xu, M.; Pirtskhalava, T.; Farr, J.N.; Weigand, B.M.; Palmer, A.K.; Weivoda, M.M.; Inman, C.L.; Ogrodnik, M.B.; Hachfeld, C.M.; Fraser, D.G.; et al. Senolytics improve physical function and increase lifespan in old age. Nat. Med. 2018, 24, 1246–1256. [Google Scholar] [CrossRef] [PubMed]
  47. Asghar, M.; Odeh, A.; Fattahi, A.J.; Henriksson, A.E.; Miglar, A.; Khosousi, S.; Svenningsson, P. Mitochondrial biogenesis, telomere length and cellular senescence in Parkinson’s disease and Lewy body dementia. Sci. Rep. 2022, 12, 17578. [Google Scholar] [CrossRef]
  48. Hornykiewicz, O. Dopamine miracle: From brain homogenate to dopamine replacement. Mov. Disord. 2002, 17, 501–508. [Google Scholar] [CrossRef]
  49. Marsden, C.D. Parkinson’s disease. Lancet 1990, 335, 948–952. [Google Scholar] [CrossRef]
  50. Brichta, L.; Greengard, P. Molecular determinants of selective dopaminergic vulnerability in Parkinson’s disease: An update. Front. Neuroanat. 2014, 8, 152. [Google Scholar] [CrossRef]
  51. Matsuda, W.; Furuta, T.; Nakamura, K.C.; Hioki, H.; Fujiyama, F.; Arai, R.; Kaneko, T. Single nigrostriatal dopaminergic neurons form widely spread and highly dense axonal arborizations in the neostriatum. J. Neurosci. 2009, 29, 444–453. [Google Scholar] [CrossRef]
  52. Moss, J.; Bolam, J.P. The Relationship between Dopaminergic Axons and Glutamatergic Synapses in the Striatum: Structural Considerations. In Dopamine Handbook; Oxford Academic: Oxford, UK, 2009. [Google Scholar]
  53. Harris, J.J.; Jolivet, R.; Attwell, D. Synaptic energy use and supply. Neuron 2012, 75, 762–777. [Google Scholar] [CrossRef] [PubMed]
  54. Pissadaki, E.K.; Bolam, J.P. The energy cost of action potential propagation in dopamine neurons: Clues to susceptibility in Parkinson’s disease. Front. Comput. Neurosci. 2013, 7, 13. [Google Scholar] [CrossRef]
  55. Surmeier, D.J.; Mercer, J.N.; Chan, C.S. Autonomous pacemakers in the basal ganglia: Who needs excitatory synapses anyway? Curr. Opin. Neurobiol. 2005, 15, 312–318. [Google Scholar] [CrossRef]
  56. Kampmann, M. Molecular and cellular mechanisms of selective vulnerability in neurodegenerative diseases. Nat. Rev. Neurosci. 2024, 25, 351–371. [Google Scholar] [CrossRef] [PubMed]
  57. Grey, M.; Dunning, C.J.; Gaspar, R.; Grey, C.; Brundin, P.; Sparr, E.; Linse, S. Acceleration of alpha-synuclein aggregation by exosomes. J. Biol. Chem. 2015, 290, 2969–2982. [Google Scholar] [CrossRef] [PubMed]
  58. Dawson, T.M. Unraveling the role of defective genes in Parkinson’s disease. Park. Relat. Disord. 2007, 13 (Suppl. 3), S248–S249. [Google Scholar] [CrossRef]
  59. Olney, J.W.; Zorumski, C.F.; Stewart, G.R.; Price, M.T.; Wang, G.J.; Labruyere, J. Excitotoxicity of L-dopa and 6-OH-dopa: Implications for Parkinson’s and Huntington’s diseases. Exp. Neurol. 1990, 108, 269–272. [Google Scholar] [CrossRef]
  60. Gonzalez-Hernandez, T.; Barroso-Chinea, P.; De La Cruz Muros, I.; Del Mar Perez-Delgado, M.; Rodriguez, M. Expression of dopamine and vesicular monoamine transporters and differential vulnerability of mesostriatal dopaminergic neurons. J. Comp. Neurol. 2004, 479, 198–215. [Google Scholar] [CrossRef]
  61. Barroso-Chinea, P.; Cruz-Muros, I.; Aymerich, M.S.; Rodriguez-Diaz, M.; Afonso-Oramas, D.; Lanciego, J.L.; Gonzalez-Hernandez, T. Striatal expression of GDNF and differential vulnerability of midbrain dopaminergic cells. Eur. J. Neurosci. 2005, 21, 1815–1827. [Google Scholar] [CrossRef]
  62. Quartu, M.; Serra, M.P.; Boi, M.; Sestu, N.; Lai, M.L.; Del Fiacco, M. Tissue distribution of neurturin, persephin and artemin in the human brainstem at fetal, neonatal and adult age. Brain Res. 2007, 1143, 102–115. [Google Scholar] [CrossRef] [PubMed]
  63. Nishio, T.; Furukawa, S.; Akiguchi, I.; Sunohara, N. Medial nigral dopamine neurons have rich neurotrophin support in humans. Neuroreport 1998, 9, 2847–2851. [Google Scholar] [CrossRef] [PubMed]
  64. Seroogy, K.B.; Lundgren, K.H.; Tran, T.M.; Guthrie, K.M.; Isackson, P.J.; Gall, C.M. Dopaminergic neurons in rat ventral midbrain express brain-derived neurotrophic factor and neurotrophin-3 mRNAs. J. Comp. Neurol. 1994, 342, 321–334. [Google Scholar] [CrossRef]
  65. Hurd, Y.L.; Pristupa, Z.B.; Herman, M.M.; Niznik, H.B.; Kleinman, J.E. The dopamine transporter and dopamine D2 receptor messenger RNAs are differentially expressed in limbic- and motor-related subpopulations of human mesencephalic neurons. Neuroscience 1994, 63, 357–362. [Google Scholar] [CrossRef]
  66. Mendez, I.; Sanchez-Pernaute, R.; Cooper, O.; Vinuela, A.; Ferrari, D.; Bjorklund, L.; Dagher, A.; Isacson, O. Cell type analysis of functional fetal dopamine cell suspension transplants in the striatum and substantia nigra of patients with Parkinson’s disease. Brain 2005, 128, 1498–1510. [Google Scholar] [CrossRef]
  67. Faucheux, B.A.; Nillesse, N.; Damier, P.; Spik, G.; Mouatt-Prigent, A.; Pierce, A.; Leveugle, B.; Kubis, N.; Hauw, J.J.; Agid, Y.; et al. Expression of lactoferrin receptors is increased in the mesencephalon of patients with Parkinson disease. Proc. Natl. Acad. Sci. USA 1995, 92, 9603–9607. [Google Scholar] [CrossRef]
  68. Kanaan, N.M.; Kordower, J.H.; Collier, T.J. Age-related changes in dopamine transporters and accumulation of 3-nitrotyrosine in rhesus monkey midbrain dopamine neurons: Relevance in selective neuronal vulnerability to degeneration. Eur. J. Neurosci. 2008, 27, 3205–3215. [Google Scholar] [CrossRef] [PubMed]
  69. Tran, J.; Anastacio, H.; Bardy, C. Genetic predispositions of Parkinson’s disease revealed in patient-derived brain cells. NPJ Park. Dis. 2020, 6, 8. [Google Scholar] [CrossRef]
  70. Brundin, P.; Melki, R. Prying into the Prion Hypothesis for Parkinson’s Disease. J. Neurosci. 2017, 37, 9808–9818. [Google Scholar] [CrossRef]
  71. Jenner, P. Oxidative mechanisms in nigral cell death in Parkinson’s disease. Mov. Disord. 1998, 13 (Suppl. 1), 24–34. [Google Scholar]
  72. Schieber, M.; Chandel, N.S. ROS function in redox signaling and oxidative stress. Curr. Biol. 2014, 24, R453–R462. [Google Scholar] [CrossRef] [PubMed]
  73. Morris, G.; Walker, A.J.; Berk, M.; Maes, M.; Puri, B.K. Cell Death Pathways: A Novel Therapeutic Approach for Neuroscientists. Mol. Neurobiol. 2018, 55, 5767–5786. [Google Scholar] [CrossRef] [PubMed]
  74. Redza-Dutordoir, M.; Averill-Bates, D.A. Activation of apoptosis signalling pathways by reactive oxygen species. Biochim. Biophys. Acta 2016, 1863, 2977–2992. [Google Scholar] [CrossRef]
  75. Bosco, D.A.; Fowler, D.M.; Zhang, Q.; Nieva, J.; Powers, E.T.; Wentworth, P., Jr.; Lerner, R.A.; Kelly, J.W. Elevated levels of oxidized cholesterol metabolites in Lewy body disease brains accelerate alpha-synuclein fibrilization. Nat. Chem. Biol. 2006, 2, 249–253. [Google Scholar] [CrossRef] [PubMed]
  76. Nakabeppu, Y.; Tsuchimoto, D.; Yamaguchi, H.; Sakumi, K. Oxidative damage in nucleic acids and Parkinson’s disease. J. Neurosci. Res. 2007, 85, 919–934. [Google Scholar] [CrossRef]
  77. Ferrer, I.; Martinez, A.; Blanco, R.; Dalfo, E.; Carmona, M. Neuropathology of sporadic Parkinson disease before the appearance of parkinsonism: Preclinical Parkinson disease. J. Neural Transm 2011, 118, 821–839. [Google Scholar] [CrossRef]
  78. Blesa, J.; Przedborski, S. Parkinson’s disease: Animal models and dopaminergic cell vulnerability. Front. Neuroanat. 2014, 8, 155. [Google Scholar] [CrossRef]
  79. Perier, C.; Bove, J.; Vila, M.; Przedborski, S. The rotenone model of Parkinson’s disease. Trends Neurosci. 2003, 26, 345–346. [Google Scholar] [CrossRef]
  80. Martin, L.J.; Pan, Y.; Price, A.C.; Sterling, W.; Copeland, N.G.; Jenkins, N.A.; Price, D.L.; Lee, M.K. Parkinson’s disease alpha-synuclein transgenic mice develop neuronal mitochondrial degeneration and cell death. J. Neurosci. 2006, 26, 41–50. [Google Scholar] [CrossRef]
  81. Hsu, L.J.; Sagara, Y.; Arroyo, A.; Rockenstein, E.; Sisk, A.; Mallory, M.; Wong, J.; Takenouchi, T.; Hashimoto, M.; Masliah, E. alpha-synuclein promotes mitochondrial deficit and oxidative stress. Am. J. Pathol. 2000, 157, 401–410. [Google Scholar] [CrossRef]
  82. Subramaniam, S.R.; Vergnes, L.; Franich, N.R.; Reue, K.; Chesselet, M.F. Region specific mitochondrial impairment in mice with widespread overexpression of alpha-synuclein. Neurobiol. Dis. 2014, 70, 204–213. [Google Scholar] [CrossRef] [PubMed]
  83. Westlund, K.N.; Denney, R.M.; Rose, R.M.; Abell, C.W. Localization of distinct monoamine oxidase A and monoamine oxidase B cell populations in human brainstem. Neuroscience 1988, 25, 439–456. [Google Scholar] [CrossRef] [PubMed]
  84. Hare, D.J.; Double, K.L. Iron and dopamine: A toxic couple. Brain 2016, 139, 1026–1035. [Google Scholar] [CrossRef]
  85. Dexter, D.T.; Wells, F.R.; Lees, A.J.; Agid, F.; Agid, Y.; Jenner, P.; Marsden, C.D. Increased nigral iron content and alterations in other metal ions occurring in brain in Parkinson’s disease. J. Neurochem. 1989, 52, 1830–1836. [Google Scholar] [CrossRef]
  86. Genoud, S.; Roberts, B.R.; Gunn, A.P.; Halliday, G.M.; Lewis, S.J.G.; Ball, H.J.; Hare, D.J.; Double, K.L. Subcellular compartmentalisation of copper, iron, manganese, and zinc in the Parkinson’s disease brain. Metallomics 2017, 9, 1447–1455. [Google Scholar] [CrossRef]
  87. Exner, N.; Lutz, A.K.; Haass, C.; Winklhofer, K.F. Mitochondrial dysfunction in Parkinson’s disease: Molecular mechanisms and pathophysiological consequences. EMBO J. 2012, 31, 3038–3062. [Google Scholar] [CrossRef] [PubMed]
  88. Harrington, K.A.; Augood, S.J.; Kingsbury, A.E.; Foster, O.J.; Emson, P.C. Dopamine transporter (Dat) and synaptic vesicle amine transporter (VMAT2) gene expression in the substantia nigra of control and Parkinson’s disease. Brain Res. Mol. Brain Res. 1996, 36, 157–162. [Google Scholar] [CrossRef]
  89. Nutt, J.G.; Carter, J.H.; Sexton, G.J. The dopamine transporter: Importance in Parkinson’s disease. Ann. Neurol. 2004, 55, 766–773. [Google Scholar] [CrossRef]
  90. Lotharius, J.; Brundin, P. Pathogenesis of Parkinson’s disease: Dopamine, vesicles and alpha-synuclein. Nat. Rev. Neurosci. 2002, 3, 932–942. [Google Scholar] [CrossRef]
  91. Sidhu, A.; Wersinger, C.; Vernier, P. alpha-Synuclein regulation of the dopaminergic transporter: A possible role in the pathogenesis of Parkinson’s disease. FEBS Lett. 2004, 565, 1–5. [Google Scholar] [CrossRef]
  92. Riederer, P.; Sofic, E.; Rausch, W.D.; Schmidt, B.; Reynolds, G.P.; Jellinger, K.; Youdim, M.B. Transition metals, ferritin, glutathione, and ascorbic acid in parkinsonian brains. J. Neurochem. 1989, 52, 515–520. [Google Scholar] [CrossRef] [PubMed]
  93. Sian, J.; Dexter, D.T.; Lees, A.J.; Daniel, S.; Agid, Y.; Javoy-Agid, F.; Jenner, P.; Marsden, C.D. Alterations in glutathione levels in Parkinson’s disease and other neurodegenerative disorders affecting basal ganglia. Ann. Neurol. 1994, 36, 348–355. [Google Scholar] [CrossRef] [PubMed]
  94. Trist, B.G.; Fifita, J.A.; Freckleton, S.E.; Hare, D.J.; Lewis, S.J.G.; Halliday, G.M.; Blair, I.P.; Double, K.L. Accumulation of dysfunctional SOD1 protein in Parkinson’s disease is not associated with mutations in the SOD1 gene. Acta Neuropathol. 2018, 135, 155–156. [Google Scholar] [CrossRef]
  95. Zeevalk, G.D.; Razmpour, R.; Bernard, L.P. Glutathione and Parkinson’s disease: Is this the elephant in the room? Biomed. Pharmacother. 2008, 62, 236–249. [Google Scholar] [CrossRef]
  96. Trist, B.G.; Davies, K.M.; Cottam, V.; Genoud, S.; Ortega, R.; Roudeau, S.; Carmona, A.; De Silva, K.; Wasinger, V.; Lewis, S.J.G.; et al. Amyotrophic lateral sclerosis-like superoxide dismutase 1 proteinopathy is associated with neuronal loss in Parkinson’s disease brain. Acta Neuropathol. 2017, 134, 113–127. [Google Scholar] [CrossRef]
  97. DelleDonne, A.; Klos, K.J.; Fujishiro, H.; Ahmed, Z.; Parisi, J.E.; Josephs, K.A.; Frigerio, R.; Burnett, M.; Wszolek, Z.K.; Uitti, R.J.; et al. Incidental Lewy body disease and preclinical Parkinson disease. Arch. Neurol. 2008, 65, 1074–1080. [Google Scholar] [CrossRef] [PubMed]
  98. Baxter, P.S.; Hardingham, G.E. Adaptive regulation of the brain’s antioxidant defences by neurons and astrocytes. Free Radic. Biol. Med. 2016, 100, 147–152. [Google Scholar] [CrossRef]
  99. Le, W.; Wu, J.; Tang, Y. Protective Microglia and Their Regulation in Parkinson’s Disease. Front. Mol. Neurosci. 2016, 9, 89. [Google Scholar] [CrossRef]
  100. Booth, H.D.E.; Hirst, W.D.; Wade-Martins, R. The Role of Astrocyte Dysfunction in Parkinson’s Disease Pathogenesis. Trends Neurosci. 2017, 40, 358–370. [Google Scholar] [CrossRef]
  101. Joe, E.H.; Choi, D.J.; An, J.; Eun, J.H.; Jou, I.; Park, S. Astrocytes, Microglia, and Parkinson’s Disease. Exp. Neurobiol. 2018, 27, 77–87. [Google Scholar] [CrossRef]
  102. Meredith, G.E.; Rademacher, D.J. MPTP mouse models of Parkinson’s disease: An update. J. Parkinsons Dis. 2011, 1, 19–33. [Google Scholar] [CrossRef]
  103. Kraytsberg, Y.; Kudryavtseva, E.; McKee, A.C.; Geula, C.; Kowall, N.W.; Khrapko, K. Mitochondrial DNA deletions are abundant and cause functional impairment in aged human substantia nigra neurons. Nat. Genet. 2006, 38, 518–520. [Google Scholar] [CrossRef]
  104. Bender, A.; Krishnan, K.J.; Morris, C.M.; Taylor, G.A.; Reeve, A.K.; Perry, R.H.; Jaros, E.; Hersheson, J.S.; Betts, J.; Klopstock, T.; et al. High levels of mitochondrial DNA deletions in substantia nigra neurons in aging and Parkinson disease. Nat. Genet. 2006, 38, 515–517. [Google Scholar] [CrossRef]
  105. Mizuno, Y.; Ohta, S.; Tanaka, M.; Takamiya, S.; Suzuki, K.; Sato, T.; Oya, H.; Ozawa, T.; Kagawa, Y. Deficiencies in complex I subunits of the respiratory chain in Parkinson’s disease. Biochem. Biophys. Res. Commun. 1989, 163, 1450–1455. [Google Scholar] [CrossRef]
  106. Schapira, A.H.; Cooper, J.M.; Dexter, D.; Clark, J.B.; Jenner, P.; Marsden, C.D. Mitochondrial complex I deficiency in Parkinson’s disease. J. Neurochem. 1990, 54, 823–827. [Google Scholar] [CrossRef]
  107. Petit, P.X.; Zamzami, N.; Vayssiere, J.L.; Mignotte, B.; Kroemer, G.; Castedo, M. Implication of mitochondria in apoptosis. Mol. Cell Biochem. 1997, 174, 185–188. [Google Scholar] [CrossRef]
  108. Singleton, A.B.; Farrer, M.; Johnson, J.; Singleton, A.; Hague, S.; Kachergus, J.; Hulihan, M.; Peuralinna, T.; Dutra, A.; Nussbaum, R.; et al. alpha-Synuclein locus triplication causes Parkinson’s disease. Science 2003, 302, 841. [Google Scholar] [CrossRef]
  109. Chartier-Harlin, M.C.; Kachergus, J.; Roumier, C.; Mouroux, V.; Douay, X.; Lincoln, S.; Levecque, C.; Larvor, L.; Andrieux, J.; Hulihan, M.; et al. Alpha-synuclein locus duplication as a cause of familial Parkinson’s disease. Lancet 2004, 364, 1167–1169. [Google Scholar] [CrossRef]
  110. Ibanez, P.; Bonnet, A.M.; Debarges, B.; Lohmann, E.; Tison, F.; Pollak, P.; Agid, Y.; Durr, A.; Brice, A. Causal relation between alpha-synuclein gene duplication and familial Parkinson’s disease. Lancet 2004, 364, 1169–1171. [Google Scholar] [CrossRef]
  111. Polymeropoulos, M.H.; Lavedan, C.; Leroy, E.; Ide, S.E.; Dehejia, A.; Dutra, A.; Pike, B.; Root, H.; Rubenstein, J.; Boyer, R.; et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science 1997, 276, 2045–2047. [Google Scholar] [CrossRef]
  112. Kamp, F.; Exner, N.; Lutz, A.K.; Wender, N.; Hegermann, J.; Brunner, B.; Nuscher, B.; Bartels, T.; Giese, A.; Beyer, K.; et al. Inhibition of mitochondrial fusion by alpha-synuclein is rescued by PINK1, Parkin and DJ-1. EMBO J. 2010, 29, 3571–3589. [Google Scholar] [CrossRef]
  113. Nakamura, K.; Nemani, V.M.; Azarbal, F.; Skibinski, G.; Levy, J.M.; Egami, K.; Munishkina, L.; Zhang, J.; Gardner, B.; Wakabayashi, J.; et al. Direct membrane association drives mitochondrial fission by the Parkinson disease-associated protein alpha-synuclein. J. Biol. Chem. 2011, 286, 20710–20726. [Google Scholar] [CrossRef]
  114. Li, L.; Nadanaciva, S.; Berger, Z.; Shen, W.; Paumier, K.; Schwartz, J.; Mou, K.; Loos, P.; Milici, A.J.; Dunlop, J.; et al. Human A53T alpha-synuclein causes reversible deficits in mitochondrial function and dynamics in primary mouse cortical neurons. PLoS ONE 2013, 8, e85815. [Google Scholar] [CrossRef]
  115. Devi, L.; Raghavendran, V.; Prabhu, B.M.; Avadhani, N.G.; Anandatheerthavarada, H.K. Mitochondrial import and accumulation of alpha-synuclein impair complex I in human dopaminergic neuronal cultures and Parkinson disease brain. J. Biol. Chem. 2008, 283, 9089–9100. [Google Scholar] [CrossRef]
  116. Rocha, E.M.; De Miranda, B.; Sanders, L.H. Alpha-synuclein: Pathology, mitochondrial dysfunction and neuroinflammation in Parkinson’s disease. Neurobiol. Dis. 2018, 109, 249–257. [Google Scholar] [CrossRef]
  117. Scarffe, L.A.; Stevens, D.A.; Dawson, V.L.; Dawson, T.M. Parkin and PINK1: Much more than mitophagy. Trends Neurosci. 2014, 37, 315–324. [Google Scholar] [CrossRef]
  118. Narendra, D.; Tanaka, A.; Suen, D.F.; Youle, R.J. Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. J. Cell Biol. 2008, 183, 795–803. [Google Scholar] [CrossRef]
  119. Pickrell, A.M.; Youle, R.J. The roles of PINK1, parkin, and mitochondrial fidelity in Parkinson’s disease. Neuron 2015, 85, 257–273. [Google Scholar] [CrossRef]
  120. Casarejos, M.J.; Menendez, J.; Solano, R.M.; Rodriguez-Navarro, J.A.; Garcia de Yebenes, J.; Mena, M.A. Susceptibility to rotenone is increased in neurons from parkin null mice and is reduced by minocycline. J. Neurochem. 2006, 97, 934–946. [Google Scholar] [CrossRef]
  121. Wang, X.; Yan, M.H.; Fujioka, H.; Liu, J.; Wilson-Delfosse, A.; Chen, S.G.; Perry, G.; Casadesus, G.; Zhu, X. LRRK2 regulates mitochondrial dynamics and function through direct interaction with DLP1. Hum. Mol. Genet. 2012, 21, 1931–1944. [Google Scholar] [CrossRef]
  122. Cleeter, M.W.; Chau, K.Y.; Gluck, C.; Mehta, A.; Hughes, D.A.; Duchen, M.; Wood, N.W.; Hardy, J.; Mark Cooper, J.; Schapira, A.H. Glucocerebrosidase inhibition causes mitochondrial dysfunction and free radical damage. Neurochem. Int. 2013, 62, 1–7. [Google Scholar] [CrossRef]
  123. Larsen, S.B.; Hanss, Z.; Kruger, R. The genetic architecture of mitochondrial dysfunction in Parkinson’s disease. Cell Tissue Res. 2018, 373, 21–37. [Google Scholar] [CrossRef]
  124. Lucchesi, M.; Biso, L.; Bonaso, M.; Longoni, B.; Buchignani, B.; Battini, R.; Santorelli, F.M.; Doccini, S.; Scarselli, M. Mitochondrial Dysfunction in Genetic and Non-Genetic Parkinson’s Disease. Int. J. Mol. Sci. 2025, 26, 4451. [Google Scholar] [CrossRef]
  125. Gautier, C.A.; Corti, O.; Brice, A. Mitochondrial dysfunctions in Parkinson’s disease. Rev. Neurol. 2014, 170, 339–343. [Google Scholar] [CrossRef]
  126. Rademacher, D.J. Potential for Therapeutic-Loaded Exosomes to Ameliorate the Pathogenic Effects of alpha-Synuclein in Parkinson’s Disease. Biomedicines 2023, 11, 1187. [Google Scholar] [CrossRef]
  127. Burre, J.; Sharma, M.; Sudhof, T.C. Cell Biology and Pathophysiology of alpha-Synuclein. Cold Spring Harb. Perspect. Med. 2018, 8, a024091. [Google Scholar] [CrossRef]
  128. Goedert, M. Alpha-synuclein and neurodegenerative diseases. Nat. Rev. Neurosci. 2001, 2, 492–501. [Google Scholar] [CrossRef]
  129. Bendor, J.T.; Logan, T.P.; Edwards, R.H. The function of alpha-synuclein. Neuron 2013, 79, 1044–1066. [Google Scholar] [CrossRef]
  130. Paciotti, S.; Bellomo, G.; Gatticchi, L.; Parnetti, L. Are We Ready for Detecting alpha-Synuclein Prone to Aggregation in Patients? The Case of “Protein-Misfolding Cyclic Amplification” and “Real-Time Quaking-Induced Conversion” as Diagnostic Tools. Front. Neurol. 2018, 9, 415. [Google Scholar] [CrossRef]
  131. Goedert, M.; Spillantini, M.G.; Del Tredici, K.; Braak, H. 100 years of Lewy pathology. Nat. Rev. Neurol. 2013, 9, 13–24. [Google Scholar] [CrossRef]
  132. Graham, S.F.; Rey, N.L.; Yilmaz, A.; Kumar, P.; Madaj, Z.; Maddens, M.; Bahado-Singh, R.O.; Becker, K.; Schulz, E.; Meyerdirk, L.K.; et al. Biochemical Profiling of the Brain and Blood Metabolome in a Mouse Model of Prodromal Parkinson’s Disease Reveals Distinct Metabolic Profiles. J. Proteome Res. 2018, 17, 2460–2469. [Google Scholar] [CrossRef]
  133. Frake, R.A.; Ricketts, T.; Menzies, F.M.; Rubinsztein, D.C. Autophagy and neurodegeneration. J. Clin. Investig. 2015, 125, 65–74. [Google Scholar] [CrossRef]
  134. Weidberg, H.; Shvets, E.; Elazar, Z. Biogenesis and cargo selectivity of autophagosomes. Annu. Rev. Biochem. 2011, 80, 125–156. [Google Scholar] [CrossRef]
  135. Cuervo, A.M.; Stefanis, L.; Fredenburg, R.; Lansbury, P.T.; Sulzer, D. Impaired degradation of mutant alpha-synuclein by chaperone-mediated autophagy. Science 2004, 305, 1292–1295. [Google Scholar] [CrossRef]
  136. Zalon, A.J.; Quiriconi, D.J.; Pitcairn, C.; Mazzulli, J.R. alpha-Synuclein: Multiple pathogenic roles in trafficking and proteostasis pathways in Parkinson’s disease. Neuroscientist 2024, 30, 612–635. [Google Scholar] [CrossRef]
  137. Ho, D.H.; Kim, H.; Nam, D.; Sim, H.; Kim, J.; Kim, H.G.; Son, I.; Seol, W. LRRK2 impairs autophagy by mediating phosphorylation of leucyl-tRNA synthetase. Cell Biochem. Funct. 2018, 36, 431–442. [Google Scholar] [CrossRef]
  138. Sidransky, E.; Nalls, M.A.; Aasly, J.O.; Aharon-Peretz, J.; Annesi, G.; Barbosa, E.R.; Bar-Shira, A.; Berg, D.; Bras, J.; Brice, A.; et al. Multicenter analysis of glucocerebrosidase mutations in Parkinson’s disease. N. Engl. J. Med. 2009, 361, 1651–1661. [Google Scholar] [CrossRef]
  139. Moors, T.E.; Paciotti, S.; Ingrassia, A.; Quadri, M.; Breedveld, G.; Tasegian, A.; Chiasserini, D.; Eusebi, P.; Duran-Pacheco, G.; Kremer, T.; et al. Characterization of Brain Lysosomal Activities in GBA-Related and Sporadic Parkinson’s Disease and Dementia with Lewy Bodies. Mol. Neurobiol. 2019, 56, 1344–1355. [Google Scholar] [CrossRef]
  140. Murphy, K.E.; Gysbers, A.M.; Abbott, S.K.; Tayebi, N.; Kim, W.S.; Sidransky, E.; Cooper, A.; Garner, B.; Halliday, G.M. Reduced glucocerebrosidase is associated with increased alpha-synuclein in sporadic Parkinson’s disease. Brain 2014, 137, 834–848. [Google Scholar] [CrossRef]
  141. Kimura, T.; Jia, J.; Kumar, S.; Choi, S.W.; Gu, Y.; Mudd, M.; Dupont, N.; Jiang, S.; Peters, R.; Farzam, F.; et al. Dedicated SNAREs and specialized TRIM cargo receptors mediate secretory autophagy. EMBO J. 2017, 36, 42–60. [Google Scholar] [CrossRef]
  142. Flavin, W.P.; Bousset, L.; Green, Z.C.; Chu, Y.; Skarpathiotis, S.; Chaney, M.J.; Kordower, J.H.; Melki, R.; Campbell, E.M. Endocytic vesicle rupture is a conserved mechanism of cellular invasion by amyloid proteins. Acta Neuropathol. 2017, 134, 629–653. [Google Scholar] [CrossRef]
  143. Burbidge, K.; Rademacher, D.J.; Mattick, J.; Zack, S.; Grillini, A.; Bousset, L.; Kwon, O.; Kubicki, K.; Simon, A.; Melki, R.; et al. LGALS3 (galectin 3) mediates an unconventional secretion of SNCA/alpha-synuclein in response to lysosomal membrane damage by the autophagic-lysosomal pathway in human midbrain dopamine neurons. Autophagy 2022, 18, 1020–1048. [Google Scholar] [CrossRef]
  144. Pickart, C.M. Mechanisms underlying ubiquitination. Annu. Rev. Biochem. 2001, 70, 503–533. [Google Scholar] [CrossRef]
  145. Voges, D.; Zwickl, P.; Baumeister, W. The 26S proteasome: A molecular machine designed for controlled proteolysis. Annu. Rev. Biochem. 1999, 68, 1015–1068. [Google Scholar] [CrossRef]
  146. Sherman, M.Y.; Goldberg, A.L. Cellular defenses against unfolded proteins: A cell biologist thinks about neurodegenerative diseases. Neuron 2001, 29, 15–32. [Google Scholar] [CrossRef]
  147. Ii, K.; Ito, H.; Tanaka, K.; Hirano, A. Immunocytochemical co-localization of the proteasome in ubiquitinated structures in neurodegenerative diseases and the elderly. J. Neuropathol. Exp. Neurol. 1997, 56, 125–131. [Google Scholar] [CrossRef]
  148. Snyder, H.; Mensah, K.; Theisler, C.; Lee, J.; Matouschek, A.; Wolozin, B. Aggregated and monomeric alpha-synuclein bind to the S6’ proteasomal protein and inhibit proteasomal function. J. Biol. Chem. 2003, 278, 11753–11759. [Google Scholar] [CrossRef]
  149. Mizuno, Y.; Hattori, N.; Mori, H.; Suzuki, T.; Tanaka, K. Parkin and Parkinson’s disease. Curr. Opin. Neurol. 2001, 14, 477–482. [Google Scholar] [CrossRef]
  150. Mori, H.; Kondo, T.; Yokochi, M.; Matsumine, H.; Nakagawa-Hattori, Y.; Miyake, T.; Suda, K.; Mizuno, Y. Pathologic and biochemical studies of juvenile parkinsonism linked to chromosome 6q. Neurology 1998, 51, 890–892. [Google Scholar] [CrossRef]
  151. Shimura, H.; Hattori, N.; Kubo, S.; Yoshikawa, M.; Kitada, T.; Matsumine, H.; Asakawa, S.; Minoshima, S.; Yamamura, Y.; Shimizu, N.; et al. Immunohistochemical and subcellular localization of Parkin protein: Absence of protein in autosomal recessive juvenile parkinsonism patients. Ann. Neurol. 1999, 45, 668–672. [Google Scholar] [CrossRef]
  152. Shimura, H.; Hattori, N.; Kubo, S.; Mizuno, Y.; Asakawa, S.; Minoshima, S.; Shimizu, N.; Iwai, K.; Chiba, T.; Tanaka, K.; et al. Familial Parkinson disease gene product, parkin, is a ubiquitin-protein ligase. Nat. Genet. 2000, 25, 302–305. [Google Scholar] [CrossRef]
  153. Leroy, E.; Boyer, R.; Auburger, G.; Leube, B.; Ulm, G.; Mezey, E.; Harta, G.; Brownstein, M.J.; Jonnalagada, S.; Chernova, T.; et al. The ubiquitin pathway in Parkinson’s disease. Nature 1998, 395, 451–452. [Google Scholar] [CrossRef]
  154. Wilkinson, K.D.; Deshpande, S.; Larsen, C.N. Comparisons of neuronal (PGP 9.5) and non-neuronal ubiquitin C-terminal hydrolases. Biochem. Soc. Trans. 1992, 20, 631–637. [Google Scholar] [CrossRef]
  155. Lowe, J.; McDermott, H.; Landon, M.; Mayer, R.J.; Wilkinson, K.D. Ubiquitin carboxyl-terminal hydrolase (PGP 9.5) is selectively present in ubiquitinated inclusion bodies characteristic of human neurodegenerative diseases. J. Pathol. 1990, 161, 153–160. [Google Scholar] [CrossRef]
  156. McNaught, K.S.; Jenner, P. Proteasomal function is impaired in substantia nigra in Parkinson’s disease. Neurosci. Lett. 2001, 297, 191–194. [Google Scholar] [CrossRef]
  157. Grace, A.A.; Bunney, B.S. Intracellular and extracellular electrophysiology of nigral dopaminergic neurons--1. Identification and characterization. Neuroscience 1983, 10, 301–315. [Google Scholar] [CrossRef]
  158. Foehring, R.C.; Zhang, X.F.; Lee, J.C.; Callaway, J.C. Endogenous calcium buffering capacity of substantia nigral dopamine neurons. J. Neurophysiol. 2009, 102, 2326–2333. [Google Scholar] [CrossRef]
  159. Zampese, E.; Wokosin, D.L.; Gonzalez-Rodriguez, P.; Guzman, J.N.; Tkatch, T.; Kondapalli, J.; Surmeier, W.C.; D’Alessandro, K.B.; De Stefani, D.; Rizzuto, R.; et al. Ca2+ channels couple spiking to mitochondrial metabolism in substantia nigra dopaminergic neurons. Sci. Adv. 2022, 8, eabp8701. [Google Scholar] [CrossRef]
  160. Nath, S.; Goodwin, J.; Engelborghs, Y.; Pountney, D.L. Raised calcium promotes alpha-synuclein aggregate formation. Mol. Cell Neurosci. 2011, 46, 516–526. [Google Scholar] [CrossRef]
  161. Goodwin, J.; Nath, S.; Engelborghs, Y.; Pountney, D.L. Raised calcium and oxidative stress cooperatively promote alpha-synuclein aggregate formation. Neurochem. Int. 2013, 62, 703–711. [Google Scholar] [CrossRef]
  162. Jain, M.K.; Bhat, R. Modulation of human alpha-synuclein aggregation by a combined effect of calcium and dopamine. Neurobiol. Dis. 2014, 63, 115–128. [Google Scholar] [CrossRef]
  163. Caraveo, G.; Auluck, P.K.; Whitesell, L.; Chung, C.Y.; Baru, V.; Mosharov, E.V.; Yan, X.; Ben-Johny, M.; Soste, M.; Picotti, P.; et al. Calcineurin determines toxic versus beneficial responses to alpha-synuclein. Proc. Natl. Acad. Sci. USA 2014, 111, E3544–E3552. [Google Scholar] [CrossRef]
  164. Gomez-Sintes, R.; Ledesma, M.D.; Boya, P. Lysosomal cell death mechanisms in aging. Ageing Res. Rev. 2016, 32, 150–168. [Google Scholar] [CrossRef]
  165. Yamada, T.; McGeer, P.L.; Baimbridge, K.G.; McGeer, E.G. Relative sparing in Parkinson’s disease of substantia nigra dopamine neurons containing calbindin-D28K. Brain Res. 1990, 526, 303–307. [Google Scholar] [CrossRef]
  166. Epstein, B.J.; Vogel, K.; Palmer, B.F. Dihydropyridine calcium channel antagonists in the management of hypertension. Drugs 2007, 67, 1309–1327. [Google Scholar] [CrossRef]
  167. Becker, C.; Jick, S.S.; Meier, C.R. Use of antihypertensives and the risk of Parkinson disease. Neurology 2008, 70, 1438–1444. [Google Scholar] [CrossRef]
  168. Qu, Y.; Li, J.; Qin, Q.; Wang, D.; Zhao, J.; An, K.; Mao, Z.; Min, Z.; Xiong, Y.; Li, J.; et al. A systematic review and meta-analysis of inflammatory biomarkers in Parkinson’s disease. NPJ Park. Dis. 2023, 9, 18. [Google Scholar] [CrossRef]
  169. Drouin-Ouellet, J.; St-Amour, I.; Saint-Pierre, M.; Lamontagne-Proulx, J.; Kriz, J.; Barker, R.A.; Cicchetti, F. Toll-like receptor expression in the blood and brain of patients and a mouse model of Parkinson’s disease. Int. J. Neuropsychopharmacol. 2014, 18. [Google Scholar] [CrossRef]
  170. Conte, C.; Ingrassia, A.; Breve, J.; Bol, J.J.; Timmermans-Huisman, E.; van Dam, A.M.; Beccari, T.; van de Berg, W.D.J. Toll-like Receptor 4 Is Upregulated in Parkinson’s Disease Patients and Co-Localizes with pSer129alphaSyn: A Possible Link with the Pathology. Cells 2023, 12, 1368. [Google Scholar] [CrossRef]
  171. Grozdanov, V.; Bliederhaeuser, C.; Ruf, W.P.; Roth, V.; Fundel-Clemens, K.; Zondler, L.; Brenner, D.; Martin-Villalba, A.; Hengerer, B.; Kassubek, J.; et al. Inflammatory dysregulation of blood monocytes in Parkinson’s disease patients. Acta Neuropathol. 2014, 128, 651–663. [Google Scholar] [CrossRef]
  172. Kosloski, L.M.; Kosmacek, E.A.; Olson, K.E.; Mosley, R.L.; Gendelman, H.E. GM-CSF induces neuroprotective and anti-inflammatory responses in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine intoxicated mice. J. Neuroimmunol. 2013, 265, 1–10. [Google Scholar] [CrossRef]
  173. Saunders, J.A.; Estes, K.A.; Kosloski, L.M.; Allen, H.E.; Dempsey, K.M.; Torres-Russotto, D.R.; Meza, J.L.; Santamaria, P.M.; Bertoni, J.M.; Murman, D.L.; et al. CD4+ regulatory and effector/memory T cell subsets profile motor dysfunction in Parkinson’s disease. J. Neuroimmune Pharmacol. 2012, 7, 927–938. [Google Scholar] [CrossRef]
  174. Kustrimovic, N.; Comi, C.; Magistrelli, L.; Rasini, E.; Legnaro, M.; Bombelli, R.; Aleksic, I.; Blandini, F.; Minafra, B.; Riboldazzi, G.; et al. Parkinson’s disease patients have a complex phenotypic and functional Th1 bias: Cross-sectional studies of CD4+ Th1/Th2/T17 and Treg in drug-naive and drug-treated patients. J. Neuroinflamm. 2018, 15, 205. [Google Scholar] [CrossRef]
  175. Holmans, P.; Moskvina, V.; Jones, L.; Sharma, M.; International Parkinson’s Disease Genomics Consortium (IPDGC); Vedernikov, A.; Buchel, F.; Saad, M.; Bras, J.M.; Bettella, F.; et al. A pathway-based analysis provides additional support for an immune-related genetic susceptibility to Parkinson’s disease. Hum. Mol. Genet. 2013, 22, 1039–1049. [Google Scholar] [CrossRef]
  176. Saiki, M.; Baker, A.; Williams-Gray, C.H.; Foltynie, T.; Goodman, R.S.; Taylor, C.J.; Compston, D.A.; Barker, R.A.; Sawcer, S.J.; Goris, A. Association of the human leucocyte antigen region with susceptibility to Parkinson’s disease. J. Neurol. Neurosurg. Psychiatry 2010, 81, 890–891. [Google Scholar] [CrossRef]
  177. McGeer, P.L.; Itagaki, S.; Boyes, B.E.; McGeer, E.G. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology 1988, 38, 1285–1291. [Google Scholar] [CrossRef]
  178. Benveniste, E.N. Inflammatory cytokines within the central nervous system: Sources, function, and mechanism of action. Am. J. Physiol. 1992, 263, C1–C16. [Google Scholar] [CrossRef]
  179. Hirsch, E.C.; Hunot, S.; Damier, P.; Faucheux, B. Glial cells and inflammation in Parkinson’s disease: A role in neurodegeneration? Ann. Neurol. 1998, 44, S115–S120. [Google Scholar] [CrossRef]
  180. Mogi, M.; Harada, M.; Kondo, T.; Riederer, P.; Inagaki, H.; Minami, M.; Nagatsu, T. Interleukin-1 beta, interleukin-6, epidermal growth factor and transforming growth factor-alpha are elevated in the brain from parkinsonian patients. Neurosci. Lett. 1994, 180, 147–150. [Google Scholar] [CrossRef]
  181. Boka, G.; Anglade, P.; Wallach, D.; Javoy-Agid, F.; Agid, Y.; Hirsch, E.C. Immunocytochemical analysis of tumor necrosis factor and its receptors in Parkinson’s disease. Neurosci. Lett. 1994, 172, 151–154. [Google Scholar] [CrossRef]
  182. Hunot, S.; Brugg, B.; Ricard, D.; Michel, P.P.; Muriel, M.P.; Ruberg, M.; Faucheux, B.A.; Agid, Y.; Hirsch, E.C. Nuclear translocation of NF-kappaB is increased in dopaminergic neurons of patients with parkinson disease. Proc. Natl. Acad. Sci. USA 1997, 94, 7531–7536. [Google Scholar] [CrossRef]
  183. Bogale, T.A.; Faustini, G.; Longhena, F.; Mitola, S.; Pizzi, M.; Bellucci, A. Alpha-Synuclein in the Regulation of Brain Endothelial and Perivascular Cells: Gaps and Future Perspectives. Front. Immunol. 2021, 12, 611761. [Google Scholar] [CrossRef]
  184. Pober, J.S.; Sessa, W.C. Evolving functions of endothelial cells in inflammation. Nat. Rev. Immunol. 2007, 7, 803–815. [Google Scholar] [CrossRef]
  185. Brochard, V.; Combadiere, B.; Prigent, A.; Laouar, Y.; Perrin, A.; Beray-Berthat, V.; Bonduelle, O.; Alvarez-Fischer, D.; Callebert, J.; Launay, J.M.; et al. Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. J. Clin. Investig. 2009, 119, 182–192. [Google Scholar] [CrossRef]
  186. Miklossy, J.; Doudet, D.D.; Schwab, C.; Yu, S.; McGeer, E.G.; McGeer, P.L. Role of ICAM-1 in persisting inflammation in Parkinson disease and MPTP monkeys. Exp. Neurol. 2006, 197, 275–283. [Google Scholar] [CrossRef]
  187. Liu, S.Y.; Qiao, H.W.; Song, T.B.; Liu, X.L.; Yao, Y.X.; Zhao, C.S.; Barret, O.; Xu, S.L.; Cai, Y.N.; Tamagnan, G.D.; et al. Brain microglia activation and peripheral adaptive immunity in Parkinson’s disease: A multimodal PET study. J. Neuroinflamm. 2022, 19, 209. [Google Scholar] [CrossRef]
  188. Haruwaka, K.; Ikegami, A.; Tachibana, Y.; Ohno, N.; Konishi, H.; Hashimoto, A.; Matsumoto, M.; Kato, D.; Ono, R.; Kiyama, H.; et al. Dual microglia effects on blood brain barrier permeability induced by systemic inflammation. Nat. Commun. 2019, 10, 5816. [Google Scholar] [CrossRef]
  189. Birch, J.; Gil, J. Senescence and the SASP: Many therapeutic avenues. Genes. Dev. 2020, 34, 1565–1576. [Google Scholar] [CrossRef]
  190. Coppé, J.P.; Desprez, P.Y.; Krtolica, A.; Campisi, J. The senescence-associated secretory phenotype: The dark side of tumor suppression. Annu. Rev. Pathol. 2010, 5, 99–118. [Google Scholar] [CrossRef]
  191. Kuilman, T.; Michaloglou, C.; Vredeveld, L.C.; Douma, S.; van Doorn, R.; Desmet, C.J.; Aarden, L.A.; Mooi, W.J.; Peeper, D.S. Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell 2008, 133, 1019–1031. [Google Scholar] [CrossRef]
  192. Kang, S.M.; Jung, H.S.; Kwon, M.J.; Lee, S.H.; Park, J.H. Testosterone Protects Pancreatic β-cells from Apoptosis and Stress-Induced Accelerated Senescence. World J. Mens. Health 2021, 39, 724–732. [Google Scholar] [CrossRef]
  193. Xue, L.; Wu, J.; Zheng, W.; Wang, P.; Li, J.; Zhang, Z.; Tong, T. Sp1 is involved in the transcriptional activation of p16INK4 by p21Waf1 in HeLa cells. FEBS Lett. 2004, 564, 199–204. [Google Scholar] [CrossRef]
  194. McHugh, D.; Gil, J. Senescence and aging: Causes, consequences, and therapeutic avenues. J. Cell Biol. 2018, 217, 65–77. [Google Scholar] [CrossRef]
  195. Acosta, J.C.; O’Loghlen, A.; Banito, A.; Guijarro, M.V.; Augert, A.; Raguz, S.; Fumagalli, M.; Da Costa, M.; Brown, C.; Popov, N.; et al. Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell 2008, 133, 1006–1018. [Google Scholar] [CrossRef]
  196. Bartkova, J.; Rezaei, N.; Liontos, M.; Karakaidos, P.; Kletsas, D.; Issaeva, N.; Vassiliou, L.V.; Kolettas, E.; Niforou, K.; Zoumpourlis, V.C.; et al. Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature 2006, 444, 633–637. [Google Scholar] [CrossRef]
  197. Deschenes-Simard, X.; Gaumont-Leclerc, M.F.; Bourdeau, V.; Lessard, F.; Moiseeva, O.; Forest, V.; Igelmann, S.; Mallette, F.A.; Saba-El-Leil, M.K.; Meloche, S.; et al. Tumor suppressor activity of the ERK/MAPK pathway by promoting selective protein degradation. Genes. Dev. 2013, 27, 900–915. [Google Scholar] [CrossRef]
  198. Eggert, T.; Wolter, K.; Ji, J.; Ma, C.; Yevsa, T.; Klotz, S.; Medina-Echeverz, J.; Longerich, T.; Forgues, M.; Reisinger, F.; et al. Distinct Functions of Senescence-Associated Immune Responses in Liver Tumor Surveillance and Tumor Progression. Cancer Cell 2016, 30, 533–547. [Google Scholar] [CrossRef]
  199. Fujita, K.; Mondal, A.M.; Horikawa, I.; Nguyen, G.H.; Kumamoto, K.; Sohn, J.J.; Bowman, E.D.; Mathe, E.A.; Schetter, A.J.; Pine, S.R.; et al. p53 isoforms Delta133p53 and p53beta are endogenous regulators of replicative cellular senescence. Nat. Cell Biol. 2009, 11, 1135–1142. [Google Scholar] [CrossRef]
  200. Haugstetter, A.M.; Loddenkemper, C.; Lenze, D.; Grone, J.; Standfuss, C.; Petersen, I.; Dorken, B.; Schmitt, C.A. Cellular senescence predicts treatment outcome in metastasised colorectal cancer. Br. J. Cancer 2010, 103, 505–509. [Google Scholar] [CrossRef]
  201. Vernier, M.; Bourdeau, V.; Gaumont-Leclerc, M.F.; Moiseeva, O.; Begin, V.; Saad, F.; Mes-Masson, A.M.; Ferbeyre, G. Regulation of E2Fs and senescence by PML nuclear bodies. Genes. Dev. 2011, 25, 41–50. [Google Scholar] [CrossRef]
  202. Ancrile, B.; Lim, K.H.; Counter, C.M. Oncogenic Ras-induced secretion of IL6 is required for tumorigenesis. Genes. Dev. 2007, 21, 1714–1719. [Google Scholar] [CrossRef]
  203. Ortiz-Montero, P.; Londono-Vallejo, A.; Vernot, J.P. Senescence-associated IL-6 and IL-8 cytokines induce a self- and cross-reinforced senescence/inflammatory milieu strengthening tumorigenic capabilities in the MCF-7 breast cancer cell line. Cell Commun. Signal 2017, 15, 17. [Google Scholar] [CrossRef]
  204. Jun, J.I.; Lau, L.F. Cellular senescence controls fibrosis in wound healing. Aging 2010, 2, 627–631. [Google Scholar] [CrossRef]
  205. Munoz-Espin, D.; Canamero, M.; Maraver, A.; Gomez-Lopez, G.; Contreras, J.; Murillo-Cuesta, S.; Rodriguez-Baeza, A.; Varela-Nieto, I.; Ruberte, J.; Collado, M.; et al. Programmed cell senescence during mammalian embryonic development. Cell 2013, 155, 1104–1118. [Google Scholar] [CrossRef]
  206. Baker, D.J.; Wijshake, T.; Tchkonia, T.; LeBrasseur, N.K.; Childs, B.G.; van de Sluis, B.; Kirkland, J.L.; van Deursen, J.M. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 2011, 479, 232–236. [Google Scholar] [CrossRef]
  207. Chinta, S.J.; Woods, G.; Demaria, M.; Rane, A.; Zou, Y.; McQuade, A.; Rajagopalan, S.; Limbad, C.; Madden, D.T.; Campisi, J.; et al. Cellular Senescence Is Induced by the Environmental Neurotoxin Paraquat and Contributes to Neuropathology Linked to Parkinson’s Disease. Cell Rep. 2018, 22, 930–940. [Google Scholar] [CrossRef]
  208. van Dijk, K.D.; Persichetti, E.; Chiasserini, D.; Eusebi, P.; Beccari, T.; Calabresi, P.; Berendse, H.W.; Parnetti, L.; van de Berg, W.D. Changes in endolysosomal enzyme activities in cerebrospinal fluid of patients with Parkinson’s disease. Mov. Disord. 2013, 28, 747–754. [Google Scholar] [CrossRef]
  209. Blum-Degen, D.; Muller, T.; Kuhn, W.; Gerlach, M.; Przuntek, H.; Riederer, P. Interleukin-1 beta and interleukin-6 are elevated in the cerebrospinal fluid of Alzheimer’s and de novo Parkinson’s disease patients. Neurosci. Lett. 1995, 202, 17–20. [Google Scholar] [CrossRef]
  210. Campbell, I.L.; Abraham, C.R.; Masliah, E.; Kemper, P.; Inglis, J.D.; Oldstone, M.B.; Mucke, L. Neurologic disease induced in transgenic mice by cerebral overexpression of interleukin 6. Proc. Natl. Acad. Sci. USA 1993, 90, 10061–10065. [Google Scholar] [CrossRef]
  211. Green, H.F.; Khosousi, S.; Svenningsson, P. Plasma IL-6 and IL-17A Correlate with Severity of Motor and Non-Motor Symptoms in Parkinson’s Disease. J. Park. Dis. 2019, 9, 705–709. [Google Scholar] [CrossRef]
  212. Mogi, M.; Harada, M.; Riederer, P.; Narabayashi, H.; Fujita, K.; Nagatsu, T. Tumor necrosis factor-α (TNF-α) increases both in the brain and in the cerebrospinal fluid from parkinsonian patients. Neurosci. Lett. 1994, 165, 208–210. [Google Scholar] [CrossRef]
  213. Choi, D.H.; Kim, Y.J.; Kim, Y.G.; Joh, T.H.; Beal, M.F.; Kim, Y.S. Role of matrix metalloproteinase 3-mediated alpha-synuclein cleavage in dopaminergic cell death. J. Biol. Chem. 2011, 286, 14168–14177. [Google Scholar] [CrossRef]
  214. Bae, E.J.; Choi, M.; Kim, J.T.; Kim, D.K.; Jung, M.K.; Kim, C.; Kim, T.K.; Lee, J.S.; Jung, B.C.; Shin, S.J.; et al. TNF-α promotes alpha-synuclein propagation through stimulation of senescence-associated lysosomal exocytosis. Exp. Mol. Med. 2022, 54, 788–800. [Google Scholar] [CrossRef]
  215. Wang, Y.; Kuca, K.; You, L.; Nepovimova, E.; Heger, Z.; Valko, M.; Adam, V.; Wu, Q.; Jomova, K. The role of cellular senescence in neurodegenerative diseases. Arch. Toxicol. 2024, 98, 2393–2408. [Google Scholar] [CrossRef]
  216. Gao, L.; Zheng, W.G.; Wu, X.K.; Du, G.H.; Qin, X.M. Baicalein Delays H2O2-Induced Astrocytic Senescence through Inhibition of Senescence-Associated Secretory Phenotype (SASP), Suppression of JAK2/STAT1/NF-κB Pathway, and Regulation of Leucine Metabolism. ACS Chem. Neurosci. 2021, 12, 2320–2335. [Google Scholar] [CrossRef]
  217. Han, X.; Zhang, T.; Liu, H.; Mi, Y.; Gou, X. Astrocyte Senescence and Alzheimer’s Disease: A Review. Front. Aging Neurosci. 2020, 12, 148. [Google Scholar] [CrossRef]
  218. Simmnacher, K.; Krach, F.; Schneider, Y.; Alecu, J.E.; Mautner, L.; Klein, P.; Roybon, L.; Prots, I.; Xiang, W.; Winner, B. Unique signatures of stress-induced senescent human astrocytes. Exp. Neurol. 2020, 334, 113466. [Google Scholar] [CrossRef]
  219. Kim, S.; Ock, J.; Kim, A.K.; Lee, H.W.; Cho, J.Y.; Kim, D.R.; Park, J.Y.; Suk, K. Neurotoxicity of microglial cathepsin D revealed by secretome analysis. J. Neurochem. 2007, 103, 2640–2650. [Google Scholar] [CrossRef]
  220. Chung, Y.C.; Kim, Y.S.; Bok, E.; Yune, T.Y.; Maeng, S.; Jin, B.K. MMP-3 contributes to nigrostriatal dopaminergic neuronal loss, BBB damage, and neuroinflammation in an MPTP mouse model of Parkinson’s disease. Mediat. Inflamm. 2013, 2013, 370526. [Google Scholar] [CrossRef]
  221. Gray, M.T.; Woulfe, J.M. Striatal blood-brain barrier permeability in Parkinson’s disease. J. Cereb. Blood Flow. Metab. 2015, 35, 747–750. [Google Scholar] [CrossRef]
  222. Cicchetti, F.; Brownell, A.L.; Williams, K.; Chen, Y.I.; Livni, E.; Isacson, O. Neuroinflammation of the nigrostriatal pathway during progressive 6-OHDA dopamine degeneration in rats monitored by immunohistochemistry and PET imaging. Eur. J. Neurosci. 2002, 15, 991–998. [Google Scholar] [CrossRef]
  223. Kim, W.G.; Mohney, R.P.; Wilson, B.; Jeohn, G.H.; Liu, B.; Hong, J.S. Regional difference in susceptibility to lipopolysaccharide-induced neurotoxicity in the rat brain: Role of microglia. J. Neurosci. 2000, 20, 6309–6316. [Google Scholar] [CrossRef]
  224. Qian, L.; Flood, P.M. Microglial cells and Parkinson’s disease. Immunol. Res. 2008, 41, 155–164. [Google Scholar] [CrossRef]
  225. Calabrese, V.; Santoro, A.; Monti, D.; Crupi, R.; Di Paola, R.; Latteri, S.; Cuzzocrea, S.; Zappia, M.; Giordano, J.; Calabrese, E.J.; et al. Aging and Parkinson’s Disease: Inflammaging, neuroinflammation and biological remodeling as key factors in pathogenesis. Free Radic. Biol. Med. 2018, 115, 80–91. [Google Scholar] [CrossRef]
  226. Kuilman, T.; Peeper, D.S. Senescence-messaging secretome: SMS-ing cellular stress. Nat. Rev. Cancer 2009, 9, 81–94. [Google Scholar] [CrossRef]
  227. Riessland, M.; Kolisnyk, B.; Kim, T.W.; Cheng, J.; Ni, J.; Pearson, J.A.; Park, E.J.; Dam, K.; Acehan, D.; Ramos-Espiritu, L.S.; et al. Loss of SATB1 Induces p21-Dependent Cellular Senescence in Post-mitotic Dopaminergic Neurons. Cell Stem Cell 2019, 25, 514–530.e8. [Google Scholar] [CrossRef]
  228. Shen, Q.Q.; Jv, X.H.; Ma, X.Z.; Li, C.; Liu, L.; Jia, W.T.; Qu, L.; Chen, L.L.; Xie, J.X. Cell senescence induced by toxic interaction between alpha-synuclein and iron precedes nigral dopaminergic neuron loss in a mouse model of Parkinson’s disease. Acta Pharmacol. Sin. 2024, 45, 268–281. [Google Scholar] [CrossRef]
  229. Sturmlechner, I.; Zhang, C.; Sine, C.C.; van Deursen, E.J.; Jeganathan, K.B.; Hamada, N.; Grasic, J.; Friedman, D.; Stutchman, J.T.; Can, I.; et al. p21 produces a bioactive secretome that places stressed cells under immunosurveillance. Science 2021, 374, eabb3420. [Google Scholar] [CrossRef]
  230. Dimri, G.P.; Lee, X.; Basile, G.; Acosta, M.; Scott, G.; Roskelley, C.; Medrano, E.E.; Linskens, M.; Rubelj, I.; Pereira-Smith, O.; et al. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc. Natl. Acad. Sci. USA 1995, 92, 9363–9367. [Google Scholar] [CrossRef]
  231. Hampel, B.; Malisan, F.; Niederegger, H.; Testi, R.; Jansen-Durr, P. Differential regulation of apoptotic cell death in senescent human cells. Exp. Gerontol. 2004, 39, 1713–1721. [Google Scholar] [CrossRef]
  232. Sharpless, N.E.; Sherr, C.J. Forging a signature of in vivo senescence. Nat. Rev. Cancer 2015, 15, 397–408. [Google Scholar] [CrossRef]
  233. Hayflick, L.; Moorhead, P.S. The serial cultivation of human diploid cell strains. Exp. Cell Res. 1961, 25, 585–621. [Google Scholar] [CrossRef]
  234. Sanokawa-Akakura, R.; Akakura, S.; Ostrakhovitch, E.A.; Tabibzadeh, S. Replicative senescence is distinguishable from DNA damage-induced senescence by increased methylation of promoter of rDNA and reduced expression of rRNA. Mech. Ageing Dev. 2019, 183, 111149. [Google Scholar] [CrossRef]
  235. Allsopp, R.C.; Chang, E.; Kashefi-Aazam, M.; Rogaev, E.I.; Piatyszek, M.A.; Shay, J.W.; Harley, C.B. Telomere shortening is associated with cell division in vitro and in vivo. Exp. Cell Res. 1995, 220, 194–200. [Google Scholar] [CrossRef]
  236. Gonzalez-Suarez, E.; Geserick, C.; Flores, J.M.; Blasco, M.A. Antagonistic effects of telomerase on cancer and aging in K5-mTert transgenic mice. Oncogene 2005, 24, 2256–2270. [Google Scholar] [CrossRef]
  237. d’Adda di Fagagna, F.; Reaper, P.M.; Clay-Farrace, L.; Fiegler, H.; Carr, P.; Von Zglinicki, T.; Saretzki, G.; Carter, N.P.; Jackson, S.P. A DNA damage checkpoint response in telomere-initiated senescence. Nature 2003, 426, 194–198. [Google Scholar] [CrossRef]
  238. Ray, P.D.; Huang, B.W.; Tsuji, Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell Signal 2012, 24, 981–990. [Google Scholar] [CrossRef]
  239. Bhayadia, R.; Schmidt, B.M.; Melk, A.; Homme, M. Senescence-Induced Oxidative Stress Causes Endothelial Dysfunction. J. Gerontol. A Biol. Sci. Med. Sci. 2016, 71, 161–169. [Google Scholar] [CrossRef]
  240. Kumari, R.; Jat, P. Mechanisms of Cellular Senescence: Cell Cycle Arrest and Senescence Associated Secretory Phenotype. Front. Cell Dev. Biol. 2021, 9, 645593. [Google Scholar] [CrossRef]
  241. Da Silva-Alvarez, S.; Picallos-Rabina, P.; Antelo-Iglesias, L.; Triana-Martinez, F.; Barreiro-Iglesias, A.; Sanchez, L.; Collado, M. The development of cell senescence. Exp. Gerontol. 2019, 128, 110742. [Google Scholar] [CrossRef]
  242. Song, T.; Gu, Y.; Hui, W.; Yang, X.; Liu, Y.; Chen, X. Oxygen-Glucose Deprivation Promoted Fibroblast Senescence and Collagen Expression via IL11. Int. J. Mol. Sci. 2022, 23, 12090. [Google Scholar] [CrossRef] [PubMed]
  243. Bielak-Zmijewska, A.; Mosieniak, G.; Sikora, E. Is DNA damage indispensable for stress-induced senescence? Mech. Ageing Dev. 2018, 170, 13–21. [Google Scholar] [CrossRef] [PubMed]
  244. Bratic, A.; Larsson, N.G. The role of mitochondria in aging. J. Clin. Investig. 2013, 123, 951–957. [Google Scholar] [CrossRef] [PubMed]
  245. Kubbutat, M.H.; Jones, S.N.; Vousden, K.H. Regulation of p53 stability by Mdm2. Nature 1997, 387, 299–303. [Google Scholar] [CrossRef]
  246. Tibbetts, R.S.; Brumbaugh, K.M.; Williams, J.M.; Sarkaria, J.N.; Cliby, W.A.; Shieh, S.Y.; Taya, Y.; Prives, C.; Abraham, R.T. A role for ATR in the DNA damage-induced phosphorylation of p53. Genes. Dev. 1999, 13, 152–157. [Google Scholar] [CrossRef]
  247. Bieging-Rolett, K.T.; Johnson, T.M.; Brady, C.A.; Beaudry, V.G.; Pathak, N.; Han, S.; Attardi, L.D. p19(Arf) is required for the cellular response to chronic DNA damage. Oncogene 2016, 35, 4414–4421. [Google Scholar] [CrossRef]
  248. Ou, H.L.; Schumacher, B. DNA damage responses and p53 in the aging process. Blood 2018, 131, 488–495. [Google Scholar] [CrossRef]
  249. Beausejour, C.M.; Krtolica, A.; Galimi, F.; Narita, M.; Lowe, S.W.; Yaswen, P.; Campisi, J. Reversal of human cellular senescence: Roles of the p53 and p16 pathways. EMBO J. 2003, 22, 4212–4222. [Google Scholar] [CrossRef]
  250. Cox, L.S. Multiple pathways control cell growth and transformation: Overlapping and independent activities of p53 and p21Cip1/WAF1/Sdi1. J. Pathol. 1997, 183, 134–140. [Google Scholar] [CrossRef]
  251. Macleod, K.F.; Sherry, N.; Hannon, G.; Beach, D.; Tokino, T.; Kinzler, K.; Vogelstein, B.; Jacks, T. p53-dependent and independent expression of p21 during cell growth, differentiation, and DNA damage. Genes. Dev. 1995, 9, 935–944. [Google Scholar] [CrossRef]
  252. Roger, L.; Tomas, F.; Gire, V. Mechanisms and Regulation of Cellular Senescence. Int. J. Mol. Sci. 2021, 22, 13173. [Google Scholar] [CrossRef] [PubMed]
  253. Serra, S.; Chetty, R. p16. J. Clin. Pathol. 2018, 71, 853–858. [Google Scholar] [CrossRef] [PubMed]
  254. Herranz, N.; Gil, J. Mechanisms and functions of cellular senescence. J. Clin. Investig. 2018, 128, 1238–1246. [Google Scholar] [CrossRef]
  255. Grison, A.; Atanasoski, S. Cyclins, Cyclin-Dependent Kinases, and Cyclin-Dependent Kinase Inhibitors in the Mouse Nervous System. Mol. Neurobiol. 2020, 57, 3206–3218. [Google Scholar] [CrossRef]
  256. Yoon, M.H.; Kang, S.M.; Lee, S.J.; Woo, T.G.; Oh, A.Y.; Park, S.; Ha, N.C.; Park, B.J. p53 induces senescence through Lamin A/C stabilization-mediated nuclear deformation. Cell Death Dis. 2019, 10, 107. [Google Scholar] [CrossRef] [PubMed]
  257. Al-Khalaf, H.H.; Aboussekhra, A. p16(INK4A) positively regulates p21WAF1 expression by suppressing AUF1-dependent mRNA decay. PLoS ONE 2013, 8, e70133. [Google Scholar] [CrossRef]
  258. Al-Khalaf, H.H.; Aboussekhra, A. p16 Controls p53 Protein Expression Through miR-dependent Destabilization of MDM2. Mol. Cancer Res. 2018, 16, 1299–1308. [Google Scholar] [CrossRef]
  259. Haigis, M.C.; Guarente, L.P. Mammalian sirtuins--emerging roles in physiology, aging, and calorie restriction. Genes. Dev. 2006, 20, 2913–2921. [Google Scholar] [CrossRef]
  260. Michan, S.; Sinclair, D. Sirtuins in mammals: Insights into their biological function. Biochem. J. 2007, 404, 1–13. [Google Scholar] [CrossRef]
  261. Yamamoto, H.; Schoonjans, K.; Auwerx, J. Sirtuin functions in health and disease. Mol. Endocrinol. 2007, 21, 1745–1755. [Google Scholar] [CrossRef]
  262. Chen, H.; Liu, X.; Zhu, W.; Chen, H.; Hu, X.; Jiang, Z.; Xu, Y.; Wang, L.; Zhou, Y.; Chen, P.; et al. SIRT1 ameliorates age-related senescence of mesenchymal stem cells via modulating telomere shelterin. Front. Aging Neurosci. 2014, 6, 103. [Google Scholar] [CrossRef] [PubMed]
  263. Hayakawa, T.; Iwai, M.; Aoki, S.; Takimoto, K.; Maruyama, M.; Maruyama, W.; Motoyama, N. SIRT1 suppresses the senescence-associated secretory phenotype through epigenetic gene regulation. PLoS ONE 2015, 10, e0116480. [Google Scholar] [CrossRef] [PubMed]
  264. Xu, C.; Wang, L.; Fozouni, P.; Evjen, G.; Chandra, V.; Jiang, J.; Lu, C.; Nicastri, M.; Bretz, C.; Winkler, J.D.; et al. SIRT1 is downregulated by autophagy in senescence and ageing. Nat. Cell Biol. 2020, 22, 1170–1179. [Google Scholar] [CrossRef] [PubMed]
  265. Lee, S.H.; Lee, J.H.; Lee, H.Y.; Min, K.J. Sirtuin signaling in cellular senescence and aging. BMB Rep. 2019, 52, 24–34. [Google Scholar] [CrossRef]
  266. Sasca, D.; Hahnel, P.S.; Szybinski, J.; Khawaja, K.; Kriege, O.; Pante, S.V.; Bullinger, L.; Strand, S.; Strand, D.; Theobald, M.; et al. SIRT1 prevents genotoxic stress-induced p53 activation in acute myeloid leukemia. Blood 2014, 124, 121–133. [Google Scholar] [CrossRef]
  267. Yuan, F.; Liu, L.; Lei, Y.; Tang, P. p53 inhibits the upregulation of sirtuin 1 expression induced by c-Myc. Oncol. Lett. 2017, 14, 4396–4402. [Google Scholar] [CrossRef]
  268. Lukasova, E.; Kovarik, A.; Kozubek, S. Consequences of Lamin B1 and Lamin B Receptor Downregulation in Senescence. Cells 2018, 7, 11. [Google Scholar] [CrossRef] [PubMed]
  269. Frediani, E.; Scavone, F.; Laurenzana, A.; Chillà, A.; Tortora, K.; Cimmino, I.; Leri, M.; Bucciantini, M.; Mangoni, M.; Fibbi, G.; et al. Olive phenols preserve lamin B1 expression reducing cGAS/STING/NFκB-mediated SASP in ionizing radiation-induced senescence. J. Cell Mol. Med. 2022, 26, 2337–2350. [Google Scholar] [CrossRef]
  270. Shah, P.P.; Donahue, G.; Otte, G.L.; Capell, B.C.; Nelson, D.M.; Cao, K.; Aggarwala, V.; Cruickshanks, H.A.; Rai, T.S.; McBryan, T.; et al. Lamin B1 depletion in senescent cells triggers large-scale changes in gene expression and the chromatin landscape. Genes. Dev. 2013, 27, 1787–1799. [Google Scholar] [CrossRef]
  271. Narita, M.; Nũnez, S.; Heard, E.; Narita, M.; Lin, A.W.; Hearn, S.A.; Spector, D.L.; Hannon, G.J.; Lowe, S.W. Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell 2003, 113, 703–716. [Google Scholar] [CrossRef]
  272. Ngai, Y.T.; Lau, D.; Mittal, P.; Hoffmann, P. Mini Review: Highlight of Recent Advances and Applications of MALDI Mass Spectrometry Imaging in 2024. Anal. Sci. Adv. 2025, 6, e70016. [Google Scholar] [CrossRef] [PubMed]
  273. Sun, Y.; Yu, N.; Zhang, J.; Yang, B. Advances in Microfluidic Single-Cell RNA Sequencing and Spatial Transcriptomics. Micromachines 2025, 16, 426. [Google Scholar] [CrossRef] [PubMed]
  274. Dehkordi, S.K.; Walker, J.; Sah, E.; Bennett, E.; Atrian, F.; Frost, B.; Woost, B.; Bennett, R.E.; Orr, T.C.; Zhou, Y.; et al. Profiling senescent cells in human brains reveals neurons with CDKN2D/p19 and tau neuropathology. Nat. Aging 2021, 1, 1107–1116. [Google Scholar] [CrossRef]
  275. Di Micco, R.; Krizhanovsky, V.; Baker, D.; d’Adda di Fagagna, F. Cellular senescence in ageing: From mechanisms to therapeutic opportunities. Nat. Rev. Mol. Cell Biol. 2021, 22, 75–95. [Google Scholar] [CrossRef] [PubMed]
  276. Burda, J.E.; Bernstein, A.M.; Sofroniew, M.V. Astrocyte roles in traumatic brain injury. Exp. Neurol. 2016, 275 Pt 3, 305–315. [Google Scholar] [CrossRef]
  277. Davalos, D.; Grutzendler, J.; Yang, G.; Kim, J.V.; Zuo, Y.; Jung, S.; Littman, D.R.; Dustin, M.L.; Gan, W.B. ATP mediates rapid microglial response to local brain injury in vivo. Nat. Neurosci. 2005, 8, 752–758. [Google Scholar] [CrossRef]
  278. Lewén, A.; Matz, P.; Chan, P.H. Free radical pathways in CNS injury. J. Neurotrauma 2000, 17, 871–890. [Google Scholar] [CrossRef]
  279. Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M.T.; Mazur, M.; Telser, J. Free radicals and antioxidants in normal physiological functions and human disease. Int. J. Biochem. Cell Biol. 2007, 39, 44–84. [Google Scholar] [CrossRef]
  280. Zhu, Y.; Tchkonia, T.; Pirtskhalava, T.; Gower, A.C.; Ding, H.; Giorgadze, N.; Palmer, A.K.; Ikeno, Y.; Hubbard, G.B.; Lenburg, M.; et al. The Achilles’ heel of senescent cells: From transcriptome to senolytic drugs. Aging Cell 2015, 14, 644–658. [Google Scholar] [CrossRef]
  281. Fan, Z.; Tong, Y.; Yang, Z.; Wang, S.; Huang, T.; Yang, D.; Ni, Q.; Zhang, M.; Li, D.; Yang, M.; et al. Inhibitor PF-04691502 works as a senolytic to regulate cellular senescence. Exp. Gerontol. 2024, 186, 112359. [Google Scholar] [CrossRef]
  282. Fuhrmann-Stroissnigg, H.; Ling, Y.Y.; Zhao, J.; McGowan, S.J.; Zhu, Y.; Brooks, R.W.; Grassi, D.; Gregg, S.Q.; Stripay, J.L.; Dorronsoro, A.; et al. Identification of HSP90 inhibitors as a novel class of senolytics. Nat. Commun. 2017, 8, 422. [Google Scholar] [CrossRef] [PubMed]
  283. Somogyvari, M.; Khatatneh, S.; Soti, C. Hsp90: From Cellular to Organismal Proteostasis. Cells 2022, 11, 2479. [Google Scholar] [CrossRef] [PubMed]
  284. Adams, J.M.; Cory, S. The Bcl-2 protein family: Arbiters of cell survival. Science 1998, 281, 1322–1326. [Google Scholar] [CrossRef] [PubMed]
  285. Chang, J.; Wang, Y.; Shao, L.; Laberge, R.M.; Demaria, M.; Campisi, J.; Janakiraman, K.; Sharpless, N.E.; Ding, S.; Feng, W.; et al. Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat. Med. 2016, 22, 78–83. [Google Scholar] [CrossRef]
  286. Yosef, R.; Pilpel, N.; Tokarsky-Amiel, R.; Biran, A.; Ovadya, Y.; Cohen, S.; Vadai, E.; Dassa, L.; Shahar, E.; Condiotti, R.; et al. Directed elimination of senescent cells by inhibition of BCL-W and BCL-XL. Nat. Commun. 2016, 7, 11190. [Google Scholar] [CrossRef]
  287. Zhu, Y.; Tchkonia, T.; Fuhrmann-Stroissnigg, H.; Dai, H.M.; Ling, Y.Y.; Stout, M.B.; Pirtskhalava, T.; Giorgadze, N.; Johnson, K.O.; Giles, C.B.; et al. Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors. Aging Cell 2016, 15, 428–435. [Google Scholar] [CrossRef]
  288. Guerrero, A.; Herranz, N.; Sun, B.; Wagner, V.; Gallage, S.; Guiho, R.; Wolter, K.; Pombo, J.; Irvine, E.E.; Innes, A.J.; et al. Cardiac glycosides are broad-spectrum senolytics. Nat. Metab. 2019, 1, 1074–1088. [Google Scholar] [CrossRef]
  289. Song, S.; Tchkonia, T.; Jiang, J.; Kirkland, J.L.; Sun, Y. Targeting Senescent Cells for a Healthier Aging: Challenges and Opportunities. Adv. Sci. 2020, 7, 2002611. [Google Scholar] [CrossRef]
  290. Ballou, L.M.; Lin, R.Z. Rapamycin and mTOR kinase inhibitors. J. Chem. Biol. 2008, 1, 27–36. [Google Scholar] [CrossRef]
  291. Li, J.; Kim, S.G.; Blenis, J. Rapamycin: One drug, many effects. Cell Metab. 2014, 19, 373–379. [Google Scholar] [CrossRef]
  292. Szaniawski, M.A.; Spivak, A.M. Senotherapeutics and HIV-1 Persistence. Curr. HIV/AIDS Rep. 2020, 17, 219–225. [Google Scholar] [CrossRef]
  293. Laberge, R.M.; Zhou, L.; Sarantos, M.R.; Rodier, F.; Freund, A.; de Keizer, P.L.; Liu, S.; Demaria, M.; Cong, Y.S.; Kapahi, P.; et al. Glucocorticoids suppress selected components of the senescence-associated secretory phenotype. Aging Cell 2012, 11, 569–578. [Google Scholar] [CrossRef] [PubMed]
  294. Moiseeva, O.; Deschenes-Simard, X.; St-Germain, E.; Igelmann, S.; Huot, G.; Cadar, A.E.; Bourdeau, V.; Pollak, M.N.; Ferbeyre, G. Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-kappaB activation. Aging Cell 2013, 12, 489–498. [Google Scholar] [CrossRef] [PubMed]
  295. Lim, H.; Park, H.; Kim, H.P. Effects of flavonoids on senescence-associated secretory phenotype formation from bleomycin-induced senescence in BJ fibroblasts. Biochem. Pharmacol. 2015, 96, 337–348. [Google Scholar] [CrossRef]
  296. Xu, M.; Tchkonia, T.; Ding, H.; Ogrodnik, M.; Lubbers, E.R.; Pirtskhalava, T.; White, T.A.; Johnson, K.O.; Stout, M.B.; Mezera, V.; et al. JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. Proc. Natl. Acad. Sci. USA 2015, 112, E6301–E6310. [Google Scholar] [CrossRef]
  297. Wan, M.; Gray-Gaillard, E.F.; Elisseeff, J.H. Cellular senescence in musculoskeletal homeostasis, diseases, and regeneration. Bone Res. 2021, 9, 41. [Google Scholar] [CrossRef] [PubMed]
  298. Alimbetov, D.; Davis, T.; Brook, A.J.; Cox, L.S.; Faragher, R.G.; Nurgozhin, T.; Zhumadilov, Z.; Kipling, D. Suppression of the senescence-associated secretory phenotype (SASP) in human fibroblasts using small molecule inhibitors of p38 MAP kinase and MK2. Biogerontology 2016, 17, 305–315. [Google Scholar] [CrossRef]
  299. Schuliga, M.; Kanwal, A.; Read, J.; Blokland, K.E.C.; Burgess, J.K.; Prele, C.M.; Mutsaers, S.E.; Grainge, C.; Thomson, C.; James, A.; et al. A cGAS-dependent response links DNA damage and senescence in alveolar epithelial cells: A potential drug target in IPF. Am. J. Physiol. Lung Cell Mol. Physiol. 2021, 321, L859–L871. [Google Scholar] [CrossRef]
  300. Shi, Y.; Zhao, L.; Wang, J.; Liu, S.; Zhang, Y.; Qin, Q. The selective NLRP3 inflammasome inhibitor MCC950 improves isoproterenol-induced cardiac dysfunction by inhibiting cardiomyocyte senescence. Eur. J. Pharmacol. 2022, 937, 175364. [Google Scholar] [CrossRef]
  301. Kelso, G.F.; Porteous, C.M.; Hughes, G.; Ledgerwood, E.C.; Gane, A.M.; Smith, R.A.; Murphy, M.P. Prevention of mitochondrial oxidative damage using targeted antioxidants. Ann. N. Y Acad. Sci. 2002, 959, 263–274. [Google Scholar] [CrossRef]
  302. Mantzorou, M.; Pavlidou, E.; Vasios, G.; Tsagalioti, E.; Giaginis, C. Effects of curcumin consumption on human chronic diseases: A narrative review of the most recent clinical data. Phytother. Res. 2018, 32, 957–975. [Google Scholar] [CrossRef] [PubMed]
  303. Ravindran, J.; Prasad, S.; Aggarwal, B.B. Curcumin and cancer cells: How many ways can curry kill tumor cells selectively? AAPS J. 2009, 11, 495–510. [Google Scholar] [CrossRef] [PubMed]
  304. Cherif, H.; Bisson, D.G.; Jarzem, P.; Weber, M.; Ouellet, J.A.; Haglund, L. Curcumin and o-Vanillin Exhibit Evidence of Senolytic Activity in Human IVD Cells In Vitro. J. Clin. Med. 2019, 8, 433. [Google Scholar] [CrossRef]
  305. Heger, M.; van Golen, R.F.; Broekgaarden, M.; Michel, M.C. The molecular basis for the pharmacokinetics and pharmacodynamics of curcumin and its metabolites in relation to cancer. Pharmacol. Rev. 2014, 66, 222–307. [Google Scholar] [CrossRef]
  306. Ragothaman, M.; Murgod, U.A.; Gururaj, G.; Kumaraswamy, S.D.; Muthane, U. Lower risk of Parkinson’s disease in an admixed population of European and Indian origins. Mov. Disord. 2003, 18, 912–914. [Google Scholar] [CrossRef] [PubMed]
  307. Commandeur, J.N.; Vermeulen, N.P. Cytotoxicity and cytoprotective activities of natural compounds. The case of curcumin. Xenobiotica 1996, 26, 667–680. [Google Scholar] [CrossRef] [PubMed]
  308. Pandey, N.; Strider, J.; Nolan, W.C.; Yan, S.X.; Galvin, J.E. Curcumin inhibits aggregation of alpha-synuclein. Acta Neuropathol. 2008, 115, 479–489. [Google Scholar] [CrossRef]
  309. Sharma, N.; Nehru, B. Curcumin affords neuroprotection and inhibits alpha-synuclein aggregation in lipopolysaccharide-induced Parkinson’s disease model. Inflammopharmacology 2018, 26, 349–360. [Google Scholar] [CrossRef]
  310. Zbarsky, V.; Datla, K.P.; Parkar, S.; Rai, D.K.; Aruoma, O.I.; Dexter, D.T. Neuroprotective properties of the natural phenolic antioxidants curcumin and naringenin but not quercetin and fisetin in a 6-OHDA model of Parkinson’s disease. Free Radic. Res. 2005, 39, 1119–1125. [Google Scholar] [CrossRef]
  311. Zhang, N.; Yan, F.; Liang, X.; Wu, M.; Shen, Y.; Chen, M.; Xu, Y.; Zou, G.; Jiang, P.; Tang, C.; et al. Localized delivery of curcumin into brain with polysorbate 80-modified cerasomes by ultrasound-targeted microbubble destruction for improved Parkinson’s disease therapy. Theranostics 2018, 8, 2264–2277. [Google Scholar] [CrossRef]
  312. Arai, Y.; Watanabe, S.; Kimira, M.; Shimoi, K.; Mochizuki, R.; Kinae, N. Dietary intakes of flavonols, flavones and isoflavones by Japanese women and the inverse correlation between quercetin intake and plasma LDL cholesterol concentration. J. Nutr. 2000, 130, 2243–2250. [Google Scholar] [CrossRef]
  313. Maher, P. How fisetin reduces the impact of age and disease on CNS function. Front. Biosci. (Sch. Ed.) 2015, 7, 58–82. [Google Scholar] [CrossRef]
  314. Khan, N.; Syed, D.N.; Ahmad, N.; Mukhtar, H. Fisetin: A dietary antioxidant for health promotion. Antioxid. Redox Signal 2013, 19, 151–162. [Google Scholar] [CrossRef]
  315. Pal, H.C.; Pearlman, R.L.; Afaq, F. Fisetin and Its Role in Chronic Diseases. Adv. Exp. Med. Biol. 2016, 928, 213–244. [Google Scholar] [CrossRef] [PubMed]
  316. Sundarraj, K.; Raghunath, A.; Perumal, E. A review on the chemotherapeutic potential of fisetin: In vitro evidences. Biomed. Pharmacother. 2018, 97, 928–940. [Google Scholar] [CrossRef]
  317. Zhu, Y.; Doornebal, E.J.; Pirtskhalava, T.; Giorgadze, N.; Wentworth, M.; Fuhrmann-Stroissnigg, H.; Niedernhofer, L.J.; Robbins, P.D.; Tchkonia, T.; Kirkland, J.L. New agents that target senescent cells: The flavone, fisetin, and the BCL-XL inhibitors, A1331852 and A1155463. Aging 2017, 9, 955–963. [Google Scholar] [CrossRef]
  318. Yousefzadeh, M.J.; Zhu, Y.; McGowan, S.J.; Angelini, L.; Fuhrmann-Stroissnigg, H.; Xu, M.; Ling, Y.Y.; Melos, K.I.; Pirtskhalava, T.; Inman, C.L.; et al. Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine 2018, 36, 18–28. [Google Scholar] [CrossRef]
  319. Watanabe, R.; Kurose, T.; Morishige, Y.; Fujimori, K. Protective Effects of Fisetin Against 6-OHDA-Induced Apoptosis by Activation of PI3K-Akt Signaling in Human Neuroblastoma SH-SY5Y Cells. Neurochem. Res. 2018, 43, 488–499. [Google Scholar] [CrossRef]
  320. Patel, M.Y.; Panchal, H.V.; Ghribi, O.; Benzeroual, K.E. The neuroprotective effect of fisetin in the MPTP model of Parkinson’s disease. J. Parkinsons Dis. 2012, 2, 287–302. [Google Scholar] [CrossRef] [PubMed]
  321. Maher, P. Protective effects of fisetin and other berry flavonoids in Parkinson’s disease. Food Funct. 2017, 8, 3033–3042. [Google Scholar] [CrossRef] [PubMed]
  322. Alikatte, K.; Palle, S.; Rajendra Kumar, J.; Pathakala, N. Fisetin Improved Rotenone-Induced Behavioral Deficits, Oxidative Changes, and Mitochondrial Dysfunctions in Rat Model of Parkinson’s Disease. J. Diet. Suppl. 2021, 18, 57–71. [Google Scholar] [CrossRef] [PubMed]
  323. Chen, T.J.; Feng, Y.; Liu, T.; Wu, T.T.; Chen, Y.J.; Li, X.; Li, Q.; Wu, Y.C. Fisetin Regulates Gut Microbiota and Exerts Neuroprotective Effect on Mouse Model of Parkinson’s Disease. Front. Neurosci. 2020, 14, 549037. [Google Scholar] [CrossRef]
  324. Lang, F.; Cohen, P. Regulation and physiological roles of serum- and glucocorticoid-induced protein kinase isoforms. Sci. STKE 2001, 2001, re17. [Google Scholar] [CrossRef]
  325. Hartmann, A.; Veldhuis, J.D.; Deuschle, M.; Standhardt, H.; Heuser, I. Twenty-four hour cortisol release profiles in patients with Alzheimer’s and Parkinson’s disease compared to normal controls: Ultradian secretory pulsatility and diurnal variation. Neurobiol. Aging 1997, 18, 285–289. [Google Scholar] [CrossRef]
  326. Kaku, K.; Shikimi, T.; Kamisaki, Y.; Shinozuka, K.; Ishino, H.; Okunishi, H.; Takaori, S. Elevation of striatal interleukin-6 and serum corticosterone contents in MPTP-treated mice. Clin. Exp. Pharmacol. Physiol. 1999, 26, 680–683. [Google Scholar] [CrossRef] [PubMed]
  327. Pratt, B.M.; McPherson, J.M. TGF-beta in the central nervous system: Potential roles in ischemic injury and neurodegenerative diseases. Cytokine Growth Factor. Rev. 1997, 8, 267–292. [Google Scholar] [CrossRef]
  328. Iwata, S.; Nomoto, M.; Morioka, H.; Miyata, A. Gene expression profiling in the midbrain of striatal 6-hydroxydopamine-injected mice. Synapse 2004, 51, 279–286. [Google Scholar] [CrossRef]
  329. Stichel, C.C.; Schoenebeck, B.; Foguet, M.; Siebertz, B.; Bader, V.; Zhu, X.R.; Lubbert, H. sgk1, a member of an RNA cluster associated with cell death in a model of Parkinson’s disease. Eur. J. Neurosci. 2005, 21, 301–316. [Google Scholar] [CrossRef]
  330. Kwon, O.C.; Song, J.J.; Yang, Y.; Kim, S.H.; Kim, J.Y.; Seok, M.J.; Hwang, I.; Yu, J.W.; Karmacharya, J.; Maeng, H.J.; et al. SGK1 inhibition in glia ameliorates pathologies and symptoms in Parkinson disease animal models. EMBO Mol. Med. 2021, 13, e13076. [Google Scholar] [CrossRef] [PubMed]
  331. Xia, M.L.; Xie, X.H.; Ding, J.H.; Du, R.H.; Hu, G. Astragaloside IV inhibits astrocyte senescence: Implication in Parkinson’s disease. J. Neuroinflamm. 2020, 17, 105. [Google Scholar] [CrossRef]
  332. Cheng, X.; Gu, J.; Zhang, M.; Yuan, J.; Zhao, B.; Jiang, J.; Jia, X. Astragaloside IV inhibits migration and invasion in human lung cancer A549 cells via regulating PKC-alpha-ERK1/2-NF-κB pathway. Int. Immunopharmacol. 2014, 23, 304–313. [Google Scholar] [CrossRef] [PubMed]
  333. Zhang, W.J.; Frei, B. Astragaloside IV inhibits NF-κB activation and inflammatory gene expression in LPS-treated mice. Mediators Inflamm. 2015, 2015, 274314. [Google Scholar] [CrossRef]
  334. Gui, D.; Huang, J.; Liu, W.; Guo, Y.; Xiao, W.; Wang, N. Astragaloside IV prevents acute kidney injury in two rodent models by inhibiting oxidative stress and apoptosis pathways. Apoptosis 2013, 18, 409–422. [Google Scholar] [CrossRef]
  335. Chan, W.S.; Durairajan, S.S.; Lu, J.H.; Wang, Y.; Xie, L.X.; Kum, W.F.; Koo, I.; Yung, K.K.; Li, M. Neuroprotective effects of Astragaloside IV in 6-hydroxydopamine-treated primary nigral cell culture. Neurochem. Int. 2009, 55, 414–422. [Google Scholar] [CrossRef]
  336. Novais, E.J.; Tran, V.A.; Johnston, S.N.; Darris, K.R.; Roupas, A.J.; Sessions, G.A.; Shapiro, I.M.; Diekman, B.O.; Risbud, M.V. Long-term treatment with senolytic drugs Dasatinib and Quercetin ameliorates age-dependent intervertebral disc degeneration in mice. Nat. Commun. 2021, 12, 5213. [Google Scholar] [CrossRef] [PubMed]
  337. Kirkland, J.L.; Tchkonia, T. Senolytic drugs: From discovery to translation. J. Intern. Med. 2020, 288, 518–536. [Google Scholar] [CrossRef]
  338. Baar, M.P.; Brandt, R.M.C.; Putavet, D.A.; Klein, J.D.D.; Derks, K.W.J.; Bourgeois, B.R.M.; Stryeck, S.; Rijksen, Y.; van Willigenburg, H.; Feijtel, D.A.; et al. Targeted Apoptosis of Senescent Cells Restores Tissue Homeostasis in Response to Chemotoxicity and Aging. Cell 2017, 169, 132–147. [Google Scholar] [CrossRef] [PubMed]
  339. Lapasset, L.; Milhavet, O.; Prieur, A.; Besnard, E.; Babled, A.; Ait-Hamou, N.; Leschik, J.; Pellestor, F.; Ramirez, J.M.; De Vos, J.; et al. Rejuvenating senescent and centenarian human cells by reprogramming through the pluripotent state. Genes. Dev. 2011, 25, 2248–2253. [Google Scholar] [CrossRef]
  340. Burslem, G.M.; Crews, C.M. Proteolysis-Targeting Chimeras as Therapeutics and Tools for Biological Discovery. Cell 2020, 181, 102–114. [Google Scholar] [CrossRef]
  341. Lai, A.C.; Crews, C.M. Induced protein degradation: An emerging drug discovery paradigm. Nat. Rev. Drug Discov. 2017, 16, 101–114. [Google Scholar] [CrossRef]
  342. Cai, Y.; Zhou, H.; Zhu, Y.; Sun, Q.; Ji, Y.; Xue, A.; Wang, Y.; Chen, W.; Yu, X.; Wang, L.; et al. Elimination of senescent cells by beta-galactosidase-targeted prodrug attenuates inflammation and restores physical function in aged mice. Cell Res. 2020, 30, 574–589. [Google Scholar] [CrossRef] [PubMed]
  343. Guerrero, A.; Guiho, R.; Herranz, N.; Uren, A.; Withers, D.J.; Martinez-Barbera, J.P.; Tietze, L.F.; Gil, J. Galactose-modified duocarmycin prodrugs as senolytics. Aging Cell 2020, 19, e13133. [Google Scholar] [CrossRef] [PubMed]
  344. Gonzalez-Gualda, E.; Paez-Ribes, M.; Lozano-Torres, B.; Macias, D.; Wilson, J.R., 3rd; Gonzalez-Lopez, C.; Ou, H.L.; Miron-Barroso, S.; Zhang, Z.; Lerida-Viso, A.; et al. Galacto-conjugation of Navitoclax as an efficient strategy to increase senolytic specificity and reduce platelet toxicity. Aging Cell 2020, 19, e13142. [Google Scholar] [CrossRef] [PubMed]
  345. Kim, K.M.; Noh, J.H.; Bodogai, M.; Martindale, J.L.; Yang, X.; Indig, F.E.; Basu, S.K.; Ohnuma, K.; Morimoto, C.; Johnson, P.F.; et al. Identification of senescent cell surface targetable protein DPP4. Genes. Dev. 2017, 31, 1529–1534. [Google Scholar] [CrossRef]
  346. Poblocka, M.; Bassey, A.L.; Smith, V.M.; Falcicchio, M.; Manso, A.S.; Althubiti, M.; Sheng, X.; Kyle, A.; Barber, R.; Frigerio, M.; et al. Targeted clearance of senescent cells using an antibody-drug conjugate against a specific membrane marker. Sci. Rep. 2021, 11, 20358. [Google Scholar] [CrossRef]
  347. Munoz-Espin, D.; Rovira, M.; Galiana, I.; Gimenez, C.; Lozano-Torres, B.; Paez-Ribes, M.; Llanos, S.; Chaib, S.; Munoz-Martin, M.; Ucero, A.C.; et al. A versatile drug delivery system targeting senescent cells. EMBO Mol. Med. 2018, 10, e9355. [Google Scholar] [CrossRef]
Figure 1. Inter-relationships between SNpc DA neurons, microglia, astrocytes, vascular endothelial cells, and T cells play a role in the pathophysiology of PD. SNpc DA neurons are vulnerable to stressors due to a state of high ATP demand, low Ca2+ buffering capacity, low anti-oxidant defenses, and low trophic support. ROS and DNA damage induce senescence in microglia, astrocytes, and SNpc DA neurons. Autocrine transmission of the senescence-associated secretory phenotype (SASP) by senescent microglia, astrocytes, and SNpc DA neurons reinforces the senescent phenotype. Nearby cells undergo senescence as a result of paracrine transmission of the SASP. Transmission of the SASP from senescent microglia and astrocytes to SNpc DA neurons induces senescence in those neurons and contributes to SNpc DA neuron cell death. Chronic inflammation causes endothelial cells to contract, forming gaps between these cells thereby allowing circulating T cells to infiltrate the brain. Infiltrating T cells contribute to the death of SNpc DA neurons by secreting chemokines and cytokines. In PD, astrocytes, microglia, and SNpc DA neurons exchange exosomes containing pathogenic α-syn, resulting in the formation of LBs and LB-like intracellular inclusions. This figure was created with BioRender.com by Rademacher, Exline, and Foecking, 2025. http://app.biorender.com/ (accessed on 21 May 2025).
Figure 1. Inter-relationships between SNpc DA neurons, microglia, astrocytes, vascular endothelial cells, and T cells play a role in the pathophysiology of PD. SNpc DA neurons are vulnerable to stressors due to a state of high ATP demand, low Ca2+ buffering capacity, low anti-oxidant defenses, and low trophic support. ROS and DNA damage induce senescence in microglia, astrocytes, and SNpc DA neurons. Autocrine transmission of the senescence-associated secretory phenotype (SASP) by senescent microglia, astrocytes, and SNpc DA neurons reinforces the senescent phenotype. Nearby cells undergo senescence as a result of paracrine transmission of the SASP. Transmission of the SASP from senescent microglia and astrocytes to SNpc DA neurons induces senescence in those neurons and contributes to SNpc DA neuron cell death. Chronic inflammation causes endothelial cells to contract, forming gaps between these cells thereby allowing circulating T cells to infiltrate the brain. Infiltrating T cells contribute to the death of SNpc DA neurons by secreting chemokines and cytokines. In PD, astrocytes, microglia, and SNpc DA neurons exchange exosomes containing pathogenic α-syn, resulting in the formation of LBs and LB-like intracellular inclusions. This figure was created with BioRender.com by Rademacher, Exline, and Foecking, 2025. http://app.biorender.com/ (accessed on 21 May 2025).
Biomedicines 13 01400 g001
Figure 2. Mechanisms of cell death induction of senolytics. The senolytic names are given in the red boxes. The red flat arrowheads indicate inhibition. Abbreviations: mTOR, mechanistic target of rapamycin; IL-1, interleukin-1; NF-κB, nuclear factor kappa-B; AKT, protein kinase B; Bcl-2, B-cell lymphoma-2; Bcl-xl, B-cell lymphoma-extra large; Bcl-w, B-cell lymphoma-w; 17-DMAG, 17-dimethylaminoethylamino; FoxO4, forkhead box subclass O protein 4; HSP90, heat shock protein 90; RTK, receptor tyrosine kinase; PI3K, phosphatidylinositol 3-kinase; P, phosphorylation; Na+/K+ ATPase, sodium/potassium adenosine triphosphatase. This figure was created with BioRender.com by Rademacher, Exline, and Foecking, 2025. http://app.biorender.com/ (accessed on 21 May 2025).
Figure 2. Mechanisms of cell death induction of senolytics. The senolytic names are given in the red boxes. The red flat arrowheads indicate inhibition. Abbreviations: mTOR, mechanistic target of rapamycin; IL-1, interleukin-1; NF-κB, nuclear factor kappa-B; AKT, protein kinase B; Bcl-2, B-cell lymphoma-2; Bcl-xl, B-cell lymphoma-extra large; Bcl-w, B-cell lymphoma-w; 17-DMAG, 17-dimethylaminoethylamino; FoxO4, forkhead box subclass O protein 4; HSP90, heat shock protein 90; RTK, receptor tyrosine kinase; PI3K, phosphatidylinositol 3-kinase; P, phosphorylation; Na+/K+ ATPase, sodium/potassium adenosine triphosphatase. This figure was created with BioRender.com by Rademacher, Exline, and Foecking, 2025. http://app.biorender.com/ (accessed on 21 May 2025).
Biomedicines 13 01400 g002
Figure 3. Mechanisms of SASP modulation by senomorphics. The names of the senomorphics are given in the red boxes. The red flat arrowheads indicate inhibition. Abbreviations: SASP, senescence-associated secretory phenotype; JAK, Janus kinase; STAT, signal transducer and activation of transcription; NLRP3, NLR family pyrin domain containing 3; H2O2, hydrogen peroxide; mTOR, mechanistic target of rapamycin; IL-1, interleukin-1; IL-1R; interleukin-1 receptor; IL-6, interleukin-6; IL-6R, interleukin-6 receptor; MitoQ, mitoquinone; NF-κB, nuclear factor kappa-B; cGAS, cyclic GMP-AMP synthase; TNF, tumor necrosis factor; TNFR, tumor necrosis factor receptor; AKT, protein kinase B; RTK, receptor tyrosine kinase; STING, stimulator of interferon genes; PI3K, phosphatidylinositol 3-kinase; P, phosphorylation. This figure was created with BioRender.com by Rademacher, Exline, and Foecking, 2025. http://app.biorender.com/ (accessed on 21 May 2025).
Figure 3. Mechanisms of SASP modulation by senomorphics. The names of the senomorphics are given in the red boxes. The red flat arrowheads indicate inhibition. Abbreviations: SASP, senescence-associated secretory phenotype; JAK, Janus kinase; STAT, signal transducer and activation of transcription; NLRP3, NLR family pyrin domain containing 3; H2O2, hydrogen peroxide; mTOR, mechanistic target of rapamycin; IL-1, interleukin-1; IL-1R; interleukin-1 receptor; IL-6, interleukin-6; IL-6R, interleukin-6 receptor; MitoQ, mitoquinone; NF-κB, nuclear factor kappa-B; cGAS, cyclic GMP-AMP synthase; TNF, tumor necrosis factor; TNFR, tumor necrosis factor receptor; AKT, protein kinase B; RTK, receptor tyrosine kinase; STING, stimulator of interferon genes; PI3K, phosphatidylinositol 3-kinase; P, phosphorylation. This figure was created with BioRender.com by Rademacher, Exline, and Foecking, 2025. http://app.biorender.com/ (accessed on 21 May 2025).
Biomedicines 13 01400 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Rademacher, D.J.; Exline, J.E.; Foecking, E.M. Role of Cellular Senescence in Parkinson’s Disease: Potential for Disease-Modification Through Senotherapy. Biomedicines 2025, 13, 1400. https://doi.org/10.3390/biomedicines13061400

AMA Style

Rademacher DJ, Exline JE, Foecking EM. Role of Cellular Senescence in Parkinson’s Disease: Potential for Disease-Modification Through Senotherapy. Biomedicines. 2025; 13(6):1400. https://doi.org/10.3390/biomedicines13061400

Chicago/Turabian Style

Rademacher, David J., Jacob E. Exline, and Eileen M. Foecking. 2025. "Role of Cellular Senescence in Parkinson’s Disease: Potential for Disease-Modification Through Senotherapy" Biomedicines 13, no. 6: 1400. https://doi.org/10.3390/biomedicines13061400

APA Style

Rademacher, D. J., Exline, J. E., & Foecking, E. M. (2025). Role of Cellular Senescence in Parkinson’s Disease: Potential for Disease-Modification Through Senotherapy. Biomedicines, 13(6), 1400. https://doi.org/10.3390/biomedicines13061400

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop