Next Article in Journal
Perceptions of Knowledge, Disease Impact and Predictive Genetic Testing in Family Members at Risk to Develop Early-Onset Alzheimer’s Disease (EOAD) and Their Levels of Suicidal Ideation: A Mixed Study
Previous Article in Journal
The Role of Telemedicine in the Treatment of Cognitive and Psychological Disorders in Parkinson’s Disease: An Overview
Previous Article in Special Issue
In Vitro and Predictive Computational Toxicology Methods for the Neurotoxic Pesticide Amitraz and Its Metabolites
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Among Gerontogens, Heavy Metals Are a Class of Their Own: A Review of the Evidence for Cellular Senescence

1
Pediatrics Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
2
Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
*
Author to whom correspondence should be addressed.
Brain Sci. 2023, 13(3), 500; https://doi.org/10.3390/brainsci13030500
Submission received: 7 February 2023 / Revised: 10 March 2023 / Accepted: 13 March 2023 / Published: 16 March 2023
(This article belongs to the Special Issue Advance in Study of Neurotoxic Chemicals in the Environment)

Abstract

:
Advancements in modern medicine have improved the quality of life across the globe and increased the average lifespan of our population by multiple decades. Current estimates predict by 2030, 12% of the global population will reach a geriatric age and live another 3–4 decades. This swelling geriatric population will place critical stress on healthcare infrastructures due to accompanying increases in age-related diseases and comorbidities. While much research focused on long-lived individuals seeks to answer questions regarding how to age healthier, there is a deficit in research investigating what aspects of our lives accelerate or exacerbate aging. In particular, heavy metals are recognized as a significant threat to human health with links to a plethora of age-related diseases, and have widespread human exposures from occupational, medical, or environmental settings. We believe heavy metals ought to be classified as a class of gerontogens (i.e., chemicals that accelerate biological aging in cells and tissues). Gerontogens may be best studied through their effects on the “Hallmarks of Aging”, nine physiological hallmarks demonstrated to occur in aged cells, tissues, and bodies. Evidence suggests that cellular senescence—a permanent growth arrest in cells—is one of the most pertinent hallmarks of aging and is a useful indicator of aging in tissues. Here, we discuss the roles of heavy metals in brain aging. We briefly discuss brain aging in general, then expand upon observations for heavy metals contributing to age-related neurodegenerative disorders. We particularly emphasize the roles and observations of cellular senescence in neurodegenerative diseases. Finally, we discuss the observations for heavy metals inducing cellular senescence. The glaring lack of knowledge about gerontogens and gerontogenic mechanisms necessitates greater research in the field, especially in the context of the global aging crisis.

1. Introduction: Two Faces to a Toxic Aging Crisis

We are experiencing an unprecedented rise in the age of our global population due to increased lifespan; a phenomenon described as an aging crisis. Estimates suggest by 2030, 12% of the world’s population will be geriatric, with this estimate reaching 16% in 2050 and 23% in 2100 [1]. This aging crisis will impact developed nations to a greater extent, where the geriatric population will reach 20% by 2030. The term “geriatric” describes an individual who has at least a 2% chance of mortality within a year, though geriatrics are colloquially defined to be individuals 65 years of age or older [2]. Such a dramatic increase in the geriatric population will place significant constraints on our healthcare systems worldwide, increasing the need for medical care, personnel, and devices to treat aging-related health conditions.
Compounding this aging issue, we are faced with global environmental pollution of innumerable chemicals that threaten our health—a separate crisis that challenges aging health in two key ways, which we refer to as a “toxic aging coin” [3]. On the heads side of this toxic aging coin, we consider how life stage (e.g., juvenile vs. middle-aged vs. geriatric) impacts the toxic effects of chemicals. On the tails side, we consider how chemicals accelerate aging, or how they act as gerontogens.
We know chemical exposures early in life can have long-lasting effects, which contribute to health status in later life, as described by the Developmental Origins of Health and Disease (DOHaD) hypothesis [4]. We recognize elderly populations are a vulnerable group in regard to health issues and threats to public health (e.g., epidemics), but we rarely recognize geriatrics as a distinct demographic for toxicological consideration. Previously, geriatrics were often dismissed in toxicology as already having deteriorating health or too close to death to consider for risk assessments. However, with increased lifespan potentially contributing 3–4 more decades of geriatric life, there is a critical need to better understand the toxicology of aging and improve risk assessment for all ages. This need is affirmed by the NIEHS in Theme 1 of their strategic plan, “Advancing Environmental Health Sciences encompasses the study of all levels of biological organization… at all stages across the lifespan, from preconception through old age” [5].
We are only just starting to understand how some environmental pollutants act as gerontogens, i.e., chemicals that induce or accelerate biological aging processes. At a clinical level, gerontogens may contribute to a broad spectrum of age-related diseases and comorbidities, affecting all organ systems, e.g., osteoporosis, cancers, diabetes, and dementias. We recognize mortality and age-related diseases are linked to or exacerbated by environmental pollutants; thus, it is imperative we better understand gerontogenic mechanisms [6,7]. To better understand gerontogens, it is essential to recognize two contexts of aging: chronological age (i.e., age according to birth year) and biological aging (i.e., cellular/tissue aging caused by gerontogenic factors). The presentation of biological aging may have traits specific to each type of cell, tissue, or organ; yet, some aging effects are consistently observed and are collectively referred to as the “Hallmarks of Aging” [8]. These hallmarks are nine distinct changes in biological processes that increase with normal aging: (1) stem cell exhaustion; (2) altered intercellular communication; (3) genomic instability; (4) telomere attrition; (5) epigenetic alterations; (6) loss of proteostasis; (7) dysregulation of nutrient sensing; (8) mitochondrial dysfunction; and (9) cellular senescence [8]. Consideration for the hallmarks of aging is further described for specific organs, such as the “Hallmarks of Brain Aging” [9]. In addition to the nine hallmarks of aging, brain aging includes (10) dysregulation of neuronal calcium homeostasis; (11) oxidative stress; (12) impaired molecular waste disposal; (13) impaired DNA repair capacity; (14) glial cell activation; and (15) impaired adaptive stress response signaling [9]. As such, these aging hallmarks become incredibly useful tools for assessing the gerontogenic effects of environmental pollutants.
Perhaps the most pertinent and readily measured aging hallmark is cellular senescence, a permanent state of cellular growth arrest with a distinct shift in metabolism and secretory phenotype. Cellular senescence represents a permanent shift in a cell’s phenotype to an aged phenotype, likely caused by mechanisms involving one or multiple other aging hallmarks. Phenotypically, senescent cells exhibit increased expression of p16INK4a and p21 following the activation of either the p53-p21 or Rb-p16INK4a pathways, respectively [10,11]. In addition to an increased expression of p16INK4a and p21, some of the most identifiable characteristics of senescent cells include the expression of senescence-associated β-Galactosidase (SA-β-Gal), mitochondrial dysfunction, increased lysosomal content, and a large, irregularly shaped cell body [12]. The accumulation of senescent cells can promote further senescence in neighboring cells through secreted factors or senescence-associated secretory phenotype (SASP) factors [13]. Importantly, senescent cells accumulate with age in all tissue types, and their selective clearance with senolytic drugs can attenuate aging-related pathology and clinical symptoms [14,15,16,17]. Hence, improving our understanding of gerontogenic mechanisms leading to cellular senescence will be imperative to improve healthy aging and diminishing adverse aging effects of chemicals.
Here, we will discuss the evidence to consider heavy metals as a class of gerontogens. Our discussion will start with identifying why heavy metals are a health concern. We will review unique heavy metal exposures to geriatric populations. Then, we will discuss what an aged brain looks like, and how heavy metals induce some brain aging effects in human and animal studies. Finally, we will review and discuss what is known about how heavy metals induce cellular senescence.

2. Materials and Methods

2.1. Search Strategy

Literature searches utilized PubMed and Google Scholar for literature relevant to heavy metals, cellular senescence, hallmarks of aging, and physiological processes of an aging brain. Searches began in October 2022 and concluded in January 2023. Searches were specific to metals and cellular senescence (e.g., “arsenic cellular senescence”) or metals and relevant biomarkers for cellular senescence (e.g., “arsenic p21 expression”). Additional searches included metals and the broad term hallmarks of aging (e.g., “arsenic hallmarks of aging”) or a metal and a specific hallmark of aging (e.g., “arsenic genomic instability”). To ensure all relevant research was found, search terms often included the abbreviation for the metal and the term “heavy metal” alongside the previously described searches (e.g., “Pb lead heavy metal cellular senescence”). This was conducted specifically in searches relevant to lead to avoid confusion with its homonyms and limit the presence of irrelevant research. Searches for current information regarding heavy metal contamination and human exposure followed a similar pattern, using the search term “heavy metals environmental contamination.” Searches for information regarding the physiology of the aging brain used terminology specific to brain regions or structures alongside the term aging and the physiological process (e.g., gray matter attrition aging, blood–brain barrier leakage aging). Searches for specific neurodegenerative diseases relevant to these diseases often followed a similar query with regards to up-to-date statistics (e.g., Alzheimer’s disease prevalence), the pathology of the disease (e.g., Alzheimer’s disease pathology), or the significance of cellular senescence or the biomarkers of senescence in these diseases (e.g., Alzheimer’s disease cellular senescence, Alzheimer’s disease p21 expression). Articles cited within relevant publications were also selected and examined.

2.2. Exclusion Strategy

Articles were excluded from searches if the model utilized was not a cell culture model, an animal model, or a study of human participants. This was performed to ensure the information discussed is wholly relevant to human health. Articles were also prioritized based on the date of publication to ensure that the information gathered and the data discussed are not outdated. Particular emphasis on publication date was given to research discussing environmental contamination and prevalence of neurodegenerative diseases.

3. Environmental Heavy Metal Pollution Poses a Significant Health Threat

“Heavy metals” refers to metals and metalloids studied in the context of environmental pollution, toxicology, and adverse effects on biological life [18]. They are part of our natural environment and may be released at toxic levels in events, such as volcanic eruptions or forest fires, but much of the concern for heavy metal pollution stems from human industry. Heavy metals pervade our societies and have innumerable valuable uses ranging from electronics, pigments and dyes, and metals to agricultural practices, and many of these industries release contaminated effluents into the environment that can lead to dangerous human exposures [19,20]. Heavy metals pollute our air, water, soil, and food supplies, and are one of the most dangerous forms of environmental pollution, as they generally cannot be metabolized or degraded into less toxic forms, and they can persist in our environment for decades [19,21,22,23,24,25]. Importantly, among the top ten chemicals on the 2022 ATSDR Hazard Priority List, the top three are heavy metals: arsenic, lead, and mercury [26]. Their neurotoxic effects are widely recognized across the age spectrum [27,28].

4. Unique Heavy Metal Exposures in Geriatric Populations

In addition to environmental pollution, geriatric populations experience some chemical exposures unique to their age group. Polypharmacy is a prominent concern in geriatric individuals contending with multiple comorbidities and deteriorating health. Though it is out of the scope of this review, studies show geriatric polypharmacy has adverse effects such as cognitive impairment, frailty, malnutrition, and mortality [29,30]. Polypharmacy presents challenges to geriatric health involving heavy metals in two key ways: (1) heavy metals are a common impurity in polymeric excipients (i.e., inert chemicals/substances required for pharmaceutical production), including lead, arsenic, mercury, chromium, nickel, cadmium, and iron, and (2) can significantly alter toxicokinetic responses to heavy metals [31].
The use of medical devices increases with age and presents unique chemical exposures, particularly metal-on-metal (MoM) joint replacements. One study estimated hip and knee replacements will have an increased incidence of 84% and 183%, respectively, from 2001 to 2026 in New Zealand [32]. MoM hip replacements fail at a greater rate than prostheses made from other materials, with the 6-year failure rate for MoM hip replacements reaching up to 50% [33]. Failed MoM joint replacements can result in metal debris released inside the body, resulting in unusually high levels of cobalt (Co) and chromium (Cr) [34]. Such internal exposures have localized (e.g., inflammation, necrosis, and osteolysis) and systemic effects (e.g., neurological effects, visual impairment, cardiomyopathy, and hypothyroidism) [34,35]. Patients have exhibited toxic levels of Co and Cr as high as 669.4 nmol/L and 664 nmol/L [36]. Metallosis caused by failed MoM joint replacements induces various toxic effects, including a host of neurological deficits (e.g., peripheral neuropathy, sensory loss, tinnitus, and gray matter atrophy) [37,38,39]. While there is no estimate for a global trend of the prevalence of MoM prosthetics, there is a clear need for an assessment of the toxic outcomes geriatric individuals experience from exposure to a wide array of medical devices.

5. Considerations for Brain Aging

To understand how heavy metals might induce brain aging, we will first review how we assess an aged brain. Several distinct behavioral changes occur in aged populations, including decreased locomotor activity, decreased cognitive abilities, impaired learning and memory abilities, and depression [40]. Many of these behaviors can be readily assessed in human or animal studies. These aging-related behaviors are accompanied by morphological and biochemical changes in the brain, such as decreased white matter volume, gray matter volume, and neurotransmitter levels [41,42]. The aged brain is often described with neurodegeneration, neuroinflammation, loss of dendritic densities, changes in synaptic plasticity, and changes in neurotransmission [43,44]. Aging-related loss of gray matter is apparent in the cerebral cortex, caudate nucleus, pallidum, amygdala, and hippocampus, while loss of white matter is observed in the anterior thalamic radiations, anterior limb of the internal capsule, and cerebellum [45]. These changes are also accompanied by decreased cortical thickness, hippocampal volume, and frontal lobe volume [46]. Aged brains exhibit an accumulation of a wide variety of senescent cells, including neurons, astrocytes, microglia, endothelial cells, and oligodendrocytes, each of which affects brain health distinctly [47,48,49]. Cellular senescence in the brain results in decreased synaptic plasticity and contributes to the overall behavioral changes associated with an aging brain. These effects can be reversed or alleviated with the application of senolytic drugs that selectively induce apoptosis in senescent cells, demonstrating the pertinence of cellular senescence to brain aging [48].

5.1. Heavy Metals and Cellular Senescence Observations in Neurodegenerative Diseases

Most often researchers consider how heavy metals contribute to age-related neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis. Many studies have reported links between heavy metal exposure and these neurodegenerative diseases [27,50,51,52]. More recently, cellular senescence has gained attention in these neurodegenerative diseases [53,54].
Alzheimer’s disease (AD), a progressive neurodegenerative disease, is the most prevalent form of dementia and is often considered a primary phenotype of brain aging. In 2022, the global number of people with AD (i.e., AD with dementia, prodromal AD, or preclinical AD) was estimated to affect over 416 million people worldwide and accounted for 22% of all people aged 50 and above [55]. AD is largely a sporadic disease, with less than 10% of cases attributed to genetic causes [56]. Behavioral changes related to AD include progressively deteriorating cognitive abilities and are most often diagnosed by a mental exam or cognitive assessment [57]. Pathologically, AD is characterized by cortical, hippocampal, and amygdala atrophy, preceded by the accumulation of amyloid beta plaques and neurofibrillary tangles [58]. Several heavy metals are linked to AD pathogenesis and progression, including iron, copper, manganese, zinc, lead, aluminum, mercury, and cadmium [59,60]. Recent evidence demonstrated increased populations of senescent neurons, astrocytes, and microglia early in AD progression [55]. Further, the brain tissue of patients who suffered from early onset AD (ages 35–50) had significantly increased expression of p16INK4a in astrocytes, with a greater burden of increased p16INK4a expression in the prefrontal cortex, an area known to atrophy during AD progression [58,61]. Cell culture studies show amyloid-β1–42 exposure induces senescence in adult neural stem cells [62]. Finally, a recent human trial reported improved AD symptoms with senolytic therapy [16]. Altogether, there is ample evidence for cellular senescence playing a pathological role in AD.
Parkinson’s disease (PD) was estimated to affect over 8.5 million people in 2019 with increasing global trends of PD burden over the previous two decades [63]. Similar to AD, PD is an idiopathic disease with approximately 10% of cases caused by genetics [64]. Heavy metals linked with PD etiology include iron, zinc, copper, lead, mercury, manganese, aluminum, arsenic, cadmium, and selenium [65,66]. PD is attributed to the death of dopaminergic neurons in the substantia nigra and loss of dopaminergic axon terminals in the striatum, which presents as motor symptoms of PD [67,68]. PD can also be classified with or without dementia, depending on if cognitive abilities are affected by the disease [69]. PD patients exhibited increased SA-β-Gal expression in cerebrospinal fluid [54]. Astrocytes cultured with paraquat, a pesticide associated with PD, exhibited increased expression of SA-β-Gal, p16INK4a mRNA, and mRNA of SASP-related cytokines (IL-1α, IL-6, and IL-8) [70,71]. PD patients also presented with other hallmarks of aging, such as loss of proteostasis, mitochondrial dysfunction, and altered cell-to-cell communication [72].
Amyotrophic lateral sclerosis (ALS) has a worldwide prevalence of 4.4 in 100,000 and has steadily increased over the last 50 years [73]. ALS has an average age of onset of 55 years with a short life expectancy (3–5 years) after diagnosis [74,75]. ALS is a rapidly progressive neurodegenerative disease characterized by the degeneration of motor neurons in the brainstem and spinal cord [74]. Epidemiological evidence points to exposure to heavy metals contributing to ALS risk, particularly lead, mercury, cadmium, and selenium [76,77]. Mouse models suggest that ALS progression is driven by cellular senescence, as disease progression was correlated with the expression of cellular senescence markers (p16INK4a, p21, and SA-β-Gal) in microglia, astrocytes, and spinal motor neurons [78]. The post mortem brain tissue of individuals who suffered from ALS had a greater proportion of cells expressing cellular senescence markers such as p16INK4a in astrocytes and p21 in both astrocytes and neurons [79].
Further details regarding exposure to heavy metals or metalloids and the development of neurodegenerative diseases can be found in Table 1. It should be noted that two metals (iron and zinc) thoroughly discussed in PD and AD were left out of this table; these metals serve physiological roles within cells/tissues that contribute to disease process, while human exposures to these metals are not known to contribute to disease. For reviews on the roles of iron and zinc in AD and PD, see reviews [80,81,82,83,84].

5.2. Age-Related BBB/NVU Impairment

The most significant changes to occur in brain aging are decreased function and impaired integrity of the blood–brain barrier (BBB) and the neurovascular unit (NVU) [117,118]. The NVU is a structure that encompasses the brain’s vasculature, composed of neurons, endothelial cells, astrocytic end feet, pericytes, and microglia, which create a physical barrier between the blood and brain parenchyma [119]. Endothelial cells and pericytes work together to secrete the basal lamina that surrounds blood vessels and to create the BBB. The BBB is a physical and metabolic barrier that creates a tightly sealed vascular wall, preventing exogenous materials from entering the brain [120]. This BBB formation makes the vascular endothelial cells of the brain phenotypically distinct from those elsewhere in the body, expressing specific tight junction proteins to form the BBB. These tight junction proteins include occludins, claudins, junctional adhesion molecules, and cytosolic proteins (e.g., zona-occludins-1, -2, and -3) [121]. The NVU and BBB gradually deteriorate with age and some brain regions exhibit greater leakage than others, such as the hippocampus (specifically CA1 and the dentate gyrus) [122,123,124,125]. Further, the olfactory bulb and circumventricular organs lack any BBB protection, including the pineal gland, neurohypophysis, area postrema, subfornical organ, median eminence, and the vascular organ of the lamina terminalis [126]. Increased permeability of the NVU will also lead to increased susceptibility to the neurotoxic effects of various chemicals, such as heavy metals.
There are many reports of heavy metals affecting BBB integrity and function. A thorough discussion of heavy metal toxicity at the BBB is beyond the scope of this paper, see references for more detailed reviews [127,128,129]. Lead (Pb) is the most often reported heavy metal for BBB toxicity and shows a higher affinity for accumulating in brain endothelial cells than other brain cell types [130]. Pb can also cross the BBB using Ca ATPase pumps and induce BBB permeability [131,132,133,134,135]. Other heavy metals observed to induce BBB permeability include cadmium, copper, mercury, and arsenic [136,137,138,139,140,141,142]. Many studies using these heavy metals have expanded their focus to assess changes in BBB proteins expressed in the vasculature, reporting decreased tight junction proteins and increased adherens proteins as indicators of BBB impairment. Decreased occludin is the tight junction protein most often reported decreased in heavy metal BBB toxicity [132,133,143,144,145]. Several studies also report metal-induced decreases in claudins and zona occludins proteins [133,142,144,145,146,147]. Finally, several reports observed elevated expression of adherens proteins (e.g., ICAM-1 and VCAM-1) in endothelial cells following heavy metal exposure, indicating a shift in endothelial cells toward an inflammatory phenotype [148,149,150]. Such an increase in adherens proteins increases interaction with circulating white blood cells and promotes their intravasation into the brain parenchyma [151,152,153].
Cellular senescence may contribute to aging-related BBB leakage and has gained a lot of attention in recent years [154,155,156,157]. Cellular senescence within the NVU or BBB is observed in the brains of patients with Alzheimer’s and Parkinson’s diseases [71,158,159,160]. Several studies with aged mice or aging-accelerated mice demonstrated increased BBB leakiness associated with senescent endothelial cells [161,162,163]. Much of the mechanisms for brain endothelial cell senescence contributing to BBB leakage remain to be elucidated. A seminal paper using an in vitro BBB model and BubR1 hylomorphic mice reported senescent endothelial cells accumulated in aged cerebrovasculature and senescent endothelial cells exhibited impaired BBB integrity with reduced tight junction coverage [161].

6. Heavy Metals Induce Cellular Senescence

Here, we present the evidence for heavy metals as a class of gerontogen. The literature for heavy-metal-induced cellular senescence specific to the brain is limited, so we broaden our focus here to heavy-metal-induced senescence in any mammalian tissue or cell type. A review of the research for heavy metals contributing to all the hallmarks of aging is beyond the scope of this review, so we limit our discussion to metals inducing cellular senescence. References to metals inducing other aging hallmarks are provided where applicable. Chromium, arsenic, cadmium, lead, copper, uranium, and silver have all been observed to induce cellular senescence in a variety of cell types and research models, whereas, zinc, nickel, and copper have limited evidence, highlighting the need for additional research. A summary of senescence markers that increased in response to heavy metal or metalloid exposure is provided in Table 2.

6.1. Arsenic

Arsenic (As) is one of the most important environmental contaminants when considering metals, given the widespread global exposure to this metal through sources such as drinking water. Doses as low as 0.25 μM As induce a nearly two-fold expression of p16 and p21 in skin cell culture models [164]. In animal models, 50 ppm As increased p16 expression even after As exposure had been terminated [165]. A human study in West Bengal, India, found peripheral blood mononuclear cells of individuals exposed to contaminated drinking water exhibited increased protein levels of p16, p21, p53, SA-β-Gal, and SASP-associated inflammatory cytokines [166]. Other studies found increased expression of SASP and SA-β-Gal in both animal and cell culture models [167,168,169]. One study observed genomic instability, telomere dysfunction, and a senescent phenotype in glioma cell culture after 48 h incubation with 4 or 8 μM As2O3 [170]. Altogether, evidence for As-induced cellular senescence is found in cell culture, animal, and human studies.

6.2. Lead

Lead pollution in air and drinking water has a long history of challenging human health. Pb exposure in animal models increased G1 cell cycle arrest in hematopoietic stem cells and increased the expression of SA-β-Gal in CD90+CD45- cells following low-dose exposure to Pb [171]. Studies utilizing rat models observed increased gene expression of SASP inflammatory cytokines (IL-1α, IL-1β, IL-6, and IL-8) in pups whose mothers had been treated with 0, 2000, or 4000 ppm lead acetate during gestation [172]. This study also reported senescence-associated changes in cells of the subgranular zone, with decreased expression of Pcna and Apex1 mRNA levels (correlated with cell proliferation), increased expression of Chek1 and Cdknc1c mRNA levels (cell cycle arrest marker), and increased expression of γH2AX protein levels (DNA damage markers). These effects indicate that Pb-induced genotoxicity contributed to senescent phenotypes in the hippocampus. Animal and cell culture models demonstrated the gerontogenic effects of Pb with evidence of premature senescence in the subgranular zone in one animal study, emphasizing the regional or localized effects of senescence.

6.3. Cadmium

Cadmium (Cd) induces senescent phenotypes and several other hallmarks of aging. Cell culture models demonstrated the activation of the p53-p21 pathway following treatment with 15 μM cadmium chloride (CdCl2) [173]. Primary neurons isolated from fetal mice exhibited increased expression of p53, p21, and p16 following 24 h exposure to 10 μM Cd [174]. This study reported an increased expression of SASP and Senescence-Associated-β-Galactosidase (SA-β-Gal). One study using zebrafish exposed to 24 μM CdCl2 reported increased expression of lipid peroxidation (MDA measured in zebrafish adult hepatic tissue) and increased SA-β-Gal expression in primary human dermal fibroblasts [197]. Luo et al. (2021) reported increased markers of genomic instability and cellular senescence in mesenchymal stromal cells derived from the bone marrow of Sprague–Dawley rats [175]. In summary, there is sufficient evidence of Cd-induced senescence in animal and cell culture models.

6.4. Chromium

Chromium (Cr), in the valence state of hexavalent chromium (Cr(VI)), readily induced cellular senescence following the activation of the p53-p21 pathway [176,177,178]. Culturing BEAS-2B lung fibroblasts with 0.5 or 1 μM Cr(VI) induced the expression of SA-β-Gal [179]. In hepatocyte cell culture, 10 nM Cr(VI) induced premature senescence with increased expression of SA-β-Gal and SASP proteins (COTL1, ENO1, and PRDX2) [180]. Hepatocytes treated with 10 nM Cr(VI) also show increased mRNA expression of inflammatory cytokines (IL-6 and IL-8) and senescence markers (p53 and p21) [181]. In the context of other hallmarks of aging, hepatocytes treated with 10 nM Cr(VI) altered mitochondrial morphology while inducing cellular senescence (confirmed by SA-β-Gal expression) [182]. A human study demonstrated occupational exposure to Cr(VI) increased blood serum levels of Apolipoprotein J/Clusterin, another aging biomarker [198]. Cr(VI) is well described to induce senescence in cell culture and aging effects in human populations.

6.5. Copper

Elevated intracellular copper (Cu) is a phenotype of senescent cells [199]. Cell culture models utilizing glioblastoma cells found Cu increased the expression of senescence-associated mRNA (ApoJ, p21, p16, and TGF-β) and proteins (p21 and p16) [183]. These changes were also observed in human lung fibroblasts with senescent morphologies [184]. Another study reported HeLa cells treated with Cu increased the expression of p53 and p21 while promoting oxidative stress and mitochondrial dysfunction [185]. This study reported Cu increased G2/M arrest, which is associated with cellular senescence [185]. Additional evidence demonstrated oxidative stress from Cu exposure induced premature senescence mediated by p38 in fetal lung fibroblasts [186]. In human lung fibroblasts, Cu induced SA-β-Gal expression and altered proteostasis [187]. Interestingly enough, treatment of Cu with the drug resveratrol decreased the expression of p16INK4a in the brains of aged mice, as well as several other hallmarks of brain aging, suggesting a non-monotonic response to Cu in brain aging [200]. While there is strong evidence of Cu inducing senescence in cell culture, a potential anti-aging property of Cu highlights the knowledge gaps that exist for metals toxicity, cellular senescence, and aging.

6.6. Uranium

Depleted uranium (DU) is the form of processed uranium remaining after uranium enrichment, maintaining roughly 60% of the radiation found in uranium ores [201]. This form of uranium is often used in military operations, medical devices, and the manufacturing of aircraft and boats, contributing to both occupational and environmental exposures [201]. DU maintains the same genotoxic effects of its base element and is well known to cross the BBB to induce neurotoxicity [202]. Drinking water exposure in animal studies has shown DU increased expression in the inflammatory genes characteristic of SASP [188]. Another animal study showed 150 mg/L DU exposure in drinking water induced lipid peroxidation in the brain and behavioral changes in 7-month-old rats after only 2 weeks of exposure [203]. There is evidence DU induces both cytotoxic and clastogenic effects [204]. Unprocessed forms of U also induced genomic instability and drove cellular senescence via ionizing radiation [205,206]. Thus, U and DU induce senescence in animal models and should be given special consideration due to their unique exposures.

6.7. Iron

Iron has a much more complex role in cellular senescence. The literature on iron inducing cellular senescence is sparse and is likely not a major concern. One paper reported iron induced cellular senescence associated with fibrotic diseases in mice, with a significant increase in p21- and p16INK4A-positive cells six days after iron delivery [196]. Another paper induced cellular senescence in microglia by overloading cell cultures with iron, resulting in an SASP phenotype [207]. The vast majority of literature pertaining to iron and cellular senescence focuses on the distinct accumulation of iron and iron dyshomeostasis in senescent cells [208,209,210]. As such, many studies have begun targeting these iron features for senolytic drugs, particularly inducing ferroptosis [211,212,213,214]. For more detailed reviews on the roles of iron dyshomeostasis and accumulation in senescent cells, see reviews [210,215,216,217].

6.8. Heavy Metals with Less Evidence for Cellular Senescence

While the aforementioned metals demonstrate a clear induction of cellular senescence or alterations to cellular physiology consistent with the hallmarks of aging, metals such as nickel, cobalt, silver, and zinc present limited evidence of these characteristics and necessitate greater research efforts.
Nickel (Ni) is often utilized to mimic hypoxic environments and induce oxidative stress to activate HIF-1α, a hypoxia-inducible transcription factor [189]. This increase in HIF-1α induces growth arrest and p21 expression in various cell lines, a strong indication of cellular senescence [218]. Additionally, Ni depolarizes the mitochondrial membrane potential and leads to mitochondrial dysfunction in HeLa cells [219].
Cobalt (Co) also acts as a hypoxia mimic and increases the expression of HIF-1α [220]. In cardiac myocytes, Co-induced HIF-1α expression increased the expression of p21, SASP, SA-β-Gal, and ROS [190]. An additional study modeled age differences in cell culture by using cells with a low or high population doubling as a substitute for chronological age. This study reported cells at both “ages” exhibited genomic instability following Co exposure [191]. Co induced SA-β-Gal expression in “older” cells, but “younger” cells exhibited decreased p38 and caspase-10 gene expression [191].
Silver (Ag) may be the most unique metal to consider as a gerontogen due to its increased use as silver nanoparticles (AgNPs) [221]. AgNPs induced oxidative stress and increased inflammatory cytokines (IL-1β, MCP-1, and MIP-2) in vitro after 24 h exposure [192]. Other studies showed AgNPs promoted a SASP phenotype and induced SA-β-Gal expression, genomic instability, and cell cycle arrest in the G2/M phase [193,194,222].
Limited evidence indicates zinc (Zn) mediated cellular senescence by inducing the overexpression of Nox1 and p21 in vascular smooth muscle cells [195]. Salazar et al. (2017) also indicated Zn accumulated within the mitochondria of these cells and promoted ROS production [195].

7. Concluding Remarks

Concerns for the health effects of heavy metals have risen over the years as public outrage concerning the environmental pollution of metals increases in intensity. Heavy metals are known to contaminate soil and water used for drinking and agricultural irrigation [21,22,23]. Additionally, heavy metal contamination is pervasive in some food sources such as seafood, crops, and, most strikingly, in baby food products [24,25,223]. Collectively, this poses a significant public health threat as heavy metals cannot be degraded or metabolized into less toxic forms and can bioaccumulate in various tissues.
A rise in aging comorbidities will likely coincide with the projected increase in human lifespan, threatening global healthcare infrastructure. While much aging research is focused on healthy aging, research into adverse factors that accelerate or exacerbate aging lags behind. Heavy metals are of particular concern, given their widespread distribution and links to many age-related diseases. Cellular senescence is a key outcome of gerontogen exposure, which can be readily assessed with simple screening assays.
This knowledge gap for gerontogenic mechanisms of heavy metals persists despite clear evidence indicating their ability to induce a variety of aging hallmarks in cell culture, animal, and human models. The localized aging effects of gerontogens demand a more subtle assessment of aging phenotypes and the comorbidities that develop from affected tissues. The threat of brain gerontogens is further compounded by a lack of knowledge concerning cellular senescence in the NVU. While there is evidence regarding senescent phenotypes in the cell types comprising the NVU, we lack an understanding of how the interplay between these components influences one’s exposure to heavy metals and gerontogens.
There is a significant need to address our limited understanding of toxic outcomes in geriatric individuals. Geriatrics experience unique toxic outcomes resulting from polypharmacy and medical devices and likely respond differently to toxicants due to their degenerating biology [224]. This is in stark contrast to a juvenile or middle-aged individual with a developing or stable biology, respectively. Despite this difference in biological contexts, there is little to no special consideration given to the toxic outcomes experienced by geriatrics.
The increasing average age of the global population will no doubt result in a concomitant increase in age-related diseases, painting a bleak picture for our impending future. To better prepare for this inevitable shift in population structure, we need significantly more research into gerontogenic mechanisms and the unique toxic outcomes in geriatric individuals.

Author Contributions

Conceptualization, J.P.W.J.; methodology, S.T.V.; writing—original draft preparation, S.T.V. and J.P.W.J.; writing—review and editing, J.P.W.J.; supervision, J.P.W.J.; project administration, J.P.W.J.; funding acquisition, J.P.W.J. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by NIEHS, grant number ES-033327.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors recognize editorial support from Sandra S. Wise and Rachel M. Speer.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. United Nations, Department of Economic and Social Affairs, Population Division. World Population Prospects 2019, Volume II: Demographic Profiles; United Nations: New York, NY, USA, 2019; p. 2.
  2. Laks, J.; Engelhardt, E. Peculiarities of Geriatric Psychiatry: A Focus on Aging and Depression. CNS Neurosci. Ther. 2010, 16, 374–379. [Google Scholar] [CrossRef]
  3. Wise, J.P., Jr. The intersection between toxicology and aging research: A toxic aging coin perspective. Front. Aging 2022, 3, 1014675. [Google Scholar] [CrossRef]
  4. Wadhwa, P.; Buss, C.; Entringer, S.; Swanson, J. Developmental Origins of Health and Disease: Brief History of the Approach and Current Focus on Epigenetic Mechanisms. Semin. Reprod. Med. 2009, 27, 358–368. [Google Scholar] [CrossRef] [Green Version]
  5. National Institute of Environmental Health Sciences. Theme One: Advancing Environmental Health Sciences. Available online: www.niehs.nih.gov/about/strategicplan/2018-2023_theme1/index.cfm (accessed on 7 February 2023).
  6. Hahad, O.; Frenis, K.; Kuntic, M.; Daiber, A.; Münzel, T. Accelerated Aging and Age-Related Diseases (CVD and Neurological) Due to Air Pollution and Traffic Noise Exposure. Int. J. Mol. Sci. 2021, 22, 2419. [Google Scholar] [CrossRef]
  7. Briggs, D. Environmental pollution and the global burden of disease. Br. Med. Bull. 2003, 68, 1–24. [Google Scholar] [CrossRef] [Green Version]
  8. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The Hallmarks of Aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Mattson, M.P.; Arumugam, T.V. Hallmarks of Brain Aging: Adaptive and Pathological Modification by Metabolic States. Cell Metab. 2018, 27, 1176–1199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Childs, B.G.; Baker, D.J.; Kirkland, J.L.; Campisi, J.; Van Deursen, J.M. Senescence and apoptosis: Dueling or complementary cell fates? EMBO Rep. 2014, 15, 1139–1153. [Google Scholar] [CrossRef] [Green Version]
  11. Baker, D.J.; Petersen, R.C. Cellular senescence in brain aging and neurodegenerative diseases: Evidence and perspectives. J. Clin. Investig. 2018, 128, 1208–1216. [Google Scholar] [CrossRef] [Green Version]
  12. Hernandez-Segura, A.; Nehme, J.; Demaria, M. Hallmarks of Cellular Senescence. Trends Cell Biol. 2018, 28, 436–453. [Google Scholar] [CrossRef]
  13. 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]
  14. Ge, M.; Hu, L.; Ao, H.; Zi, M.; Kong, Q.; He, Y. Senolytic targets and new strategies for clearing senescent cells. Mech. Ageing Dev. 2021, 195, 111468. [Google Scholar] [CrossRef]
  15. Chaib, S.; Tchkonia, T.; Kirkland, J.L. Cellular senescence and senolytics: The path to the clinic. Nat. Med. 2022, 28, 1556–1568. [Google Scholar] [CrossRef] [PubMed]
  16. Gonzales, M.M.; Garbarino, V.R.; Zilli, E.M.; Petersen, R.C.; Kirkland, J.L.; Tchkonia, T.; Musi, N.; Seshadri, S.; Craft, S.; Orr, M.E. Senolytic Therapy to Modulate the Progression of Alzheimer’s Disease (SToMP-AD): A Pilot Clinical Trial. J. Prev. Alzheimer’s Dis. 2022, 9, 22–29. [Google Scholar] [CrossRef] [PubMed]
  17. Zhu, Y.; Prata, L.G.L.; Gerdes, E.O.W.; Netto, J.M.E.; Pirtskhalava, T.; Giorgadze, N.; Tripathi, U.; Inman, C.L.; Johnson, K.O.; Xue, A.; et al. Orally-active, clinically-translatable senolytics restore α-Klotho in mice and humans. Ebiomedicine 2022, 77, 103912. [Google Scholar] [CrossRef] [PubMed]
  18. Ali, H.; Khan, E. What are heavy metals? Long-standing controversy over the scientific use of the term ‘heavy metals’—Proposal of a comprehensive definition. Toxicol. Environ. Chem. 2018, 100, 6–19. [Google Scholar] [CrossRef]
  19. Briffa, J.; Sinagra, E.; Blundell, R. Heavy metal pollution in the environment and their toxicological effects on humans. Heliyon 2020, 6, e04691. [Google Scholar] [CrossRef] [PubMed]
  20. Hsu, M.; Selvaraj, K.; Agoramoorthy, G. Taiwan’s industrial heavy metal pollution threatens terrestrial biota. Environ. Pollut. 2006, 143, 327–334. [Google Scholar] [CrossRef]
  21. Wang, Z.; Luo, P.; Zha, X.; Xu, C.; Kang, S.; Zhou, M.; Nover, D.; Wang, Y. Overview assessment of risk evaluation and treatment technologies for heavy metal pollution of water and soil. J. Clean. Prod. 2022, 379, 134043. [Google Scholar] [CrossRef]
  22. Pieper, K.J.; Tang, M.; Edwards, M.A. Flint Water Crisis Caused By Interrupted Corrosion Control: Investigating “Ground Zero” Home. Environ. Sci. Technol. 2017, 51, 2007–2014. [Google Scholar] [CrossRef]
  23. Rehman, K.; Fatima, F.; Waheed, I.; Akash, M.S.H. Prevalence of exposure of heavy metals and their impact on health consequences. J. Cell. Biochem. 2017, 119, 157–184. [Google Scholar] [CrossRef] [PubMed]
  24. Zhang, M.; Sun, X.; Xu, J. Heavy metal pollution in the East China Sea: A review. Mar. Pollut. Bull. 2020, 159, 111473. [Google Scholar] [CrossRef] [PubMed]
  25. Khan, A.; Khan, S.; Khan, M.A.; Qamar, Z.; Waqas, M. The uptake and bioaccumulation of heavy metals by food plants, their effects on plants nutrients, and associated health risk: A review. ESPR 2015, 22, 13772–13799. [Google Scholar] [CrossRef] [PubMed]
  26. Agency for Toxic Substances and Disease Registry (ATSDR). Substance Priority List. Available online: www.atsdr.cdc.gov/spl/index.html#2022spl (accessed on 7 February 2023).
  27. Ijomone, O.M.; Ifenatuoha, C.W.; Aluko, O.M.; Ijomone, O.K.; Aschner, M. The aging brain: Impact of heavy metal neurotoxicity. Crit. Rev. Toxicol. 2020, 50, 801–814. [Google Scholar] [CrossRef] [PubMed]
  28. Vellingiri, B.; Suriyanarayanan, A.; Selvaraj, P.; Abraham, K.S.; Pasha, Y.; Winster, H.; Gopalakrishnan, A.V.; G, S.; Reddy, J.K.; Ayyadurai, N.; et al. Role of heavy metals (copper (Cu), arsenic (As), cadmium (Cd), iron (Fe) and lithium (Li)) induced neurotoxicity. Chemosphere 2022, 301, 134625. [Google Scholar] [CrossRef]
  29. Khezrian, M.; McNeil, C.J.; Murray, A.; Myint, P.K. An overview of prevalence, determinants and health outcomes of polypharmacy. Ther. Adv. Drug Saf. 2020, 11, 2042098620933741. [Google Scholar] [CrossRef]
  30. Pazan, F.; Wehling, M. Polypharmacy in older adults: A narrative review of definitions, epidemiology and consequences. Eur. Geriatr. Med. 2021, 12, 443–452. [Google Scholar] [CrossRef]
  31. Omidian, H.; Babanejad, N.; Mfoafo, K.; Bertol, C.; Omidi, Y. Toxicity of the polymeric excipients in geriatric polypharmacy. Int. J. Pharm. 2022, 622, 121901. [Google Scholar] [CrossRef]
  32. Hooper, G.; Lee, A.J.-J.; Rothwell, A.; Frampton, C. Current trends and projections in the utilisation rates of hip and knee replacement in New Zealand from 2001 to 2026. N. Z. Med. J. 2014, 127, 82–93. [Google Scholar] [PubMed]
  33. Rising, J.P.; Reynolds, I.S.; Sedrakyan, A. Delays and Difficulties in Assessing Metal-on-Metal Hip Implants. N. Engl. J. Med. 2012, 367, e1. [Google Scholar] [CrossRef] [PubMed]
  34. Campbell, J.R.; Estey, M.P. Metal release from hip prostheses: Cobalt and chromium toxicity and the role of the clinical laboratory. Clin. Chem. Lab. Med. 2012, 51, 213–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Mao, X.; Wong, A.A.; Crawford, R.W. Cobalt toxicity—An emerging clinical problem in patients with metal-on-metal hip prostheses? Med. J. Aust. 2011, 194, 649–651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Green, B.; Griffiths, E.; Almond, S. Neuropsychiatric symptoms following metal-on-metal implant failure with cobalt and chromium toxicity. BMC Psychiatry 2017, 17, 33. [Google Scholar] [CrossRef] [Green Version]
  37. Ikeda, T.; Takahashi, K.; Kabata, T.; Sakagoshi, D.; Tomita, K.; Yamada, M. Polyneuropathy caused by cobalt-chromium metallosis after total hip replacement. Muscle Nerve 2010, 42, 140–143. [Google Scholar] [CrossRef]
  38. Catalani, S.; Rizzetti, M.C.; Padovani, A.; Apostoli, P. Neurotoxicity of cobalt. Hum. Exp. Toxicol. 2021, 31, 421–437. [Google Scholar] [CrossRef]
  39. Wise, J.P.; Young, J.L.; Cai, J.; Cai, L. Current understanding of hexavalent chromium [Cr(VI)] neurotoxicity and new perspectives. Environ. Int. 2021, 158, 106877. [Google Scholar] [CrossRef]
  40. Yankner, B.A.; Lu, T.; Loerch, P. The Aging Brain. Annu. Rev. Pathol. Mech. Dis. 2008, 3, 41–66. [Google Scholar] [CrossRef]
  41. de Mooij, S.M.; Henson, R.N.; Waldorp, L.J.; Kievit, R.A. Age Differentiation within Gray Matter, White Matter, and between Memory and White Matter in an Adult Life Span Cohort. J. Neurosci. 2018, 38, 5826–5836. [Google Scholar] [CrossRef] [Green Version]
  42. Ge, Y.; Grossman, R.I.; Babb, J.S.; Rabin, M.L.; Mannon, L.J.; Kolson, D.L. Age-Related Total Gray Matter and White Matter Changes in Normal Adult Brain. Part I: Volumetric MR Imaging Analysis. AJNR Am. J. Neuroradiol. 2002, 23, 1327–1333. [Google Scholar]
  43. Mora, F.; Segovia, G.; del Arco, A. Aging, plasticity and environmental enrichment: Structural changes and neurotransmitter dynamics in several areas of the brain. Brain Res. Rev. 2007, 55, 78–88. [Google Scholar] [CrossRef]
  44. Godbout, J.P.; Johnson, R.W. Age and Neuroinflammation: A Lifetime of Psychoneuroimmune Consequences. Immunol. Allergy Clin. North Am. 2009, 29, 321–337. [Google Scholar] [CrossRef] [PubMed]
  45. Giorgio, A.; Santelli, L.; Tomassini, V.; Bosnell, R.; Smith, S.; De Stefano, N.; Johansen-Berg, H. Age-related changes in grey and white matter structure throughout adulthood. Neuroimage 2010, 51, 943–951. [Google Scholar] [CrossRef] [Green Version]
  46. Lockhart, S.N.; DeCarli, C. Structural Imaging Measures of Brain Aging. Neuropsychol. Rev. 2014, 24, 271–289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Salminen, A.; Ojala, J.; Kaarniranta, K.; Haapasalo, A.; Hiltunen, M.; Soininen, H. Astrocytes in the aging brain express characteristics of senescence-associated secretory phenotype. Eur. J. Neurosci. 2011, 34, 3–11. [Google Scholar] [CrossRef]
  48. Sikora, E.; Bielak-Zmijewska, A.; Dudkowska, M.; Krzystyniak, A.; Mosieniak, G.; Wesierska, M.; Wlodarczyk, J. Cellular Senescence in Brain Aging. Front. Aging Neurosci. 2021, 13, 646924. [Google Scholar] [CrossRef] [PubMed]
  49. Norden, D.M.; Godbout, J.P. Review: Microglia of the aged brain: Primed to be activated and resistant to regulation. Neuropathol. Appl. Neurobiol. 2013, 39, 19–34. [Google Scholar] [CrossRef]
  50. Lee, H.J.; Park, M.K.; Seo, Y.R. Pathogenic Mechanisms of Heavy Metal Induced-Alzheimer’s Disease. Toxicol. Environ. Health Sci. 2018, 10, 1–10. [Google Scholar] [CrossRef]
  51. Woimant, F.; Trocello, J.-M. Disorders of heavy metals. Handb. Clin. Neurol. 2014, 120, 851–864. [Google Scholar] [CrossRef]
  52. Cavaleri, F. Review of Amyotrophic Lateral Sclerosis, Parkinson’s and Alzheimer’s diseases helps further define pathology of the novel paradigm for Alzheimer’s with heavy metals as primary disease cause. Med. Hypotheses 2015, 85, 779–790. [Google Scholar] [CrossRef]
  53. Boccardi, V.; Pelini, L.; Ercolani, S.; Ruggiero, C.; Mecocci, P. From cellular senescence to Alzheimer’s disease: The role of telomere shortening. Ageing Res. Rev. 2015, 22, 1–8. [Google Scholar] [CrossRef]
  54. Van Dijk, K.D.; Persichetti, E.; Chiasserini, D.; Eusebi, P.; Beccari, T.; Calabresi, P.; Berendse, H.W.; Parnetti, L.; Van De Berg, W.D.J. Changes in endolysosomal enzyme activities in cerebrospinal fluid of patients with Parkinson’s disease. Mov. Disord. 2013, 28, 747–754. [Google Scholar] [CrossRef]
  55. Gustavsson, A.; Norton, N.; Fast, T.; Frölich, L.; Georges, J.; Holzapfel, D.; Kirabali, T.; Krolak-Salmon, P.; Rossini, P.M.; Ferretti, M.T.; et al. Global estimates on the number of persons across the Alzheimer’s disease continuum. Alzheimer’s Dement. 2022, 19, 658–670. [Google Scholar] [CrossRef]
  56. Zhang, L.; Chen, C.; Mak, M.S.; Lu, J.; Wu, Z.; Chen, Q.; Han, Y.; Li, Y.; Pi, R. Advance of sporadic Alzheimer’s disease animal models. Med. Res. Rev. 2019, 40, 431–458. [Google Scholar] [CrossRef]
  57. Webster, S.J.; Bachstetter, A.; Nelson, P.T.; Schmitt, F.A.; Van Eldik, L.J. Using mice to model Alzheimer’s dementia: An overview of the clinical disease and the preclinical behavioral changes in 10 mouse models. Front. Genet. 2014, 5, 88. [Google Scholar] [CrossRef] [Green Version]
  58. De Ture, M.A.; Dickson, D.W. The neuropathological diagnosis of Alzheimer’s disease. Mol. Neurodegener. 2019, 14, 32. [Google Scholar] [CrossRef] [Green Version]
  59. Kabir, T.; Uddin, S.; Zaman, S.; Begum, Y.; Ashraf, G.; Bin-Jumah, M.N.; Bungau, S.G.; Mousa, S.A.; Abdel-Daim, M.M. Molecular Mechanisms of Metal Toxicity in the Pathogenesis of Alzheimer’s Disease. Mol. Neurobiol. 2020, 58, 1–20. [Google Scholar] [CrossRef]
  60. Bakulski, K.M.; Seo, Y.A.; Hickman, R.C.; Brandt, D.; Vadari, H.S.; Hu, H.; Park, S.K. Heavy Metals Exposure and Alzheimer’s Disease and Related Dementias. J. Alzheimer’s Dis. 2020, 76, 1215–1242. [Google Scholar] [CrossRef]
  61. Bhat, R.; Crowe, E.P.; Bitto, A.; Moh, M.; Katsetos, C.D.; Garcia, F.U.; Johnson, F.B.; Trojanowski, J.Q.; Sell, C.; Torres, C. Astrocyte Senescence as a Component of Alzheimer’s Disease. PLoS ONE 2012, 7, e45069. [Google Scholar] [CrossRef] [PubMed]
  62. He, N.; Jin, W.-L.; Lok, K.-H.; Wang, Y.; Yin, M.; Wang, Z.-J. Amyloid-β1–42 oligomer accelerates senescence in adult hippocampal neural stem/progenitor cells via formylpeptide receptor 2. Cell Death Dis. 2013, 4, e924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Ou, Z.; Pan, J.; Tang, S.; Duan, D.; Yu, D.; Nong, H.; Wang, Z. Global Trends in the Incidence, Prevalence, and Years Lived with Disability of Parkinson’s Disease in 204 Countries/Territories From 1990 to 2019. Front. Public Health 2021, 9, 776847. [Google Scholar] [CrossRef] [PubMed]
  64. Emamzadeh, F.N.; Surguchov, A. Parkinson’s Disease: Biomarkers, Treatment, and Risk Factors. Front. Neurosci. 2018, 12, 612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Raj, K.; Kaur, P.; Gupta, G.; Singh, S. Metals associated neurodegeneration in Parkinson’s disease: Insight to physiological, pathological mechanisms and management. Neurosci. Lett. 2021, 753, 135873. [Google Scholar] [CrossRef]
  66. Pyatha, S.; Kim, H.; Lee, D.; Kim, K. Association between Heavy Metal Exposure and Parkinson’s Disease: A Review of the Mechanisms Related to Oxidative Stress. Antioxidants 2022, 11, 2467. [Google Scholar] [CrossRef]
  67. Przedborski, S. The two-century journey of Parkinson disease research. Nat. Rev. Neurosci. 2017, 18, 251–259. [Google Scholar] [CrossRef] [PubMed]
  68. Bloem, B.R.; Okun, M.S.; Klein, C. Parkinson’s disease. Lancet 2021, 397, 2284–2303. [Google Scholar] [CrossRef]
  69. Aarsland, D.; Andersen, K.; Larsen, J.P.; Lolk, A.; Kragh-Sørensen, P. Prevalence and characteristics of dementia in Parkinson disease: An 8-year prospective study. Arch. Neurol. 2003, 60, 387–392. [Google Scholar] [CrossRef] [Green Version]
  70. Berry, C.M.; La Vecchia, C.; Nicotera, P. Paraquat and Parkinson’s disease. Cell Death Differ. 2010, 17, 1115–1125. [Google Scholar] [CrossRef] [Green Version]
  71. Chinta, S.J.; Woods, G.; Demaria, M.; Rane, A.; Zou, Y.; McQuade, A.; Rajagopalan, S.; Limbad, C.; Madden, D.T.; Campisi, J.; et al. Cellular Senescence Is Induced by the Environmental Neurotoxin Paraquat and Contributes to Neuropathology Linked to Parkinson’s Disease. Cell Rep. 2018, 22, 930–940. [Google Scholar] [CrossRef] [Green Version]
  72. Poewe, W.; Seppi, K.; Tanner, C.M.; Halliday, G.M.; Brundin, P.; Volkmann, J.; Schrag, A.E.; Lang, A.E. Parkinson disease. Nat. Rev. Dis. Primers 2017, 3, 17013. [Google Scholar] [CrossRef]
  73. Xu, L.; Liu, T.; Liu, L.; Yao, X.; Chen, L.; Fan, D.; Zhan, S.; Wang, S. Global variation in prevalence and incidence of amyotrophic lateral sclerosis: A systematic review and meta-analysis. J. Neurol. 2019, 267, 944–953. [Google Scholar] [CrossRef] [PubMed]
  74. Taylor, J.P.; Brown, R.H., Jr.; Cleveland, D.W. Decoding ALS: From genes to mechanism. Nature 2016, 539, 197–206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Orsini, M.; Oliveira, A.B.; Nascimento, O.J.; Reis, C.H.M.; Leite, M.A.A.; De Souza, J.A.; Pupe, C.; De Souza, O.G.; Bastos, V.H.; De Freitas, M.R.; et al. Amyotrophic Lateral Sclerosis: New Perpectives and Update. Neurol. Int. 2015, 7, 39–47. [Google Scholar] [CrossRef] [Green Version]
  76. Filippini, T.; Tesauro, M.; Fiore, M.; Malagoli, C.; Consonni, M.; Violi, F.; Iacuzio, L.; Arcolin, E.; Conti, G.O.; Cristaldi, A.; et al. Environmental and Occupational Risk Factors of Amyotrophic Lateral Sclerosis: A Population-Based Case-Control Study. Int. J. Environ. Res. Public Health 2020, 17, 2882. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Newell, M.E.; Adhikari, S.; Halden, R.U. Systematic and state-of the science review of the role of environmental factors in Amyotrophic Lateral Sclerosis (ALS) or Lou Gehrig’s Disease. Sci. Total. Environ. 2021, 817, 152504. [Google Scholar] [CrossRef]
  78. Trias, E.; Beilby, P.R.; Kovacs, M.; Ibarburu, S.; Varela, V.; Barreto-Núñez, R.; Bradford, S.C.; Beckman, J.S.; Barbeito, L. Emergence of Microglia Bearing Senescence Markers During Paralysis Progression in a Rat Model of Inherited ALS. Front. Aging Neurosci. 2019, 11, 42. [Google Scholar] [CrossRef] [Green Version]
  79. Vazquez-Villasenor, I.; Garwood, C.J.; Heath, P.R.; Simpson, J.E.; Ince, P.G.; Wharton, S.B. Expression of p16 and p21 in the frontal association cortex of ALS/MND brains suggests neuronal cell cycle dysregulation and astrocyte senescence in early stages of the disease. Neuropathol. Appl. Neurobiol. 2020, 46, 171–185. [Google Scholar] [CrossRef]
  80. Xie, Z.; Wu, H.; Zhao, J. Multifunctional roles of zinc in Alzheimer’s disease. Neurotoxicology 2020, 80, 112–123. [Google Scholar] [CrossRef] [PubMed]
  81. Silvestri, L.; Camaschella, C. A potential pathogenetic role of iron in Alzheimer’s disease. J. Cell. Mol. Med. 2008, 12, 1548–1550. [Google Scholar] [CrossRef]
  82. Mochizuki, H.; Choong, C.-J.; Baba, K. Parkinson’s disease and iron. J. Neural Transm. 2020, 127, 181–187. [Google Scholar] [CrossRef]
  83. Wojtunik-Kulesza, K.; Oniszczuk, A.; Waksmundzka-Hajnos, M. An attempt to elucidate the role of iron and zinc ions in development of Alzheimer’s and Parkinson’s diseases. Biomed. Pharmacother. 2019, 111, 1277–1289. [Google Scholar] [CrossRef]
  84. Peng, Y.; Chang, X.; Lang, M. Iron Homeostasis Disorder and Alzheimer’s Disease. Int. J. Mol. Sci. 2021, 22, 12442. [Google Scholar] [CrossRef]
  85. Felix, K.; Manna, S.K.; Wise, K.; Barr, J.; Ramesh, G.T. Low levels of arsenite activates nuclear factor-κB and activator protein-1 in immortalized mesencephalic cells. J. Biochem. Mol. Toxicol. 2005, 19, 67–138. [Google Scholar] [CrossRef] [Green Version]
  86. Piao, F.; Ma, N.; Hiraku, Y.; Murata, M.; Oikawa, S.; Cheng, F.; Zhong, L.; Yamauchi, T.; Kawanishi, S.; Yokoyama, K. Oxidative DNA Damage in Relation to Neurotoxicity in the Brain of Mice Exposed to Arsenic at Environmentally Relevant Levels. J. Occup. Health 2005, 4, 371–470. [Google Scholar] [CrossRef] [Green Version]
  87. Chin-Chan, M.; Navarro-Yepes, J.; Quintanilla-Vega, B. Environmental pollutants as risk factors for neurodegenerative disorders: Alzheimer and Parkinson diseases. Front. Cell. Neurosci. 2015, 9, 124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. O’Bryant, S.E.; Edwards, M.; Menon, C.V.; Gong, G.; Barber, R. Long-Term Low-Level Arsenic Exposure Is Associated with Poorer Neuropsychological Functioning: A Project FRONTIER Study. Int. J. Environ. Res. Public Health 2011, 8, 861–874. [Google Scholar] [CrossRef] [PubMed]
  89. Dani, S.U. Arsenic for the fool: An exponential connection. Sci. Total. Environ. 2010, 408, 1842–1846. [Google Scholar] [CrossRef] [PubMed]
  90. Lee, C.-P.; Zhu, C.-H.; Su, C.-C. Increased prevalence of Parkinson’s disease in soils with high arsenic levels. Park. Relat. Disord. 2021, 88, 19–23. [Google Scholar] [CrossRef]
  91. De Benedetti, S.; Lucchini, G.; Del Bo’, C.; Deon, V.; Marocchi, A.; Penco, S.; Lunetta, C.; Gianazza, E.; Bonomi, F.; Iametti, S. Blood trace metals in a sporadic amyotrophic lateral sclerosis geographical cluster. Biometals 2017, 30, 355–365. [Google Scholar] [CrossRef]
  92. Lidsky, T.I.; Schneider, J.S. Lead neurotoxicity in children: Basic mechanisms and clinical correlates. Brain 2003, 126, 5–19. [Google Scholar] [CrossRef] [Green Version]
  93. Coon, S.; Stark, A.; Peterson, E.; Gloi, A.; Kortsha, G.; Pounds, J.; Chettle, D.; Gorell, J. Whole-Body Lifetime Occupational Lead Exposure and Risk of Parkinson’s Disease. Environ. Health Perspect. 2006, 114, 1872–1876. [Google Scholar] [CrossRef] [Green Version]
  94. Roos, P.M.; Vesterberg, O.; Syversen, T.; Flaten, T.P.; Nordberg, M. Metal Concentrations in Cerebrospinal Fluid and Blood Plasma from Patients with Amyotrophic Lateral Sclerosis. Biol. Trace Elem. Res. 2013, 151, 159–170. [Google Scholar] [CrossRef] [Green Version]
  95. Wu, J.; Basha, R.; Brock, B.; Cox, D.P.; Cardozo-Pelaez, F.; McPherson, C.A.; Harry, J.; Rice, D.C.; Maloney, B.; Chen, D.; et al. Alzheimer’s Disease (AD)-Like Pathology in Aged Monkeys after Infantile Exposure to Environmental Metal Lead (Pb): Evidence for a Developmental Origin and Environmental Link for AD. J. Neurosci. 2008, 28, 3–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Basha, M.R.; Wei, W.; Bakheet, S.A.; Benitez, N.; Siddiqi, H.K.; Ge, Y.-W.; Lahiri, D.K.; Zawia, N.H. The Fetal Basis of Amyloidogenesis: Exposure to Lead and Latent Overexpression of Amyloid Precursor Protein and β-Amyloid in the Aging Brain. J. Neurosci. 2005, 25, 823–829. [Google Scholar] [CrossRef] [Green Version]
  97. Li, N.; Liu, F.; Song, L.; Zhang, P.; Qiao, M.; Zhao, Q.; Li, W. The effects of early life Pb exposure on the expression of IL1-β, TNF-α and Aβ in cerebral cortex of mouse pups. J. Trace Elem. Med. Biol. 2014, 28, 100–104. [Google Scholar] [CrossRef]
  98. Kala, S.V.; Jadhav, A.L. Low Level Lead Exposure Decreases In Vivo Release of Dopamine in the Rat Nucleus Accumbens: A Microdialysis Study. J. Neurochem. 2002, 65, 1631–1635. [Google Scholar] [CrossRef]
  99. Sheykhansari, S.; Kozielski, K.; Bill, J.; Sitti, M.; Gemmati, D.; Zamboni, P.; Singh, A.V. Redox metals homeostasis in multiple sclerosis and amyotrophic lateral sclerosis: A review. Cell Death Dis. 2018, 9, 348. [Google Scholar] [CrossRef]
  100. Okuda, B.; Iwamoto, Y.; Tachibana, H.; Sugita, M. Parkinsonism after acute cadmium poisoning. Clin. Neurol. Neurosurg. 1997, 99, 263–265. [Google Scholar] [CrossRef] [PubMed]
  101. Gellein, K.; Garruto, R.M.; Syversen, T.; Sjøbakk, T.E.; Flaten, T.P. Concentrations of Cd, Co, Cu, Fe, Mn, Rb, V, and Zn in Formalin-Fixed Brain Tissue in Amyotrophic Lateral Sclerosis and Parkinsonism-Dementia Complex of Guam Determined by High-Resolution ICP-MS. Biol. Trace Elem. Res. 2003, 96, 39–60. [Google Scholar] [CrossRef]
  102. Min, J.-Y.; Min, K.-B. Blood cadmium levels and Alzheimer’s disease mortality risk in older US adults. Environ. Health 2016, 15, 69. [Google Scholar] [CrossRef] [Green Version]
  103. Strumylaite, L.; Kregzdyte, R.; Kucikiene, O.; Baranauskiene, D.; Simakauskiene, V.; Naginiene, R.; Damuleviciene, G.; Lesauskaite, V.; Zemaitiene, R. Alzheimer’s Disease Association with Metals and Metalloids Concentration in Blood and Urine. Int. J. Environ. Res. Public Health 2022, 19, 7309. [Google Scholar] [CrossRef] [PubMed]
  104. Hsu, H.-W.; Bondy, S.C.; Kitazawa, M. Environmental and Dietary Exposure to Copper and Its Cellular Mechanisms Linking to Alzheimer’s Disease. Toxicol. Sci. 2018, 163, 338–345. [Google Scholar] [CrossRef]
  105. Lovell, M.; Robertson, J.; Teesdale, W.; Campbell, J.; Markesbery, W. Copper, iron and zinc in Alzheimer’s disease senile plaques. J. Neurol. Sci. 1998, 158, 47–52. [Google Scholar] [CrossRef]
  106. Litwin, T.; Gromadzka, G.; Szpak, G.; Jabłonka-Salach, K.; Bulska, E.; Członkowska, A. Brain metal accumulation in Wilson’s disease. J. Neurol. Sci. 2013, 329, 55–58. [Google Scholar] [CrossRef]
  107. Xiao, G.; Fan, Q.; Wang, X.; Zhou, B. Huntington disease arises from a combinatory toxicity of polyglutamine and copper binding. Proc. Natl. Acad. Sci. USA 2013, 110, 14995–15000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Tarohda, T.; Ishida, Y.; Kawai, K.; Yamamoto, M.; Amano, R. Regional distributions of manganese, iron, copper, and zinc in the brains of 6-hydroxydopamine-induced parkinsonian rats. Anal. Bioanal. Chem. 2005, 383, 224–234. [Google Scholar] [CrossRef] [PubMed]
  109. Dinocourt, C.; Legrand, M.; Dublineau, I.; Lestaevel, P. The neurotoxicology of uranium. Toxicology 2015, 337, 58–71. [Google Scholar] [CrossRef] [PubMed]
  110. Lu, M.; Li, H.; Li, Y.; Lu, Y.; Wang, H.; Wang, X. Exploring the Toxicology of Depleted Uranium with Caenorhabditis elegans. ACS Omega 2020, 5, 12119–12125. [Google Scholar] [CrossRef] [PubMed]
  111. Song, X.; Kenston, S.S.F.; Kong, L.; Zhao, J. Molecular mechanisms of nickel induced neurotoxicity and chemoprevention. Toxicology 2017, 392, 47–54. [Google Scholar] [CrossRef]
  112. El Brouzi, M.Y.; Lamtai, M.; Zghari, O.; Ouakki, S.; Azizoun, I.; El Hessni, A.; Mesfioui, A.; Ouichou, A. Intrahippocampal Effects of Nickel Injection on the Affective and Cognitive Response in Wistar Rat: Potential Role of Oxidative Stress. Biol. Trace Elem. Res. 2020, 199, 3382–3392. [Google Scholar] [CrossRef]
  113. Lamtai, M.; Chaibat, J.; Ouakki, S.; Zghari, O.; Mesfioui, A.; El Hessni, A.; Rifi, E.-H.; Marmouzi, I.; Essamri, A.; Ouichou, A. Effect of Chronic Administration of Nickel on Affective and Cognitive Behavior in Male and Female Rats: Possible Implication of Oxidative Stress Pathway. Brain Sci. 2018, 8, 141. [Google Scholar] [CrossRef] [Green Version]
  114. Adedara, I.A.; Adegbosin, A.N.; Abiola, M.A.; Odunewu, A.A.; Owoeye, O.; Owumi, S.E.; Farombi, E.O. Neurobehavioural and biochemical responses associated with exposure to binary waterborne mixtures of zinc and nickel in rats. Environ. Toxicol. Pharmacol. 2020, 73, 103294. [Google Scholar] [CrossRef] [PubMed]
  115. Zheng, F.; Li, Y.; Zhang, F.; Sun, Y.; Zheng, C.; Luo, Z.; Wang, Y.-L.; Aschner, M.; Zheng, H.; Lin, L.; et al. Cobalt induces neurodegenerative damages through Pin1 inactivation in mice and human neuroglioma cells. J. Hazard. Mater. 2021, 419, 126378. [Google Scholar] [CrossRef] [PubMed]
  116. Narayanan, S.E.; Rehuman, N.A.; Harilal, S.; Vincent, A.; Rajamma, R.G.; Behl, T.; Uddin, M.S.; Ashraf, G.M.; Mathew, B. Molecular mechanism of zinc neurotoxicity in Alzheimer’s disease. Environ. Sci. Pollut. Res. 2020, 27, 43542–43552. [Google Scholar] [CrossRef] [PubMed]
  117. Simpson, J.E.; Wharton, S.B.; Cooper, J.; Gelsthorpe, C.; Baxter, L.; Forster, G.; Shaw, P.; Savva, G.; Matthews, F.; Brayne, C.; et al. Alterations of the blood–brain barrier in cerebral white matter lesions in the ageing brain. Neurosci. Lett. 2010, 486, 246–251. [Google Scholar] [CrossRef]
  118. Zlokovic, B.V. The Blood-Brain Barrier in Health and Chronic Neurodegenerative Disorders. Neuron 2008, 57, 178–201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Giuliani, A.; Sivilia, S.; Baldassarro, V.A.; Gusciglio, M.; Lorenzini, L.; Sannia, M.; Calzà, L.; Giardino, L. Age-Related Changes of the Neurovascular Unit in the Cerebral Cortex of Alzheimer Disease Mouse Models: A Neuroanatomical and Molecular Study. J. Neuropathol. Exp. Neurol. 2019, 78, 101–112. [Google Scholar] [CrossRef] [Green Version]
  120. Banks, W.A.; Reed, M.J.; Logsdon, A.F.; Rhea, E.M.; Erickson, M.A. Healthy aging and the blood–brain barrier. Nat. Aging 2021, 1, 243–254. [Google Scholar] [CrossRef] [PubMed]
  121. Alahmari, A. Blood-Brain Barrier Overview: Structural and Functional Correlation. Neural Plast. 2021, 2021, 6564585. [Google Scholar] [CrossRef] [PubMed]
  122. Zeevi, N.; Pachter, J.; McCullough, L.D.; Wolfson, L.; Kuchel, G. The Blood-Brain Barrier: Geriatric Relevance of a Critical Brain-Body Interface. J. Am. Geriatr. Soc. 2010, 58, 1749–1757. [Google Scholar] [CrossRef] [PubMed]
  123. Montagne, A.; Barnes, S.R.; Sweeney, M.D.; Halliday, M.R.; Sagare, A.P.; Zhao, Z.; Toga, A.W.; Jacobs, R.E.; Liu, C.Y.; Amezcua, L.; et al. Blood-Brain Barrier Breakdown in the Aging Human Hippocampus. Neuron 2015, 85, 296–302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Pandya, V.A.; Patani, R. Region-specific vulnerability in neurodegeneration: Lessons from normal ageing. Ageing Res. Rev. 2021, 67, 101311. [Google Scholar] [CrossRef]
  125. Ohene, Y.; Harrison, I.F.; Evans, P.G.; Thomas, D.L.; Lythgoe, M.F.; Wells, J.A. Increased blood–brain barrier permeability to water in the aging brain detected using noninvasive multi-TE ASL MRI. Magn. Reson. Med. 2020, 85, 326–333. [Google Scholar] [CrossRef] [PubMed]
  126. Ganong, W.F. Circumventricular Organs: Definition and Role In The Regulation Of Endocrine And Autonomic Function. Clin. Exp. Pharmacol. Physiol. 2000, 27, 422–427. [Google Scholar] [CrossRef]
  127. Zheng, W.; Aschner, M.; Ghersi-Egea, J.-F. Brain barrier systems: A new frontier in metal neurotoxicological research. Toxicol. Appl. Pharmacol. 2003, 192, 1–11. [Google Scholar] [CrossRef] [Green Version]
  128. Olung, N.F.; Aluko, O.M.; Jeje, S.O.; Adeagbo, A.S.; Ijomone, O.M. Vascular Dysfunction in the Brain; Implications for Heavy Metal Exposures. Curr. Hypertens. Rev. 2021, 17, 5–13. [Google Scholar] [CrossRef] [PubMed]
  129. Kim, J.-H.; Byun, H.-M.; Chung, E.-C.; Chung, H.-Y.; Bae, O.-N. Loss of Integrity: Impairment of the Blood-brain Barrier in Heavy Metal-associated Ischemic Stroke. Toxicol. Res. 2013, 29, 157–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Toews, A.D.; Kolber, A.; Hayward, J.; Krigman, M.R.; Morell, P. Experimental lead encephalopathy in the suckling rat: Con-centration of lead in cellular fractions enriched in brain capillaries. Brain Res. 1978, 147, 131–138. [Google Scholar] [CrossRef]
  131. Sanders, T.; Liu, Y.; Buchner, V.; Tchounwoy, P.B. Neurotoxic effects of lead toxicity: A Review. Rev. Environ. Health 2009, 24, 15–45. [Google Scholar] [CrossRef] [Green Version]
  132. Strużyńska, L.; Walski, M.; Gadamski, R.; Dabrowska-Bouta, B.; Rafałowska, U. Lead-induced abnormalities in blood-brain barrier permeability in experimental chronic toxicity. Mol. Chem. Neuropathol. 1997, 31, 207–224. [Google Scholar] [CrossRef]
  133. Balbuena, P.; Li, W.; Ehrich, M. Assessments of tight junction proteins occludin, claudin 5 and scaffold proteins ZO1 and ZO2 in endothelial cells of the rat blood–brain barrier: Cellular responses to neurotoxicants malathion and lead acetate. Neurotoxicology 2011, 32, 58–67. [Google Scholar] [CrossRef]
  134. Hossain, M.A.; Russell, J.C.; Miknyoczki, S.; Ruggeri, B.; Lal, B.; Laterra, J. Vascular endothelial growth factor mediates vasogenic edema in acute lead encephalopathy. Ann. Neurol. 2004, 55, 660–667. [Google Scholar] [CrossRef]
  135. Wang, Q.; Luo, W.; Zheng, W.; Liu, Y.; Xu, H.; Zheng, G.; Dai, Z.; Zhang, W.; Chen, Y.; Chen, J. Iron supplement prevents lead-induced disruption of the blood–brain barrier during rat development. Toxicol. Appl. Pharmacol. 2007, 219, 33–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Shukla, A.; Shukla, G.S.; Srimal, R.C. Cadmium-induced alterations in blood- brain barrier permeability and its possible correlation with decreased microvessel antioxidant potential in rat. Hum. Exp. Toxicol. 1996, 15, 400–405. [Google Scholar] [CrossRef]
  137. Peterson, E.W.; Cardoso, E.R. The blood-brain battier following experimental subarachnoid hemorrhage. Part 2: Response to mercuric chloride infusion. J. Neurosurg. 1983, 58, 345–351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Chen, S.-C.; Tsai, M.-H.; Wang, H.-J.; Yu, H.-S.; Chang, L.W. Vascular permeability alterations induced by arsenic. Hum. Exp. Toxicol. 2004, 23, 1–7. [Google Scholar] [CrossRef] [PubMed]
  139. Sharma, H.S.; Hussain, S.; Schlager, J.; Ali, S.F.; Sharma, A. Influence of nanoparticles on blood-brain barrier permeability and brain edema formation in rats. In Brain Edema XIV; Springer: Berlin/Heidelberg, Germany, 2010; pp. 359–364. [Google Scholar] [CrossRef]
  140. Foidl, B.M.; Humpel, C. Chronic treatment with five vascular risk factors causes cerebral amyloid angiopathy but no Alzheimer pathology in C57BL6 mice. Brain, Behav. Immun. 2019, 78, 52–64. [Google Scholar] [CrossRef]
  141. Chang, L.W.; Hartmann, H.A. Blood-brain barrier dysfunction in experimental mercury intoxication. Acta Neuropathol. 1972, 21, 179–184. [Google Scholar] [CrossRef]
  142. Chou, Y.; Tsai, C.-H.; Ueng, K.-C.; Tian, T.-Y.; Chen, S.-C.; Yeh, H.-I. Endothelial gap junctions are down-regulated by arsenic and stimulate angiogenesis in vitro. Chem. Res. Toxicol. 2007, 16, 460–468. [Google Scholar] [CrossRef]
  143. Song, H.; Zheng, G.; Shen, X.-F.; Liu, X.-Q.; Luo, W.-J.; Chen, J.-Y. Role of activation of nonreceptor tyrosine kinase Src vis chaperone GRP78. Toxicol. Sci. 2014, 138, 393–402. [Google Scholar] [CrossRef] [Green Version]
  144. Liu, X.; Su, P.; Meng, S.; Aschner, M.; Cao, Y.; Luo, W.; Zheng, G.; Liu, M. Role of matrix metalloproteinase-2/9 (MMP2/9) in lead-induced changes in an in vitro blood-brain barrier model. Int. J. Biol. Sci. 2017, 13, 1351–1360. [Google Scholar] [CrossRef] [Green Version]
  145. Manthari, R.K.; Tikka, C.; Ommati, M.M.; Niu, R.; Sun, Z.; Wang, J.; Zhang, J.; Wang, J. Arsenic induces autophagy in developmental mouse cerebral cortex and hippocampus by inhibiting PI3K/Akt/mTOR signaling pathway: Involvement of blood-brain barrier’s tight junction proteins. Arch. Toxicol. 2018, 92, 3255–3275. [Google Scholar] [CrossRef]
  146. Wang, J.; Chen, J.; Tang, Z.; Li, Y.; Hu, L.; Pan, J. The Effects of Copper on Brain Microvascular Endothelial Cells and Claudin Via Apoptosis and Oxidative Stress. Biol. Trace Elem. Res. 2016, 174, 132–141. [Google Scholar] [CrossRef]
  147. Branca, J.J.V.; Maresca, M.; Morucci, G.; Mello, T.; Becatti, M.; Pazzagli, L.; Colzi, I.; Gonnelli, C.; Carrino, D.; Paternostro, F.; et al. Effects of Cadmium on ZO-1 Tight Junction Integrity of the Blood Brain Barrier. Int. J. Mol. Sci. 2019, 20, 6010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Wei, H.; Zhang, W.-J.; LeBoeuf, R.; Frei, B. Copper induces-and copper chelation by tetrathiomolybdate inhibits-endothelial activation in vitro. Redox Rep. 2013, 19, 40–48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Takahashi, T.; Fujimura, M.; Koyama, M.; Kanazawa, M.; Usuki, F.; Nishizawa, M.; Shimohata, T. Methylmercury Causes Blood-Brain Barrier Damage in Rats via Upregulation of Vascular Endothelial Growth Factor Expression. PLoS ONE 2017, 12, e0170623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Jeong, E.M.; Moon, C.H.; Kim, C.S.; Lee, S.H.; Baik, E.J.; Moon, C.K.; Jung, Y.S. Cadmium stimulates the expression of ICAM-1 via NF-κB activation in cerebrovascular endothelial cells. Biochem. Biophys. Res. Commun. 2004, 320, 887–892. [Google Scholar] [CrossRef]
  151. Williams, A.J.; Wei, H.H.; Dave, J.R.; Tortella, F.C. Acute and delayed neuroinflammatory response following experimental penetrating ballistic brain injury in the rat. J. Neuroinflamm. 2007, 4, 17. [Google Scholar] [CrossRef] [Green Version]
  152. Yi, J.-H.; Park, S.-W.; Brooks, N.; Lang, B.T.; Vemuganti, R. PPARγ agonist rosiglitazone is neuroprotective after traumatic brain injury via anti-inflammatory and anti-oxidative mechanisms. Brain Res. 2008, 1244, 164–172. [Google Scholar] [CrossRef] [Green Version]
  153. Kenne, E.; Erlandsson, A.; Lindbom, L.; Hillered, L.; Clausen, F. Neutrophil depletion reduces edema formation and tissue loss following traumatic brain injury in mice. J. Neuroinflamm. 2012, 9, 17. [Google Scholar] [CrossRef] [Green Version]
  154. Sahu, M.R.; Rani, L.; Subba, R.; Mondal, A.C. Cellular senescence in the aging brain: A promising target for neurodegenerative diseases. Mech. Ageing Dev. 2022, 204, 111675. [Google Scholar] [CrossRef]
  155. Rojas-Vázquez, S.; Blasco-Chamarro, L.; López-Fabuel, I.; Martínez-Máñez, R.; Fariñas, I. Vascular Senescence: A Potential Bridge Between Physiological Aging and Neurogenic Decline. Front. Neurosci. 2021, 15, 666881. [Google Scholar] [CrossRef] [PubMed]
  156. Graves, S.I.; Baker, D.J. Implicating endothelial cell senescence to dysfunction in the ageing and diseased brain. BCPT 2020, 127, 102–110. [Google Scholar] [CrossRef] [Green Version]
  157. Finger, C.E.; Moreno-Gonzalez, I.; Gutierrez, A.; Moruno-Manchon, J.F.; McCullough, L.D. Age-related immune alterations and cerebrovascular inflammation. Mol. Psychiatry 2022, 27, 803–818. [Google Scholar] [CrossRef] [PubMed]
  158. Marques, F.; Sousa, J.C.; Sousa, N.; Palha, J.A. Blood–brain-barriers in aging and in Alzheimer’s disease. Mol. Neurodegener. 2013, 8, 38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Bryant, A.G.; Hu, M.; Carlyle, B.C.; Arnold, S.E.; Frosch, M.P.; Das, S.; Hyman, B.T.; Bennett, R.E. Cerebrovascular Senescence Is Associated with Tau Pathology in Alzheimer’s Disease. Front. Neurol. 2020, 11, 575953. [Google Scholar] [CrossRef] [PubMed]
  160. Garwood, C.J.; Simpson, J.E.; Al Mashhadi, S.; Axe, C.; Wilson, S.; Heath, P.R.; Shaw, P.J.; Matthews, F.E.; Brayne, C.; Ince, P.G.; et al. DNA damage response and senescence in endothelial cells of human cerebral cortex and relation to Alzheimer’s neuropathology progression: A population-based study in the Medical Research Council Cognitive Function and Ageing Study (MRC-CFAS) cohort. Neuropathol. Appl. Neurobiol. 2014, 40, 802–814. [Google Scholar] [CrossRef]
  161. Yamazaki, Y.; Baker, D.J.; Tachibana, M.; Liu, C.-C.; Van Deursen, J.M.; Brott, T.G.; Bu, G.; Kanekiyo, T. Vascular Cell Senescence Contributes to Blood–Brain Barrier Breakdown. Stroke 2016, 47, 1068–1077. [Google Scholar] [CrossRef]
  162. Pelegrí, C.; Canudas, A.M.; del Valle, J.; Casadesus, G.; Smith, M.A.; Camins, A.; Pallàs, M.; Vilaplana, J. Increased permeability of blood–brain barrier on the hippocampus of a murine model of senescence. Mech. Ageing Dev. 2007, 128, 522–528. [Google Scholar] [CrossRef]
  163. Valle, J.; Duran-Vilaregut, J.; Manich, G.; Camins, A.; Pallàs, M.; Vilaplana, J.; Pelegrí, C. Time-course of blood–brain barrier disruption in senescence-accelerated mouse prone 8 (SAMP8) mice. Int. J. Dev. Neurosci. 2009, 27, 47–52. [Google Scholar] [CrossRef]
  164. Zeng, Q.; Du, S.; Xu, Y.; Yang, F.; Wu, L.; Wang, N.; Zhang, S.; Wei, S.; Wang, G.; Zhang, S.; et al. Assessing the Potential Value and Mechanism of Kaji-Ichigoside F1 on Arsenite-Induced Skin Cell Senescence. Oxidative Med. Cell. Longev. 2022, 2022, 9574473. [Google Scholar] [CrossRef]
  165. Sorrentino, J.A.; Krishnamurthy, J.; Tilley, S.; Alb, J.G.; Burd, C.E.; Sharpless, N. p16INK4a reporter mice reveal age-promoting effects of environmental toxicants. J. Clin. Investig. 2014, 124, 169–173. [Google Scholar] [CrossRef] [PubMed]
  166. Chatterjee, D.; Bhattacharjee, P.; Sau, T.J.; Das, J.K.; Sarma, N.; Bandyopadhyay, A.K.; Roy, S.S.; Giri, A.K. Arsenic exposure through drinking water leads to senescence and alteration of telomere length in humans: A case-control study in West Bengal, India. Mol. Carcinog. 2015, 54, 800–809. [Google Scholar] [CrossRef] [PubMed]
  167. Chung, Y.-P.; Chen, Y.-W.; Weng, T.-I.; Yang, R.-S.; Liu, S.-H. Arsenic induces human chondrocyte senescence and accelerates rat articular cartilage aging. Arch. Toxicol. 2020, 94, 89–101. [Google Scholar] [CrossRef]
  168. Yamaguchi, Y.; Madhyastha, H.; Madhyastha, R.; Choijookhuu, N.; Hishikawa, Y.; Pengjam, Y.; Nakajima, Y.; Maruyama, M. Arsenic acid inhibits proliferation of skin fibroblasts, and increases cellular senescence through ROS mediated MST1-FOXO signaling pathway. J. Toxicol. Sci. 2016, 41, 105–113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Okamura, K.; Sato, M.; Suzuki, T.; Nohara, K. Inorganic arsenic exposure-induced premature senescence and senescence-associated secretory phenotype (SASP) in human hepatic stellate cells. Toxicol. Appl. Pharmacol. 2022, 454, 116231. [Google Scholar] [CrossRef]
  170. Cheng, Y.; Li, Y.; Ma, C.; Song, Y.; Xu, H.; Yu, H.; Xu, S.; Mu, Q.; Li, H.; Chen, Y.; et al. Arsenic trioxide inhibits glioma cell growth through induction of telomerase displacement and telomere dysfunction. Oncotarget 2016, 7, 12682–12692. [Google Scholar] [CrossRef] [Green Version]
  171. Liu, J.; Jia, D.-Y.; Cai, S.-Z.; Li, C.-P.; Zhang, M.-S.; Zhang, Y.-Y.; Yan, C.-H.; Wang, Y.-P. Mitochondria defects are involved in lead-acetate-induced adult hematopoietic stem cell decline. Toxicol. Lett. 2015, 235, 37–44. [Google Scholar] [CrossRef] [PubMed]
  172. Yamashita, R.; Takahashi, Y.; Takashima, K.; Okano, H.; Ojiro, R.; Tang, Q.; Kikuchi, S.; Kobayashi, M.; Ogawa, B.; Jin, M.; et al. Induction of cellular senescence as a late effect and BDNF-TrkB signaling-mediated ameliorating effect on disruption of hippocampal neurogenesis after developmental exposure to lead acetate in rats. Toxicology 2021, 456, 152782. [Google Scholar] [CrossRef]
  173. Chatterjee, S.; Kundu, S.; Sengupta, S.; Bhattacharyya, A. Divergence to apoptosis from ROS induced cell cycle arrest: Effect of cadmium. Mutat. Res. Mol. Mech. Mutagen. 2009, 663, 22–31. [Google Scholar] [CrossRef]
  174. Wang, T.; Yuan, Y.; Zou, H.; Yang, J.; Zhao, S.; Ma, Y.; Wang, Y.; Bian, J.; Liu, X.; Gu, J.; et al. The ER stress regulator Bip mediates cadmium-induced autophagy and neuronal senescence. Sci. Rep. 2016, 6, 38091. [Google Scholar] [CrossRef] [Green Version]
  175. Luo, H.; Gu, R.; Ouyang, H.; Wang, L.; Shi, S.; Ji, Y.; Bao, B.; Liao, G.; Xu, B. Cadmium exposure induces osteoporosis through cellular senescence, associated with activation of NF-κB pathway and mitochondrial dysfunction. Environ. Pollut. 2021, 290, 118043. [Google Scholar] [CrossRef]
  176. Zhang, Y.; Zhang, Y.; Zhong, C.; Xiao, F. Cr(VI) induces premature senescence through ROS-mediated p53 pathway in L-02 hepatocytes. Sci. Rep. 2016, 6, 34578. [Google Scholar] [CrossRef]
  177. Xiao, F.; Feng, X.; Zeng, M.; Guan, L.; Hu, Q.; Zhong, C. Hexavalent chromium induces energy metabolism disturbance and p53-dependent cell cycle arrest via reactive oxygen species in L-02 hepatocytes. Mol. Cell. Biochem. 2012, 371, 65–76. [Google Scholar] [CrossRef] [PubMed]
  178. Sivakumar, K.K.; Stanley, J.A.; Arosh, J.A.; Pepling, M.E.; Burghardt, R.C.; Banu, S.K. Prenatal exposure to chromium induces early reproductive senescence by increasing germ cell apoptosis and advancing germ cell cyst breakdown in the F1 offspring. Dev. Biol. 2014, 388, 22–34. [Google Scholar] [CrossRef] [Green Version]
  179. Val, M.M.; Mendes, L.A.; Alarcão, A.; Carvalho, L.; Carreira, I.M.; Rodrigues, C.; Alpoim, M.C. Senescent bronchial fibroblasts induced to senescence by Cr(VI) promote epithelial–mesenchymal transition when co-cultured with bronchial epithelial cells in the presence of Cr(VI). Mutagenesis 2015, 30, 277–286. [Google Scholar] [CrossRef]
  180. Ma, Y.; Liang, Y.; Liang, N.; Zhang, Y.; Xiao, F. Identification and functional analysis of senescence-associated secretory phenotype of premature senescent hepatocytes induced by hexavalent chromium. Ecotoxicol. Environ. Saf. 2021, 211, 111908. [Google Scholar] [CrossRef]
  181. Liang, Y.; Liang, N.; Ma, Y.; Tang, S.; Ye, S.; Xiao, F. Role of Clusterin/NF-κB in the secretion of senescence-associated secretory phenotype in Cr(VI)-induced premature senescent L-02 hepatocytes. Ecotoxicol. Environ. Saf. 2021, 219, 112343. [Google Scholar] [CrossRef] [PubMed]
  182. Li, S.; Ma, Y.; Liang, Y.; Liang, N.; Ye, S.; Xiao, F. The role of PKA/PP2B-mediated Drp1 phosphorylation and the subsequent EGFR inhibition in Cr(VI)-induced premature senescence. Ecotoxicol. Environ. Saf. 2021, 218, 112300. [Google Scholar] [CrossRef] [PubMed]
  183. Li, Y.; Hu, J.; Guan, F.; Song, L.; Fan, R.; Zhu, H.; Hu, X.; Shen, E.; Yang, B. Copper induces cellular senescence in human glioblastoma multiforme cells through downregulation of Bmi-1. Oncol. Rep. 2013, 29, 1805–1810. [Google Scholar] [CrossRef] [Green Version]
  184. Matos, L.; Gouveia, A.; Almeida, H. Copper ability to induce premature senescence in human fibroblasts. Age 2012, 34, 783–794. [Google Scholar] [CrossRef] [Green Version]
  185. Chen, S.-Y.; Liu, S.-T.; Lin, W.-R.; Lin, C.-K.; Huang, S.-M. The Mechanisms Underlying the Cytotoxic Effects of Copper Via Differentiated Embryonic Chondrocyte Gene 1. Int. J. Mol. Sci. 2019, 20, 5225. [Google Scholar] [CrossRef] [Green Version]
  186. Boilan, E.; Winant, V.; Dumortier, E.; Piret, J.-P.; Bonfitto, F.; Osiewacz, H.D.; Debacq-Chainiaux, F.; Toussaint, O. Role of p38MAPK and oxidative stress in copper-induced senescence. Age 2013, 35, 2255–2271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Matos, L.; Gouveia, A.M.; Almeida, H. Resveratrol Attenuates Copper-Induced Senescence by Improving Cellular Proteostasis. Oxidative Med. Cell. Longev. 2017, 2017, 3793817. [Google Scholar] [CrossRef] [Green Version]
  188. Dublineau, I.; Grandcolas, L.; Grison, S.; Baudelin, C.; Paquet, F.; Voisin, P.; Aigueperse, J.; Gourmelon, P. Modifications of Inflammatory Pathways in Rat Intestine Following Chronic Ingestion of Depleted Uranium. Toxicol. Sci. 2007, 98, 458–468. [Google Scholar] [CrossRef] [PubMed]
  189. Luczak, M.W.; Zhitkovich, A. Nickel-induced HIF-1α promotes growth arrest and senescence in normal human cells but lacks toxic effects in transformed cells. Toxicol. Appl. Pharmacol. 2017, 331, 94–100. [Google Scholar] [CrossRef] [PubMed]
  190. Lewinska, A.; Adamczyk-Grochala, J.; Bloniarz, D.; Horeczy, B.; Zurek, S.; Kurowicki, A.; Woloszczuk-Gebicka, B.; Widenka, K.; Wnuk, M. Remifentanil preconditioning protects against hypoxia-induced senescence and necroptosis in human cardiac myocytes in vitro. Aging 2020, 12, 13924–13938. [Google Scholar] [CrossRef]
  191. Papageorgiou, I.; Yin, Z.; Ladon, D.; Baird, D.; Lewis, A.C.; Sood, A.; Newson, R.; Learmonth, I.D.; Case, C.P. Genotoxic effects of particles of surgical cobalt chrome alloy on human cells of different age in vitro. Mutat. Res. Mol. Mech. Mutagen. 2007, 619, 45–58. [Google Scholar] [CrossRef]
  192. Braakhuis, H.M.; Gosens, I.; Krystek, P.; Boere, J.A.F.; Cassee, F.R.; Fokkens, P.H.B.; Post, J.A.; Van Loveren, H.; Park, M.V.D.Z. Particle size dependent deposition and pulmonary inflammation after short-term inhalation of silver nanoparticles. Part. Fibre Toxicol. 2014, 11, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Chen, B.; Chai, Q.; Xu, S.; Li, Q.; Wu, T.; Chen, S.; Wu, L. Silver nanoparticle-activated COX2/PGE2 axis involves alteration of lung cellular senescence in vitro and in vivo. Ecotoxicol. Environ. Saf. 2020, 204, 111070. [Google Scholar] [CrossRef]
  194. Mytych, J.; Pacyk, K.; Pepek, M.; Zebrowski, J.; Lewinska, A.; Wnuk, M. Nanoparticle-mediated decrease of lamin B1 pools promotes a TRF protein-based adaptive response in cultured cells. Biomaterials 2015, 53, 107–116. [Google Scholar] [CrossRef]
  195. Salazar, G.; Huang, J.; Feresin, R.; Zhao, Y.; Griendling, K. Zinc regulates Nox1 expression through a NF-κB and mitochondrial ROS dependent mechanism to induce senescence of vascular smooth muscle cells. Free. Radic. Biol. Med. 2017, 108, 225–235. [Google Scholar] [CrossRef] [PubMed]
  196. Maus, M.; López-Polo, V.; Lafarga, M.; Aguilera, M.; De Lama, E.; Meyer, K.; Manonelles, A.; Sola, A.; Martinez, C.L.; López-Alonso, I.; et al. Iron accumulation drives fibrosis, senescence, and the senescence-associated secretory phenotype. bioRxiv 2022. [Google Scholar] [CrossRef]
  197. Kim, J.-Y.; Kim, S.-J.; Bae, M.A.; Cho, K.-H. Cadmium exposure exacerbates severe hyperlipidemia and fatty liver changes in zebrafish via impairment of high-density lipoproteins functionality. Toxicol. Vitr. 2018, 47, 249–258. [Google Scholar] [CrossRef]
  198. Alexopoulos, E.; Cominos, X.; Trougakos, I.P.; Lourda, M.; Gonos, E.S.; Makropoulos, V. Biological Monitoring of Hexavalent Chromium and Serum Levels of the Senescence Biomarker Apolipoprotein J/Clusterin in Welders. Bioinorg. Chem. Appl. 2008, 2008, 420578. [Google Scholar] [CrossRef] [PubMed]
  199. Masaldan, S.; Clatworthy, S.A.; Gamell, C.; Smith, Z.M.; Francis, P.S.; Denoyer, D.; Meggyesy, P.M.; La Fontaine, S.; Cater, M.A. Copper accumulation in senescent cells: Interplay between copper transporters and impaired autophagy. Redox Biol. 2018, 16, 322–331. [Google Scholar] [CrossRef]
  200. Pal, K.; Raghuram, G.V.; Dsouza, J.; Shinde, S.; Jadhav, V.; Shaikh, A.; Rane, B.; Tandel, H.; Kondhalkar, D.; Chaudhary, S.; et al. A pro-oxidant combination of resveratrol and copper down-regulates multiple biological hallmarks of ageing and neurodegeneration in mice. Sci. Rep. 2022, 12, 17209. [Google Scholar] [CrossRef]
  201. Bleise, A.; Danesi, P.; Burkart, W. Properties, use and health effects of depleted uranium (DU): A general overview. J. Environ. Radioact. 2003, 64, 93–112. [Google Scholar] [CrossRef]
  202. Briner, W. The Toxicity of Depleted Uranium. Int. J. Environ. Res. Public Health 2010, 7, 303–313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Briner, W.; Murray, J. Effects of short-term and long-term depleted uranium exposure on open-field behavior and brain lipid oxidation in rats. Neurotoxicol. Teratol. 2005, 27, 135–144. [Google Scholar] [CrossRef] [PubMed]
  204. Wise, S.S.; Thompson, W.D.; Aboueissa, A.-M.; Mason, M.D.; Wise, J.P., Sr. Particulate Depleted Uranium Is Cytotoxic and Clastogenic to Human Lung Cells. Chem. Res. Toxicol. 2007, 20, 815–820. [Google Scholar] [CrossRef] [PubMed]
  205. Streffer, C.; Kryscio, A.; Müller, W.-U.; Kotschy-Lang, N. Genomic instability in uranium miners after high radiation expo-sures. Int. Congr. Ser. 2002, 1225, 223–227. [Google Scholar] [CrossRef]
  206. Wang, Q.-Q.; Yin, G.; Huang, J.-R.; Xi, S.-J.; Qian, F.; Lee, R.-X.; Peng, X.-C.; Tang, F.-R. Ionizing Radiation-Induced Brain Cell Aging and the Potential Underlying Molecular Mechanisms. Cells 2021, 10, 3570. [Google Scholar] [CrossRef]
  207. Angelova, D.M.; Brown, D.R. Altered Processing of β-Amyloid in SH-SY5Y Cells Induced by Model Senescent Microglia. ACS Chem. Neurosci. 2018, 9, 3137–3152. [Google Scholar] [CrossRef]
  208. Killilea, D.W.; Wong, S.L.; Cahaya, H.S.; Atamna, H.; Ames, B.N. Iron Accumulation during Cellular Senescence. Ann. N. Y. Acad. Sci. 2004, 1019, 365–367. [Google Scholar] [CrossRef] [PubMed]
  209. Masaldan, S.; Clatworthy, S.A.; Gamell, C.; Meggyesy, P.M.; Rigopoulos, A.-T.; Haupt, S.; Haupt, Y.; Denoyer, D.; Adlard, P.A.; Bush, A.I.; et al. Iron accumulation in senescent cells is coupled with impaired ferritinophagy and inhibition of ferroptosis. Redox Biol. 2018, 14, 100–115. [Google Scholar] [CrossRef]
  210. Masaldan, S.; Belaidi, A.A.; Ayton, S.; Bush, A.I. Cellular Senescence and Iron Dyshomeostasis in Alzheimer’s Disease. Pharmaceuticals 2019, 12, 93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  211. Zhang, Y.; Wang, Z.; Li, X.; Wang, L.; Yin, M.; Wang, L.; Chen, N.; Fan, C.; Song, H. Dietary Iron Oxide Nanoparticles Delay Aging and Ameliorate Neurodegeneration in Drosophila. Adv. Mater. 2015, 28, 1387–1393. [Google Scholar] [CrossRef] [PubMed]
  212. Admasu, T.D.; Kim, K.; Rae, M.; Avelar, R.; Gonciarz, R.L.; Rebbaa, A.; de Magalhães, J.P.; Renslo, A.R.; Stolzing, A.; Sharma, A. Selective ablation of primary and paracrine senescent cells by targeting iron dyshomeostasis. Cell Rep. 2023, 42, 112058. [Google Scholar] [CrossRef] [PubMed]
  213. Go, S.; Kang, M.; Kwon, S.P.; Jung, M.; Jeon, O.H.; Kim, B. The Senolytic Drug JQ1 Removes Senescent Cells via Ferroptosis. Tissue Eng. Regen. Med. 2021, 18, 841–850. [Google Scholar] [CrossRef] [PubMed]
  214. Li, J.; Bi, Z.; Wang, L.; Xia, Y.; Xie, Y.; Liu, Y. Recent Advances in Strategies for Imaging Detection and Intervention of Cellular Senescence. Chembiochem 2023, 24, e202200364. [Google Scholar] [CrossRef] [PubMed]
  215. Chen, W.J.; Kung, G.P.; Gnana-Prakasam, J.P. Role of Iron in Aging Related Diseases. Antioxidants 2022, 11, 865. [Google Scholar] [CrossRef]
  216. Tian, Y.; Tian, Y.; Yuan, Z.; Zeng, Y.; Wang, S.; Fan, X.; Yang, D.; Yang, M. Iron Metabolism in Aging and Age-Related Diseases. Int. J. Mol. Sci. 2022, 23, 3612. [Google Scholar] [CrossRef]
  217. Ward, R.J.; Zucca, F.A.; Duyn, J.H.; Crichton, R.R.; Zecca, L. The role of iron in brain ageing and neurodegenerative disorders. Lancet Neurol. 2014, 13, 1045–1060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Ouyang, W.; Zhang, D.; Li, J.; Verma, U.N.; Costa, M.; Huang, C. Soluble and insoluble nickel compounds exert a differential inhibitory effect on cell growth through IKKα-dependent cyclin D1 down-regulation. J. Cell. Physiol. 2009, 218, 205–214. [Google Scholar] [CrossRef] [Green Version]
  219. Ma, C.; Song, M.; Zhang, Y.; Yan, M.; Zhang, M.; Bi, H. Nickel nanowires induce cell cycle arrest and apoptosis by generation of reactive oxygen species in HeLa cells. Toxicol. Rep. 2014, 1, 114–121. [Google Scholar] [CrossRef] [Green Version]
  220. Petrova, E.; Pavlova, E.; Tinkov, A.A.; Ajsuvakova, O.P.; Skalny, A.V.; Rashev, P.; Vladov, I.; Gluhcheva, Y. Cobalt accumulation and iron-regulatory protein profile expression in immature mouse brain after perinatal exposure to cobalt chloride. Chem. Interact. 2020, 329, 109217. [Google Scholar] [CrossRef] [PubMed]
  221. Rai, M.; Yadav, A.; Gade, A. Silver nanoparticles as a new generation of antimicrobials. Biotechnol. Adv. 2009, 27, 76–83. [Google Scholar] [CrossRef]
  222. Silva, R.M.; Anderson, D.S.; Peake, J.; Edwards, P.C.; Patchin, E.S.; Guo, T.; Gordon, T.; Chen, L.C.; Sun, X.; Van Winkle, L.S.; et al. Aerosolized Silver Nanoparticles in the Rat Lung and Pulmonary Responses over Time. Toxicol. Pathol. 2016, 44, 673–686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Tripathi, R.; Raghunath, R.; Sastry, V.; Krishnamoorthy, T. Daily intake of heavy metals by infants through milk and milk products. Sci. Total. Environ. 1999, 227, 229–235. [Google Scholar] [CrossRef]
  224. Fedarko, N.S. The Biology of Aging and Frailty. Clin. Geriatr. Med. 2011, 27, 27–37. [Google Scholar] [CrossRef] [Green Version]
Table 1. Associations between heavy metals/metalloids and neurodegenerative diseases.
Table 1. Associations between heavy metals/metalloids and neurodegenerative diseases.
Metal or
Metalloid
Demographics of
Human Study
Animal Model
(Species/Strain)
ExposureAssociated Neurodegenerative Disease(s)ObservationsMechanistic DetailsRefs.
Arsenic133 men and 301 women older than 40 years, living in Cochran or Parmer County, TexasN/A2.19–15.26 μg/L for 1–80 yearsADDecreased language scores, visuospatial skills, and executive functioning.As exposure induces ROS generation and DNA damage, leading to broad neurodegeneration [85,86]. Dimethyl arsenic increases expression of Aβ following APP upregulation, suggesting a mechanism related to AD [87].[88]
Prevalence of AD and other dementias compared to average As concentration in topsoil from eleven European countriesN/A7–18 ppmADDiagnosis and prevalence within country. Dementia Composite Index (product of AD prevalence and mortality rank within group) plotted against As soil concentration.[89]
Prevalence of PD patients in Taiwan compared to As soil contaminationN/A2.18–11.47 mg/kg in soilPDDiagnosis of PD correlated with As soil concentration using Local Moran’s I and spatial regression analysis.[90]
6 ALS patients (3 men, 3 women)N/AN.R.ALSSerum As concentration had a strong positive correlation with disease duration.[91]
Lead121 PD patients and 414 controlsN/ADetermined via participant interviewPDPD diagnosis and regression, indicating a two-fold increase in PD diagnosis following Pb exposure.Pb crosses the BBB by substituting for divalent metals [92]. Pb enters neurons and astroglia, inducing mitochondrial damage and initiating C-caspase cascade activation [92]. This disrupts metabolism, alters neurotransmission, and induces neurodegeneration [92]. Pb is linked to AD as it is shown to increase Aβ accumulation and reduce Aβ clearance [87].[93]
17 patients with a definitive ALS diagnosis aged 47–85 years old from Oslo, Norway; 10 controls were aged 26–77 years oldN/AN.R.ALSICP-MS indicates a significantly greater CSF concentration of Pb in ALS patients compared to controls.[94]
V/AMacaca fascicularis1.5 mg/kg lead acetate during infancy (until day 400), assessed at age 23ADIncreased APP expression, elevated Aβ levels; decreased DNA methyltransferase and higher oxidative DNA damage[95]
N/ALong–Evans hooded rats, 24 h old200 ppm lead acetate for 20 days or 18–20 monthsADUpregulation of APP and increased expression of Aβ.[96]
N/AKunming MicePrenatal exposure to 0.1%, 0.5%, or 1% lead acetate, lasting until weaningADAltered performance on Morris water maze. Increased immunoreactivity in cerebral cortex. Increased expression of Aβ.[97]
N/ALong–Evans hooded rats50 ppm lead acetate in drinking water for 90 daysPDLoss of basal dopamine, 3,4-dihydroxyphenylace tic acid, and homovanillic acid levels.[98]
Cadmium64-year-old manN/AAcute (60 min) inhalationPD3 months after exposure patient exhibited with bradykinesia, stooped posture, cogwheel muscle rigidity, short-stepped gait, and disturbed freezing and righting reflexes.Cd is associated with ALS pathogenesis by reducing Cu/Zn SOD enzymes, disrupting BBB integrity, and inducing glutamate toxicity [99]. Cd induced oxidative stress and inflammation in rat cortex and hippocampus, and increased synthesis of Aβ, resembling AD pathology [87].[100]
Fixed brain tissue from 8 ALS, 4 PDC, and 5 control subjects who lived in Guam.N/AN.R.ALSGray and white matter from ALS patients had elevated concentrations of Cd in all regions except for the basal ganglia.[101]
4064 participants from the 1999–2004 NHANES aged greater than 60 years with available blood Cd data. Participants included 51 individuals who suffered AD mortality.N/AN.R.ADGreater blood Cd concentration is strongly associated with increased risk of AD mortality.[102]
17 patients with a definitive ALS diagnosis aged 47–85 years old from Oslo, Norway; 10 controls were aged 26–77 years oldN/AN.R.ALSICP-MS indicates a significantly greater CSF concentration of Cd in ALS patients compared to controls.[94]
Chromium6 ALS patients
(3 men, 3 women)
N/AN.R.ALSWhole blood Cr was higher in ALS patients when compared to reference values of Italian population. Whole blood Cr concentration is also correlated with disease duration in this study.No exact mechanism has been elucidated for Cr(VI) neurotoxicity. Lipid peroxidation, apoptosis, altered acetylcholinesterase, and inflammation observed in Cr(VI) neurotoxicity across multiple taxa [39].[91]
AD patients (12 men, 41 women) aged 68–94 years old, controls (100 men, 117 women) were patients at the same hospital being treated for optical disordersN/AN.R.ADMultivariable logistic regression and odds ratios indicate an association between high blood concentrations of Cr and AD pathology.[103]
Copper9 AD patients and 5 controls (post mortem)N/AN.R.ADPresence of neuritic plaques correlated with increased Cu in senile plaque rims and cores, total senile plaques, and neuropil (measured using micro-PIXE).Excess Cu induced mitochondrial release of ROS and activated microglia, resembling pathology in MS [99]. Cu accumulation in the spinal cord increased lipid peroxidation and may be a mechanism for ALS [99]. Cu neurotoxicity resembles AD by increasing ROS and inflammation while downregulating LRP1, resulting in reduced Aβ clearance [104]. Cu complexes with Aβ to form aggregates and impair mitochondrial function [104].[105]
15 WD patients and 5 controls; controls were autopsy samples collected from individuals who died in car accidents but received no brain trauma.N/AN.R.WDICP-MS demonstrated that Cu accumulation is ubiquitous in WD neuropathology, increasing nearly 8-fold compared to controls[106]
17 patients with a definitive ALS diagnosis aged 47–85 years old from Oslo, Norway. 10 controls were aged 26–77 years oldN/AN.R.ALSICP-MS indicates a significantly greater CSF concentration of Cu in ALS patients compared to controls.[94]
N/AD. melanogaster (P463, P463/Ctr1B RNAi, P463/DmATP7 OE, P463/DmATP7 RNAi)0.25 mM copper chloride for 24 hHDCu reduction rescued HD phenotypes in D. melanogaster (eclosion rate, shortened lifespan, impaired mobility, and degeneration of eyes and brain), whereas Cu accumulation exacerbated phenotypes. Overexpression of Cu transporters reduces protein aggregation, while silencing transporters increases protein aggregation. Feeding with Cu induced D. melanogaster HD phenotypes, while cotreatment with a chelator rescued this phenotype.[107]
Wistar rat
(6-OHDA induced PD)
0.01 μg/g drinking waterPDIncreases in Cu concentrations in substantia nigra, globulus pallidus, putamen, and amygdala in 6-OHDA-treated rats compared to controls.[108]
Uranium17 patients with a definitive ALS diagnosis aged 47–85 years old from Oslo, Norway. 10 controls were aged 26–77 years oldN/AN.R.ALSICP-MS indicates a significantly greater CSF concentration of U in ALS patients compared to controls.U neurotoxicity is not well defined. U induced neuroinflammation, oxidative stress, cell death, and altered neuronal signaling [109].[94]
N/AC. elegans (NL5901, BZ555)1 μM DU for 1–3 daysPDC. elegans treated with DU exhibited increased aggregation of α-synuclein at days 3, 5, and 7. C. elegans exhibited shrinking of dopaminergic neurons at day 3, indicating chronic DU exposure induces neurodegeneration.[110]
NickelN/AMale Wistar rats aged 12–15 weeks2 μL of 300 μM nickel chloride intracerebral injectionNSIncreased behaviors demonstrating anxiety and depression, as well as decreased spatial learning abilities and memory.Multiple models show Ni accumulated in the cerebral cortex and whole brain [111]. Ni induced oxidative stress, decreased acetylcholinesterase, demyelination, and cell death in the brain [111].[112]
N/AMale and female Wistar rats0.25–0.5 mg/kg nickel chloride IP injectionNSIncreased behaviors demonstrating anxiety and depression. Worsened spatial memory.[113]
N/A60 male Wistar rats aged 9 weeks old150 μg/L nickel chlorideNSDecreased locomotor abilities and an increase in anxiety-like behaviors. Elevated levels of Ni in the cerebrum, cerebellum, and liver. Decreased CAT, GPx, GST, and AChE activity. Increased GSH activity. Degeneration of cerebellar cortical, Purkinje, and CA3 pyramidal neurons and granule cells of dentate gyrus.[114]
Cobalt6 ALS patients
(3 men, 3 women)
N/AN.R.ALSWhole blood Co was higher in ALS patients when compared to reference values of Italian population.Co induced neurotoxicity through DNA damage, caspase activation, increased ROS, altered mitochondrial function, apoptosis, and necrosis in neural cells [38]. Co alters neurotransmission by blocking Ca channels [38].[91]
17 patients with a definitive ALS diagnosis aged 47–85 years old from Oslo, Norway. 10 controls were aged 26–77 years oldN/AN.R.ALSICP-MS indicates a significantly greater CSF concentration of Co in ALS patients compared to controls.[94]
N/AMale C57BL/6J mice aged 8 weeks and 48 weeks4–16 mg/kg/day IP injection for 30 daysADSignificant Co accumulation in blood, cerebellar cortex, and hippocampus following treatment. Increased expression of Aβ1-42, APP, and GSK3β. Worsened performance in cognitive and memory tasks (Morris water maze).[115]
Zinc9 AD patients and 5 controls (post mortem)N/AN.R.ADPresence of neuritic plaques correlated with increased Cu in senile plaque rims and cores, total senile plaques, and neuropil (measured using micro-PIXE).Reduced clearance of Zn from glutamatergic synapses leads to amyloid oligomerization and tau protein phosphorylation in the cortex and hippocampus [116]. Zn increases aggregation rate of Aβ [116]. Zn-induced ROS is associated with lipid peroxidation in substantia nigra and mitochondrial dysfunction, associated with PD and ALS [116].[105]
6 ALS patients
(3 men, 3 women)
N/AN.R.ALSWhole blood Zn was higher in ALS patients when compared to reference values of Italian population.[91]
Fixed brain tissue from 8 ALS, 4 PDC, and 5 control subjects who lived in Guam.N/AN.R.PDGray matter from PDC patients had significantly increased Zn concentrations.[101]
17 patients with a definitive ALS diagnosis aged 47–85 years old from Oslo, Norway. 10 controls were aged 26–77 years oldN/AN.R.ALSICP-MS indicates a significantly greater CSF concentration of Zn in ALS patients compared to controls.[94]
N/AWistar Rat
(6-OHDA induced PD)
0.005 μg/g drinking waterPDIncreased Zn concentrations in substantia nigra, globulus pallidus, putamen, amygdala in 6-OHDA-treated rats compared to controls.[108]
N.R.—Not Reported, AD—Alzheimer’s Disease, PD—Parkinson’s Disease, PDC—Parkinson’s–Dementia Complex, ALS—Amyotrophic Lateral Sclerosis, WD—Wilson’s Disease, HD—Huntington’s Disease, NS—Non-Specific Symptoms of Neurodegeneration, CSF—Cerebrospinal Fluid, ICP-MS—Inductively Coupled Plasma Mass Spectrometry, IP—Intraperitoneal, ROS—Reactive Oxygen Species, LRP1—Low-Density Lipoprotein Receptor-Related Protein 1, SOD—Superoxide Dismutase.
Table 2. Summary of Senescence Markers Increased After Heavy Metal of Metalloid Exposure.
Table 2. Summary of Senescence Markers Increased After Heavy Metal of Metalloid Exposure.
Metal/MetalloidsModel
(Organ/Tissue Type)
Doses
(Compound)
Duration of ExposureSenescence Markers with Increased ExpressionMethod of Detection *Refs.
ArsenicHuman Immortalized Keratinocytes, HaCaT0.05–0.25 μM
(Sodium Arsenite)
24–72 hp16INK4a, p21, p53, SA-β-Gal, SASP (IL-1α, IL-6, IL-8, TGF-β1, MMP1, MMP3, EGF, VGF)WB, qRT-PCR, ELISA, FC[164]
Hairless SKH1-E p16Luc+ Mice
(Total Body)
50 ppm
(Sodium Arsenite)
12 monthsp16INK4aTBLI[165]
Human Participants (Peripheral Blood Mononuclear Cells)30–620 μg/LHuman exposure > 10 yearsp21, SA-β-Gal, SASP (IL-6, IL-8, MMP1, MMP3)WB, ICC, EILSA, MMP1 and MMP3 Kit[166]
Human Articular Chondrocyte Cells, HC-a1–5 μM
(Arsenic Trioxide)
24 hp16, p21, p53, SA-β-Gal, SASP (IL-1α, IL-1β, TGF-β, TNF-α, CCL2, PAI-1, MMP13)WB, qRT-PCR, ICC, FC[167]
4-Week-Old Male Wistar Rats
(Articular Cartilage)
0.05–0.5 ppm
(Arsenic Trioxide)
36 weeksp16, p21, p53, SA-β-Gal, SASP (IL-1α, IL-1β, TGF-β, TNF-α, CCL2, PAI-1, MMP13)WB, ICC, qRT-PCR[167]
Mouse Skin Fibroblasts, m5S10 ppm
(Arsenic Acid)
16 hSA-β-GalICC[168]
Human Hepatic Stellate, LX-22–7.5 μM
(Sodium Arsenite)
24–144 hp21, SASP (IL-1β, IL-18, MMP1, MMP3, CXCL1)WB, qRT-PCR, ICC[169]
Human Glioblastoma, U872–8 μM
(Arsenic Trioxide)
48 h–2 weeksp21, p53, SA-β-GalWB, ICC[170]
Human Glioblastoma, U2512–8 μM
(Arsenic Trioxide)
48 h–2 weeksSA-β-GalICC[170]
Human Glioma, SHG-44 cells2–8 μM
(Arsenic Trioxide)
48 h–2 weeksSA-β-GalICC[170]
Rat Glioma, C62–8 μM
(Arsenic Trioxide)
48 h–2 weeksSA-β-GalICC[170]
Lead8-Week-Old Sprague–Dawley Rats (Hematopoietic Stem Cells)200–600 mg/kg
(Lead Acetate)
4 weeksSA-β-GalICC[171]
Female Sprague–Dawley Rats
(Brain)
1238–2548 ppm
(Lead [II] Acetate Trihydrate)
34 weeksSASP (IL-1α, IL-1β, IL-6, IL-8, TGF-β1, TGF-β2, TGF-β3, TNF)qRT-PCR[172]
Cadmium6–8-Week-Old Male Swiss Albino Mice (Spleen)15 μM
(Cadmium Chloride)
6–18 hp21, p53, SA-β-Gal, SASPWB[173]
Rat Pheochromocytoma Cells, PC125–20 μM6–48 hp16, p21, p53, SA-β-Gal, SASP (IL-1α, IL-6)WB, ICC, qRT-PCR[174]
Primary Human Dermal Fibroblasts, HDF12–24 μM
(Cadmium Chloride)
4 passagesSA-β-GalICC[138]
3–4-Week-Old Female Sprague–Dawley Rats (Bone Marrow)50 mg/L
(Cadmium Chloride)
4 weeksSA-β-GalICC[175]
Primary Bone Marrow Mesenchymal Stem Cells10 μM
(Cadmium Chloride)
3 h–15 daysp16, p21, SA-β-Gal, SASP (IL-1α, IL-1β, TGF-β, VEGF, CXCL-1)WB, ICC[175]
ChromiumHuman Hepatocytes, L-0210 nM
(Potassium Dichromate)
24 h twice a week for 4 weeksp21, p53, SA-β-Gal, SMP30, SASP (IL-1, IL-6, IL-8, G-CSF, bFGF, MMP3)WB, ICC, qRT-PCR, ELISA[176]
Human Hepatocytes, L-024–16 μM
(Potassium Dichromate)
24–48 hp53WB[177]
Prenatal Rat (Ovaries)25 ppm
(Potassium Dichromate)
5 daysp53IHC, IF[178]
Human Bronchial Fibroblasts, BEAS-2B0.5–2 μM
(Potassium Dichromate)
4 weeksSA-β-GalICC[179]
Human Hepatocytes, L-0210 nM
(Potassium Dichromate)
3x 24 h treatments per week for 4 weeksSMP30, SA-β-Gal, SASP (ENO1, PRDX2, COTL1, CLU)WB, ICC, qRT-PCR, ELISA[180]
Human Hepatocytes, L-0210 nm
(Potassium Dichromate)
24 h treatment every other day, for 4 weeksp21, p53, SA-β-Gal, SASP (IL-6, IL-8, GM-CSF, CXCL1, MCP-1, CLU)WB, ICC, qRT-PCR, ELISA[181]
Human Hepatocytes, L-0210 nM3x 24 h treatments a week for 4 weeksSA-β-GalICC[182]
CopperGlioblastoma Multiforme Cells, U87-MG100–750 μM
(Copper Sulfate)
24 hp16, p21, p53, SA-β-Gal, SASP (TGF-β1, CLU)WB, ICC, qRT-PCR[183]
Human Diploid Fibroblasts, WI-38250–1000 μM
(Copper Sulfate)
24 hp21, SA-β-Gal, SASP (TGF-β1)WB, ICC, qRT-PCR[184]
Cervical Cancer Cells, HeLa10–500 μM
(Copper Sulfate)
8–16 hp21, p53, SA-β-GalWB, qRT-PCR[185]
Human Embryonic Kidney, HEK29310–500 μM
(Copper Sulfate)
8 hp53WB[185]
Human Pulmonary Adenocarcinoma, A54910–500 μM
(Copper Sulfate)
8 hp53WB[185]
Human Bronchial Epithelial, BEAS-2B10–500 μM
(Copper Sulfate)
8 hp53WB[185]
Human Endometrial Adenocarcinoma, HEC-1-A10–500 μM
(Copper Sulfate)
8 hp53WB[185]
Human Diploid Fibroblasts, WI-38500 μM
(Copper Sulfate)
24 hp16, p21, SA-β-Gal, SASP (TGF-β1, CTGF)ICC, qRT-PCR[186]
Human Diploid Fibroblasts, WI-38350 μM
(Copper Sulfate)
24 hp21, SA-β-Gal, SASP (TGF-β1)WB, ICC, qRT-PCR[187]
Uranium12-Week-Old Male Sprague–Dawley Rats (Ileal Mucosa)40 mg/L
(Depleted Uranium)
3–9 monthsSASP (IL- 1β, CCL2, TGF-β1, IL-10, IFNγ, COX2, PGE2)qRT-PCR[188]
NickelHuman Diploid Fibroblasts, IMR90300 μM–1 mM
(Nickle Chloride)
48 hp16, p21, p53WB[189]
CobaltPrimary Human Cardiac Cells, HCM200 μM
(Cobalt Chloride)
48 hp21, SA-β-Gal, SASP (IL-8)WB, ICC[190]
Primary Human Skin Fibroblasts0.0005:1–10,000 particle volume (μm3)/cell
(Cobalt Chrome)
6 h–15 daysSA-β-GalICC[191]
SilverMale Fischer Rats (Broncho-alveolar lavage fluid)167 μg/m3–179 μg/m36 h/day for 4 consecutive daysSASP (IL-1β, MCP-1, MIP-2)Bio-Plex Pro Assay[192]
Normal Human Lung Fibroblast, MRC50.4 μg/mL Ag+;
1–4 μg/mL AgNP
10 daysp21, SA-β-Gal, SASP (CSF2, CXCL3, CXCL5, CXCL8, IL- 1β, IL-6, MMP1, MMP2, MMP3, VEGFC, COX2, PGE2)WB, ICC, IB, IF, RNA-seq[193]
Nine-Week-Old Male C57BL/6 Mice
(Lung)
0.7 mg/m3 AgNP30 min/day for 45 daysSA-β-GalIHC[193]
Human Dermal Fibroblasts, HDFa100 μg/mL AgNP24–144 hp21, p53, SA-β-GalWB, ICC, IF[194]
Renal Cell Carcinoma, ACHN100 μg/mL AgNP24–144 hp21, p53, SA-β-GalWB, ICC, IF[194]
Cervical Cancer Cells, HeLa100 μg/mL AgNP24–144 hp21, p53, SA-β-GalWB, ICC, IF[194]
ZincVascular Smooth Muscle Cells isolated from Sprague–Dawley rat thoracic aortas (Endothelial)10–100 μM12 hp21, SA-β-GalWB, ICC, Plate-Based Fluorescence[195]
IronC57BL/6 mice (lung)Intratracheal bleomycin, induced iron chelation6 days–2 weeksp21, p16INK4aIHC[196]
Human Microglia Cells, SV40 Immortalized500 μM ferric ammonium citrate2 weeksIL-1β, IL-6, IL-8, KV1.3Cell Morphology, WB, ELISA
* WB—Western Blot, ICC—Immunocytochemistry, IF—Immunofluorescence, qRT-PCR—Quantitative Real-Time Polymerase Chain Reaction, RNA-seq—RNA sequencing, ELISA—Enzyme-Linked Immunosorbent Assay, TBLI—Total Body Luciferase Imaging.
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

Vielee, S.T.; Wise, J.P., Jr. Among Gerontogens, Heavy Metals Are a Class of Their Own: A Review of the Evidence for Cellular Senescence. Brain Sci. 2023, 13, 500. https://doi.org/10.3390/brainsci13030500

AMA Style

Vielee ST, Wise JP Jr. Among Gerontogens, Heavy Metals Are a Class of Their Own: A Review of the Evidence for Cellular Senescence. Brain Sciences. 2023; 13(3):500. https://doi.org/10.3390/brainsci13030500

Chicago/Turabian Style

Vielee, Samuel T., and John P. Wise, Jr. 2023. "Among Gerontogens, Heavy Metals Are a Class of Their Own: A Review of the Evidence for Cellular Senescence" Brain Sciences 13, no. 3: 500. https://doi.org/10.3390/brainsci13030500

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