Next Article in Journal
GramSeq-DTA: A Grammar-Based Drug–Target Affinity Prediction Approach Fusing Gene Expression Information
Previous Article in Journal
Exploring the CNOT1(800–999) HEAT Domain and Its Interactions with Tristetraprolin (TTP) as Revealed by Hydrogen/Deuterium Exchange Mass Spectrometry
Previous Article in Special Issue
Possible Anti-Aging and Anti-Stress Effects of Long-Term Transcendental Meditation Practice: Differences in Gene Expression, EEG Correlates of Cognitive Function, and Hair Steroids
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathological and Inflammatory Consequences of Aging

by
Mario García-Domínguez
1,2,3
1
Program of Immunology and Immunotherapy, CIMA-Universidad de Navarra, 31008 Pamplona, Spain
2
Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, 31008 Pamplona, Spain
3
Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
Biomolecules 2025, 15(3), 404; https://doi.org/10.3390/biom15030404
Submission received: 25 February 2025 / Revised: 8 March 2025 / Accepted: 10 March 2025 / Published: 12 March 2025
(This article belongs to the Special Issue Molecular Advances in Mechanism and Regulation of Lifespan and Aging)

Abstract

:
Aging is a complex, progressive, and irreversible biological process that entails numerous structural and functional changes in the organism. These changes affect all bodily systems, reducing their ability to respond and adapt to the environment. Chronic inflammation is one of the key factors driving the development of age-related diseases, ultimately causing a substantial decline in the functional abilities of older individuals. This persistent inflammatory state (commonly known as “inflammaging”) is characterized by elevated levels of pro-inflammatory cytokines, an increase in oxidative stress, and a perturbation of immune homeostasis. Several factors, including cellular senescence, contribute to this inflammatory milieu, thereby amplifying conditions such as cardiovascular disease, neurodegeneration, and metabolic disorders. Exploring the mechanisms of chronic inflammation in aging is essential for developing targeted interventions aimed at promoting healthy aging. This review explains the strong connection between aging and chronic inflammation, highlighting potential therapeutic approaches like pharmacological treatments, dietary strategies, and lifestyle changes.

1. Introduction

The process of aging constitutes a multifaceted and intricate phenomenon characterized by a gradual deterioration of several biological functions [1,2], thereby rendering humans more susceptible to a range of age-associated diseases [3]. This mechanism involves many molecular, cellular, and systemic alterations that progressively impair overall functionality [4,5,6]. At the molecular level, aging is linked to genomic instability [7], resulting in the accumulation of DNA damage [8]. Consequently, the DNA damage leads to telomere shortening [9] and epigenetic alterations that regulate gene expression [10]. On the other hand, aging is linked to strong reduction in proteostasis, which impairs cells’ ability to maintain a functional proteome [11]. Additionally, mitochondrial dysfunction occurs, leading to a decrease in energy production and an increased generation of reactive oxygen species (ROS) that damage cellular structures [12].
These changes result in cellular senescence, a state in which cells cease to divide but remain metabolically active [13]. Senescent cells exhibit a senescence-associated secretory phenotype (SASP), characterized by the secretion of pro-inflammatory cytokines (such as IL-1α, IL-1β, IL-6, IL-8, and IL-18), growth factors (e.g., VEGF, TGF-β, GM-CSF, IGFBP-2, and GDF-15), and proteases [14,15,16]. The accumulation of senescent cells within tissues is now widely recognized as a key driver of aging and the development of many conditions like cardiovascular diseases, neurodegeneration, and cancer [17,18,19]. The sustained secretion of the mentioned SASP factors occupies an essential position in the formation and continuation of a chronic inflammatory milieu; this environment contributes to both peripheral and central inflammatory mechanisms [20].
At the peripheral level, the release of SASP mediators significantly amplifies systemic inflammation, triggering a cascade of physiopathological events that disrupt normal tissue homeostasis [21]. However, this persistent inflammatory state causes structural and functional alterations in tissues, driving pathological remodeling in organs such as the heart, liver, and kidneys. Pro-inflammatory mediators promote fibrosis and ischemic injury by inducing maladaptive repair processes while simultaneously impairing the regenerative processes necessary for maintaining tissue integrity. As a result, this situation creates a cycle of dysfunction that accelerates tissue damage and hinders proper recovery [22,23]. This biological process, known as inflammaging, is characterized by chronic low-grade inflammation associated with aging, sustained by the SASP mediators [24,25]. This condition not only perpetuates tissue damage but also amplifies the degenerative changes associated with aging, establishing a vicious cycle of inflammation and dysfunction that impedes tissue repair and accelerates the progression of age-related diseases.
At the central nervous system (CNS) level, SASP mediators can exert significant effects on the brain. One of the major consequences is the disruption of the blood–brain barrier (BBB), a critical defense mechanism that protects the brain from harmful substances present in the bloodstream [26]. The prolonged inflammation caused by SASP mediators can compromise the integrity of the BBB; thus, it facilitates the infiltration of peripheral immune cells, toxins, and pro-inflammatory cytokines into the CNS [27]. This dysregulation within the CNS not only initiates neuroinflammation but also strongly activates microglial cells. In their activated state, these cells release additional pro-inflammatory cytokines, further exacerbating neurodegenerative processes linked to aging such as Parkinson’s and Alzheimer’s diseases [28,29]. Microglial activation induces several effects, like synaptic dysfunction, neuronal cell death, and disruptions in neuronal signaling [30,31]. This aberrant inflammatory response may accelerate neuronal loss, impair cognitive function, and evoke the accumulation of toxic protein aggregates, such as β-amyloid plaques in Alzheimer’s disease and α-synuclein in Parkinson’s disease [32,33].
This review will provide a comprehensive analysis of the pathophysiological consequences of aging, emphasizing the molecular, cellular, and systemic alterations that drive age-related deterioration. Furthermore, this review will investigate the inflammatory processes linked to aging, both at the peripheral and central levels. Particular emphasis will be placed on the underlying mechanisms that drive inflammation, including immune cell activation, oxidative stress, and the accumulation of senescent cells. By synthesizing current research findings, this review aims to provide a deeper understanding of the interplay between aging and inflammation, shedding light on potential therapeutic interventions to mitigate the adverse effects of these processes.

2. Aging: Physiopathological Consequences

Aging is a process that leads to functional physiological changes, including increased susceptibility to many chronic conditions [3]. The complex physiopathological processes linked to aging influence multiple body systems, such as the cardiovascular, nervous, immune, and musculoskeletal systems, thereby elevating the risk of neurodegeneration, cardiovascular diseases, and metabolic disorders, among others [34]. Gaining a deep understanding of the physiological consequences of aging is essential for developing effective therapeutic approaches that not only support healthy aging but also mitigate the impact of age-related diseases. As the population ages, it becomes increasingly important to identify and address the underlying biological mechanisms that contribute to the decline in organ function, immune system efficiency, and cognitive abilities. By prioritizing these processes, researchers can design interventions that promote longevity, enhance quality of life, and reduce the burden of chronic diseases, ultimately helping older individuals maintain their independence and well-being as they age [35].

2.1. Nervous System

The human body undergoes numerous alterations within its CNS throughout the aging process, leading to changes in cognitive functions, memory capabilities, and overall brain health status [36]. A key factor in this process is the increase in oxidative stress. On a molecular level, oxidative stress is critical, as aging neurons accumulate ROS due to mitochondrial dysfunction [37]. The brain experiences elevated levels of malondialdehyde (MDA) along with lipid peroxidation products such as 4-hydroxy-2-nonenal (4-HNE), which induce DNA damage and promote lipid peroxidation and protein misfolding factors involved in neurodegenerative processes [38,39].
Telomere shortening is a fundamental marker of aging that amplifies neuronal vulnerability by restricting the regenerative potential of neural stem cells and reducing synaptic plasticity while also inhibiting neurogenesis in the hippocampus, a crucial brain region for learning and memory [40,41]. Conversely, the balance of proteostasis, involving protein synthesis, folding, and degradation, is disrupted during aging as the functional efficiency of both the ubiquitin–proteasome system and autophagy declines [42]. This deficiency facilitates the accumulation of protein aggregates such as β-amyloid plaques seen in Alzheimer’s disease (Figure 1) and α-synuclein aggregates found in Parkinson’s disease [32,33].
Furthermore, reduced levels of neurotransmitters such as dopamine, acetylcholine, and serotonin can disrupt signal transmission, resulting in several problems with motor coordination, mood regulation, and cognitive function [43]. The myelin sheath is likewise affected by dysfunctions in oligodendrocytes, resulting in delayed neural conduction and increased susceptibility to demyelinating diseases [44]. Neuroinflammation, primarily driven by activated microglia and astrocytes, accelerates neural aging through the release of various pro-inflammatory cytokines such as IL-6, IL-1β, and TNF-α [45]. These cytokines can impair synaptic function and contribute to neuronal degeneration [46]. Furthermore, aging influences several signaling pathways that are fundamental for normal neuronal function, including the PI3K-AKT and MAPK pathways [47]. Additionally, the age-related decline in BBB integrity aggravates neurodegenerative conditions by facilitating the infiltration of peripheral immune cells and toxins into the central nervous system, triggering chronic inflammation and neuronal injury [27].
On the other hand, the buildup of advanced glycation end-products (AGEs) also negatively impacts neuronal function by causing oxidative stress and persistent inflammation [48]. Finally, the meninges, the protective layers surrounding the brain and spinal cord, undergo age-related changes in gene expression that may modify the brain’s microenvironment and contribute to age-related neurological decline [49].

2.2. Cardiovascular System

The aging process profoundly affects the cardiovascular system through a complex interplay of molecular and cellular mechanisms, resulting in structural and functional alterations that compromise both cardiac and vascular integrity [50]. At the molecular level, collagen accumulation and impaired cardiomyocyte function play a critical role in the development of left ventricular hypertrophy and myocardial fibrosis [51]. Additionally, mitochondrial dysfunction, which involves decreased ATP production and the increased generation of ROS, disrupts cellular energy balance and worsens oxidative stress [52]. The blood vessels also experience significant transformations, such as reduced nitric oxide availability due to lower activity of endothelial nitric oxide synthase (eNOS) and increased nitric oxide depletion from ROS [53]. Chronic low-grade inflammation, driven by the SASP phenotype, along with impaired angiogenesis resulting from diminished vascular endothelial growth factor (VEGF) signaling, further exacerbates cardiovascular dysfunction [54].
Telomere shortening in vascular cells accelerates cellular senescence and increases the risk of apoptosis [55]. Epigenetic alterations, including changes in DNA methylation, histone modifications, and dysregulated microRNA expression, modulate gene activity linked to oxidative stress, fibrosis, and inflammation [56]. The accumulation of AGEs promotes the crosslinking of extracellular matrix proteins, leading to increased arterial stiffness and diastolic dysfunction [57]. Additionally, impaired protein maintenance and autophagy lead to the accumulation of misfolded proteins and damaged organelles, accelerating cellular dysfunction and disease progression [58].

2.3. Gastrointestinal System

The aging process has a significant effect on the gastrointestinal (GI) system, leading to functional decline and a higher risk of diseases through various molecular mechanisms [59]. One major factor is the degeneration of neuromuscular function, which includes the loss of interstitial cells of Cajal and a reduction in acetylcholine signaling, resulting in impaired peristalsis and contributing to constipation [60,61]. Esophageal issues, particularly the weakening of the lower esophageal sphincter, are influenced by changes in nitric oxide signaling, which raises the risk of gastroesophageal reflux disease (GERD) [62]. Furthermore, gastric acid secretion decreases due to atrophic gastritis, which is frequently linked to inflammation caused by Helicobacter pylori and an increase in pro-inflammatory cytokines like IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IFN-γ, and TNF-α [63]. The reduction in acid secretion can interfere with nutrient absorption [64].
In the pancreas, mitochondrial dysfunction and endoplasmic reticulum stress lead to a significant reduction in enzyme secretion from the pancreatic islets, resulting in the malabsorption of macronutrients [65,66]. The liver also suffers from decreased blood flow and reduced activity of cytochrome P450 enzymes, especially CYP3A4, which affects drug metabolism and raises the risk of medication toxicity [67]. On the other hand, the colon undergoes structural weakening due to strong alterations in extracellular matrix remodeling, driven by matrix metalloproteinases, thereby increasing the risk of diverticulosis and inflammatory complications [68]. The risk of colorectal cancer also rises due to accumulated DNA damage, telomere shortening, and many mutations in tumor suppressor genes such as TP53, worsened by the chronic activation of NF-κB and the overproduction of prostaglandin E2 [69,70]. Moreover, polypharmacy can disrupt gut health, as NSAIDs inhibit COX-1 and raise the risk of ulcers, while antibiotics can lead to dysbiosis, increasing susceptibility to Clostridioides difficile infections [71,72]. Dehydration, resulting from reduced sensitivity of hypothalamic osmoreceptors, exacerbates colonic transit, leading to more severe constipation and increased systemic inflammation [73].
Ultimately, the gut–brain axis plays a critical role in the aging process, impacting cognitive function, mental health, and overall well-being [74]. The gut microbiome, a complex community of trillions of microorganisms residing in the digestive tract, promotes bidirectional communication with the brain via the gut–brain axis, an interconnected network that includes the vagus nerve, immune system, and neuroactive compounds such as neurotransmitters and short-chain fatty acids (SCFAs) [75]. With age, the gut microbiome undergoes profound changes, often exhibiting decreased diversity due to factors such as an imbalanced diet, medication use (particularly antibiotics), and decreased physical activity [76]. Alterations in the gut microbiota, characterized by a strong reduction in Bifidobacteria and Lactobacillus populations, activate Toll-like receptor (TLR) pathways and compromise gut barrier integrity by downregulating tight junction proteins such as occludin and claudin-1 [77,78,79]. This situation contributes to systemic inflammation, immune dysregulation, and cognitive decline [80]. Additionally, the gut microbiota regulates the production of some neurotransmitters, including serotonin and dopamine, which participate in mood regulation and cognitive function [81]. Finally, aging is associated with a reduction in gut barrier integrity, resulting in increased intestinal permeability, which enables harmful substances to enter the bloodstream and trigger immune responses that could accelerate neurodegeneration [82].

2.4. Respiratory System

As individuals age, the respiratory system undergoes several physiological changes that progressively reduce pulmonary function, increasing susceptibility to respiratory illnesses and impairing oxygenation efficiency [83,84]. The primary age-related alteration is the gradual loss of lung elasticity, driven by structural modifications in elastin and collagen fibers within lung tissue [85], due to the deposition of collagen and the degradation of elastin fibers. This change is caused by an imbalance between matrix metalloproteinases (MMPs), enzymes responsible for degrading extracellular matrix (ECM) components, and their tissue inhibitors (TIMPs) [86], which disrupt the structural integrity of the lungs, leading to decreased lung compliance and a reduction in alveolar surface area [87].
In the alveolar region, aging is associated with a reduction in the production of surfactants (such as dipalmitoylphosphatidylcholine—DPPC) and several surfactant proteins [88]. This leads to an altered composition of the surfactant, resulting in a less efficient reduction in surface tension in the alveolar sacs. Consequently, this alteration in surfactant composition not only compromises alveolar integrity but also hinders effective gas exchange by reducing the surface area [89].
The chest wall becomes increasingly rigid due to costal cartilage calcification and structural changes in the intercostal muscles, limiting thoracic expansion and decreasing the capacity for deep inhalation [90]. The diaphragm may also experience functional decline due to age-related sarcopenia, further weakening inspiratory and expiratory forces [91]. Moreover, aging negatively impacts pulmonary immune defense mechanisms, including a significant reduction in mucociliary clearance, thereby increasing vulnerability to respiratory infections such as pneumonia and chronic obstructive pulmonary disease (COPD) [92]. Finally, the sensitivity of central and peripheral chemoreceptors responsible for detecting fluctuations in blood oxygen and carbon dioxide levels diminishes with age, leading to a blunted ventilatory response to hypoxia and hypercapnia [93].

2.5. Urogenital System

In the urinary tract, age-related changes manifest as a decrease in renal mass, particularly in the renal cortex, leading to a reduction in the number of functioning nephrons [94,95]. This decline in the nephron population contributes to a gradual decrease in the glomerular filtration rate and renal blood flow, impairing the kidneys’ ability to concentrate urine, regulate fluid balance, and maintain electrolyte homeostasis [96]. As a result, older adults may experience diminished renal clearance of waste products, making them more susceptible to dehydration, drug accumulation, and electrolyte imbalances, which can further exacerbate comorbid conditions such as hypertension and diabetes [97,98]. Additionally, the dysregulation of the renin–angiotensin–aldosterone system (RAAS) and endothelial dysfunction, marked by reduced nitric oxide production, further compromise renal function in older individuals [99,100].
The bladder also undergoes significant structural and functional changes with aging. A decrease in detrusor muscle mass, along with increased collagen deposition in the bladder wall, leads to reduced bladder compliance and impaired contractility [101]. These alterations, combined with numerous age-related neurological changes affecting autonomic bladder control, contribute to lower bladder capacity and an increased prevalence of urinary symptoms like frequency, urgency, nocturia, and incomplete emptying [102]. Additionally, age-related weakening of the urethral sphincter and pelvic floor muscles in men and women increases the risk of urinary incontinence, which can impact the overall quality of life and psychological well-being [103,104].
In men, the prostate gland commonly enlarges with age due to benign prostatic hyperplasia (BPH), which may produce urinary obstruction, hesitancy, a weak stream, and post-void dribbling [105]. In BPH, the overexpression of HSP70 subfamily members contributes to cell survival, proliferation, and epithelial–mesenchymal transition, promoting prostate enlargement [106]. Oxidative stress plays a fundamental role, with a considerable accumulation of ROS resulting in mitochondrial dysfunction and, subsequently, cellular damage [107]. Additionally, the activation of the CXCL12/CXCR4 axis moderates prostate myofibroblast phenoconversion through non-canonical EGFR/MEK/ERK signaling, contributing to tissue fibrosis [108]. If BPH is left untreated, severe cases lead to chronic urinary retention and an increased risk of urinary tract infections, bladder stones, and kidney damage [109,110,111]. In contrast, the urogenital system in older women undergoes numerous changes, particularly post menopause, due to reducing estrogen levels [112]. This hormonal shift significantly contributes to the atrophy of the vaginal and urethral epithelia, leading to dyspareunia, vaginal dryness, increased susceptibility to bacterial and fungal infections, and decreased urethral resistance, thereby increasing the risk of stress and incontinence [113]. The loss of estrogen also accelerates connective tissue degeneration, increasing the risk of pelvic organ prolapse [114].
The impact of aging extends to the reproductive system, where a progressive decline in fertility is observed in both sexes [115]. In men, testicular function diminishes, resulting in reduced testosterone production, a lower sperm count, and a decrease in sperm motility, which can contribute to subfertility [116]. Although older men retain reproductive potential throughout life, these changes may lead to decreased sexual function, including reduced libido and an increased prevalence of erectile dysfunction, often influenced by vascular, neurological, and several metabolic factors [117,118]. Women experience a more abrupt transition with menopause, marked by the cessation of ovarian function and reproductive capability [119]. The depletion of ovarian follicles leads to a substantial decline in estrogen and progesterone levels, which may reduce antioxidant capacity, thereby increasing oxidative damage. This process promotes a pro-inflammatory state characterized by elevated cytokine production, including IL-6 and TNF-α, with systemic effects on bone density and cardiovascular function [119,120,121].

2.6. Endocrine System

The aging process profoundly affects the endocrine system, resulting in significant physiological and biochemical changes that disrupt homeostasis, metabolism, and overall health [122]. At a molecular level, aging results in the dysregulation of hormonal signaling pathways, often manifested by diminished hormone production, reduced receptor sensitivity, and impaired feedback systems [123]. A critical factor in this decline is the gradual deterioration of hypothalamic function, which leads to a decrease in the secretion of regulatory hormones such as gonadotropin-releasing hormone (GnRH), growth hormone-releasing hormone (GHRH), and corticotropin-releasing hormone (CRH) [124]. This reduction leads to lower levels of peripheral hormones, including growth hormone (GH), insulin-like growth factor 1 (IGF-1), and sex steroid hormones (androgens, estrogens, and progestogens) [125].
Several factors, such as cellular senescence, oxidative stress, and inflammaging, further worsen endocrine dysfunction by impairing the responsiveness of endocrine glands [126]. Furthermore, aging induces certain post-translational modifications (PTMs), leading to alterations in proteins that affect cellular signaling, enzymatic activity, and hormone receptor functionality, further contributing to endocrine decline [127]. These PTMs include glycation, a non-enzymatic process in which reducing sugars, such as glucose and fructose, covalently bind to proteins, lipids, or nucleic acids [128]. This process results in the formation of advanced glycation end-products (AGEs), a diverse group of molecular complexes that progressively accumulate with age [129]. AGEs play a key role in cellular dysfunction, particularly in the endocrine system, as they can modify hormone receptors, disrupt their normal functioning, and alter the structure of circulating hormones [130].
The aging of pancreatic β-cells is associated with mitochondrial impairment, endoplasmic reticulum stress, and epigenetic changes, which collectively diminish insulin secretion and heighten the risk of developing type 2 diabetes [131]. In the adrenal glands, age-related modifications include disrupted cortisol secretion due to altered sensitivity to adrenocorticotropic hormone (ACTH), exacerbating stress responses and metabolic irregularities [132]. Similarly, the reduction in dehydroepiandrosterone (DHEA) secretion negatively impacts immune function and tissue repair [133]. On the other hand, changes in thyroid function, particularly the reduced bioconversion of thyroxine (T4) to triiodothyronine (T3), lead to metabolic slowdowns and greater vulnerability to age-related illnesses [134].

2.7. Musculoskeletal System

The aging process evokes a cascade of structural and functional changes that significantly reduce mobility, strength, and overall quality of life [135]. These modifications are driven by a complex interplay of mechanisms that together contribute to the progressive decline in musculoskeletal health. Among the most prominent of these factors is oxidative stress. Oxidative damage affects mitochondria, decreasing ATP production and impairing muscle function while reducing cellular energy. Moreover, the decline in repair mechanisms with age worsens the buildup of damaged molecules, organelles, and tissues [136,137].
In skeletal muscle, one of the hallmark consequences of aging is sarcopenia, characterized by the progressive loss of muscle mass and function [138]. Sarcopenia is driven by several factors, including a decrease in the number and size of muscle fibers, particularly fast-twitch type II fibers, which are responsible for rapid, high-intensity muscle contractions. This decline in muscle fibers results in reduced strength and endurance, making daily activities more difficult [139]. The ability of muscle tissue to repair itself also diminishes with age, due in part to a decline in satellite cell function [140]. Satellite cells play a critical role in muscle regeneration and repair; however, with advancing age, their ability to proliferate and differentiate into new muscle fibers decreases substantially [141]. Aging also leads to an imbalance between muscle protein synthesis and protein degradation. The reduced biosynthesis of muscle proteins and the increased breakdown of existing muscle proteins contribute to muscle atrophy [142]. Moreover, age-related hormonal changes, including a reduction in GH and testosterone levels, further impair muscle anabolism and regeneration, intensifying the overall loss of muscle mass and function [143].
Simultaneously, aging has significant consequences for bone health. In bone tissue, aging leads to an imbalance between bone formation and resorption, favoring bone loss and increasing the risk of fractures [144]. Osteoblasts, the cells responsible for bone formation, decrease in number and activity with age, while osteoclasts, the cells responsible for breaking down bone, either remain unchanged or increase in activity [145]. Oxidative stress is a key factor in this process, where increased levels of ROS cause cellular damage and induce apoptosis in osteocytes [146]. Simultaneously, there is a reduction in the osteogenic capacity of bone marrow stromal cells, indicated by a reduced expression of Runx2, an essential regulator of osteoblast differentiation [147]. The alteration in bone metabolism is also influenced by age-related changes in systemic hormones, such as estrogen, testosterone, and growth hormone, as well as local growth factors that regulate bone turnover [148,149]. Another contributor to age-related bone fragility is the accumulation of AGEs in collagen fibers within the bone matrix, leading to the increased rigidity and decreased elasticity of collagen. This process causes the bone matrix to become more brittle and more prone to fractures, further weakening bone strength [150].
The ECM also undergoes several changes with age in both muscle and bone [151,152]. In muscle tissue, the ECM becomes increasingly fibrotic, which reduces its elasticity and impairs its ability to support muscle regeneration and contraction [153]. Muscle fibrosis limits the capacity of muscle tissue to contract efficiently, contributing to reduced strength and function [154]. Additionally, changes in the composition and function of MMPs contribute to the remodeling of muscle tissue and the degradation of some structural proteins [155]. Similarly, in bone tissue, aging leads to diverse alterations in collagen crosslinking and biomineralization patterns, influencing the mechanical properties of bone [156]. These changes in the bone ECM are driven by shifts in the expression and activity of MMPs, further contributing to the increased fragility of bones in older individuals [157].
In muscle and bone tissues, epigenetic changes can disrupt the production of essential proteins necessary for tissue regeneration and maintenance, accelerating the decline in tissue function [158]. The accumulation of senescent cells in muscle and bone tissues releases several pro-inflammatory cytokines and MMPs, which aggravate tissue degeneration and impair the repair capacity of the musculoskeletal system [159].
Finally, as individuals age, the connection between the brain and muscles becomes progressively crucial for maintaining overall physical health and functional independence [160]. The brain regulates muscle movement through a network of neurons that transmit electrical signals from the CNS to activate muscle fibers and coordinate voluntary movements. However, with aging, this communication system may become less efficient [161]. The motor cortex, which is responsible for planning and executing movement, may experience a reduction in both the quantity and effectiveness of neural connections, leading to slower reaction times, impaired coordination, and a decreased ability to perform complex motor tasks [162]. The decline in neural and muscular functions elevates the risk of falls, diminishes mobility, and adversely impacts quality of life [163].

3. Chronic Inflammation in Aging

The relationship between aging and peripheral inflammation represents a complex and multifactorial process, with many molecular mechanisms contributing to a prolonged state of chronic low-grade inflammation, also called inflammaging [24,25,164,165,166]. In contrast to acute inflammation, which is a transient response to infection or injury [167], inflammaging is a persistent, low-grade inflammatory state that develops as a result of the combined influence of internal and external factors accumulated throughout life. This process is characterized by sustained immune pathway activation, the increased production of pro-inflammatory cytokines, and the dysregulation of immune homeostasis, all of which contribute to the progressive functional decline associated with aging [168].
Aging affects multiple peripheral organs, including the liver, adipose tissue, skeletal muscles, and gastrointestinal tract, all of which play a crucial role in modulating systemic inflammation [17,18,19]. The progressive dysfunction of these organs with age is primarily caused by molecular and cellular alterations, including oxidative stress, genomic instability, epigenetic changes, mitochondrial impairment, and cellular senescence [1,2,169]. All of these create an inflammatory microenvironment that evokes tissue damage, ultimately contributing to the onset and progression of many age-related diseases, including cardiovascular disorders, neurodegenerative conditions, and cancer [170,171,172]. At the molecular level, inflammaging involves a complex network of inflammatory mediators, including cytokines, acute-phase proteins, and DAMPs, which activate various intracellular signaling pathways [7,8,9,10,11,12,173].
A defining feature of inflammaging is the SASP phenotype. As aging occurs, senescent cells accumulate in several tissues, promoting a pro-inflammatory environment that supports immune system activation and drives tissue remodeling [13,14,15,16,174]. An additional factor in inflammaging is gut microbiota dysbiosis, which has become increasingly recognized as a significant regulator of systemic inflammation in aging individuals [175]. Age-related alterations in gut microbiota composition can result in increased intestinal permeability, facilitating the translocation of bacterial endotoxins, such as lipopolysaccharide (LPS), into the circulation [176]. This process, called metabolic endotoxemia, triggers sustained immune cell activation, further enhancing systemic inflammation [177].
Nevertheless, chronic inflammation in the CNS of older adults is primarily mediated by the accumulation of SASP mediators, which can cross the BBB and activate glial cells, thereby contributing to the detrimental effects of the inflammatory process. These factors not only exacerbate the neuroinflammatory response but also create a cycle of sustained immune activation that further impairs brain health, making it a significant factor in age-related neurological disorders [26,27].

3.1. Peripheral Inflammation

Chronic inflammation is considered one of the hallmarks of aging, and a key player in this process is the accumulation of senescent cells [20]. Cellular senescence is a state in which cells cease proliferation, frequently in response to many stressors, including DNA damage, telomere shortening, and oxidative stress [7,8,9,10,11,12]. These cells show high metabolic activity; they lose their ability to replicate, which makes them resistant to apoptotic signals [13]. Senescent cells (Figure 2) adopt an active state, undergoing some changes in gene expression rather than remaining inactive, thereby promoting the senescence-associated secretory phenotype (SASP). Senescent cells secrete several pro-inflammatory cytokines, including IL-6 and TNF-α, both of which play essential roles in inflammation and immune responses [178,179]. In addition to IL-6 and TNF-α, SASP factors encompass other molecules, such as IL-1β and IL-8, chemokines (e.g., CCL2 and CCL4), MMPs (e.g., MMP-3 and MMP-13), and growth factors (e.g., TGF-β and IGF-1), amplifying local inflammation and contributing to tissue remodeling and degradation [178,180,181,182]. As people age, the activity of the ubiquitin–proteasome system (UPS) decreases, leading to the strong accumulation of polyubiquitinated proteins. This decline is associated with structural changes in the proteasome, the reduced expression of proteasome subunits, and oxidative damage to proteasomal components [183]. In parallel, both autophagy and chaperone-mediated autophagy (CMA) show age-related dysfunctions, leading to a decreased formation of autophagic vacuoles and delayed fusion of autophagosomes with lysosomes, worsening the inflammatory process [184]. Furthermore, the accumulation of ROS stimulates the DNA damage response (DDR) pathway, leading to the stabilization of p53 and the upregulation of p16 and p21 [185]. The activation of p16 and p21 leads to a permanent and stable cell cycle arrest, which is a critical characteristic of senescent cells [186]. This growth arrest is mediated through the inhibition of cyclin-dependent kinases, particularly CDK4/6, leading to the inhibition of retinoblastoma protein (Rb) phosphorylation, a vital regulator of the G1/S transition in the cell cycle [187]. Furthermore, the activation of TLRs and the NLRP3 inflammasome by DAMPs released from senescent cells further amplifies the inflammatory response [188].
A key characteristic of SASP mediators is their capacity to evoke autocrine and paracrine signaling, where the signals released by senescent cells affect healthy cells [185,189]. For instance, the pro-inflammatory cytokines secreted by senescent cells can promote the activation of immune cells, such as macrophages and T lymphocytes, which in turn release more pro-inflammatory molecules, exacerbating the inflammatory environment [190,191]. The presence of SASP mediators in some tissues, such as adipose tissue, skeletal muscle, liver, and the vasculature, has been linked to several major age-related diseases [192]. In atherosclerosis, senescent cells located in the blood vessel walls contribute to the chronic inflammation that underlies plaque formation [14]. Furthermore, in osteoarthritis, senescent cells situated in the cartilage secrete factors that degrade the ECM, leading to joint dysfunction and pain [193]. Although senescence serves as a protective function by preventing the proliferation of damaged cells, its presence has detrimental long-term effects. The aging immune system becomes less efficient at clearing senescent cells, which leads to the exacerbation of inflammation [194]. In this context, aging significantly influences the clearance of damaged cells. The reduced effectiveness of immunosurveillance leads to a decreased ability of the body to identify and eliminate damaged cells [195].
A critical molecular pathway that contributes significantly to aging-related inflammation is the involvement of the NF-κB transcription factor, which plays a crucial role in the immune system’s response to stress, injury, and infection [196]. While this response is protective in younger individuals, its persistent activation in the elderly leads to an overzealous immune reaction that contributes to chronic low-grade inflammation [197]. NF-κB, activated by several stimuli (such as pro-inflammatory cytokines and ROS), triggers a cascade of downstream signaling events that enhance the production of pro-inflammatory mediators and adhesion molecules in neutrophils (such as E-selectin and VCAM-1) [198]. Conversely, one of the most consequential adverse effects of prolonged activation is the dysregulation and functional impairment of regulatory CD4+ CD25+ T cells (Tregs), which are essential for maintaining immune balance by controlling excessive immune responses [199]. With advancing age, Tregs experience a decline in function, reducing their ability to regulate immune activation. This dysfunction in Tregs leads to the breakdown of the finely tuned regulation of immune responses, promoting an imbalance in CD4+ T-cell activity and exacerbating tissue damage [199,200]. Additionally, CD4+ CD25+ Tregs are susceptible to mitochondrial dysfunction, which is characterized by decreased mitochondrial protein levels and impaired oxidative phosphorylation, along with increased mitochondrial oxidative stress [201]. Tregs undergo alterations in mitochondrial dynamics, including a reduction in mitochondrial mass, a compromised membrane potential, and the accumulation of damaged mitochondria with abnormal morphology, resulting in instability in FoxP3 expression and a reduced capacity to sustain immune homeostasis [202]. Furthermore, mitochondrial dysfunction in aging Tregs is further amplified by impairments in mitophagy (the process responsible for eliminating damaged mitochondria), leading to a self-perpetuating cycle of oxidative damage and cellular senescence [201].
Nevertheless, the aging immune system undergoes additional functional alterations, commonly referred to as immunosenescence. This process encompasses the reduction in the efficiency and capacity of immune responses [203]. In the innate immune system, macrophage and neutrophil activity decreases due to reduced signaling from GM-CSF and impaired TLRs function in monocytes, macrophages, and dendritic cells (DCs). Therefore, these immune cells become less efficient at detecting and responding to pathogens, resulting in a delayed immune response [204]. Likewise, the adaptive immune system suffers a decline in T- and B-cell functionality, compromising their ability to recognize specific pathogens and generate many specific immune responses [205]. This impairment manifests as a reduced ability to produce antibodies after vaccination or infection, making older individuals more susceptible to many infections, including influenza and pneumonia [206].
On the other hand, the molecular mechanisms driving age-related inflammation and vascular dysfunction involve a network of interconnected pathways, each contributing to the multifaceted phenotype of aging. At the epigenetic level, DNA methylation undergoes significant changes with age, marked by global hypomethylation and site-specific hypermethylation [207]. These alterations lead to the activation of numerous pro-inflammatory genes, such as IL-6, TNF-α, and COX-2, while simultaneously suppressing anti-inflammatory factors like IL-10 and TGF-β [208,209]. Histone modifications are vital in chromatin remodeling during aging, with a strong reduction in H3K9 trimethylation and an increase in H4K16 acetylation being particularly notable [210,211,212]. Moreover, the expression of non-coding RNAs, especially microRNAs, is altered with age. For instance, the upregulation of miR-21 promotes vascular inflammation by targeting PTEN and activating the AKT pathway, while the downregulation of miR-126 impairs endothelial function by reducing VEGF signaling [213,214].
The interaction between inflammation and obesity in aging creates a network of molecular processes that significantly impacts health and longevity. Obesity, a metabolic disorder in the elderly [215], induces adipocyte hypertrophy and hyperplasia, leading to a significant increase in the secretion of pro-inflammatory adipokines such as leptin and resistin [216,217]. These adipokines initiate a cascade of inflammatory events, activating mainly mast cells and CD4+ T cells to release numerous pro-inflammatory cytokines, including IL-6, IL-12, IL-17, IL-18, and TNF-α [218]. Simultaneously, the expansion of adipose tissue linked to obesity results in localized hypoxia, activating the NF-κB pathway and further enhancing the production of pro-inflammatory mediators [219]. Furthermore, excess adipose tissue acts as an endocrine organ, secreting bioactive molecules that disturb the metabolic balance, including altered lipid metabolism and increased circulating free fatty acids, exacerbating oxidative stress and mitochondrial dysfunction in immune cells [220]. The cumulative effect of these processes establishes a self-perpetuating cycle of inflammation and metabolic dysregulation, known as adipaging, which not only accelerates the aging process but also elevates the risk of age-related diseases such as type 2 diabetes [221].

3.2. Central Inflammation

The detrimental influence of SASP mediators on the CNS arises through intricate molecular mechanisms that undermine the structural and functional integrity of the BBB. The BBB is a highly specialized and selective permeability barrier composed of endothelial cells interconnected by tight junctions, primarily consisting of transmembrane proteins such as claudins, occludin, and zonula occludens (ZO) proteins, supported by pericytes and astrocytes [222]. Under physiological conditions, this barrier plays an essential role in maintaining CNS homeostasis by regulating the exchange of ions, nutrients, and signaling molecules while restricting the entry of circulating immune cells and neurotoxic agents [223]. However, chronic exposure of the BBB to SASP factors impairs its selective permeability, resulting in widespread neurovascular dysfunction [224]. As the BBB integrity declines with age, peripheral inflammatory mediators and immune cells are able to infiltrate the CNS [225].
Among the key SASP factors implicated in BBB breakdown are a range of pro-inflammatory cytokines, including IL-1β, IL-6, and TNF-α, in addition to MMPs (such as MMP-2 and MMP-9), which directly degrade tight junction proteins [226]. This disruption facilitates the increased permeability of the BBB, allowing the infiltration of peripheral immune cells, ROS, and additional inflammatory mediators into the CNS parenchyma [227]. The extravasation of these harmful agents exacerbates neuroinflammation by triggering the activation of resident microglial cells, which play a crucial role in the innate immune response within the CNS [228]. Microglial activation is mainly mediated via TLR signaling cascades and the NF-κB transcriptional pathway, leading to a feedforward loop of pro-inflammatory cytokine release that reinforces neuronal damage [229]. The mentioned cytokines activate microglia through TLR4, resulting in the nuclear translocation of NF-κB and subsequent amplification of inflammatory responses [230]. Simultaneously, the activation of the NLRP3 inflammasome in microglia enhances the release of mature IL-1β, perpetuating neuroinflammation [231].
A consequence of sustained microglial activation (Figure 3) is the excessive production of reactive oxygen and nitrogen species (ROS and RNS), mainly mediated by inducible nitric oxide synthase (iNOS) and NADPH oxidase [232,233]. This overproduction creates a highly pro-inflammatory and cytotoxic environment within the CNS, disrupting cellular homeostasis. The accumulation of ROS exacerbates mitochondrial dysfunction, leading to impaired ATP synthesis and energy deficits in neurons. The energy deficiency compromises several neuronal processes, including synaptic transmission, plasticity, and cell viability [234]. Oxidative stress disrupts several redox-sensitive signaling pathways, triggering molecular damage like DNA oxidation, lipid peroxidation, and protein misfolding [235].
In addition to mitochondrial dysfunction, the interplay between neuroinflammation and oxidative stress exacerbates excitotoxicity. Overactivated microglia evoke excessive glutamate release while simultaneously impairing its reuptake by astrocytes, resulting in glutamate accumulation within the synaptic cleft [236]. This causes the overactivation of NMDA and AMPA receptors, leading to an aberrant influx of calcium into neurons [237]. The increased intracellular calcium activates multiple neurotoxic pathways, including the stimulation of calpains and caspases (proteolytic enzymes that degrade essential cellular components and drive apoptotic and necrotic cell death) [238,239].
At the molecular level, elevated calcium influx disrupts calcium-dependent signaling cascades, exacerbating mitochondrial stress and stimulating the additional release of ROS [240]. This feedback loop perpetuates oxidative damage and cellular dysfunction, creating a self-sustaining cycle of neurodegeneration. Moreover, neuronal death amplifies microglial activation, exacerbating inflammation and oxidative stress, which in turn accelerates disease progression [241].
With respect to astroglia, this cell type plays a key role in the inflammatory processes within the CNS. Astrocytes are crucial in driving chronic neuroinflammation, thereby disturbing homeostasis and increasing the vulnerability of neurons to degeneration and cell death [242]. Astrocytes, the most abundant glial cells in the CNS, contribute to neuroinflammation via the activation of fundamental signaling pathways. Two major transcription factors, NF-κB and AHR, are central players in this regulatory network. NF-κB is a rapidly inducible factor that governs the expression of many pro-inflammatory genes, including cytokines (such as IL-6 and TNF-α), chemokines (such as CCL2 and CXCL10), and adhesion molecules (e.g., ICAM-1) [243]. In contrast, AHR has been identified as a potential negative regulator of NF-κB-mediated inflammation, modulating the intensity of inflammatory responses through strong interactions with cytochrome P450 enzymes [244]. The dysregulation of these pathways in astrocytes contributes to a persistent pro-inflammatory state, exacerbating neurodegeneration [245]. Moreover, astrocytes respond to these cytokines, further amplifying inflammatory signaling through JAK/STAT pathways [246].
Oxidative stress is another critical component of age-related neuroinflammation. The accumulation of ROS due to mitochondrial dysfunction leads to the oxidative damage of cellular components, such as lipids, proteins, and DNA [12]. Aged microglia and astrocytes exhibit impaired antioxidant responses, further exacerbating ROS accumulation [247,248]. This oxidative stress not only directly damages neurons but also activates NF-κβ, thereby sustaining the inflammatory cycle. Dysfunctional mitochondria in aging CNS cells exhibit reduced oxidative phosphorylation efficiency, increased mitochondrial DNA mutations, and enhanced cytochrome c release, triggering apoptotic pathways [249].
Aging-related inflammation also affects neurotransmitter systems, disrupting synaptic plasticity and neuronal communication. Pro-inflammatory cytokines modulate neurotransmitter release and receptor expression, leading to excitotoxicity and impaired synaptic function. For example, TNF-α has been shown to disrupt glutamate homeostasis by decreasing the expression of various glutamate transporters (GLT-1 and GLAST) in astrocytes, leading to elevated extracellular glutamate levels and facilitating excitotoxic neuronal damage through excessive NMDA receptor stimulation [250]. Persistent neuroinflammatory and oxidative conditions alter synaptic plasticity by modulating the expression and functionality of key synaptic proteins, including postsynaptic density protein 95 (PSD-95), synaptophysin, and glutamatergic receptor subunits [251]. The breakdown of these proteins impairs long-term potentiation (LTP) and long-term depression (LTD), critical mechanisms underlying learning and memory [252,253].
Moreover, chronic neuroinflammation affects dendritic spine morphology, contributing to the synaptic loss and cognitive decline observed in numerous neurodegenerative diseases [254]. Beyond synaptic dysfunction, the sustained presence of SASP mediators promotes τ hyperphosphorylation, a hallmark of Alzheimer’s disease, through the dysregulation of kinases such as glycogen synthase kinase 3β (GSK-3β) and cyclin-dependent kinase 5 (CDK5). Hyperphosphorylated τ aggregates into insoluble neurofibrillary tangles (NFTs), breaking axonal transport and neuronal communication [255,256]. In Parkinson’s disease, SASP-mediated inflammation enhances α-synuclein misfolding and aggregation via impaired autophagy–lysosomal degradation and proteasomal dysfunction, leading to the accumulation of oligomers that intensify dopaminergic neurodegeneration [257,258]. The persistence of misfolded protein aggregates, coupled with chronic inflammatory and oxidative stress conditions, drives neuronal apoptosis through both caspase-dependent and caspase-independent pathways [259]. Mitochondrial outer membrane permeabilization (MOMP), mediated by the pro-apoptotic Bcl-2 family proteins BAX and BAK, initiates apoptosis by releasing cytochrome c and activating caspase-3 and caspase-9 [260]. Additionally, the excessive activation of PARP-1 in response to DNA damage contributes to parthanatos, a form of programmed cell death distinct from classical apoptosis, further exacerbating neuronal loss [261]. In contrast to classical apoptosis, which is marked by well-defined morphological and biochemical hallmarks such as cell shrinkage, chromatin condensation, and membrane blebbing [262], parthanatos is characterized by a unique series of cellular events, including the overproduction of PAR chains, mitochondrial dysfunction, and the release of apoptosis-inducing factor (AIF) from mitochondria, which subsequently translocates to the nucleus to trigger chromatin condensation and DNA fragmentation [263]. Parthanatos causes irreversible neuronal damage and worsens neuronal loss, particularly in neurodegenerative diseases and acute neuronal injuries, where it interacts with other cell death pathways to accelerate tissue degeneration [264].

4. Current Anti-Inflammatory Strategies for Aging

Adopting a healthy lifestyle has long been recognized as one of the most effective strategies for maintaining overall well-being and reducing the impact of aging. A growing body of research supports the idea that some lifestyle factors (e.g., proper nutrition, moderate physical activity, and mental stability) contribute to the mitigation of the aging process. A well-balanced and sufficient nutrient intake has been shown to positively impact aging, with numerous studies suggesting that nutritional factors play an integral role in the modulation of age-related molecular mechanisms. Long-term adherence to a diet rich in polyphenols, naturally occurring compounds in fruits and vegetables, has been particularly associated with significant improvements in intestinal permeability [265,266]. This effect may help maintain the integrity of the intestinal barrier, thereby reducing systemic inflammation [267]. Moreover, polyphenolic compounds have been demonstrated to activate many antioxidant and anti-inflammatory pathways, thus mitigating oxidative stress and promoting cellular resilience [268].
On the other hand, research has revealed that the intake of specific probiotic strains, particularly Lactobacillus pentosus var. plantarum C29, can reduce SASP factors [269]. The modulation of the gut microbiota through probiotic supplementation results in a reduction in pro-inflammatory cytokine levels and has the potential to decrease the expression of age-associated biomarkers, including tumor suppressor proteins like p16 and p53 [270]. These markers are crucial for cellular senescence and apoptosis, and their regulation has been linked to slowing age-associated cellular dysfunction [271].
Similarly, the intake of polyunsaturated fatty acids (PUFAs), particularly ω-3 fatty acids, has been shown to reduce the levels of pro-inflammatory cytokines [272]. It is thought that reducing inflammation helps preserve metabolic and immune balance, thereby supporting healthier aging [273]. The anti-inflammatory effects of PUFAs extend beyond their role in lipid metabolism and influence gene expression by activating some nuclear receptors such as peroxisome proliferator-activated receptors (PPARs), which regulate lipid homeostasis and inflammatory signaling pathways [274].
Furthermore, the frequent intake of essential vitamins such as vitamin C and vitamin E has been demonstrated to enhance immune cell functions in older adults. Vitamin C, a significant antioxidant, is vital for the proper function of neutrophils, which are critical to immune defense [275]. Specifically, vitamin C facilitates neutrophil chemotaxis and phagocytosis, thereby augmenting the body’s ability to fight infections [276]. Vitamin E, known for its potent antioxidant properties, also plays a fundamental role in stabilizing cellular membranes and protecting immune cells from oxidative damage [277].
Beyond these vitamins, mineral supplementation, particularly zinc, has been identified as a critical factor in supporting immune homeostasis during aging [278]. Zinc is involved in the regulation of a variety of immune processes, including the differentiation and function of T cells [279]. Studies indicate that zinc supplementation can increase the naive T-cell population, which is essential for the body’s ability to respond to new pathogens [280]. Furthermore, zinc contributes to balancing the activity of CD4+ T cells, enhancing the immune response and reducing the negative impact of age-related immune dysfunction [281].
Exercise is widely recognized as a highly effective and accessible strategy for mitigating the aging process, functioning through complex biological mechanisms that counteract DNA damage, oxidative stress, and the overall decline in cellular function that often accompanies aging [282]. Recent studies have underscored its role in delaying the aging process at the molecular level, especially through its capacity to regulate telomere length and the activity of crucial enzymes such as telomerase [283]. Research examining middle-aged marathon runners and triathletes has shown that these individuals exhibit significantly higher telomerase activity along with longer telomeres in their circulating white blood cells compared to their sedentary counterparts. This finding suggests that endurance exercise not only helps maintain telomere integrity but may also promote the rejuvenation of immune cells, enhancing overall immune function and mitigating age-related declines in immune response [284]. This evidence indicates that endurance exercise helps preserve telomere integrity and rejuvenates immune cells, improving immune function and delaying age-related immune decline. The underlying mechanisms likely involve reducing oxidative stress, modulating inflammation, and activating genes that promote cell survival and repair [285,286]. In contrast, resistance training offers benefits for combatting aging at the cellular level, with studies showing significant improvements in the health of adipose tissue in older and obese patients [287]. It was discovered that resistance exercise led to a reduction in the number of cells expressing p16, a protein marker commonly associated with cellular senescence [288]. By reducing the number of p16-expressing cells in thigh adipose tissue, resistance training appears to foster a more youthful and functional tissue environment, potentially by promoting the turnover of damaged cells and alleviating inflammation in adipose tissue [289].
Emerging research has increasingly highlighted the pro-inflammatory cytokine network as a crucial target for anti-aging interventions, focusing on the potential to alleviate chronic inflammation, which is one of the key drivers of age-related diseases and cellular dysfunction [290]. In light of this, various anti-inflammatory compounds have garnered significant attention for their ability to counteract inflammation, promote cellular health, and potentially delay the onset of age-related decline. Among these, metformin, a widely used medication for managing type 2 diabetes, has emerged as a promising candidate for promoting healthy aging [291]. Metformin has been shown to influence the IKK/NF-κB pathway, a primary regulator of inflammation, as well as the GPX7/NRF2 axis, which is essential for cellular defense against oxidative stress and the regulation of antioxidant responses [292,293]. Recent research has revealed that metformin interacts with PEN2, an identified target involved in age-related inflammation [294]. By modulating all of these pathways, metformin reduces inflammation and promotes healthier aging, potentially extending lifespan [291]. Similarly, aspirin reduces oxidative damage, maintains tissue function, promotes cellular regeneration, and eliminates SASP mediators, thereby presenting a potential therapeutic strategy for age-related tissue degeneration [295].
Finally, among the broad spectrum of cellular anti-aging therapies, senolytics (agents designed to selectively eliminate senescent cells) emerge as one of the most promising yet highly debated strategies in gerontology [296]. Since their discovery in 2015 [297], several compounds have moved from initial discovery to clinical trials, resulting in considerable enthusiasm within the scientific community. Senolytic drugs can be classified into phytochemicals and proprietary compounds, differing in their origin, composition, regulatory oversight, and mechanisms of action. Phytochemicals are naturally occurring compounds derived from plants, often associated with potential health benefits and therapeutic properties. They are found in several foods, herbal medicines, and dietary supplements, with their biological effects typically attributed to the synergistic interactions of multiple bioactive constituents [297,298,299]. In contrast, proprietary drugs are synthetic compounds developed by pharmaceutical companies. These drugs undergo thorough testing, including clinical trials, to assess their safety, efficacy, and standardized dosing before gaining regulatory approval [299,300].
Among the most studied senolytic agents are dasatinib and quercetin. Dasatinib has shown effectiveness in eliminating senescent human adipocyte progenitor cells, which reside in adipose tissue and contribute to age-related metabolic dysfunction [301,302]. Quercetin has proven potent in clearing senescent endothelial cells, which line the blood vessels, as well as bone marrow stem cells in murine models [303]. These two drugs constitute a combination therapy that has shown significant efficacy in targeting various types of senescent cells, thereby reducing inflammation, enhancing tissue function, and alleviating the burden of age-related diseases in animal models [301,304,305,306]. Fenofibrate, primarily used for cholesterol management, has emerged as a promising therapeutic candidate due to its ability to clear senescent cells by upregulating PPARα [307]. Ultimately, taurine and spermidine have emerged as promising senolytic agents in the context of aging, demonstrating potential in promoting cellular homeostasis and mitigating age-related decline. Taurine, a conditionally essential amino acid, has been shown to enhance mitochondrial function, reduce oxidative stress, and modulate inflammatory responses, thereby contributing to the clearance of senescent cells [308]. Similarly, spermidine, a polyamine present in youthful cells, has been strongly implicated in autophagy induction, chromatin stabilization, and the suppression of pro-inflammatory pathways, all of which are fundamental for maintaining cellular integrity and delaying age-associated pathologies [309].

5. Conclusions

In conclusion, this review presents a thorough analysis of the complex biological processes that underpin aging and its connection to various pathological conditions. Aging encompasses several intricate physiological changes that significantly impact the body’s ability to maintain homeostasis. This loss of homeostasis plays a role in the development of chronic diseases commonly associated with aging, such as cardiovascular disease and neurodegenerative disorders, by disrupting cellular activities, impairing the body’s ability to repair tissues, and increasing its vulnerability to inflammation and oxidative stress.
One of the key themes explored in this article is the role of inflammation in the aging process. Chronic low-grade inflammation, also known as inflammaging, is highlighted as a hallmark of aging that drives the progression of these age-related diseases. Unlike the acute, short-lived inflammation that arises from injury or infection, inflammaging is a sustained, low-grade inflammation that can inflict lasting damage on the body. This chronic inflammatory state accelerates the decline of cellular and tissue function by inducing tissue damage and impairing the body’s capacity for self-repair.
Furthermore, this article explores the connection between aging and immune system dysfunction, which plays a key role in the initiation and progression of diseases in older adults. Immunosenescence, the progressive deterioration of immune function, impairs the body’s ability to protect against pathogens and injuries. As the immune system weakens, older individuals become more susceptible to infections, autoimmune disorders, and cancer. These processes not only induce inflammation but also result in the accumulation of dysfunctional cells, thereby exacerbating the pathological effects of aging.
This research highlights the importance of identifying therapeutic strategies to mitigate the effects of aging and chronic inflammation. Targeting specific inflammatory mediators and promoting immune rejuvenation could play a fundamental role in extending healthspan (the period of life spent in good health). Furthermore, delaying cellular senescence, the process by which cells lose their ability to divide and function, could help maintain tissue integrity and enhance overall health in aging populations.
Finally, this review calls for a more cohesive healthcare approach for the aging population, one that addresses not only the symptoms of age-related diseases but also the root causes of aging itself. By focusing on the fundamental mechanisms of aging and chronic inflammation and implementing personalized interventions, it is possible to encourage healthier aging, enhance quality of life, and increase the resilience of older adults.

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 author declares no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
4-HNE4-hydroxy-2-nonenal
ACTHAdrenocorticotropic hormone
AGEAdvanced glycation end-product
AHRAryl hydrocarbon receptor
AIFApoptosis-inducing factor
AKT/PKBProtein kinase B
AMPAAlpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
ATPAdenosine triphosphate
BAKBcl-2 homologous antagonist/killer
BAXBcl-2-associated X protein
BBBBlood–brain barrier
Bcl-2B-cell lymphoma 2
BPHBenign prostatic hyperplasia
CCL2C-C motif chemokine ligand 2
CCL4C-C motif chemokine ligand 4
CD25Cluster of differentiation 25
CD4Cluster of differentiation 4
CDK4/6Cyclin-dependent kinases 4 and 6
CDK5Cyclin-dependent kinase 5
CMAChaperone-mediated autophagy
CNSCentral nervous system
COPDChronic obstructive pulmonary disease
COX-1Cyclooxygenase 1
COX-2Cyclooxygenase 2
CRHCorticotropin-releasing hormone
CXCL10C-X-C motif chemokine ligand 10
CXCL12C-X-C motif chemokine ligand 12
CXCR4C-X-C chemokine receptor type 4
CYP3A4Cytochrome P450 3A4
DAMPsDamage-associated molecular patterns
DCDendritic cell
DDRDNA damage response
DHEADehydroepiandrosterone
DNADeoxyribonucleic acid
DPPCDipalmitoylphosphatidylcholine
ECMExtracellular matrix
EGFREpidermal growth factor receptor
eNOSEndothelial nitric oxide synthase
ERKExtracellular signal-regulated kinase 1/2
FoxP3Forkhead box P3
GDF-15Growth differentiation factor 15
GERDGastroesophageal reflux disease
GHGrowth hormone
GHRHGrowth hormone-releasing hormone
GIGastrointestinal system
GLASTGlutamate aspartate transporter
GLT-1Glutamate transporter 1
GM-CSFGranulocyte–macrophage colony-stimulating factor
GnRHGonadotropin-releasing hormone
GPX7Glutathione peroxidase 7
GSK-3βGlycogen synthase kinase 3 beta
HSP7070 kDa heat shock protein
ICAM-1Intercellular adhesion molecule 1
IFN-γInterferon gamma
IGF-1Insulin-like growth factor 1
IGFBP-2Insulin-like growth factor binding protein 2
IKKIκB Kinase
IL-10Interleukin 10
IL-12Interleukin 12
IL-17Interleukin 17
IL-18Interleukin 18
IL-1αInterleukin 1 alpha
IL-1βInterleukin 1 beta
IL-2Interleukin 2
IL-4Interleukin 4
IL-6Interleukin 6
IL-8Interleukin 8
iNOSInducible nitric oxide synthase
LPSLipopolysaccharide
LTDLong-term depression
LTPLong-term potentiation
MAPKMitogen-activated protein kinase
MDAMalondialdehyde
MEKMitogen-activated protein kinase kinase
MMPMatrix metalloproteinase
MMP-13Matrix metalloproteinase 13
MMP-2Matrix metalloproteinase 2
MMP-3Matrix metalloproteinase 3
MMP-9Matrix metalloproteinase 9
MOMPMitochondrial outer membrane permeabilization
NADPHNicotinamide adenine dinucleotide phosphate (reduced form)
NFTNeurofibrillary tangle
NF-κBNuclear factor kappa-light-chain-enhancer of activated B cells
NLRP3NLR family pyrin domain containing 3
NMDAN-methyl-D-aspartate
NRF2Nuclear factor erythroid 2-related factor 2
NSAIDNon-steroidal anti-inflammatory drug
p16Cyclin-dependent kinase inhibitor 2A
p21Cyclin-dependent kinase inhibitor 1
P450Cytochrome P450
p53Tumor protein P53
PARPoly (ADP-ribose)
PARP-1Poly (ADP-ribose) polymerase 1
PEN2Presenilin enhancer 2
PI3KPhosphoinositide 3-kinase
PPARPeroxisome proliferator-activated receptor
PPARαPeroxisome proliferator-activated receptor alpha
PSD-95Postsynaptic density protein 95
PTENPhosphatase and tensin homolog
PTMPost-translational modification
PUFAPolyunsaturated fatty acid
RAASRenin–angiotensin–aldosterone system
RbRetinoblastoma
RNARibonucleic acid
RNSReactive nitrogen species
ROSReactive oxygen species
Runx2Runt-related transcription factor 2
SASPSenescence-associated secretory phenotype
SCFAShort-chain fatty acid
T3Triiodothyronine
T4Thyroxine
TGF-βTransforming growth factor beta
TIMPTissue inhibitor of metalloproteinase
TLRToll-like receptor
TLR4Toll-like receptor 4
TNFRTumor necrosis factor receptor
TNF-αTumor necrosis factor alpha
TP53Tumor protein p53
TregRegulatory T cell
UPSUbiquitin–proteasome system
VCAM-1Vascular cell adhesion molecule 1
VEGFVascular endothelial growth factor
ZOZonula occludens

References

  1. Guo, J.; Huang, X.; Dou, L.; Yan, M.; Shen, T.; Tang, W.; Li, J. Aging and aging-related diseases: From molecular mechanisms to interventions and treatments. Signal Transduct. Target. Ther. 2022, 7, 391. [Google Scholar] [CrossRef]
  2. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. Hallmarks of aging: An expanding universe. Cell 2023, 186, 243–278. [Google Scholar] [CrossRef] [PubMed]
  3. Jaul, E.; Barron, J. Age-Related Diseases and Clinical and Public Health Implications for the 85 Years Old and Over Population. Front. Public Health 2017, 5, 335. [Google Scholar] [CrossRef] [PubMed]
  4. Li, Y.; Tian, X.; Luo, J.; Bao, T.; Wang, S.; Wu, X. Molecular mechanisms of aging and anti-aging strategies. Cell Commun. Signal. 2024, 22, 285. [Google Scholar] [CrossRef]
  5. da Silva, P.F.L.; Schumacher, B. Principles of the Molecular and Cellular Mechanisms of Aging. J. Investig. Dermatol. 2021, 141, 951–960. [Google Scholar] [CrossRef] [PubMed]
  6. Preston, J.; Biddell, B. The physiology of ageing and how these changes affect older people. Medicine 2020, 49, 1–5. [Google Scholar] [CrossRef]
  7. Vijg, J.; Suh, Y. Genome instability and aging. Annu. Rev. Physiol. 2013, 75, 645–668. [Google Scholar] [CrossRef]
  8. Schumacher, B.; Pothof, J.; Vijg, J.; Hoeijmakers, J.H.J. The central role of DNA damage in the ageing process. Nature 2021, 592, 695–703. [Google Scholar] [CrossRef]
  9. Rossiello, F.; Jurk, D.; Passos, J.F.; d’Adda di Fagagna, F. Telomere dysfunction in ageing and age-related diseases. Nat. Cell Biol. 2022, 24, 135–147. [Google Scholar] [CrossRef]
  10. Dhar, P.; Moodithaya, S.S.; Patil, P. Epigenetic alterations-The silent indicator for early aging and age-associated health-risks. Aging Med. 2022, 5, 287–293. [Google Scholar] [CrossRef]
  11. Hipp, M.S.; Kasturi, P.; Hartl, F.U. The proteostasis network and its decline in ageing. Nat. Rev. Mol. Cell Biol. 2019, 20, 421–435. [Google Scholar] [CrossRef] [PubMed]
  12. Srivastava, S. The Mitochondrial Basis of Aging and Age-Related Disorders. Genes 2017, 8, 398. [Google Scholar] [CrossRef]
  13. Zhang, L.; Pitcher, L.E.; Yousefzadeh, M.J.; Niedernhofer, L.J.; Robbins, P.D.; Zhu, Y. Cellular senescence: A key therapeutic target in aging and diseases. J. Clin. Investig. 2022, 132, e158450. [Google Scholar] [CrossRef] [PubMed]
  14. Sun, Y.; Wang, X.; Liu, T.; Zhu, X.; Pan, X. The multifaceted role of the SASP in atherosclerosis: From mechanisms to therapeutic opportunities. Cell Biosci. 2022, 12, 74. [Google Scholar] [CrossRef] [PubMed]
  15. Evans, D.S.; Young, D.; Tanaka, T.; Basisty, N.; Bandinelli, S.; Ferrucci, L.; Campisi, J.; Schilling, B. Proteomic Analysis of the Senescence-Associated Secretory Phenotype: GDF-15, IGFBP-2, and Cystatin-C Are Associated with Multiple Aging Traits. J. Gerontol. A Biol. Sci. Med. Sci. 2024, 79, glad265. [Google Scholar] [CrossRef] [PubMed]
  16. Basisty, N.; Kale, A.; Jeon, O.H.; Kuehnemann, C.; Payne, T.; Rao, C.; Holtz, A.; Shah, S.; Sharma, V.; Ferrucci, L.; et al. A proteomic atlas of senescence-associated secretomes for aging biomarker development. PLoS Biol. 2020, 18, e3000599. [Google Scholar] [CrossRef]
  17. Hu, C.; Zhang, X.; Teng, T.; Ma, Z.G.; Tang, Q.Z. Cellular Senescence in Cardiovascular Diseases: A Systematic Review. Aging Dis. 2022, 13, 103–128. [Google Scholar] [CrossRef]
  18. Martínez-Cué, C.; Rueda, N. Cellular Senescence in Neurodegenerative Diseases. Front. Cell. Neurosci. 2020, 14, 16. [Google Scholar] [CrossRef]
  19. Schmitt, C.A.; Wang, B.; Demaria, M. Senescence and cancer—role and therapeutic opportunities. Nat. Rev. Clin. Oncol. 2022, 19, 619–636. [Google Scholar] [CrossRef]
  20. Escobar, A.P.; Bonansco, C.; Cruz, G.; Dagnino-Subiabre, A.; Fuenzalida, M.; Negrón, I.; Sotomayor-Zárate, R.; Martínez-Pinto, J.; Jorquera, G. Central and Peripheral Inflammation: A Common Factor Causing Addictive and Neurological Disorders and Aging-Related Pathologies. Int. J. Mol. Sci. 2023, 24, 10083. [Google Scholar] [CrossRef]
  21. Li, X.; Li, C.; Zhang, W.; Wang, Y.; Qian, P.; Huang, H. Inflammation and aging: Signaling pathways and intervention therapies. Signal Transduct. Target Ther. 2023, 8, 239. [Google Scholar] [CrossRef] [PubMed]
  22. Vannella, K.M.; Wynn, T.A. Mechanisms of Organ Injury and Repair by Macrophages. Annu. Rev. Physiol. 2017, 79, 593–617. [Google Scholar] [CrossRef] [PubMed]
  23. Scalise, R.F.M.; De Sarro, R.; Caracciolo, A.; Lauro, R.; Squadrito, F.; Carerj, S.; Bitto, A.; Micari, A.; Bella, G.D.; Costa, F.; et al. Fibrosis after Myocardial Infarction: An Overview on Cellular Processes, Molecular Pathways, Clinical Evaluation and Prognostic Value. Med. Sci. 2021, 9, 16. [Google Scholar] [CrossRef] [PubMed]
  24. Franceschi, C.; Garagnani, P.; Parini, P.; Giuliani, C.; Santoro, A. Inflammaging: A new immune-metabolic viewpoint for age-related diseases. Nat. Rev. Endocrinol. 2018, 14, 576–590. [Google Scholar] [CrossRef]
  25. Dugan, B.; Conway, J.; Duggal, N.A. Inflammaging as a target for healthy ageing. Age Ageing 2023, 52, afac328. [Google Scholar] [CrossRef]
  26. Wu, D.; Chen, Q.; Chen, X.; Han, F.; Chen, Z.; Wang, Y. The blood-brain barrier: Structure, regulation, and drug delivery. Signal. Transduct. Target. Ther. 2023, 8, 217. [Google Scholar] [CrossRef]
  27. Silva, B.A.; Farías, M.I.; Miglietta, E.A.; Leal, M.C.; Ávalos, J.C.; Pitossi, F.J.; Ferrari, C.C. Understanding the role of the blood brain barrier and peripheral inflammation on behavior and pathology on ongoing confined cortical lesions. Mult. Scler. Relat. Disord. 2022, 57, 103346. [Google Scholar] [CrossRef]
  28. Miao, J.; Ma, H.; Yang, Y.; Liao, Y.; Lin, C.; Zheng, J.; Yu, M.; Lan, J. Microglia in Alzheimer’s disease: Pathogenesis, mechanisms, and therapeutic potentials. Front. Aging Neurosci. 2023, 15, 1201982. [Google Scholar] [CrossRef]
  29. Ho, M.S. Microglia in Parkinson’s Disease. Adv. Exp. Med. Biol. 2019, 1175, 335–353. [Google Scholar]
  30. Gao, C.; Jiang, J.; Tan, Y.; Chen, S. Microglia in neurodegenerative diseases: Mechanism and potential therapeutic targets. Signal Transduct. Target Ther. 2023, 8, 359. [Google Scholar] [CrossRef]
  31. Xu, Y.; Gao, W.; Sun, Y.; Wu, M. New insight on microglia activation in neurodegenerative diseases and therapeutics. Front. Neurosci. 2023, 17, 1308345. [Google Scholar] [CrossRef]
  32. Zhang, Y.; Chen, H.; Li, R.; Sterling, K.; Song, W. Amyloid β-based therapy for Alzheimer’s disease: Challenges, successes and future. Signal Transduct. Target. Ther. 2023, 8, 248. [Google Scholar] [CrossRef]
  33. Calabresi, P.; Mechelli, A.; Natale, G.; Volpicelli-Daley, L.; Di Lazzaro, G.; Ghiglieri, V. Alpha-synuclein in Parkinson’s disease and other synucleinopathies: From overt neurodegeneration back to early synaptic dysfunction. Cell Death Dis 2023, 14, 176. [Google Scholar] [CrossRef] [PubMed]
  34. Tian, Y.E.; Cropley, V.; Maier, A.B.; Lautenschlager, N.T.; Breakspear, M.; Zalesky, A. Heterogeneous aging across multiple organ systems and prediction of chronic disease and mortality. Nat. Med. 2023, 29, 1221–1231. [Google Scholar] [CrossRef] [PubMed]
  35. Khalil, R.; Diab-Assaf, M.; Lemaitre, J.M. Emerging Therapeutic Approaches to Target the Dark Side of Senescent Cells: New Hopes to Treat Aging as a Disease and to Delay Age-Related Pathologies. Cells 2023, 12, 915. [Google Scholar] [CrossRef] [PubMed]
  36. Kesidou, E.; Theotokis, P.; Damianidou, O.; Boziki, M.; Konstantinidou, N.; Taloumtzis, C.; Sintila, S.A.; Grigoriadis, P.; Evangelopoulos, M.E.; Bakirtzis, C.; et al. CNS Ageing in Health and Neurodegenerative Disorders. J. Clin. Med. 2023, 12, 2255. [Google Scholar] [CrossRef]
  37. Salim, S. Oxidative Stress and the Central Nervous System. J. Pharmacol. Exp. Ther. 2017, 360, 201–205. [Google Scholar] [CrossRef]
  38. Jové, M.; Mota-Martorell, N.; Pradas, I.; Martín-Gari, M.; Ayala, V.; Pamplona, R. The Advanced Lipoxidation End-Product Malondialdehyde-Lysine in Aging and Longevity. Antioxidants 2020, 9, 1132. [Google Scholar] [CrossRef]
  39. Li, Y.; Zhao, T.; Li, J.; Xia, M.; Li, Y.; Wang, X.; Liu, C.; Zheng, T.; Chen, R.; Kan, D.; et al. Oxidative Stress and 4-hydroxy-2-nonenal (4-HNE): Implications in the Pathogenesis and Treatment of Aging-related Diseases. J. Immunol. Res. 2022, 2022, 2233906. [Google Scholar] [CrossRef]
  40. Liu, M.Y.; Nemes, A.; Zhou, Q.G. The Emerging Roles for Telomerase in the Central Nervous System. Front. Mol. Neurosci. 2018, 11, 160. [Google Scholar] [CrossRef]
  41. Palmos, A.B.; Duarte, R.R.R.; Smeeth, D.M.; Hedges, E.C.; Nixon, D.F.; Thuret, S.; Powell, T.R. Telomere length and human hippocampal neurogenesis. Neuropsychopharmacology 2020, 45, 2239–2247. [Google Scholar] [CrossRef] [PubMed]
  42. Hetz, C. Adapting the proteostasis capacity to sustain brain healthspan. Cell 2021, 184, 1545–1560. [Google Scholar] [CrossRef] [PubMed]
  43. Lee, J.; Kim, H.J. Normal Aging Induces Changes in the Brain and Neurodegeneration Progress: Review of the Structural, Biochemical, Metabolic, Cellular, and Molecular Changes. Front. Aging Neurosci. 2022, 14, 931536. [Google Scholar] [CrossRef] [PubMed]
  44. Chapman, T.W.; Hill, R.A. Myelin plasticity in adulthood and aging. Neurosci. Lett. 2020, 715, 134645. [Google Scholar] [CrossRef]
  45. Latham, A.S.; Moreno, J.A.; Geer, C.E. Biological agents and the aging brain: Glial inflammation and neurotoxic signaling. Front. Aging 2023, 4, 1244149. [Google Scholar] [CrossRef]
  46. Zhang, W.; Xiao, D.; Mao, Q.; Xia, H. Role of neuroinflammation in neurodegeneration development. Signal. Transduct. Target. Ther. 2023, 8, 267. [Google Scholar] [CrossRef]
  47. Liu, Y.; Liu, Q.; Zhang, Z.; Yang, Y.; Zhou, Y.; Yan, H.; Wang, X.; Li, X.; Zhao, J.; Hu, J.; et al. The regulatory role of PI3K in ageing-related diseases. Ageing Res. Rev. 2023, 88, 101963. [Google Scholar] [CrossRef]
  48. Byun, K.; Yoo, Y.; Son, M.; Lee, J.; Jeong, G.B.; Park, Y.M.; Salekdeh, G.H.; Lee, B. Advanced glycation end-products produced systemically and by macrophages: A common contributor to inflammation and degenerative diseases. Pharmacol. Ther. 2017, 177, 44–55. [Google Scholar] [CrossRef]
  49. Neutzner, M.; Kohler, C.; Frank, S.; Killer, H.E.; Neutzner, A. Impact of aging on meningeal gene expression. Fluids Barriers CNS 2023, 20, 12. [Google Scholar] [CrossRef]
  50. Singam, N.S.V.; Fine, C.; Fleg, J.L. Cardiac changes associated with vascular aging. Clin. Cardiol. 2020, 43, 92–98. [Google Scholar] [CrossRef]
  51. Anwar, I.; Wang, X.; Pratt, R.E.; Dzau, V.J.; Hodgkinson, C.P. The impact of aging on cardiac repair and regeneration. J. Biol. Chem. 2024, 300, 107682. [Google Scholar] [CrossRef] [PubMed]
  52. Sagar, S.; Gustafsson, A.B. Cardiovascular aging: The mitochondrial influence. J. Cardiovasc. Aging 2023, 3, 33. [Google Scholar] [CrossRef] [PubMed]
  53. González, L.D.M.; Romero-Orjuela, S.P.; Rabeya, F.J.; Del Castillo, V.; Echeverri, D. Age and vascular aging: An unexplored frontier. Front. Cardiovasc. Med. 2023, 10, 1278795. [Google Scholar] [CrossRef] [PubMed]
  54. Han, Y.; Kim, S.Y. Endothelial senescence in vascular diseases: Current understanding and future opportunities in senotherapeutics. Exp. Mol. Med. 2023, 55, 1–12. [Google Scholar] [CrossRef]
  55. De Meyer, T.; Nawrot, T.; Bekaert, S.; De Buyzere, M.L.; Rietzschel, E.R.; Andrés, V. Telomere Length as Cardiovascular Aging Biomarker: JACC Review Topic of the Week. J. Am. Coll. Cardiol. 2018, 72, 805–813. [Google Scholar] [CrossRef]
  56. Herman, A.B.; Occean, J.R.; Sen, P. Epigenetic dysregulation in cardiovascular aging and disease. J. Cardiovasc. Aging 2021, 1, 10. [Google Scholar] [CrossRef]
  57. Kim, M.; Kim, H.L.; Lim, W.H.; Seo, J.B.; Kim, S.H.; Kim, M.A.; Zo, J.H. Association between arterial stiffness and left ventricular diastolic function: A large population-based cross-sectional study. Front. Cardiovasc. Med. 2022, 9, 1001248. [Google Scholar] [CrossRef]
  58. Miyamoto, S. Autophagy and cardiac aging. Cell Death Differ. 2019, 26, 653–664. [Google Scholar] [CrossRef]
  59. Soenen, S.; Rayner, C.K.; Jones, K.L.; Horowitz, M. The ageing gastrointestinal tract. Curr. Opin. Clin. Nutr. Metab. Care 2016, 19, 12–18. [Google Scholar] [CrossRef]
  60. Sun, T.; Li, D.; Hu, S.; Huang, L.; Sun, H.; Yang, S.; Wu, B.; Ji, F.; Zhou, D. Aging-dependent decrease in the numbers of enteric neurons, interstitial cells of Cajal and expression of connexin43 in various regions of gastrointestinal tract. Aging 2018, 10, 3851–3865. [Google Scholar] [CrossRef]
  61. Nguyen, T.T.; Baumann, P.; Tüscher, O.; Schick, S.; Endres, K. The Aging Enteric Nervous System. Int. J. Mol. Sci. 2023, 24, 9471. [Google Scholar] [CrossRef] [PubMed]
  62. Kurin, M.; Fass, R. Management of Gastroesophageal Reflux Disease in the Elderly Patient. Drugs Aging 2019, 36, 1073–1081. [Google Scholar] [CrossRef] [PubMed]
  63. Yu, B.; Xiang, L.; Peppelenbosch, M.P.; Fuhler, G.M. Overlapping cytokines in H. pylori infection and gastric cancer: A tandem meta-analysis. Front. Immunol. 2023, 14, 1125658. [Google Scholar] [CrossRef] [PubMed]
  64. Skokowski, J.; Vashist, Y.; Girnyi, S.; Cwalinski, T.; Mocarski, P.; Antropoli, C.; Brillantino, A.; Boccardi, V.; Goyal, A.; Ciarleglio, F.A.; et al. The Aging Stomach: Clinical Implications of H. pylori Infection in Older Adults-Challenges and Strategies for Improved Management. Int. J. Mol. Sci. 2024, 25, 12826. [Google Scholar] [CrossRef]
  65. Amorim, J.A.; Coppotelli, G.; Rolo, A.P.; Palmeira, C.M.; Ross, J.M.; Sinclair, D.A. Mitochondrial and metabolic dysfunction in ageing and age-related diseases. Nat. Rev. Endocrinol. 2022, 18, 243–258. [Google Scholar] [CrossRef]
  66. Mbara, K.C.; Fotsing, M.C.D.; Ndinteh, D.T.; Mbeb, C.N.; Nwagwu, C.S.; Khan, R.; Mokhetho, K.C.; Baijnath, H.; Nlooto, M.; Mokhele, S.; et al. Endoplasmic reticulum stress in pancreatic β-cell dysfunction: The potential therapeutic role of dietary flavonoids. Curr. Res. Pharmacol. Drug Discov. 2024, 6, 100184. [Google Scholar] [CrossRef]
  67. Konstandi, M.; Johnson, E.O. Age-related modifications in CYP-dependent drug metabolism: Role of stress. Front. Endocrinol. 2023, 14, 1143835. [Google Scholar] [CrossRef]
  68. Wang, Q.; Qi, Y.; Shen, W.; Xu, J.; Wang, L.; Chen, S.; Hou, T.; Si, J. The Aged Intestine: Performance and Rejuvenation. Aging Dis. 2021, 12, 1693–1712. [Google Scholar] [CrossRef]
  69. Yue, T.; Chen, S.; Zhu, J.; Guo, S.; Huang, Z.; Wang, P.; Zuo, S.; Liu, Y. The aging-related risk signature in colorectal cancer. Aging 2021, 13, 7330–7349. [Google Scholar] [CrossRef]
  70. Pretzsch, E.; Nieß, H.; Bösch, F.; Westphalen, C.B.; Jacob, S.; Neumann, J.; Werner, J.; Heinemann, V.; Angele, M.K. Age and metastasis—How age influences metastatic spread in cancer. Colorectal cancer as a model. Cancer Epidemiol. 2022, 77, 102112. [Google Scholar] [CrossRef]
  71. Sohail, R.; Mathew, M.; Patel, K.K.; Reddy, S.A.; Haider, Z.; Naria, M.; Habib, A.; Abdin, Z.U.; Razzaq Chaudhry, W.; Akbar, A. Effects of Non-steroidal Anti-inflammatory Drugs (NSAIDs) and Gastroprotective NSAIDs on the Gastrointestinal Tract: A Narrative Review. Cureus 2023, 15, e37080. [Google Scholar] [CrossRef] [PubMed]
  72. Jump, R.L. Clostridium difficile infection in older adults. Aging Health 2013, 9, 403–414. [Google Scholar] [CrossRef] [PubMed]
  73. Li, S.; Xiao, X.; Zhang, X. Hydration Status in Older Adults: Current Knowledge and Future Challenges. Nutrients 2023, 15, 2609. [Google Scholar] [CrossRef]
  74. Rutsch, A.; Kantsjö, J.B.; Ronchi, F. The Gut-Brain Axis: How Microbiota and Host Inflammasome Influence Brain Physiology and Pathology. Front. Immunol. 2020, 11, 604179. [Google Scholar] [CrossRef] [PubMed]
  75. Afzaal, M.; Saeed, F.; Shah, Y.A.; Hussain, M.; Rabail, R.; Socol, C.T.; Hassoun, A.; Pateiro, M.; Lorenzo, J.M.; Rusu, A.V.; et al. Human gut microbiota in health and disease: Unveiling the relationship. Front. Microbiol. 2022, 13, 999001. [Google Scholar] [CrossRef]
  76. Wu, Y.L.; Xu, J.; Rong, X.Y.; Wang, F.; Wang, H.J.; Zhao, C. Gut microbiota alterations and health status in aging adults: From correlation to causation. Aging Med. 2021, 4, 206–213. [Google Scholar] [CrossRef]
  77. Kossowska, M.; Olejniczak, S.; Karbowiak, M.; Mosiej, W.; Zielińska, D.; Brzezicka, A. The Interplay between Gut Microbiota and Cognitive Functioning in the Healthy Aging Population: A Systematic Review. Nutrients 2024, 16, 852. [Google Scholar] [CrossRef]
  78. Zhang, Y.; Wei, S.; Zhang, H.; Jo, Y.; Kang, J.S.; Ha, K.T.; Joo, J.; Lee, H.J.; Ryu, D. Gut microbiota-generated metabolites: Missing puzzles to hosts’ health, diseases, and aging. BMB Rep. 2024, 57, 207–215. [Google Scholar] [CrossRef]
  79. Menezes, A.A.; Shah, Z.A. A Review of the Consequences of Gut Microbiota in Neurodegenerative Disorders and Aging. Brain Sci. 2024, 14, 1224. [Google Scholar] [CrossRef]
  80. Ghosh, T.S.; Shanahan, F.; O’Toole, P.W. The gut microbiome as a modulator of healthy ageing. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 565–584. [Google Scholar] [CrossRef]
  81. Chen, Y.; Xu, J.; Chen, Y. Regulation of Neurotransmitters by the Gut Microbiota and Effects on Cognition in Neurological Disorders. Nutrients 2021, 13, 2099. [Google Scholar] [CrossRef] [PubMed]
  82. Salazar, A.M.; Aparicio, R.; Clark, R.I.; Rera, M.; Walker, D.W. Intestinal barrier dysfunction: An evolutionarily conserved hallmark of aging. Dis. Model Mech. 2023, 16, dmm049969. [Google Scholar] [CrossRef] [PubMed]
  83. Lalley, P.M. The aging respiratory system--pulmonary structure, function and neural control. Respir. Physiol. Neurobiol. 2013, 187, 199–210. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, Y.; Huang, X.; Luo, G.; Xu, Y.; Deng, X.; Lin, Y.; Wang, Z.; Zhou, S.; Wang, S.; Chen, H.; et al. The aging lung: Microenvironment, mechanisms, and diseases. Front. Immunol. 2024, 15, 1383503. [Google Scholar] [CrossRef]
  85. Ulldemolins, A.; Narciso, M.; Sanz-Fraile, H.; Otero, J.; Farré, R.; Gavara, N.; Almendros, I. Effects of aging on the biomechanical properties of the lung extracellular matrix: Dependence on tissular stretch. Front. Cell. Dev. Biol. 2024, 12, 1381470. [Google Scholar] [CrossRef]
  86. Vitenberga, Z.; Pilmane, M. Age-related lung tissue remodeling due to the local distribution of MMP-2, TIMP-2, TGF-β and Hsp70. Biotech. Histochem. 2018, 93, 239–248. [Google Scholar] [CrossRef]
  87. Torrelles, J.B.; Restrepo, B.I.; Bai, Y.; Ross, C.; Schlesinger, L.S.; Turner, J. The Impact of Aging on the Lung Alveolar Environment, Predetermining Susceptibility to Respiratory Infections. Front. Aging. 2022, 3, 818700. [Google Scholar] [CrossRef]
  88. Ratjen, F.; Rehn, B.; Costabel, U.; Bruch, J. Age-dependency of surfactant phospholipids and surfactant protein A in bronchoalveolar lavage fluid of children without bronchopulmonary disease. Eur. Respir. J. 1996, 9, 328–333. [Google Scholar] [CrossRef]
  89. Hentschel, R.; Bohlin, K.; van Kaam, A.; Fuchs, H.; Danhaive, O. Surfactant replacement therapy: From biological basis to current clinical practice. Pediatr. Res. 2020, 88, 176–183. [Google Scholar] [CrossRef]
  90. Adachi, D.; Yamada, M.; Nishiguchi, S.; Fukutani, N.; Hotta, T.; Tashiro, Y.; Morino, S.; Shirooka, H.; Nozaki, Y.; Hirata, H.; et al. Age-related decline in chest wall mobility: A cross-sectional study among community-dwelling elderly women. J. Am. Osteopath. Assoc. 2015, 115, 384–389. [Google Scholar] [CrossRef]
  91. Yamada, T.; Minami, T.; Shinohara, T.; Ouchi, S.; Mabuchi, S.; Yoshino, S.; Emoto, K.; Nakagawa, K.; Yoshimi, K.; Saito, M.; et al. The Impact of Ageing on Diaphragm Function and Maximal Inspiratory Pressure: A Cross-Sectional Ultrasound Study. Diagnostics 2025, 15, 163. [Google Scholar] [CrossRef] [PubMed]
  92. Shaikh, S.B.; Goracci, C.; Tjitropranoto, A.; Rahman, I. Impact of aging on immune function in the pathogenesis of pulmonary diseases: Potential for therapeutic targets. Expert Rev. Respir. Med. 2023, 17, 351–364. [Google Scholar] [CrossRef] [PubMed]
  93. Paleczny, B.; Niewiński, P.; Rydlewska, A.; Piepoli, M.F.; Borodulin-Nadzieja, L.; Jankowska, E.A.; Ponikowska, B. Age-related reflex responses from peripheral and central chemoreceptors in healthy men. Clin. Auton. Res. 2014, 24, 285–296. [Google Scholar] [CrossRef] [PubMed]
  94. Vetterlein, M.W.; Jindal, T.; Becker, A.; Regier, M.; Kluth, L.A.; Tilki, D.; Chun, F.K. Small renal masses in the elderly: Contemporary treatment approaches and comparative oncological outcomes of nonsurgical and surgical strategies. Investig. Clin. Urol. 2016, 57, 231–239. [Google Scholar] [CrossRef]
  95. Alam, R.; Yerrapragada, A.; Wlajnitz, T.; Watts, E.; Pallauf, M.; Enikeev, D.; Chang, P.; Wagner, A.A.; McKiernan, J.M.; Pierorazio, P.M.; et al. Evaluation of Growth Rates for Small Renal Masses in Elderly Patients Undergoing Active Surveillance. Eur. Urol. Open Sci. 2023, 50, 78–84. [Google Scholar] [CrossRef]
  96. Wang, Y.; Zhang, W.; Qian, T.; Sun, H.; Xu, Q.; Hou, X.; Hu, W.; Zhang, G.; Drummond, G.R.; Sobey, C.G.; et al. Reduced renal function may explain the higher prevalence of hyperuricemia in older people. Sci. Rep. 2021, 11, 1302. [Google Scholar] [CrossRef]
  97. Ray, N.; Reddy, P.H. Structural and physiological changes of the kidney with age and its impact on chronic conditions and COVID-19. Ageing Res. Rev. 2023, 88, 101932. [Google Scholar] [CrossRef]
  98. Dybiec, J.; Szlagor, M.; Młynarska, E.; Rysz, J.; Franczyk, B. Structural and Functional Changes in Aging Kidneys. Int. J. Mol. Sci. 2022, 23, 15435. [Google Scholar] [CrossRef]
  99. Monteonofrio, L.; Florio, M.C.; AlGhatrif, M.; Lakatta, E.G.; Capogrossi, M.C. Aging- and gender-related modulation of RAAS: Potential implications in COVID-19 disease. Vasc. Biol. 2020, 3, R1–R14. [Google Scholar] [CrossRef]
  100. Li, M.; Wang, D.; Liu, Z.; Huang, Y.; Zhang, Q.; Pan, C.; Lin, Y.; Sun, L.; Zheng, Y. Assessing the effects of aging on the renal endothelial cell landscape using single-cell RNA sequencing. Front. Genet. 2023, 14, 1175716. [Google Scholar] [CrossRef]
  101. Nishii, H. A Review of Aging and the Lower Urinary Tract: The Future of Urology. Int. Neurourol. J. 2021, 25, 273–284. [Google Scholar] [CrossRef]
  102. Lee, J.; Yoo, S.; Cho, M.C.; Jeong, H.; Choo, M.S.; Son, H. Significance of a decrease in the proportion of detrusor muscle to bladder wall for non-invasive diagnosis of detrusor underactivity in men with lower urinary tract symptoms. Sci. Rep. 2022, 12, 5237. [Google Scholar] [CrossRef] [PubMed]
  103. Hardy, C.C.; Korstanje, R. Aging and urinary control: Alterations in the brain-bladder axis. Aging Cell 2023, 22, e13990. [Google Scholar] [CrossRef] [PubMed]
  104. Batmani, S.; Jalali, R.; Mohammadi, M.; Bokaee, S. Prevalence and factors related to urinary incontinence in older adults women worldwide: A comprehensive systematic review and meta-analysis of observational studies. BMC Geriatr. 2021, 21, 212. [Google Scholar] [CrossRef] [PubMed]
  105. Sayegh, N.; Gross, K. Benign prostatic hyperplasia: A global challenge of the ageing population. Lancet Healthy Longev. 2022, 3, e725–e726. [Google Scholar] [CrossRef]
  106. Fu, X.; Liu, H.; Liu, J.; DiSanto, M.E.; Zhang, X. The Role of Heat Shock Protein 70 Subfamily in the Hyperplastic Prostate: From Molecular Mechanisms to Therapeutic Opportunities. Cells 2022, 11, 2052. [Google Scholar] [CrossRef]
  107. Olascoaga, S.; Castañeda-Sánchez, J.I.; Königsberg, M.; Gutierrez, H.; López-Diazguerrero, N.E. Oxidative stress-induced gene expression changes in prostate epithelial cells in vitro reveal a robust signature of normal prostatic senescence and aging. Biogerontology 2024, 25, 1145–1169. [Google Scholar] [CrossRef]
  108. Rodríguez-Nieves, J.A.; Patalano, S.C.; Almanza, D.; Gharaee-Kermani, M.; Macoska, J.A. CXCL12/CXCR4 Axis Activation Mediates Prostate Myofibroblast Phenoconversion through Non-Canonical EGFR/MEK/ERK Signaling. PLoS ONE 2016, 11, e0159490. [Google Scholar] [CrossRef]
  109. Rodriguez-Mañas, L. Urinary tract infections in the elderly: A review of disease characteristics and current treatment options. Drugs Context. 2020, 9, 2020-4-13. [Google Scholar] [CrossRef]
  110. Maresca, G.; Mc Clinton, S.; Swami, S.; El-Mokadem, I.; Donaldson, J.F. Do men with bladder stones benefit from treatment of benign prostatic obstruction? BJU Int. 2022, 130, 619–627. [Google Scholar] [CrossRef]
  111. Liu, P.; Quinn, R.R.; Lam, N.N.; Elliott, M.J.; Xu, Y.; James, M.T.; Manns, B.; Ravani, P. Accounting for Age in the Definition of Chronic Kidney Disease. JAMA Intern. Med. 2021, 181, 1359–1366. [Google Scholar] [CrossRef] [PubMed]
  112. Calleja-Agius, J.; Brincat, M.P. The urogenital system and the menopause. Climacteric 2015, 18 (Suppl. S1), 18–22. [Google Scholar] [CrossRef] [PubMed]
  113. Liu, Y.; Li, C. Hormone Therapy and Biological Aging in Postmenopausal Women. JAMA Netw. Open 2024, 7, e2430839. [Google Scholar] [CrossRef] [PubMed]
  114. Critchlow, A.J.; Hiam, D.; Williams, R.; Scott, D.; Lamon, S. The role of estrogen in female skeletal muscle aging: A systematic review. Maturitas 2023, 178, 107844. [Google Scholar] [CrossRef]
  115. Wei, S.; Tang, W.; Chen, D.; Xiong, J.; Xue, L.; Dai, Y.; Guo, Y.; Wu, C.; Dai, J.; Wu, M.; et al. Multiomics insights into the female reproductive aging. Ageing Res. Rev. 2024, 95, 102245. [Google Scholar] [CrossRef]
  116. Dong, S.; Chen, C.; Zhang, J.; Gao, Y.; Zeng, X.; Zhang, X. Testicular aging, male fertility and beyond. Front. Endocrinol. 2022, 13, 1012119. [Google Scholar] [CrossRef]
  117. Kaufman, J.M.; Lapauw, B.; Mahmoud, A.; T’Sjoen, G.; Huhtaniemi, I.T. Aging and the Male Reproductive System. Endocr. Rev. 2019, 40, 906–972. [Google Scholar] [CrossRef]
  118. Jimbo, M.; Kunisaki, J.; Ghaed, M.; Yu, V.; Flores, H.A.; Hotaling, J.M. Fertility in the aging male: A systematic review. Fertil. Steril. 2022, 118, 1022–1034. [Google Scholar] [CrossRef]
  119. Davis, S.R.; Pinkerton, J.; Santoro, N.; Simoncini, T. Menopause-Biology, consequences, supportive care, and therapeutic options. Cell 2023, 186, 4038–4058. [Google Scholar] [CrossRef]
  120. Andrews, R.; Lacey, A.; Bache, K.; Kidd, E.J. The role of menopausal symptoms on future health and longevity: A systematic scoping review of longitudinal evidence. Maturitas 2024, 190, 108130. [Google Scholar] [CrossRef]
  121. Bi, J.; Zhang, C.; Lu, C.; Mo, C.; Zeng, J.; Yao, M.; Jia, B.; Liu, Z.; Yuan, P.; Xu, S. Age-related bone diseases: Role of inflammaging. J. Autoimmun. 2024, 143, 103169. [Google Scholar] [CrossRef] [PubMed]
  122. Xing, Y.; Xuan, F.; Wang, K.; Zhang, H. Aging under endocrine hormone regulation. Front. Endocrinol. 2023, 14, 1223529. [Google Scholar] [CrossRef] [PubMed]
  123. van den Beld, A.W.; Kaufman, J.M.; Zillikens, M.C.; Lamberts, S.W.J.; Egan, J.M.; van der Lely, A.J. The physiology of endocrine systems with ageing. Lancet Diabetes Endocrinol. 2018, 6, 647–658. [Google Scholar] [CrossRef] [PubMed]
  124. Hajdarovic, K.H.; Yu, D.; Webb, A.E. Understanding the aging hypothalamus, one cell at a time. Trends Neurosci. 2022, 45, 942–954. [Google Scholar] [CrossRef]
  125. Pataky, M.W.; Young, W.F.; Nair, K.S. Hormonal and Metabolic Changes of Aging and the Influence of Lifestyle Modifications. Mayo Clin. Proc. 2021, 96, 788–814. [Google Scholar] [CrossRef]
  126. Cappola, A.R.; Auchus, R.J.; El-Hajj Fuleihan, G.; Handelsman, D.J.; Kalyani, R.R.; McClung, M.; Stuenkel, C.A.; Thorner, M.O.; Verbalis, J.G. Hormones and Aging: An Endocrine Society Scientific Statement. J. Clin. Endocrinol Metab. 2023, 108, 1835–1874. [Google Scholar] [CrossRef]
  127. Santos, A.L.; Lindner, A.B. Protein Posttranslational Modifications: Roles in Aging and Age-Related Disease. Oxid. Med. Cell. Longev. 2017, 2017, 5716409. [Google Scholar] [CrossRef]
  128. Kim, C.S.; Park, S.; Kim, J. The role of glycation in the pathogenesis of aging and its prevention through herbal products and physical exercise. J. Exerc. Nutr. Biochem. 2017, 21, 55–61. [Google Scholar] [CrossRef]
  129. Chaudhuri, J.; Bains, Y.; Guha, S.; Kahn, A.; Hall, D.; Bose, N.; Gugliucci, A.; Kapahi, P. The Role of Advanced Glycation End Products in Aging and Metabolic Diseases: Bridging Association and Causality. Cell. Metab. 2018, 28, 337–352. [Google Scholar] [CrossRef]
  130. Gill, V.; Kumar, V.; Singh, K.; Kumar, A.; Kim, J.J. Advanced Glycation End Products (AGEs) May Be a Striking Link Between Modern Diet and Health. Biomolecules 2019, 9, 888. [Google Scholar] [CrossRef]
  131. Tudurí, E.; Soriano, S.; Almagro, L.; Montanya, E.; Alonso-Magdalena, P.; Nadal, Á.; Quesada, I. The pancreatic β-cell in ageing: Implications in age-related diabetes. Ageing Res. Rev. 2022, 80, 101674. [Google Scholar] [CrossRef] [PubMed]
  132. Warde, K.M.; Smith, L.J.; Basham, K.J. Age-related Changes in the Adrenal Cortex: Insights and Implications. J. Endocr. Soc. 2023, 7, bvad097. [Google Scholar] [CrossRef] [PubMed]
  133. Powrie, Y.S.L.; Smith, C. Central intracrine DHEA synthesis in ageing-related neuroinflammation and neurodegeneration: Therapeutic potential? J. Neuroinflamm. 2018, 15, 289. [Google Scholar] [CrossRef] [PubMed]
  134. van Heemst, D. The ageing thyroid: Implications for longevity and patient care. Nat. Rev. Endocrinol. 2024, 20, 5–15. [Google Scholar] [CrossRef]
  135. Cai, Y.; Han, Z.; Cheng, H.; Li, H.; Wang, K.; Chen, J.; Liu, Z.X.; Xie, Y.; Lin, Y.; Zhou, S.; et al. The impact of ageing mechanisms on musculoskeletal system diseases in the elderly. Front. Immunol. 2024, 15, 1405621. [Google Scholar] [CrossRef]
  136. Xu, H.; Brown, J.L.; Bhaskaran, S.; Van Remmen, H. Reactive oxygen species in the pathogenesis of sarcopenia. Free Radic. Biol. Med. 2025, 227, 446–458. [Google Scholar] [CrossRef]
  137. Marques-Carvalho, A.; Kim, H.N.; Almeida, M. The role of reactive oxygen species in bone cell physiology and pathophysiology. Bone Rep. 2023, 19, 101664. [Google Scholar] [CrossRef]
  138. Dao, T.; Green, A.E.; Kim, Y.A.; Bae, S.J.; Ha, K.T.; Gariani, K.; Lee, M.R.; Menzies, K.J.; Ryu, D. Sarcopenia and Muscle Aging: A Brief Overview. Endocrinol. Metab. 2020, 35, 716–732. [Google Scholar] [CrossRef]
  139. Cho, M.R.; Lee, S.; Song, S.K. A Review of Sarcopenia Pathophysiology, Diagnosis, Treatment and Future Direction. J. Korean Med. Sci. 2022, 37, e146. [Google Scholar] [CrossRef]
  140. Muñoz-Cánoves, P.; Neves, J.; Sousa-Victor, P. Understanding muscle regenerative decline with aging: New approaches to bring back youthfulness to aged stem cells. FEBS J. 2020, 287, 406–416. [Google Scholar] [CrossRef]
  141. Sousa-Victor, P.; García-Prat, L.; Muñoz-Cánoves, P. Control of satellite cell function in muscle regeneration and its disruption in ageing. Nat. Rev. Mol. Cell. Biol. 2022, 23, 204–226. [Google Scholar] [CrossRef] [PubMed]
  142. Bodine, S.C.; Sinha, I.; Sweeney, H.L. Mechanisms of Skeletal Muscle Atrophy and Molecular Circuitry of Stem Cell Fate in Skeletal Muscle Regeneration and Aging. J. Gerontol. A Biol. Sci. Med. Sci. 2023, 78, 14–18. [Google Scholar] [CrossRef] [PubMed]
  143. Gupta, P.; Kumar, S. Sarcopenia and Endocrine Ageing: Are They Related? Cureus 2022, 14, e28787. [Google Scholar] [CrossRef] [PubMed]
  144. Ravazzano, L.; Colaianni, G.; Tarakanova, A.; Xiao, Y.B.; Grano, M.; Libonati, F. Multiscale and multidisciplinary analysis of aging processes in bone. NPJ Aging 2024, 10, 28. [Google Scholar] [CrossRef]
  145. Becerikli, M.; Jaurich, H.; Schira, J.; Schulte, M.; Döbele, C.; Wallner, C.; Abraham, S.; Wagner, J.M.; Dadras, M.; Kneser, U.; et al. Age-dependent alterations in osteoblast and osteoclast activity in human cancellous bone. J. Cell. Mol. Med. 2017, 21, 2773–2781. [Google Scholar] [CrossRef]
  146. Werner, S.L.; Sharma, R.; Woodruff, K.; Horn, D.; Harris, S.E.; Gorin, Y.; Lee, D.Y.; Hua, R.; Gu, S.; Fajardo, R.J.; et al. CSF-1 in Osteocytes Inhibits Nox4-mediated Oxidative Stress and Promotes Normal Bone Homeostasis. JBMR Plus 2019, 4, e10080. [Google Scholar] [CrossRef]
  147. Cheng, Y.H.; Liu, S.F.; Dong, J.C.; Bian, Q. Transcriptomic alterations underline aging of osteogenic bone marrow stromal cells. World J. Stem Cells 2021, 13, 128–138. [Google Scholar] [CrossRef]
  148. Cannarella, R.; Barbagallo, F.; Condorelli, R.A.; Aversa, A.; La Vignera, S.; Calogero, A.E. Osteoporosis from an Endocrine Perspective: The Role of Hormonal Changes in the Elderly. J. Clin. Med. 2019, 8, 1564. [Google Scholar] [CrossRef]
  149. Mills, E.G.; Yang, L.; Nielsen, M.F.; Kassem, M.; Dhillo, W.S.; Comninos, A.N. The Relationship Between Bone and Reproductive Hormones Beyond Estrogens and Androgens. Endocr. Rev. 2021, 42, 691–719. [Google Scholar] [CrossRef]
  150. Vashishth, D.; Dhaliwal, R.; Rubin, M. AGEs (Advanced Glycation End-products) in bone come of age. Bone 2025, 190, 117301. [Google Scholar] [CrossRef]
  151. Rahmati, M.; Nalesso, G.; Mobasheri, A.; Mozafari, M. Aging and osteoarthritis: Central role of the extracellular matrix. Ageing Res. Rev. 2017, 40, 20–30. [Google Scholar] [CrossRef]
  152. Sinha, U.; Malis, V.; Chen, J.S.; Csapo, R.; Kinugasa, R.; Narici, M.V.; Sinha, S. Role of the Extracellular Matrix in Loss of Muscle Force With Age and Unloading Using Magnetic Resonance Imaging, Biochemical Analysis, and Computational Models. Front. Physiol. 2020, 11, 626. [Google Scholar] [CrossRef] [PubMed]
  153. Petrocelli, J.J.; McKenzie, A.I.; de Hart, N.M.M.P.; Reidy, P.T.; Mahmassani, Z.S.; Keeble, A.R.; Kaput, K.L.; Wahl, M.P.; Rondina, M.T.; Marcus, R.L.; et al. Disuse-induced muscle fibrosis, cellular senescence, and senescence-associated secretory phenotype in older adults are alleviated during re-ambulation with metformin pre-treatment. Aging Cell 2023, 22, e13936. [Google Scholar] [CrossRef] [PubMed]
  154. Tieland, M.; Trouwborst, I.; Clark, B.C. Skeletal muscle performance and ageing. J. Cachexia Sarcopenia Muscle 2018, 9, 3–19. [Google Scholar] [CrossRef] [PubMed]
  155. Chen, W.J.; Lin, I.H.; Lee, C.W.; Chen, Y.F. Aged Skeletal Muscle Retains the Ability to Remodel Extracellular Matrix for Degradation of Collagen Deposition after Muscle Injury. Int. J. Mol. Sci. 2021, 22, 2123. [Google Scholar] [CrossRef]
  156. Kanazawa, Y.; Takahashi, T.; Inoue, T.; Nagano, M.; Koinuma, S.; Eiyo, H.; Tamura, Y.; Miyachi, R.; Iida, N.; Miyahara, K.; et al. Effects of Aging on Intramuscular Collagen-Related Factors After Injury to Mouse Tibialis Anterior Muscle. Int. J. Mol. Sci. 2025, 26, 801. [Google Scholar] [CrossRef]
  157. Bektan Kanat, B.; Ulugerger Avci, G.; Bayramlar, O.F.; Unal, D.; Sonmez, O.; Bolayirli, I.M.; Doventas, A.; Erdincler, D.S.; Yavuzer, H. Matrix Metalloproteinases/Tissue Inhibitors of Metalloproteinases Ratio: A Biomarker of Bone Resorption in Geriatric Osteoporosis? Ann. Geriatr. Med. Res. 2023, 27, 151–157. [Google Scholar] [CrossRef]
  158. Fuggle, N.R.; Laskou, F.; Harvey, N.C.; Dennison, E.M. A review of epigenetics and its association with ageing of muscle and bone. Maturitas 2022, 165, 12–17. [Google Scholar] [CrossRef]
  159. Gerosa, L.; Malvandi, A.M.; Malavolta, M.; Provinciali, M.; Lombardi, G. Exploring cellular senescence in the musculoskeletal system: Any insights for biomarkers discovery? Ageing Res. Rev. 2023, 88, 101943. [Google Scholar] [CrossRef]
  160. Li, Z.; Wu, X.; Yan, Z.; Cui, Y.; Liu, Y.; Cui, S.; Wang, Y.; Liu, T. Unveiling the muscle-brain axis: A bidirectional mendelian randomization study investigating the causal relationship between sarcopenia-related traits and brain aging. Arch. Gerontol. Geriatr. 2024, 123, 105412. [Google Scholar] [CrossRef]
  161. Kumar, A.; Vaca-Dempere, M.; Mortimer, T.; Deryagin, O.; Smith, J.G.; Petrus, P.; Koronowski, K.B.; Greco, C.M.; Segalés, J.; Andrés, E.; et al. Brain-muscle communication prevents muscle aging by maintaining daily physiology. Science 2024, 384, 563–572. [Google Scholar] [CrossRef] [PubMed]
  162. Bhattacharjee, S.; Kashyap, R.; Abualait, T.; Annabel Chen, S.H.; Yoo, W.K.; Bashir, S. The Role of Primary Motor Cortex: More Than Movement Execution. J. Mot. Behav. 2021, 53, 258–274. [Google Scholar] [CrossRef] [PubMed]
  163. Han, X.; Ashraf, M.; Tipparaju, S.M.; Xuan, W. Muscle-Brain crosstalk in cognitive impairment. Front. Aging Neurosci. 2023, 15, 1221653. [Google Scholar] [CrossRef] [PubMed]
  164. Saleh, Z.; Mirzazadeh, S.; Mirzaei, F.; Heidarnejad, K.; Meri, S.; Kalantar, K. Alterations in metabolic pathways: A bridge between aging and weaker innate immune response. Front. Aging 2024, 5, 1358330. [Google Scholar] [CrossRef]
  165. Bachmann, M.C.; Bellalta, S.; Basoalto, R.; Gómez-Valenzuela, F.; Jalil, Y.; Lépez, M.; Matamoros, A.; von Bernhardi, R. The Challenge by Multiple Environmental and Biological Factors Induce Inflammation in Aging: Their Role in the Promotion of Chronic Disease. Front. Immunol. 2020, 11, 570083. [Google Scholar] [CrossRef]
  166. Chung, H.Y.; Kim, D.H.; Lee, E.K.; Chung, K.W.; Chung, S.; Lee, B.; Seo, A.Y.; Chung, J.H.; Jung, Y.S.; Im, E.; et al. Redefining Chronic Inflammation in Aging and Age-Related Diseases: Proposal of the Senoinflammation Concept. Aging Dis. 2019, 10, 367–382. [Google Scholar] [CrossRef]
  167. Soliman, A.M.; Barreda, D.R. Acute Inflammation in Tissue Healing. Int. J. Mol. Sci. 2022, 24, 641. [Google Scholar] [CrossRef]
  168. Santoro, A.; Bientinesi, E.; Monti, D. Immunosenescence and inflammaging in the aging process: Age-related diseases or longevity? Ageing Res. Rev. 2021, 71, 101422. [Google Scholar] [CrossRef]
  169. 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]
  170. Abdellatif, M.; Rainer, P.P.; Sedej, S.; Kroemer, G. Hallmarks of cardiovascular ageing. Nat. Rev. Cardiol. 2023, 20, 754–777. [Google Scholar] [CrossRef]
  171. Hou, Y.; Dan, X.; Babbar, M.; Wei, Y.; Hasselbalch, S.G.; Croteau, D.L.; Bohr, V.A. Ageing as a risk factor for neurodegenerative disease. Nat. Rev. Neurol. 2019, 15, 565–581. [Google Scholar] [CrossRef] [PubMed]
  172. López-Otín, C.; Pietrocola, F.; Roiz-Valle, D.; Galluzzi, L.; Kroemer, G. Meta-hallmarks of aging and cancer. Cell Metab. 2023, 35, 12–35. [Google Scholar] [CrossRef] [PubMed]
  173. Chung, H.Y.; Sung, B.; Jung, K.J.; Zou, Y.; Yu, B.P. The molecular inflammatory process in aging. Antioxid. Redox Signal. 2006, 8, 572–581. [Google Scholar] [CrossRef] [PubMed]
  174. Fulop, T.; Larbi, A.; Pawelec, G.; Khalil, A.; Cohen, A.A.; Hirokawa, K.; Witkowski, J.M.; Franceschi, C. Immunology of Aging: The Birth of Inflammaging. Clin. Rev. Allergy Immunol. 2023, 64, 109–122. [Google Scholar] [CrossRef]
  175. Molinero, N.; Antón-Fernández, A.; Hernández, F.; Ávila, J.; Bartolomé, B.; Moreno-Arribas, M.V. Gut Microbiota, an Additional Hallmark of Human Aging and Neurodegeneration. Neuroscience 2023, 518, 141–161. [Google Scholar] [CrossRef]
  176. Bosco, N.; Noti, M. The aging gut microbiome and its impact on host immunity. Genes Immun. 2021, 22, 289–303. [Google Scholar] [CrossRef]
  177. Chmielarz, M.; Sobieszczańska, B.; Środa-Pomianek, K. Metabolic Endotoxemia: From the Gut to Neurodegeneration. Int. J. Mol. Sci. 2024, 25, 7006. [Google Scholar] [CrossRef]
  178. Tylutka, A.; Walas, Ł.; Zembron-Lacny, A. Level of IL-6, TNF, and IL-1β and age-related diseases: A systematic review and meta-analysis. Front. Immunol. 2024, 15, 1330386. [Google Scholar] [CrossRef]
  179. Davizon-Castillo, P.; McMahon, B.; Aguila, S.; Bark, D.; Ashworth, K.; Allawzi, A.; Campbell, R.A.; Montenont, E.; Nemkov, T.; D’Alessandro, A.; et al. TNF-α-driven inflammation and mitochondrial dysfunction define the platelet hyperreactivity of aging. Blood 2019, 134, 727–740. [Google Scholar] [CrossRef]
  180. Rea, I.M.; Gibson, D.S.; McGilligan, V.; McNerlan, S.E.; Alexander, H.D.; Ross, O.A. Age and Age-Related Diseases: Role of Inflammation Triggers and Cytokines. Front. Immunol. 2018, 9, 586. [Google Scholar] [CrossRef]
  181. Tominaga, K.; Suzuki, H.I. TGF-β Signaling in Cellular Senescence and Aging-Related Pathology. Int. J. Mol. Sci. 2019, 20, 5002. [Google Scholar] [CrossRef] [PubMed]
  182. Freitas-Rodríguez, S.; Folgueras, A.R.; López-Otín, C. The role of matrix metalloproteinases in aging: Tissue remodeling and beyond. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 2015–2025. [Google Scholar] [CrossRef] [PubMed]
  183. Neves, D.; Duarte-Pereira, S.; Matos, S.; Silva, R.M. Proteostasis networks in aging: Novel insights from text-mining approaches. Biogerontology 2023, 24, 555–562. [Google Scholar] [CrossRef] [PubMed]
  184. Ruano, D. Proteostasis Dysfunction in Aged Mammalian Cells. The Stressful Role of Inflammation. Front. Mol. Biosci. 2021, 8, 658742. [Google Scholar] [CrossRef]
  185. 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]
  186. Huang, W.; Hickson, L.J.; Eirin, A.; Kirkland, J.L.; Lerman, L.O. Cellular senescence: The good, the bad and the unknown. Nat. Rev. Nephrol. 2022, 18, 611–627. [Google Scholar] [CrossRef]
  187. Bonelli, M.; La Monica, S.; Fumarola, C.; Alfieri, R. Multiple effects of CDK4/6 inhibition in cancer: From cell cycle arrest to immunomodulation. Biochem. Pharmacol. 2019, 170, 113676. [Google Scholar] [CrossRef]
  188. Liang, R.; Qi, X.; Cai, Q.; Niu, L.; Huang, X.; Zhang, D.; Ling, J.; Wu, Y.; Chen, Y.; Yang, P.; et al. The role of NLRP3 inflammasome in aging and age-related diseases. Immun. Ageing 2024, 21, 14. [Google Scholar] [CrossRef]
  189. Ohtani, N. The roles and mechanisms of senescence-associated secretory phenotype (SASP): Can it be controlled by senolysis? Inflamm. Regen. 2022, 42, 11. [Google Scholar] [CrossRef]
  190. Elder, S.S.; Emmerson, E. Senescent cells and macrophages: Key players for regeneration? Open Biol. 2020, 10, 200309. [Google Scholar] [CrossRef]
  191. Marin, I.; Serrano, M.; Pietrocola, F. Recent insights into the crosstalk between senescent cells and CD8 T lymphocytes. NPJ Aging 2023, 9, 8. [Google Scholar] [CrossRef] [PubMed]
  192. Wang, B.; Han, J.; Elisseeff, J.H.; Demaria, M. The senescence-associated secretory phenotype and its physiological and pathological implications. Nat. Rev. Mol. Cell. Biol. 2024, 25, 958–978. [Google Scholar] [CrossRef] [PubMed]
  193. Coryell, P.R.; Diekman, B.O.; Loeser, R.F. Mechanisms and therapeutic implications of cellular senescence in osteoarthritis. Nat. Rev. Rheumatol. 2021, 17, 47–57. [Google Scholar] [CrossRef] [PubMed]
  194. Lee, K.A.; Flores, R.R.; Jang, I.H.; Saathoff, A.; Robbins, P.D. Immune Senescence, Immunosenescence and Aging. Front. Aging 2022, 3, 900028. [Google Scholar] [CrossRef]
  195. Perez-Lanzon, M.; Zitvogel, L.; Kroemer, G. Failure of immunosurveillance accelerates aging. Oncoimmunology 2019, 8, e1575117. [Google Scholar] [CrossRef]
  196. Haga, M.; Okada, M. Systems approaches to investigate the role of NF-κB signaling in aging. Biochem. J. 2022, 479, 161–183. [Google Scholar] [CrossRef]
  197. Saavedra, D.; Añé-Kourí, A.L.; Barzilai, N.; Caruso, C.; Cho, K.H.; Fontana, L.; Franceschi, C.; Frasca, D.; Ledón, N.; Niedernhofer, L.J.; et al. Aging and chronic inflammation: Highlights from a multidisciplinary workshop. Immun. Ageing 2023, 20, 25. [Google Scholar] [CrossRef]
  198. Coleman, P.R.; Chang, G.; Hutas, G.; Grimshaw, M.; Vadas, M.A.; Gamble, J.R. Age-associated stresses induce an anti-inflammatory senescent phenotype in endothelial cells. Aging 2013, 5, 913–924. [Google Scholar] [CrossRef]
  199. Palatella, M.; Guillaume, S.M.; Linterman, M.A.; Huehn, J. The dark side of Tregs during aging. Front. Immunol. 2022, 13, 940705. [Google Scholar] [CrossRef]
  200. Hou, P.F.; Zhu, L.J.; Chen, X.Y.; Qiu, Z.Q. Age-related changes in CD4+CD25+FOXP3+ regulatory T cells and their relationship with lung cancer. PLoS ONE 2017, 12, e0173048. [Google Scholar] [CrossRef]
  201. Escrig-Larena, J.I.; Delgado-Pulido, S.; Mittelbrunn, M. Mitochondria during T cell aging. Semin. Immunol. 2023, 69, 101808. [Google Scholar] [CrossRef] [PubMed]
  202. Gill, J.S.; Bansal, B.; Guo, K.; Huang, F.; Singh, H.; Hur, J.; Khan, N.; Mathur, R. Mitochondrial Oxidative Stress Regulates FOXP3+ T-Cell Activity and CD4-Mediated Inflammation in Older Adults with Frailty. Int. J. Mol. Sci. 2024, 25, 6235. [Google Scholar] [CrossRef] [PubMed]
  203. Liu, Z.; Liang, Q.; Ren, Y.; Guo, C.; Ge, X.; Wang, L.; Cheng, Q.; Luo, P.; Zhang, Y.; Han, X. Immunosenescence: Molecular mechanisms and diseases. Signal Transduct. Target Ther. 2023, 8, 200. [Google Scholar] [CrossRef] [PubMed]
  204. Shaw, A.C.; Goldstein, D.R.; Montgomery, R.R. Age-dependent dysregulation of innate immunity. Nat. Rev. Immunol. 2013, 13, 875–887. [Google Scholar] [CrossRef]
  205. Kasler, H.; Verdin, E. How inflammaging diminishes adaptive immunity. Nat. Aging 2021, 1, 24–25. [Google Scholar] [CrossRef]
  206. Quiros-Roldan, E.; Sottini, A.; Natali, P.G.; Imberti, L. The Impact of Immune System Aging on Infectious Diseases. Microorganisms 2024, 12, 775. [Google Scholar] [CrossRef]
  207. Wilkinson, G.S.; Adams, D.M.; Haghani, A.; Lu, A.T.; Zoller, J.; Breeze, C.E.; Arnold, B.D.; Ball, H.C.; Carter, G.G.; Cooper, L.N.; et al. DNA methylation predicts age and provides insight into exceptional longevity of bats. Nat. Commun. 2021, 12, 1615. [Google Scholar] [CrossRef]
  208. Nardini, C.; Moreau, J.F.; Gensous, N.; Ravaioli, F.; Garagnani, P.; Bacalini, M.G. The epigenetics of inflammaging: The contribution of age-related heterochromatin loss and locus-specific remodelling and the modulation by environmental stimuli. Semin. Immunol. 2018, 40, 49–60. [Google Scholar] [CrossRef]
  209. Alimohammadi, M.; Makaremi, S.; Rahimi, A.; Asghariazar, V.; Taghadosi, M.; Safarzadeh, E. DNA methylation changes and inflammaging in aging-associated diseases. Epigenomics 2022, 14, 965–986. [Google Scholar] [CrossRef]
  210. de Lima Camillo, L.P.; Asif, M.H.; Horvath, S.; Larschan, E.; Singh, R. Histone mark age of human tissues and cell types. Sci. Adv. 2025, 11, eadk9373. [Google Scholar] [CrossRef]
  211. Mrabti, C.; Yang, N.; Desdín-Micó, G.; Alonso-Calleja, A.; Vílchez-Acosta, A.; Pico, S.; Parras, A.; Piao, Y.; Schoenfeldt, L.; Luo, S.; et al. Loss of H3K9 trimethylation leads to premature aging. bioRxiv 2024, 604929. [Google Scholar]
  212. Zhang, X.; Liu, H.; Zhou, J.Q.; Krick, S.; Barnes, J.W.; Thannickal, V.J.; Sanders, Y.Y. Modulation of H4K16Ac levels reduces pro-fibrotic gene expression and mitigates lung fibrosis in aged mice. Theranostics 2022, 12, 530–541. [Google Scholar] [CrossRef] [PubMed]
  213. Borja-Gonzalez, M.; Casas-Martinez, J.C.; McDonagh, B.; Goljanek-Whysall, K. Inflamma-miR-21 Negatively Regulates Myogenesis during Ageing. Antioxidants 2020, 9, 345. [Google Scholar] [CrossRef] [PubMed]
  214. Wang, L.; Lee, A.Y.; Wigg, J.P.; Peshavariya, H.; Liu, P.; Zhang, H. miR-126 Regulation of Angiogenesis in Age-Related Macular Degeneration in CNV Mouse Model. Int. J. Mol. Sci. 2016, 17, 895. [Google Scholar] [CrossRef]
  215. Malenfant, J.H.; Batsis, J.A. Obesity in the geriatric population—A global health perspective. J. Glob. Health Rep. 2019, 3, e2019045. [Google Scholar] [CrossRef]
  216. Nieva-Vazquez, A.; Pérez-Fuentes, R.; Torres-Rasgado, E.; López-López, J.G.; Romero, J.R. Serum resistin levels are associated with adiposity and insulin sensitivity in obese Hispanic subjects. Metab. Syndr. Relat. Disord. 2014, 12, 143–148. [Google Scholar] [CrossRef]
  217. Obradovic, M.; Sudar-Milovanovic, E.; Soskic, S.; Essack, M.; Arya, S.; Stewart, A.J.; Gojobori, T.; Isenovic, E.R. Leptin and Obesity: Role and Clinical Implication. Front. Endocrinol. 2021, 12, 585887. [Google Scholar] [CrossRef]
  218. Mancuso, P. The role of adipokines in chronic inflammation. Immunotargets Ther. 2016, 5, 47–56. [Google Scholar] [CrossRef]
  219. Mirabelli, M.; Misiti, R.; Sicilia, L.; Brunetti, F.S.; Chiefari, E.; Brunetti, A.; Foti, D.P. Hypoxia in Human Obesity: New Insights from Inflammation towards Insulin Resistance-A Narrative Review. Int. J. Mol. Sci. 2024, 25, 9802. [Google Scholar] [CrossRef]
  220. Nerstedt, A.; Smith, U. The impact of cellular senescence in human adipose tissue. J. Cell Commun. Signal. 2023, 17, 563–573. [Google Scholar] [CrossRef]
  221. Pérez, L.M.; Pareja-Galeano, H.; Sanchis-Gomar, F.; Emanuele, E.; Lucia, A.; Gálvez, B.G. ‘Adipaging’: Ageing and obesity share biological hallmarks related to a dysfunctional adipose tissue. J. Physiol. 2016, 594, 3187–3207. [Google Scholar] [CrossRef] [PubMed]
  222. Kadry, H.; Noorani, B.; Cucullo, L. A blood-brain barrier overview on structure, function, impairment, and biomarkers of integrity. Fluids Barriers CNS 2020, 17, 69. [Google Scholar] [CrossRef] [PubMed]
  223. Sweeney, M.D.; Zhao, Z.; Montagne, A.; Nelson, A.R.; Zlokovic, B.V. Blood-Brain Barrier: From Physiology to Disease and Back. Physiol. Rev. 2019, 99, 21–78. [Google Scholar] [CrossRef] [PubMed]
  224. Knopp, R.C.; Erickson, M.A.; Rhea, E.M.; Reed, M.J.; Banks, W.A. Cellular senescence and the blood-brain barrier: Implications for aging and age-related diseases. Exp. Biol. Med. 2023, 248, 399–411. [Google Scholar] [CrossRef]
  225. Knox, E.G.; Aburto, M.R.; Clarke, G.; Cryan, J.F.; O’Driscoll, C.M. The blood-brain barrier in aging and neurodegeneration. Mol. Psychiatry 2022, 27, 2659–2673. [Google Scholar] [CrossRef]
  226. Rempe, R.G.; Hartz, A.M.S.; Bauer, B. Matrix metalloproteinases in the brain and blood-brain barrier: Versatile breakers and makers. J. Cereb. Blood Flow Metab. 2016, 36, 1481–1507. [Google Scholar] [CrossRef]
  227. Varatharaj, A.; Galea, I. The blood-brain barrier in systemic inflammation. Brain Behav. Immun. 2017, 60, 1–12. [Google Scholar] [CrossRef]
  228. Li, Q.; Barres, B.A. Microglia and macrophages in brain homeostasis and disease. Nat. Rev. Immunol. 2018, 18, 225–242. [Google Scholar] [CrossRef]
  229. Anilkumar, S.; Wright-Jin, E. NF-κB as an Inducible Regulator of Inflammation in the Central Nervous System. Cells 2024, 13, 485. [Google Scholar] [CrossRef]
  230. Peng, J.; Xie, Y.; Pang, J.; Wu, Y.; Zhou, J.; Gu, L.; Guo, K.; Zhang, L.; Xie, B.; Yin, S.; et al. Toll-like receptor 4-mediated microglial inflammation exacerbates early white matter injury following experimental subarachnoid hemorrhage. J. Neurochem. 2023, 166, 280–293. [Google Scholar] [CrossRef]
  231. Moraes, C.A.; Hottz, E.D.; Dos Santos Ornellas, D.; Adesse, D.; de Azevedo, C.T.; d’Avila, J.C.; Zaverucha-do-Valle, C.; Maron-Gutierrez, T.; Barbosa, H.S.; Bozza, P.T.; et al. Microglial NLRP3 Inflammasome Induces Excitatory Synaptic Loss Through IL-1β-Enriched Microvesicle Release: Implications for Sepsis-Associated Encephalopathy. Mol. Neurobiol. 2023, 60, 481–494. [Google Scholar] [CrossRef] [PubMed]
  232. Nieman, A.N.; Li, G.; Zahn, N.M.; Mian, M.Y.; Mikulsky, B.N.; Hoffman, D.A.; Wilcox, T.M.; Kehoe, A.S.; Luecke, I.W.; Poe, M.M.; et al. Targeting Nitric Oxide Production in Microglia with Novel Imidazodiazepines for Nonsedative Pain Treatment. ACS Chem. Neurosci. 2020, 11, 2019–2030. [Google Scholar] [CrossRef] [PubMed]
  233. Di Filippo, M.; de Iure, A.; Giampà, C.; Chiasserini, D.; Tozzi, A.; Orvietani, P.L.; Ghiglieri, V.; Tantucci, M.; Durante, V.; Quiroga-Varela, A.; et al. Persistent activation of microglia and NADPH oxidase [corrected] drive hippocampal dysfunction in experimental multiple sclerosis. Sci. Rep. 2016, 6, 20926. [Google Scholar]
  234. Smith, A.N.; Shaughness, M.; Collier, S.; Hopkins, D.; Byrnes, K.R. Therapeutic targeting of microglia mediated oxidative stress after neurotrauma. Front. Med. 2022, 9, 1034692. [Google Scholar] [CrossRef]
  235. Kishi, S.; Nagasu, H.; Kidokoro, K.; Kashihara, N. Oxidative stress and the role of redox signalling in chronic kidney disease. Nat. Rev. Nephrol. 2024, 20, 101–119. [Google Scholar] [CrossRef]
  236. Zhu, G.; Wang, X.; Chen, L.; Lenahan, C.; Fu, Z.; Fang, Y.; Yu, W. Crosstalk Between the Oxidative Stress and Glia Cells After Stroke: From Mechanism to Therapies. Front. Immunol. 2022, 13, 852416. [Google Scholar] [CrossRef]
  237. Nicosia, N.; Giovenzana, M.; Misztak, P.; Mingardi, J.; Musazzi, L. Glutamate-Mediated Excitotoxicity in the Pathogenesis and Treatment of Neurodevelopmental and Adult Mental Disorders. Int. J. Mol. Sci. 2024, 25, 6521. [Google Scholar] [CrossRef]
  238. Troy, C.M.; Akpan, N.; Jean, Y.Y. Regulation of caspases in the nervous system implications for functions in health and disease. Prog. Mol. Biol. Transl Sci. 2011, 99, 265–305. [Google Scholar]
  239. Zaman, V.; Drasites, K.P.; Myatich, A.; Shams, R.; Shields, D.C.; Matzelle, D.; Haque, A.; Banik, N.L. Inhibition of Calpain Attenuates Degeneration of Substantia Nigra Neurons in the Rotenone Rat Model of Parkinson’s Disease. Int. J. Mol. Sci. 2022, 23, 13849. [Google Scholar] [CrossRef]
  240. Görlach, A.; Bertram, K.; Hudecova, S.; Krizanova, O. Calcium and ROS: A mutual interplay. Redox Biol. 2015, 6, 260–271. [Google Scholar] [CrossRef]
  241. Li, Y.; Li, Y.J.; Zhu, Z.Q. To re-examine the intersection of microglial activation and neuroinflammation in neurodegenerative diseases from the perspective of pyroptosis. Front. Aging Neurosci. 2023, 15, 1284214. [Google Scholar] [CrossRef]
  242. Verkhratsky, A.; Nedergaard, M. Physiology of Astroglia. Physiol. Rev. 2018, 98, 239–389. [Google Scholar] [CrossRef]
  243. Jong Huat, T.; Camats-Perna, J.; Newcombe, E.A.; Onraet, T.; Campbell, D.; Sucic, J.T.; Martini, A.; Forner, S.; Mirzaei, M.; Poon, W.; et al. The impact of astrocytic NF-κB on healthy and Alzheimer’s disease brains. Sci. Rep. 2024, 14, 14305. [Google Scholar] [CrossRef] [PubMed]
  244. Rothhammer, V.; Kenison, J.E.; Li, Z.; Tjon, E.; Takenaka, M.C.; Chao, C.C.; Alves de Lima, K.; Borucki, D.M.; Kaye, J.; Quintana, F.J. Aryl Hydrocarbon Receptor Activation in Astrocytes by Laquinimod Ameliorates Autoimmune Inflammation in the CNS. Neurol. Neuroimmunol. Neuroinflamm. 2021, 8, e946. [Google Scholar] [CrossRef] [PubMed]
  245. Patani, R.; Hardingham, G.E.; Liddelow, S.A. Functional roles of reactive astrocytes in neuroinflammation and neurodegeneration. Nat. Rev. Neurol. 2023, 19, 7395–7409. [Google Scholar] [CrossRef] [PubMed]
  246. Ageeva, T.; Rizvanov, A.; Mukhamedshina, Y. NF-κB and JAK/STAT Signaling Pathways as Crucial Regulators of Neuroinflammation and Astrocyte Modulation in Spinal Cord Injury. Cells 2024, 13, 581. [Google Scholar] [CrossRef]
  247. Simpson, D.S.A.; Oliver, P.L. ROS Generation in Microglia: Understanding Oxidative Stress and Inflammation in Neurodegenerative Disease. Antioxidants 2020, 9, 743. [Google Scholar] [CrossRef]
  248. Chen, Y.; Qin, C.; Huang, J.; Tang, X.; Liu, C.; Huang, K.; Xu, J.; Guo, G.; Tong, A.; Zhou, L. The role of astrocytes in oxidative stress of central nervous system: A mixed blessing. Cell Prolif. 2020, 53, e12781. [Google Scholar] [CrossRef]
  249. Qin, P.; Sun, Y.; Li, L. Mitochondrial dysfunction in chronic neuroinflammatory diseases (Review). Int. J. Mol. Med. 2024, 53, 47. [Google Scholar] [CrossRef]
  250. Olmos, G.; Lladó, J. Tumor necrosis factor alpha: A link between neuroinflammation and excitotoxicity. Mediat. Inflamm. 2014, 2014, 861231. [Google Scholar] [CrossRef]
  251. Mottahedin, A.; Ardalan, M.; Chumak, T.; Riebe, I.; Ek, J.; Mallard, C. Effect of Neuroinflammation on Synaptic Organization and Function in the Developing Brain: Implications for Neurodevelopmental and Neurodegenerative Disorders. Front. Cell Neurosci. 2017, 11, 190. [Google Scholar] [CrossRef] [PubMed]
  252. Golia, M.T.; Poggini, S.; Alboni, S.; Garofalo, S.; Ciano Albanese, N.; Viglione, A.; Ajmone-Cat, M.A.; St-Pierre, A.; Brunello, N.; Limatola, C.; et al. Interplay between inflammation and neural plasticity: Both immune activation and suppression impair LTP and BDNF expression. Brain Behav. Immun. 2019, 81, 484–494. [Google Scholar] [CrossRef]
  253. Jurcau, M.C.; Jurcau, A.; Cristian, A.; Hogea, V.O.; Diaconu, R.G.; Nunkoo, V.S. Inflammaging and Brain Aging. Int. J. Mol. Sci. 2024, 25, 10535. [Google Scholar] [CrossRef] [PubMed]
  254. Akhgari, A.; Michel, T.M.; Vafaee, M.S. Dendritic spines and their role in the pathogenesis of neurodevelopmental and neurological disorders. Rev. Neurosci. 2024, 35, 489–502. [Google Scholar] [CrossRef] [PubMed]
  255. Rodríguez-Martín, T.; Cuchillo-Ibáñez, I.; Noble, W.; Nyenya, F.; Anderton, B.H.; Hanger, D.P. Tau phosphorylation affects its axonal transport and degradation. Neurobiol. Aging 2013, 34, 2146–2157. [Google Scholar] [CrossRef]
  256. Gaikwad, S.; Senapati, S.; Haque, M.A.; Kayed, R. Senescence, brain inflammation, and oligomeric tau drive cognitive decline in Alzheimer’s disease: Evidence from clinical and preclinical studies. Alzheimers Dement. 2024, 20, 709–727. [Google Scholar] [CrossRef]
  257. Mehra, S.; Sahay, S.; Maji, S.K. α-Synuclein misfolding and aggregation: Implications in Parkinson’s disease pathogenesis. Biochim. Biophys. Acta Proteins Proteom. 2019, 1867, 890–908. [Google Scholar] [CrossRef]
  258. Sepúlveda, D.; Cisternas-Olmedo, M.; Arcos, J.; Nassif, M.; Vidal, R.L. Contribution of Autophagy-Lysosomal Pathway in the Exosomal Secretion of Alpha-Synuclein and Its Impact in the Progression of Parkinson’s Disease. Front. Mol. Neurosci. 2022, 15, 805087. [Google Scholar] [CrossRef]
  259. Koszła, O.; Sołek, P. Misfolding and aggregation in neurodegenerative diseases: Protein quality control machinery as potential therapeutic clearance pathways. Cell Commun. Signal. 2024, 22, 421. [Google Scholar] [CrossRef]
  260. Sabirzhanov, B.; Makarevich, O.; Barrett, J.; Jackson, I.L.; Faden, A.I.; Stoica, B.A. Down-Regulation of miR-23a-3p Mediates Irradiation-Induced Neuronal Apoptosis. Int. J. Mol. Sci. 2020, 21, 3695. [Google Scholar] [CrossRef]
  261. Yang, L.; Guttman, L.; Dawson, V.L.; Dawson, T.M. Parthanatos: Mechanisms, modulation, and therapeutic prospects in neurodegenerative disease and stroke. Biochem. Pharmacol. 2024, 228, 116174. [Google Scholar] [CrossRef] [PubMed]
  262. Mustafa, M.; Ahmad, R.; Tantry, I.Q.; Ahmad, W.; Siddiqui, S.; Alam, M.; Abbas, K.; Moinuddin; Hassan, M. I.; Habib, S.; Islam, S. Apoptosis: A Comprehensive Overview of Signaling Pathways, Morphological Changes, and Physiological Significance and Therapeutic Implications. Cells 2024, 13, 1838. [Google Scholar] [CrossRef] [PubMed]
  263. Huang, P.; Chen, G.; Jin, W.; Mao, K.; Wan, H.; He, Y. Molecular Mechanisms of Parthanatos and Its Role in Diverse Diseases. Int. J. Mol. Sci. 2022, 23, 7292. [Google Scholar] [CrossRef] [PubMed]
  264. Zhang, J.; Hu, X.; Geng, Y.; Xiang, L.; Wu, Y.; Li, Y.; Yang, L.; Zhou, K. Exploring the role of parthanatos in CNS injury: Molecular insights and therapeutic approaches. J. Adv. Res. 2024, S2090-1232, 00174–00177. [Google Scholar] [CrossRef]
  265. Rana, A.; Samtiya, M.; Dhewa, T.; Mishra, V.; Aluko, R.E. Health benefits of polyphenols: A concise review. J. Food Biochem. 2022, 46, e14264. [Google Scholar] [CrossRef]
  266. Zhang, W.; Qi, S.; Xue, X.; Al Naggar, Y.; Wu, L.; Wang, K. Understanding the Gastrointestinal Protective Effects of Polyphenols using Foodomics-Based Approaches. Front. Immunol. 2021, 12, 671150. [Google Scholar] [CrossRef]
  267. Wilson, S.M.; Oliver, A.; Larke, J.A.; Naveja, J.J.; Alkan, Z.; Awika, J.M.; Stephensen, C.B.; Lemay, D.G. Fine-Scale Dietary Polyphenol Intake Is Associated with Systemic and Gastrointestinal Inflammation in Healthy Adults. J. Nutr. 2024, 154, 3286–3297. [Google Scholar] [CrossRef]
  268. Sun, S.; Liu, Z.; Lin, M.; Gao, N.; Wang, X. Polyphenols in health and food processing: Antibacterial, anti-inflammatory, and antioxidant insights. Front. Nutr. 2024, 11, 1456730. [Google Scholar] [CrossRef]
  269. Sharma, R.; Padwad, Y. Probiotic bacteria as modulators of cellular senescence: Emerging concepts and opportunities. Gut Microbes 2020, 11, 335–349. [Google Scholar] [CrossRef]
  270. Hamamah, S.; Lobiuc, A.; Covasa, M. Antioxidant Role of Probiotics in Inflammation-Induced Colorectal Cancer. Int. J. Mol. Sci. 2024, 25, 9026. [Google Scholar] [CrossRef]
  271. Boyajian, J.L.; Ghebretatios, M.; Schaly, S.; Islam, P.; Prakash, S. Microbiome and Human Aging: Probiotic and Prebiotic Potentials in Longevity, Skin Health and Cellular Senescence. Nutrients 2021, 13, 4550. [Google Scholar] [CrossRef] [PubMed]
  272. Li, Q.; Zhang, Q.; Wang, M.; Zhao, S.; Xu, G.; Li, J. n-3 polyunsaturated fatty acids prevent disruption of epithelial barrier function induced by proinflammatory cytokines. Mol. Immunol. 2008, 45, 1356–1365. [Google Scholar] [CrossRef] [PubMed]
  273. Xiong, Y.; Li, X.; Liu, J.; Luo, P.; Zhang, H.; Zhou, H.; Ling, X.; Zhang, M.; Liang, Y.; Chen, Q.; et al. Omega-3 PUFAs slow organ aging through promoting energy metabolism. Pharmacol. Res. 2024, 208, 107384. [Google Scholar] [CrossRef] [PubMed]
  274. Ahmadi, A.R.; Shirani, F.; Abiri, B.; Siavash, M.; Haghighi, S.; Akbari, M. Impact of omega-3 fatty acids supplementation on the gene expression of peroxisome proliferator activated receptors-γ, α and fibroblast growth factor-21 serum levels in patients with various presentation of metabolic conditions: A GRADE assessed systematic review and dose-response meta-analysis of clinical trials. Front. Nutr. 2023, 10, 1202688. [Google Scholar]
  275. See, X.Z.; Yeo, W.S.; Saptoro, A. A comprehensive review and recent advances of vitamin C: Overview, functions, sources, applications, market survey and processes. Process Saf. Environ. Prot. 2024, 206, 108–129. [Google Scholar] [CrossRef]
  276. Moore, A.; Khanna, D. The Role of Vitamin C in Human Immunity and Its Treatment Potential Against COVID-19: A Review Article. Cureus 2023, 15, e33740. [Google Scholar] [CrossRef]
  277. Chu, C.C.; Chew, S.C.; Liew, W.C.; Nyam, K.L. Review article vitamin E: A multi-functional ingredient for health enhancement and food preservation. J. Food Meas. Charact. 2023, 17, 6144–6156. [Google Scholar] [CrossRef]
  278. Jin, D.; Wei, X.; He, Y.; Zhong, L.; Lu, H.; Lan, J.; Wei, Y.; Liu, Z.; Liu, H. The nutritional roles of zinc for immune system and COVID-19 patients. Front. Nutr. 2024, 11, 1385591. [Google Scholar] [CrossRef]
  279. Wessels, I.; Maywald, M.; Rink, L. Zinc as a Gatekeeper of Immune Function. Nutrients 2017, 9, 1286. [Google Scholar] [CrossRef]
  280. Narayan, S.; Dalal, R.; Rizvi, Z.A.; Awasthi, A. Zinc dampens antitumor immunity by promoting Foxp3+ regulatory T cells. Front. Immunol. 2024, 15, 1389387. [Google Scholar] [CrossRef]
  281. Sergi, C.M. The Role of Zinc in the T-Cell Metabolism in Infection Requires Further Investigation—An Opinion. Front. Immunol. 2022, 13, 865504. [Google Scholar] [CrossRef]
  282. Di Lorito, C.; Long, A.; Byrne, A.; Harwood, R.H.; Gladman, J.R.F.; Schneider, S.; Logan, P.; Bosco, A.; van der Wardt, V. Exercise interventions for older adults: A systematic review of meta-analyses. J. Sport Health Sci. 2021, 10, 29–47. [Google Scholar] [CrossRef] [PubMed]
  283. Hernández-Álvarez, D.; Rosado-Pérez, J.; Gavia-García, G.; Arista-Ugalde, T.L.; Aguiñiga-Sánchez, I.; Santiago-Osorio, E.; Mendoza-Núñez, V.M. Aging, Physical Exercise, Telomeres, and Sarcopenia: A Narrative Review. Biomedicines 2023, 11, 598. [Google Scholar] [CrossRef] [PubMed]
  284. Mathur, S.; Ardestani, A.; Parker, B.; Cappizzi, J.; Polk, D.; Thompson, P.D. Telomere length and cardiorespiratory fitness in marathon runners. J. Investig. Med. 2013, 61, 613–615. [Google Scholar] [CrossRef] [PubMed]
  285. Knez, W.L.; Coombes, J.S.; Jenkins, D.G. Ultra-endurance exercise and oxidative damage: Implications for cardiovascular health. Sports Med. 2006, 36, 429–441. [Google Scholar] [CrossRef]
  286. Samjoo, I.A.; Safdar, A.; Hamadeh, M.J.; Raha, S.; Tarnopolsky, M.A. The effect of endurance exercise on both skeletal muscle and systemic oxidative stress in previously sedentary obese men. Nutr. Diabetes 2013, 3, e88. [Google Scholar] [CrossRef]
  287. Tang, F.; Wang, W.; Wang, Y.; Lee, Y.; Lou, Q. Moderate resistance training reduces intermuscular adipose tissue and risk factors of atherosclerotic cardiovascular disease for elderly patients with type 2 diabetes. Diabetes Obes. Metab. 2024, 26, 3418–3428. [Google Scholar] [CrossRef]
  288. Goodenough, C.G.; Wogksch, M.D.; Kundu, M.; Lear, M.; Thomas, P.G.; Srivastava, D.K.; Wang, Z.; Armstrong, G.T.; Hudson, M.M.; Robison, L.L.; et al. Associations between exercise capacity, p16INK4a expression and inflammation among adult survivors of childhood cancer. Front. Oncol. 2022, 12, 1014661. [Google Scholar] [CrossRef]
  289. Justice, J.N.; Gregory, H.; Tchkonia, T.; LeBrasseur, N.K.; Kirkland, J.L.; Kritchevsky, S.B.; Nicklas, B.J. Cellular Senescence Biomarker p16INK4a+ Cell Burden in Thigh Adipose is Associated With Poor Physical Function in Older Women. J. Gerontol. A Biol. Sci. Med. Sci. 2018, 73, 939–945. [Google Scholar] [CrossRef]
  290. Thanapairoje, K.; Junsiritrakhoon, S.; Wichaiyo, S.; Osman, M.A.; Supharattanasitthi, W. Anti-ageing effects of FDA-approved medicines: A focused review. J. Basic Clin. Physiol. Pharmacol. 2023, 34, 277–289. [Google Scholar] [CrossRef]
  291. Mohammed, I.; Hollenberg, M.D.; Ding, H.; Triggle, C.R. A Critical Review of the Evidence That Metformin Is a Putative Anti-Aging Drug That Enhances Healthspan and Extends Lifespan. Front. Endocrinol. 2021, 12, 718942. [Google Scholar] [CrossRef] [PubMed]
  292. Moiseeva, O.; Deschênes-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-κB activation. Aging Cell 2013, 12, 489–498. [Google Scholar] [CrossRef] [PubMed]
  293. Fang, J.; Yang, J.; Wu, X.; Zhang, G.; Li, T.; Wang, X.; Zhang, H.; Wang, C.C.; Liu, G.H.; Wang, L. Metformin alleviates human cellular aging by upregulating the endoplasmic reticulum glutathione peroxidase 7. Aging Cell 2018, 17, e12765. [Google Scholar] [CrossRef] [PubMed]
  294. Liu, L.; Patnana, P.K.; Nimmagadda, S.C. Low-dose metformin and PEN2-dependent lysosomal AMPK activation: Benefits outnumber side effects. Signal Transduct. Target Ther. 2022, 7, 178. [Google Scholar] [CrossRef]
  295. Lushchak, O.; Piskovatska, V.; Strilbytska, O.; Kindrat, I.; Stefanyshyn, N.; Koliada, A.; Bubalo, V.; Storey, K.B.; Vaiserman, A. Aspirin as a Potential Geroprotector: Experimental Data and Clinical Evidence. Adv. Exp. Med. Biol. 2021, 1286, 145–161. [Google Scholar]
  296. Lelarge, V.; Capelle, R.; Oger, F.; Mathieu, T.; Le Calvé, B. Senolytics: From pharmacological inhibitors to immunotherapies, a promising future for patients’ treatment. NPJ Aging 2024, 10, 12. [Google Scholar] [CrossRef]
  297. Luís, C.; Maduro, A.T.; Pereira, P.; Mendes, J.J.; Soares, R.; Ramalho, R. Nutritional senolytics and senomorphics: Implications to immune cells metabolism and aging—From theory to practice. Front. Nutr. 2022, 9, 958563. [Google Scholar] [CrossRef]
  298. Martel, J.; Ojcius, D.M.; Wu, C.Y.; Peng, H.H.; Voisin, L.; Perfettini, J.L.; Ko, Y.F.; Young, J.D. Emerging use of senolytics and senomorphics against aging and chronic diseases. Med. Res. Rev. 2020, 40, 2114–2131. [Google Scholar] [CrossRef]
  299. Lagoumtzi, S.M.; Chondrogianni, N. Senolytics and senomorphics: Natural and synthetic therapeutics in the treatment of aging and chronic diseases. Free Radic. Biol. Med. 2021, 171, 169–190. [Google Scholar] [CrossRef]
  300. Power, H.; Valtchev, P.; Dehghani, F.; Schindeler, A. Strategies for senolytic drug discovery. Aging Cell 2023, 22, e13948. [Google Scholar] [CrossRef]
  301. Islam, M.T.; Tuday, E.; Allen, S.; Kim, J.; Trott, D.W.; Holland, W.L.; Donato, A.J.; Lesniewski, L.A. Senolytic drugs, dasatinib and quercetin, attenuate adipose tissue inflammation, and ameliorate metabolic function in old age. Aging Cell 2023, 22, e13767. [Google Scholar] [CrossRef] [PubMed]
  302. Suvakov, S.; Cubro, H.; White, W.M.; Butler Tobah, Y.S.; Weissgerber, T.L.; Jordan, K.L.; Zhu, X.Y.; Woollard, J.R.; Chebib, F.T.; Milic, N.M.; et al. Targeting senescence improves angiogenic potential of adipose-derived mesenchymal stem cells in patients with preeclampsia. Biol. Sex Differ. 2019, 10, 49. [Google Scholar] [CrossRef] [PubMed]
  303. Dagher, O.; Mury, P.; Thorin-Trescases, N.; Noly, P.E.; Thorin, E.; Carrier, M. Therapeutic Potential of Quercetin to Alleviate Endothelial Dysfunction in Age-Related Cardiovascular Diseases. Front. Cardiovasc. Med. 2021, 8, 658400. [Google Scholar] [CrossRef] [PubMed]
  304. Saccon, T.D.; Nagpal, R.; Yadav, H.; Cavalcante, M.B.; Nunes, A.D.C.; Schneider, A.; Gesing, A.; Hughes, B.; Yousefzadeh, M.; Tchkonia, T.; et al. Senolytic Combination of Dasatinib and Quercetin Alleviates Intestinal Senescence and Inflammation and Modulates the Gut Microbiome in Aged Mice. J. Gerontol. A Biol. Sci. Med. Sci. 2021, 76, 1895–1905. [Google Scholar] [CrossRef]
  305. Ota, H.; Kodama, A. Dasatinib plus quercetin attenuates some frailty characteristics in SAMP10 mice. Sci. Rep. 2022, 12, 2425. [Google Scholar] [CrossRef]
  306. Zhao, J.; Zheng, L.; Dai, G.; Sun, Y.; He, R.; Liu, Z.; Jin, Y.; Wu, T.; Hu, J.; Cao, Y.; et al. Senolytics cocktail dasatinib and quercetin alleviate chondrocyte senescence and facet joint osteoarthritis in mice. Spine J. 2025, 25, 184–198. [Google Scholar] [CrossRef]
  307. Nogueira-Recalde, U.; Lorenzo-Gómez, I.; Blanco, F.J.; Loza, M.I.; Grassi, D.; Shirinsky, V.; Shirinsky, I.; Lotz, M.; Robbins, P.D.; Domínguez, E.; et al. Fibrates as drugs with senolytic and autophagic activity for osteoarthritis therapy. EBioMedicine 2019, 45, 588–605. [Google Scholar] [CrossRef]
  308. Sheikh, A.; Iqbal, M. Taurine as a potential anti-ageing therapy: The key to reversing the ageing process? Short communication. Ann. Med. Surg. 2023, 85, 3759–3760. [Google Scholar] [CrossRef]
  309. Ni, Y.Q.; Liu, Y.S. New Insights into the Roles and Mechanisms of Spermidine in Aging and Age-Related Diseases. Aging Dis. 2021, 12, 1948–1963. [Google Scholar] [CrossRef]
Figure 1. Cellular mechanisms that occur in the CNS neurons of individuals with Alzheimer’s disease. Abbreviations: NMDA (N-methyl-D-aspartate) and ROS (reactive oxygen species).
Figure 1. Cellular mechanisms that occur in the CNS neurons of individuals with Alzheimer’s disease. Abbreviations: NMDA (N-methyl-D-aspartate) and ROS (reactive oxygen species).
Biomolecules 15 00404 g001
Figure 2. The process through which senescent cells secrete SASP mediators in response to damage caused by oxidative stress, telomere shortening, and DNA impairment. Abbreviations: DNA (deoxyribonucleic acid), IL-1β (Interleukin 1 beta), IL-6 (Interleukin 6), IL-8 (Interleukin 8), TNF-α (tumor necrosis factor alpha), CCL2 (C-C motif chemokine ligand 2), CCL4 (C-C motif chemokine ligand 4), MMP-3 (matrix metalloproteinase 3), MMP-13 (matrix metalloproteinase 13), TGF-β (transforming growth factor beta), and IGF-1 (insulin-like growth factor 1).
Figure 2. The process through which senescent cells secrete SASP mediators in response to damage caused by oxidative stress, telomere shortening, and DNA impairment. Abbreviations: DNA (deoxyribonucleic acid), IL-1β (Interleukin 1 beta), IL-6 (Interleukin 6), IL-8 (Interleukin 8), TNF-α (tumor necrosis factor alpha), CCL2 (C-C motif chemokine ligand 2), CCL4 (C-C motif chemokine ligand 4), MMP-3 (matrix metalloproteinase 3), MMP-13 (matrix metalloproteinase 13), TGF-β (transforming growth factor beta), and IGF-1 (insulin-like growth factor 1).
Biomolecules 15 00404 g002
Figure 3. Microglial activation induced by SASP mediators in resting-state microglia. The activation of TLR4 receptors by SASP factors stimulates microglial activation and the release of various pro-inflammatory cytokines. Abbreviations: IL-1α (Interleukin 1 alpha), IL-1β (Interleukin 1 beta), IL-12 (Interleukin 12), IL-23 (Interleukin 23), TNF-α (tumor necrosis factor alpha), iNOS (inducible nitric oxide synthase), and NADPH (nicotinamide adenine dinucleotide phosphate—reduced form).
Figure 3. Microglial activation induced by SASP mediators in resting-state microglia. The activation of TLR4 receptors by SASP factors stimulates microglial activation and the release of various pro-inflammatory cytokines. Abbreviations: IL-1α (Interleukin 1 alpha), IL-1β (Interleukin 1 beta), IL-12 (Interleukin 12), IL-23 (Interleukin 23), TNF-α (tumor necrosis factor alpha), iNOS (inducible nitric oxide synthase), and NADPH (nicotinamide adenine dinucleotide phosphate—reduced form).
Biomolecules 15 00404 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

García-Domínguez, M. Pathological and Inflammatory Consequences of Aging. Biomolecules 2025, 15, 404. https://doi.org/10.3390/biom15030404

AMA Style

García-Domínguez M. Pathological and Inflammatory Consequences of Aging. Biomolecules. 2025; 15(3):404. https://doi.org/10.3390/biom15030404

Chicago/Turabian Style

García-Domínguez, Mario. 2025. "Pathological and Inflammatory Consequences of Aging" Biomolecules 15, no. 3: 404. https://doi.org/10.3390/biom15030404

APA Style

García-Domínguez, M. (2025). Pathological and Inflammatory Consequences of Aging. Biomolecules, 15(3), 404. https://doi.org/10.3390/biom15030404

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