Next Article in Journal
Neurochemical Aspects of the Role of Thirst in Body Fluid Homeostasis and Their Significance in Health and Disease: A Literature Review
Previous Article in Journal
Genome-Wide Identification of ATP-Binding Cassette (ABC) Transporter Gene Family and Their Expression Analysis in Response to Anthocyanin Transportation in the Fruit Peel of Eggplant (Solanum melongena L.)
Previous Article in Special Issue
Analyzing Molecular Determinants of Nanodrugs’ Cytotoxic Effects
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Effects of Microplastics and Nanoplastics Exposure on Neurogenesis: Are Thymidine Analogs a Good Option to Study Such Effects?

by
Mercè Encinas
and
Joaquin Martí Clúa
*
Unidad de Citología e Histología, Departament de Biologia Cel·Lular, de Fisiologia i d’Immunologia, Facultad de Biociencias, Institut de Neurociències, Universidad Autónoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(16), 7845; https://doi.org/10.3390/ijms26167845
Submission received: 11 July 2025 / Revised: 10 August 2025 / Accepted: 12 August 2025 / Published: 14 August 2025
(This article belongs to the Special Issue Molecular Research on Nanotoxicology)

Abstract

An important disadvantage of plastics is their fragmentation into smaller particles, classified according to size as microplastics and nanoplastics. These plastic particles persist for extended periods in aerial, terrestrial, and aquatic ecosystems and can be incorporated into animal bodies through various routes, including inhalation, dermal contact, and the food chain. The accumulation of these debris generates toxicity on several organs, including the nervous system. In this review article, I will cover the detrimental consequences of plastic exposure on the nervous system, the impact of microplastics and nanoplastics on the genesis of neurons both in the embryonic period as well as in adulthood, and the reliability of 5-bromo-2′-deoxyuridine (BrdU) labeling as a tool to analyze the effect of microplastic and nanoplastic exposure on the proliferative behavior of neuronal precursors. BrdU is a marker of DNA synthesis. It is widely used to identify proliferating neuroblasts and follow their fate during embryonic, perinatal, and adult neurogenesis. However, the use of BrdU labeling for analyzing neurogenesis may be inaccurate due to pitfalls and limitations. This is because BrdU exposure can induce apoptosis, cellular senescence, and alterations in DNA methylation. Interestingly, these cellular events also occur following exposure to plastic particles.

1. Introduction

Plastic is a word that originally meant pliable and easily shaped. The first plastic was developed in the mid-19th century by John Wesley Hyatt as a substitute for ivory. Since then, plastics have been widely used in modern industry around the world. Nowadays, almost all aspects of daily life involve plastics. They have revolutionized industries such as food packaging, construction, medical equipment, and electronics [1]. According to the International Union of Pure and Applied Chemistry, plastic is defined as a polymeric material. There are seven major plastic species: polypropylene, high- and low-density polyethylene, polyvinyl chloride, polyethylene terephthalate, polystyrene, and polyurethane. They are produced from nonrenewable petroleum and natural gas [2]. To these polymers, several additives are added to impart or enhance some physical and chemical properties, including resistance to solar irradiation, ductility, and color.
The use of plastic goods presents both advantages and disadvantages. Among their advantages are their low cost, light weight, durability, and resistance to mechanical damage, corrosion, and adverse weather conditions. However, their disadvantages mainly arise from overproduction and inadequate recycling practices. In 2022, global plastic production exceeded 400 million tons. If this current trend persists, it is presumable that plastic production will triple, and about 12 billion metric tons will be released into the environment by 2050 [3]. Several studies have revealed that only 18% of plastics are recycled, and 24% are incinerated. The remaining 58% is returned to the environment as waste, where they persist for a long time [4,5]. It is unknown how long plastic waste has been present in the ecosystems. It is believed that, due to their slow degradation, plastics may persist for centuries or even millennia [6].
Other major disadvantages of plastics are their additives and fragmentation. There are several molecules classified as plastic additives, including stabilizers, fillers, and cross-linking agents. These chemicals can accumulate within host organisms, resulting in adverse biological effects [7]. In this context, it has been observed that phthalates are related to low fertility in humans and premature development in women. Similarly, bisphenol A and polybrominated diphenyl ethers are associated with endocrine disruption and respiratory toxicity [8].
On the other hand, the breakage of plastics into small debris is another serious issue in our society. Plastic fragments are categorized into macroplastics (fragments larger than 5 mm), microplastics (particles smaller than 5 mm), and nanoplastics (particles smaller than 0.1 µm). The latter two are generated either through primary industrial processes or via the degradation of macroplastics by various physical, chemical, and biological mechanisms [9]. Due to their small size and physicochemical properties, microplastics and nanoplastics become bioavailable and have the potential to bioaccumulate. These persistent and ubiquitous contaminants appear in high concentrations in terrestrial and aquatic ecosystems, including oceans, soils, and plants [10,11]. Microplastics and nanoplastics can serve as carriers for several environmental contaminants, including organic pollutants, antibiotics, and pathogenic microorganisms [12,13]. Furthermore, plastic debris has detrimental effects on both plants and animals. In animals, plastic particle accumulation occurs through inhalation, dermal exposure, and ingestion of contaminated foodstuffs of both animal and vegetable origin [14,15]. After exposure, microplastics and nanoplastics are found in numerous human biological specimens, including plasma of blood, stools, saliva, and mother’s milk, as well as in the lung, liver, and brain. Moreover, previous studies have indicated that accumulation of these pollutants produces toxicity in different tissues and organs of invertebrates and vertebrates, including the gastrointestinal tract, liver, kidney, lung, and nervous system [11,15,16,17].
In the ambit of neurogenesis, a pivotal issue in both embryonic and adult neurogenesis research is the reliable identification, in histological sections, of those neuroblasts engaged in DNA synthesis [18]. The possibility of tagging proliferative neuron precursors and dating timetables of neurogenesis began with the application of tritiated thymidine and subsequent autoradiography in fixed samples [19,20]. This procedure is arduous, requires technical expertise to ensure the handling of samples, and needs a well equipped laboratory. These limitations were overcome with the use of thymidine analogs. Several pyrimidine analogs have been used: 5-bromo-2′-deoxyuridine (BrdU), 5-chloro-2′-deoxyuridine, 5-iodo-2′-deoxyuridine, and 5-ethynyl-2′-deoxyuridine. Recently, novel nucleotide analogs such as (2′S)-2′-deoxy-2′-fluoro-5-ethynyluridine, 5- (azidomethyl) -2′-deoxyuridine, and 5-vinyl-2′-deoxyuridine have been developed. These analogs share a common feature: all incorporate into nascent DNA [21]. Of all these, BrdU is the most widely used. This synthesized bromide-labeled base analog is chemically and structurally distinct from thymidine. BrdU incorporates a completely foreign atom (Br) into replicating DNA by substituting bromouracil for thymidine [18]. Moreover, this halopyrimidine crosses the blood–brain barrier (BBB) via nucleoside transporters and is permanently integrated into newly synthesized DNA during the S-phase [21,22]. Once incorporated into nascent DNA, BrdU remains stable for prolonged periods of time, and it will be passed down to daughter neuroblasts following the mitotic phase of the cell cycle [23].
In this review, I will cover the following items: (i) the harmful effects of plastic particle exposure on the central nervous system (CNS), (ii) the impact of microplastics and nanoplastics on the genesis of neurons both in the embryonic period as well as in adulthood, and (iii) the reliability of BrdU labeling as a tool to analyze the effect of microplastic and nanoplastic exposure on the proliferative behavior of neuronal precursors. In this section, I will also demonstrate that both plastic debris and BrdU induce apoptosis, cellular senescence, and alterations in DNA methylation. Therefore, the use of this marker may not be appropriate for assessing the effects of plastic particles.

2. Harmful Effects of Plastic Particle Exposure on the Central Nervous System

Plastic particles are found in aquatic, terrestrial, and aerial areas. These debris produce cardiovascular, hepatic, and renal injury [24,25]. The nervous system is also affected. In this context, previous research has provided evidence of the transplacental transfer of plastic particles to the fetal brain following exposure of pregnant dams. Plastics were found in the cerebellum, hippocampus, striatum, prefrontal cortex, hippocampus, substantia nigra, and pituitary. The cerebellum, hippocampus, striatum, and prefrontal cortex were the regions containing the highest accumulation of plastic particles [26]. In addition, plastic particles have been shown to cross the BBB and can subsequently accumulate in both gray and white matter.
An in vivo study revealed that plastic particles are found in the mouse brain 120 min after exposure [11]. The mechanisms by which microplastics and nanoplastics cross the BBB are not yet fully understood. At least two mechanisms have been proposed: The first of them has proposed that plastic debris increases the permeability of the BBB through the impairment of the zonulae occludentes located in the endothelial cells [10]. The second mechanism, using computer models, suggests that the presence of a biomolecular corona around plastic particles is needed for their ability to cross the BBB. The second mechanism, using computer models, suggests that the presence of a biomolecular corona around plastic particles is needed for their ability to cross the BBB. The same study indicates that cholesterol molecules promote the ability of plastic particles to cross the BBB [11].
When located in the brain, plastic debris produces deleterious effects through several mechanisms such as: (i) oxidative stress, (ii) inhibition of acetylcholinesterase, (iii) mitochondrial malfunction, and (iv) inflammatory reactions. In the first mechanism, it has been reported that exposure to microplastics and nanoplastics in mice increases the production of reactive oxygen species and malondialdehyde, while simultaneously decreasing glutathione levels in the CNS [27,28]. The same study also demonstrated that learning and memory activity were altered in the Morris water maze paradigm [27]. Interestingly, excessive levels of reactive oxygen species are negatively related to alterations in neural development during perinatal life [29]. Another experiment has reported that plastic particle exposure promotes alterations in the activities and levels of superoxide dismutase, catalase, and malondialdehyde in the brain of the Oryzias javanicus, which suggests that plastic debris generates an imbalance between the production of reactive oxygen species and the capacity of the cellular antioxidant system to ameliorate that production [30]. In the annelid Eisenia foetida, on the other hand, it has been reported that plastic debris produces modifications in the content of malondialdehyde as well as in the catalase activity [31].
Acetylcholinesterase is a cholinergic enzyme involved in the breakdown of acetylcholine into choline and acetate. Because acetylcholine is a neurotransmitter that regulates motor neuron function of the gray matter, the inhibition of acetylcholinesterase activity is a signal of neurotoxicity [32]. Several studies have reported that exposure to microplastics and nanoplastics inhibits acetylcholinesterase activity in various marine organisms, a process associated with the disruption of normal CNS [30,33,34]. The effects of plastic particles on acetylcholinesterase activity are not limited to aquatic species but have also been observed in terrestrial mammals. In this context, treatment with plastic particles has been shown to alter the activity of this hydrolase in both the liver and the brain [35].
Another important issue related to microplastic and nanoplastic exposure is the effect of these pollutants on mitochondrial function. This cytoplasmic organelle is involved in critical cellular functions, including aerobic respiration to generate ATP. The effects of plastic particle exposure on neuronal mitochondrial function have been shown to depend on particle size. Several studies have indicated that nanoplastics exert greater effects than microplastics, likely due to their higher propensity to accumulate in both the inner and outer mitochondrial membranes [36,37]. In this context, exposure to plastic particles has been found to impair mitochondrial oxidative phosphorylation in dopaminergic-differentiated SH-SY5Y cells, leading to dysfunctions in mitochondrial membrane potential, oxygen consumption rate, and ATP production via the AMPK/ULK1 pathway [38]. Consistent with these findings, nanoplastic exposure in rodents has been associated with loss of Nissl substance and neuronal depletion in the pars compacta of the substantia nigra and the striatum, attributed to mitochondrial dysfunction [39]. Moreover, in zebrafish neurons, plastic particle exposure has been shown to cause mitochondrial impairment, which is magnified due to the low copy number of mitochondrial DNA and the expression of the mRNA of genes associated with the inhibition of mitochondrial fusion, activation of the mitochondrial division and mitophagy, and reduced copy number [40].
Exposure to microplastics and nanoplastics has also been associated with inflammatory reactions. In vitro studies using the human microglial HMC-3 cell line have shown that plastic particles alter the expression of immune cell clusters, while in vivo experiments in mice have demonstrated that exposure leads to changes in the expression of microglial differentiation markers, accompanied by activation of the NF-κB pathway and increased levels of pro-inflammatory cytokines in the hippocampus, cerebral cortex, and cerebellum [41]. Furthermore, in an Alzheimer’s disease mouse model (APP/PS1 double-transgenic mice), nanoplastic exposure exacerbates cognitive impairment and promotes neuroinflammation [42]. Moreover, administration of nanoplastics—but not microplastics—has been shown to induce overexpression of glial fibrillary acidic protein (GFAP) in the cornu ammonis 3 of the hippocampus [43]. In addition, only nanoplastics have been reported to cause a shift in quiescent microglia to a reactive phenotype in both the cerebral cortex and the CA3 region of the hippocampus, leading to the loss of Nissl bodies and neuronal injury [10,43].

3. Detrimental Consequences of Plastic Particle Exposure on the Production of Neurons Both in the Prenatal and Adult Life

During the embryonic and perinatal development of the CNS, neural stem cells produce astrocytes and oligodendrocytes as well as several types of neurons. In this specialized process, neural stem cells originate from neuroepithelial cells located in the neural tube [44]. Neurons are produced from prenatal life until early postnatal stages, with only a few neurogenetic regions remaining active in adulthood. In most adult mammals, neural stem cells are found in two brain regions: (i) the dentate gyrus in the hippocampus, where newly produced granule cells move a short distance from the subgranular cell layer to the granular cell layer, and (ii) the ventricular–subventricular area lining the lateral ventricles, where newly generated neurons migrate to the olfactory bulb via the rostral migratory stream. Neural stem cells, derived from embryonic radial glia, are involved in the generation of neurons, astrocytes, and oligodendrocytes [45,46,47].
As indicated above, a transplacental transfer of plastic particles to the fetal brain has been demonstrated following maternal exposure. Furthermore, previous studies have shown that plastic particles can cross the BBB and accumulate in neuroblasts, neurons, glial cells, and endothelial cells. Once located in the brain parenchyma, microplastics and nanoplastics exert deleterious effects in both invertebrate and vertebrate species. It has been revealed that plastic particles alter prenatal and adult neurogenesis (Figure 1). For instance, exposure of sea urchins to microplastic particles during embryonic and larval stages resulted in an important depletion of serotonergic sensory neurons in the apical organ of these echinoderms [48]. Similarly, in zebrafish, microplastic exposure during both embryonic and larval stages decreased the expression of two key neurodevelopmental genes: neurogenin1 (involved in specifying neuronal differentiation) and olig2 (involved in oligodendrocyte specification and differentiation). Additionally, the same study also reported a reduction in the number of PCNA (involved in the control of DNA replication)—positive cells and SOX2 (involved in maintaining stem cell pluripotency)—positive cells, indicating impaired proliferative activity of neural progenitors and a reduction in the neural stem cell pool [49].
On the other hand, the effects of microplastics and nanoplastics are also observed in the perinatal period. In proliferating neural stem cells cultured from a 5-day-old mouse subventricular zone and exposed to plastic particles, a substantial reduction in the number of phosphorylated histone H3 and 5-ethynyl-2′-deoxyuridine-reactive cells exists. The cytotoxic effects also affected the generation of neurons and oligodendrocytes [50]. Furthermore, perinatal exposure of mice to nanoplastics leads to neuronal depletion in the cornu ammonis regions 1 and 3, as well as impaired neurogenesis in the dentate gyrus. Interestingly, this effect was not seen after microplastic treatment. Despite that, both microplastics and nanoplastics modify the dendritic spines morphology in the cornus ammonis 1, but only microplastics reduce the spines density in this area of the hippocampus [43].
In another set of experiments, it was demonstrated that proliferating neuronal precursors are vulnerable to the effects of nanoplastic exposure. Consistent with this, in vitro studies have shown that the negative impact of plastic particles on the proliferative dynamics of C17.2 neuronal progenitor cells results from G1 cell cycle arrest. Additionally, nanoplastic treatment increased the expression of cell cycle inhibitors such as p21 and p27, while decreasing the expression of cyclin D. The study also reported that nanoplastic exposure reduced neural stem cell proliferation in the dentate gyrus of the hippocampus, leading to impaired neurogenesis in adult mice and decreased expression of the neural stem cell marker, nestin [28].
In the ventricular–subventricular area lining zone, a transient increase has been observed in the number of proliferating neural stem cells, proliferating neuroblasts, and doublecortin-positive cells [51]. The same study also reported that new interneurons migrate from the ventricular–subventricular zone to the olfactory bulb via the rostral migratory stream.

4. Plastic Particles Exposure and Neurogenesis in the Context of the BrdU-Labeling

Plastic particles are found in all ecosystems. These environmental pollutants can enter the animal body following three routes: skin (cosmetics and textiles), inhalation (exposure to textiles, synthetic rubber tires, and plastic covers), and ingestion (consumption of contaminated plants and animals, and products such as toothpaste, beer, and mineral water contained in plastic bottles) [52,53]. The accumulation of these pollutants in the CNS produces a myriad of negative effects, including apoptosis, senescence, and alterations in DNA methylation. The same cellular events are also produced due to BrdU exposure.

4.1. Plastic Particles and 5-Bromo-2′-Deoxyuridine Induce Apoptosis

Several studies have indicated the induction of apoptotic cellular events in nerve cells following plastic particle exposure. In this regard, it has been shown that exposure of adult zebrafish to 2 mg/L of plastic particles increases the expression of the apoptotic genes, caspase-3, caspase-9, and caspase-8, in the brain [54]. Furthermore, it was also revealed that mixed neuronal populations isolated from the prenatal mouse cortex present increased expression of cleaved caspase-3 protein after a two-day exposure to 100 mg/L of nanoplastics [55]. Similarly, exposure of cortical neuron cultures to 1 mg/L or 10 mg/L of plastic particles led to a significant increase in cleaved caspase-3-immunoreactive neurons [56]. In addition, exposure to polystyrene nanoparticles for 24 h or 48 h induces apoptosis in mouse Neuro-2a neuroblastoma cells and human HLA-G-positive choriocarcinoma cells. In the same study, a significant increase in apoptotic thalamic neurons was seen in C57BL mouse fetuses following maternal exposure to 1 mg/day of polystyrene nanoparticles administered via intragastric gavage for 17 consecutive days [57].
An important aspect to consider is the relationship between apoptosis and cell proliferation following plastic particle exposure. It has been reported that polystyrene microplastics can simultaneously induce both apoptotic events and cell proliferation. In other words, the activation of apoptosis may trigger a compensatory proliferative response in surrounding healthy cells to maintain tissue homeostasis [56]. In this context, it has been shown that pro-apoptotic caspases can activate the c-Jun N-terminal kinase signaling pathway, which promotes the proliferation of neighboring cells to replace those lost through apoptosis [58]. Similarly, it has been reported that embryonic brain damage induces the proliferation of neural precursors via an apoptosis-induced mechanism [59].
Since the introduction of the first monoclonal antibody against BrdU [60], several antibodies are commercially available [21,22]. This has led to the development of several immunocytochemical methods for detecting BrdU incorporated into replicating DNA. It has provided valuable insights into the cellular mechanisms involved in embryonic and perinatal development of the CNS. These procedures have been used to analyze various processes, including cell cycle kinetics, developmental timetables, and migration and cell lineage in a wide range of species, including mammals [61,62]. Despite these advantages, the possibility of false BrdU labeling or the incorrect interpretation of such labeling should be considered. In this context, the controversy is generated when BrdU labeling can be explained by processes unrelated to cell proliferation, i.e., when a neuroblast is undergoing an apoptotic cellular event.
During embryonic and perinatal development, the neuronal types that populate the nervous system are produced according to strict neurogenetic timetables. These times of neuron origin can be traced via the administration of BrdU [63,64]. In some cases, an excessive number of neurons is generated, leading to the formation of incorrect connections with their target cells. Subsequently, a proportion of these neurons undergo apoptosis as a homeostatic mechanism to adjust their final numbers [65,66]. Cell death is also a normal process in the adult neurogenetic regions, such as the dentate gyrus of the hippocampus and the ventricular–subventricular area, where a significant proportion of newly originated neurons undergo apoptosis before reaching maturity [67,68].
Neurons are fully differentiated cells. They are in the G0 phase of the cell cycle. However, several studies have reported that, in response to damage, neurons initiate DNA synthesis without completing cytokinesis [69,70]. In line with these findings, it has been indicated that homocysteine exposure in cultured rat cerebral cortex neurons induces apoptotic cellular events, triggers DNA synthesis, and leads to BrdU incorporation into neuronal nuclei [71]. On the other hand, it has been reported that, following cerebral hypoxia/ischemia, apoptotic neurons in the cornus ammonis 1 region of the hippocampus can reinitiate DNA synthesis and incorporate BrdU into their nuclei [72]. These findings reveal that, after injury, the incorporation of BrdU into a nucleus is not evidence of neurogenesis because BrdU labeling can also occur when nerve cells are undergoing an apoptotic cellular event. In addition to that, it has been reported that a single injection of BrdU at doses ranging from 100 to 300 µg/g leads to the activation of apoptotic events in the rat embryonic cerebellum [73,74].
Collectively, these studies suggest that, in the absence of appropriate controls, BrdU-labeling may not be an appropriate tool for assessing the effects of plastic particles exposure on neurogenesis.

4.2. Plastic Particles and 5-Bromo-2’-Deoxyuridine Induce Senescence

Senescence is a dynamic multistep physiological process through which the cell cycle is irreversibly arrested. This cellular event is a protective mechanism to maintain homeostasis and avoid the replication of aged or injured cells that are under certain stress conditions [75]. Cellular senescence can occur at any stage of life, from prenatal period to adulthood [76]. However, the accumulation of senescent cells has been related to several neurological diseases, including amyotrophic lateral sclerosis, Parkinson’s disease, and Alzheimer’s disease. In this context, the senescence in glial cells, neurons, and neural stem cells has been related to these neurodegenerative disorders [42].
The widespread use of plastic products has led to an alarming increase in microplastics and nanoplastics in the environment. These debris can cross biological barriers and induce premature senescence in various cell types and organs. For instance, Wang et al. [77] reported that plastic particles induce senescence in cardiomyocytes by causing mitochondrial oxidative stress and extrusion of mitochondrial DNA into the cytoplasm, which elicits a strong inflammatory response. Additionally, plastic debris promotes senescence of endothelial cells through oxidative stress, accompanied by increased expression of nicotinamide adenine dinucleotide phosphate oxidase and downregulation of sirtuin, a DAD+ dependent deacetylase that regulates several processes by modifying gene expression through histone deacetylation [78]. Plastic particles also promote premature senescence in two human lung-related cell lines (A549 and BEAS-2B) as well as in the mouse lung by deregulating the balance between intracellular reactive oxygen species and the antioxidant system of lung cells [79]. Additionally, senescence has been shown in the cultured alveolar epithelial cell line MLE12 and in rat lung cells following microplastic treatment, involving a molecular mechanism where circ_kif2cb promotes the expression of non-coding RNAs, the miR-346-3p [80]. On the other hand, plastic debris has also been revealed to induce senescence in cultured rat renal interstitial fibroblasts (NRK-49F cells), epithelial cells from human proximal tubular (HK-2 cells), and in nephrocytes of mice via the klotho/Wnt/β-catenin signaling pathway [81]. Moreover, senescence following exposure to plastic fragments has been reported in cultured C17.2 neuronal progenitor cells derived from the embryonic mouse cerebellum, as well as in proliferative neural stem cells located in the adult mouse hippocampus. In both cell types, microplastics increased the expression of the cell cycle arrest markers, p16, p21, and p27, while decreasing cyclin D expression. In both proliferating cell types, plastic particles produce cellular senescence by triggering cell cycle arrest at the G1 phase through mitochondrial dysfunction [28].
Based on the above-mentioned findings, it can be concluded that exposure to microplastics and nanoplastics induces senescence in various cell types. Interestingly, the incorporation of BrdU into DNA also triggers a wide range of detrimental alterations in the DNA double helix, including the induction of cellular senescence. In this context, studies have found that BrdU exposure induces senescence-like morphological changes in cultured embryonic fibroblasts as well as in immortalized human colon carcinoma cell lines (HCT116 and HCT116/80S14), even in the absence of functional p16, p21, and p53 [82]. In addition to these phenotypic alterations, BrdU treatment upregulates the expression of senescence-associated proteins such as p16, p21, p53, and the mortality marker mortalin. Similarly, BrdU exposure in neurosphere cultures derived from mice during the perinatal period has been shown to induce senescence in neural stem and progenitor cells, mediated by the activation of p53 and components of the retinoblastoma protein pathway [83].
Collectively, these findings demonstrate that both microplastics and BrdU induce cellular senescence. Notably, BrdU exposure has been shown to promote the expression of key senescence-associated proteins, including p16, p21, and p53 proteins that are similarly upregulated following exposure to microplastics and nanoplastics. Despite these parallels, the mechanism involved in the induction of senescence is not well understood. Some studies suggest that chromatin decondensation, driven by BrdU incorporation into scaffold or nuclear matrix attachment region sequences, may serve as an initial trigger for senescence [84]. Furthermore, two distinct mechanisms have been proposed to explain BrdU-induced senescence: (i) BrdU acts as a DNA intercalating molecule, preferentially inserting at adenine/thymidine-rich regions, thereby making genes with high A/T content particularly susceptible to its deleterious effects [85], and (ii) BrdU disrupts gene expression by targeting a highly conserved domain within the N-terminal tail of histone H2B, thereby altering nucleosome positioning and chromatin structure [86].
Current results have indicated that both plastic particles and BrdU promote cellular senescence and reinforce the evidence that this halopyrimidine analog may not be a reliable marker for identifying neuroblasts undergoing DNA synthesis.

4.3. Plastic Particles and 5-Bromo-2′-Deoxyuridine Alter DNA Methylation

In eukaryotes, methylation of DNA is a chemical modification in which methyl groups are added to the DNA molecule. Specifically, a methyl group is transferred to the 5-carbon position of a cytosine to form 5-methylcytosine. This chemical reaction is regulated by a family of enzymes known as DNA methyltransferases [87]. DNA methylation plays a crucial role as an epigenetic mechanism by regulating gene expression, primarily through the inhibition of transcription factors binding to DNA. This process is involved in several biological processes, including X-chromosome inactivation, chromatin structure regulation, neuronal activity, DNA repair, and the maintenance of cell identity [88].
Microplastics and nanoplastics can cross the nuclear envelope and directly interact with DNA, leading to damage. Previous studies have reported DNA degradation following microplastics exposure in mussels [89] and demersal fish species [90]. Additionally, evidence suggests that plastic particle exposure may alter DNA methylation patterns, although the direction and extent of these changes appear to be species-dependent [91]. For instance, DNA hypomethylation has been seen in zebrafish following microplastics exposure [92], with similar findings reported in mussels [93]. In contrast, an increase in DNA methylation has been reported in the blood cells of rats, with the degree of methylation rising in a dose-dependent manner after microplastics exposure [94]. A proposed mechanism suggests that DNA oxidation can promote DNA methylation through the polymerase ß-DNMTs 3b during base excision repair [95].
The halogenated nucleotide BrdU is known to induce DNA demethylation. The mechanisms by which BrdU affects DNA methylation are thought to resemble those of 5-aza-2′-deoxycytidine, a DNA demethylating agent. In this context, it has been proposed that BrdU produces DNA demethylation by reducing the expression of methyltransferases [96]. Although the precise mechanism remains unclear, the distinct chemical structure of BrdU compared to endogenous thymidine likely plays a key role. Specifically, this synthetic halogenated pyrimidine incorporates a bromine atom (Br) into replicating DNA when bromouracil is substituted for thymidine during DNA synthesis [18]. Moreover, the addition of exogenous BrdU can disrupt the cellular nucleotide pool. When BrdU levels are excessive, or when the ratio of deoxycytidine triphosphate (dCTP) to BrdU triphosphate decreases, the conversion of nucleotide triphosphates to deoxynucleotide triphosphates becomes inhibited. Under these conditions of nucleotide pool imbalance, BrdU can be incorporated into DNA opposite guanine, rather than its typical pairing with adenine [97]. Based on these findings, it is reasonable to assume that genes containing bromosubstituted DNA may be transcribed incorrectly into RNA, ultimately leading to the production of defective or altered proteins, including the DNA methyltransferases.
Another mechanism involved in DNA demethylation following BrdU exposure is related to nucleosome destabilization, which results from modifications in heterochromatin organization and gene expression [86]. These changes can create conditions in which BrdU is incorporated into CpG islands–DNA regions where a cytosine is followed by a guanine–substituting for cytidine and thereby leading to the loss of CpG methylation. Interestingly, other thymidine analogs, such as CldU and IdU, have also been shown to induce DNA demethylation [96].
Again, these results support the notion that BrdU labeling after plastics particle exposure may introduce artifacts that could lead to misinterpretation of experimental data. In the context of neurogenesis, an important issue is to have a confident identification of those cell precursors engaged in the S-phase of the cell cycle. BrdU tagging is widely used to evidence cell proliferation and neurogenesis in the developing and adult nervous system. Methodological problems with BrdU-labeling can be overcome through co-labeling BrdU with cell cycle markers (PCNA, mini-chromosome maintenance protein-2, Ki67 and phosphohistone-H3) or immature neuron markers (polysialylated neuronal cell adhesion molecule and doublecortin). Another approach is the use of intracranial injection of retroviral vectors to tag proliferating neuronal precursors [98,99].

5. Conclusions and Future Perspectives

This review outlines the detrimental effects of microplastic and nanoplastic exposure on the central nervous system, with a particular focus on their impact on neurogenesis during both prenatal and postnatal development. This is a critical concern, as plastic particles are capable of crossing both the placental barrier and the blood–brain barrier. This report also has implications for interpreting the effects of plastic debris exposure on neurogenesis when BrdU is used as a marker to identify neuron precursors engaged in the S-phase of the cell cycle. This exogenous nucleoside has yielded valuable insights into central nervous system development under various experimental approaches. Nevertheless, the impact of BrdU incorporation into DNA is often overlooked. This is an important issue when a single high dose or repeated doses of this thymidine analog are administered. The controversy arises when BrdU-positive neuroblasts can be attributed to processes unrelated to cell division, such as apoptosis, cellular senescence, or alterations in DNA methylation. Notably, these cellular events also occur following exposure to microplastics and nanoplastics. Therefore, data obtained using BrdU should be interpreted with caution, and appropriate controls are essential to ensure that BrdU labeling accurately reflects the fraction of neuroblasts engaged in DNA synthesis. Based on previous reports [73,74], it is proposed that, to label proliferating neuroblasts, a single BrdU pulse at a dose below 100 µg/g body weight should be administered.

Author Contributions

M.E. and J.M.C. were involved in the writing of this article. J.M.C. provided overall supervision, conceptualization, and reviewed the structure of the article. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
BrdU5-bromo-2’-deoxyuridine
CNSCentral Nervous System
BBBBlood–brain barrier

References

  1. Baby, M.G.; Gerritse, J.; Beltran-Sanahuja, A.; Wolter, H.; Rohais, S.; Romero-Sarmiento, M.-F. Aging of Plastics and Microplastics in the Environment: A Review on Influencing Factors, Quantification Methods, Challenges, and Future Perspectives. Environ. Sci. Pollut. Res. 2024, 32, 1009–1042. [Google Scholar] [CrossRef]
  2. Chamas, A.; Moon, H.; Zheng, J.; Qiu, Y.; Tabassum, T.; Jang, J.H.; Abu-Omar, M.; Scott, S.L.; Suh, S. Degradation Rates of Plastics in the Environment. ACS Sustain. Chem. Eng. 2020, 8, 3494–3511. [Google Scholar] [CrossRef]
  3. Usman, S.; Abdull Razis, A.F.; Shaari, K.; Azmai, M.N.A.; Saad, M.Z.; Mat Isa, N.; Nazarudin, M.F. The Burden of Microplastics Pollution and Contending Policies and Regulations. Int. J. Environ. Res. Public Health 2022, 19, 6773. [Google Scholar] [CrossRef]
  4. Geyer, R.; Jambeck, J.R.; Law, K.L. Production, Use, and Fate of All Plastics Ever Made. Sci. Adv. 2017, 3, e1700782. [Google Scholar] [CrossRef]
  5. Fernandes, R.; Martins, R.; Marques, C. Critical Review on Microplastics Characterisation in Aquatic Environments: Recent Trends in the Last 10 Years. Anal. Methods 2025, 17, 1415–1427. [Google Scholar] [CrossRef]
  6. Andrady, A.L. Microplastics in the marine environment. Mar. Pollut. Bull. 2011, 62, 1596–1605. [Google Scholar] [CrossRef]
  7. Wang, F.; Zhang, M.; Sha, W.; Wang, Y.; Hao, H.; Dou, Y.; Li, Y. Sorption Behavior and Mechanisms of Organic Contaminants to Nano and Microplastics. Molecules 2020, 25, 1827. [Google Scholar] [CrossRef] [PubMed]
  8. Zhang, L.; He, Y.; Jiang, L.; Shi, Y.; Hao, L.; Huang, L.; Lyu, M.; Wang, S. Plastic Additives as a New Threat to the Global Environment: Research Status, Remediation Strategies and Perspectives. Environ. Res. 2024, 263, 120007. [Google Scholar] [CrossRef]
  9. Gigault, J.; Ter Halle, A.; Baudrimont, M.; Pascal, P.-Y.; Gauffre, F.; Phi, T.-L.; El Hadri, H.; Grassl, B.; Reynaud, S. Current Opinion: What Is a Nanoplastic? Environ. Pollut. 2018, 235, 1030–1034. [Google Scholar] [CrossRef] [PubMed]
  10. Shan, S.; Zhang, Y.; Zhao, H.; Zeng, T.; Zhao, X. Polystyrene Nanoplastics Penetrate across the Blood-Brain Barrier and Induce Activation of Microglia in the Brain of Mice. Chemosphere 2022, 298, 134261. [Google Scholar] [CrossRef]
  11. Kopatz, V.; Wen, K.; Kovács, T.; Keimowitz, A.S.; Pichler, V.; Widder, J.; Vethaak, A.D.; Hollóczki, O.; Kenner, L. Micro- and Nanoplastics Breach the Blood–Brain Barrier (BBB): Biomolecular Corona’s Role Revealed. Nanomaterials 2023, 13, 1404. [Google Scholar] [CrossRef]
  12. Li, J.; Zhang, K.; Zhang, H. Adsorption of Antibiotics on Microplastics. Environ. Pollut. 2018, 237, 460–467. [Google Scholar] [CrossRef]
  13. Gerdes, Z.; Ogonowski, M.; Nybom, I.; Ek, C.; Adolfsson-Erici, M.; Barth, A.; Gorokhova, E. Microplastic-Mediated Transport of PCBs? A Depuration Study with Daphnia Magna. PLoS ONE 2019, 14, e0205378. [Google Scholar] [CrossRef] [PubMed]
  14. Toussaint, B.; Raffael, B.; Angers-Loustau, A.; Gilliland, D.; Kestens, V.; Petrillo, M.; Rio-Echevarria, I.M.; Van den Eede, G. Review of Micro- and Nanoplastic Contamination in the Food Chain. Food Addit. Contam. Part A 2019, 36, 639–673. [Google Scholar] [CrossRef] [PubMed]
  15. Barceló, D.; Picó, Y.; Alfarhan, A.H. Microplastics: Detection in Human Samples, Cell Line Studies, and Health Impacts. Environ. Toxicol. Pharmacol. 2023, 101, 104204. [Google Scholar] [CrossRef]
  16. Nihart, A.J.; Garcia, M.A.; El Hayek, E.; Liu, R.; Olewine, M.; Kingston, J.D.; Castillo, E.F.; Gullapalli, R.R.; Howard, T.; Bleske, B.; et al. Bioaccumulation of Microplastics in Decedent Human Brains. Nat. Med. 2025, 31, 1114–1119. [Google Scholar] [CrossRef]
  17. Ghosh, A.; Gorain, B. Mechanistic Insight of Neurodegeneration due to Micro/Nano-Plastic-Induced Gut Dysbiosis. Arch. Toxicol. 2025, 99, 83–101. [Google Scholar] [CrossRef] [PubMed]
  18. Duque, A.; Rakic, P. Different Effects of Bromodeoxyuridine and [3H]Thymidine Incorporation into DNA on Cell Proliferation, Position, and Fate. J. Neurosci. 2011, 31, 15205–15217. [Google Scholar] [CrossRef]
  19. Martí, J.; Wills, K.V.; Ghetti, B.; Bayer, S.A. A Combined Immunohistochemical and Autoradiographic Method to Detect Midbrain Dopaminergic Neurons and Determine Their Time of Origin. Brain Res. Protoc. 2002, 9, 197–205. [Google Scholar] [CrossRef]
  20. Martí, J.; Santa-Cruz, M.C.; Bayer, S.A.; Ghetti, B.; Hervás, J.P. Purkinje Cell Age-Distribution in Fissures and in Foliar Crowns: A Comparative Study in the Weaver Cerebellum. Brain Struct. Funct. 2007, 212, 347–357. [Google Scholar] [CrossRef]
  21. Solius, G.M.; Maltsev, D.I.; Belousov, V.V.; Podgorny, O.V. Recent Advances in Nucleotide Analogue-Based Techniques for Tracking Dividing Stem Cells: An Overview. J. Biol. Chem. 2021, 297, 101345. [Google Scholar] [CrossRef] [PubMed]
  22. Martí-Clúa, J. 5-Bromo-2′-Deoxyuridine Labeling: Historical Perspectives, Factors Influencing the Detection, Toxicity, and Its Implications in the Neurogenesis. Neural Regen. Res. 2024, 19, 302–308. [Google Scholar] [CrossRef]
  23. Lehner, B.; Sandner, B.; Marschallinger, J.; Lehner, C.; Furtner, T.; Couillard-Despres, S.; Rivera, F.J.; Brockhoff, G.; Bauer, H.-C.; Weidner, N.; et al. The Dark Side of BrdU in Neural Stem Cell Biology: Detrimental Effects on Cell Cycle, Differentiation and Survival. Cell Tissue Res. 2011, 345, 313–328. [Google Scholar] [CrossRef]
  24. Li, Z.; Zhu, S.; Liu, Q.; Wei, J.; Jin, Y.; Wang, X.; Zhang, L. Polystyrene Microplastics Cause Cardiac Fibrosis by Activating Wnt/β-Catenin Signaling Pathway and Promoting Cardiomyocyte Apoptosis in Rats. Environ. Pollut. 2020, 265, 115025. [Google Scholar] [CrossRef]
  25. Dong, C.-D.; Chen, C.-W.; Chen, Y.-C.; Chen, H.-H.; Lee, J.-S.; Lin, C.-H. Polystyrene Microplastic Particles: In Vitro Pulmonary Toxicity Assessment. J. Hazard. Mater. 2020, 385, 121575. [Google Scholar] [CrossRef] [PubMed]
  26. Zhang, Y.; Tian, L.; Chen, J.; Liu, X.; Li, K.; Liu, H.; Lai, W.; Shi, Y.; Lin, B.; Xi, Z. Selective Bioaccumulation of Polystyrene Nanoplastics in Fetal Rat Brain and Damage to Myelin Development. Ecotoxicol. Environ. Saf. 2024, 278, 116393. [Google Scholar] [CrossRef]
  27. Wang, S.; Han, Q.; Wei, Z.; Wang, Y.; Xie, J.; Chen, M. Polystyrene Microplastics Affect Learning and Memory in Mice by Inducing Oxidative Stress and Decreasing the Level of Acetylcholine. Food Chem. Toxicol. 2022, 162, 112904. [Google Scholar] [CrossRef] [PubMed]
  28. Yang, S.; Lee, S.; Lee, Y.; Cho, J.-H.; Kim, S.H.; Ha, E.-S.; Jung, Y.-S.; Chung, H.Y.; Kim, M.-S.; Kim, H.S.; et al. Cationic Nanoplastic Causes Mitochondrial Dysfunction in Neural Progenitor Cells and Impairs Hippocampal Neurogenesis. Free Radic. Biol. Med. 2023, 208, 194–210. [Google Scholar] [CrossRef]
  29. Nishimura, Y.; Kanda, Y.; Sone, H.; Aoyama, H. Oxidative Stress as a Common Key Event in Developmental Neurotoxicity. Oxidative Med. Cell. Longev. 2021, 2021, 6685204. [Google Scholar] [CrossRef]
  30. Usman, S.; Abdull Razis, A.F.; Shaari, K.; Amal, M.N.A.; Saad, M.Z.; Mat Isa, N.; Nazarudin, M.F. Polystyrene Microplastics Exposure: An Insight into Multiple Organ Histological Alterations, Oxidative Stress and Neurotoxicity in Javanese Medaka Fish (Oryzias Javanicus Bleeker, 1854). Int. J. Environ. Res. Public Health 2021, 18, 9449. [Google Scholar] [CrossRef]
  31. Chen, Y.; Liu, X.; Leng, Y.; Wang, J. Defense Responses in Earthworms (Eisenia fetida) Exposed to Low-Density Polyethylene Microplastics in Soils. Ecotoxicol. Environ. Saf. 2020, 187, 109788. [Google Scholar] [CrossRef] [PubMed]
  32. Rodríguez-Ithurralde, D.; Maruri, A.; Rodríguez, X. Motor Neurone Acetylcholinesterase Release Precedes Neurotoxicity Caused by Systemic Administration of Excitatory Amino Acids and Strychnine. J. Neurol. Sci. 1998, 160, S80–S86. [Google Scholar] [CrossRef] [PubMed]
  33. Ding, J.; Zhang, S.; Razanajatovo, R.M.; Zou, H.; Zhu, W. Accumulation, Tissue Distribution, and Biochemical Effects of Polystyrene Microplastics in the Freshwater Fish Red Tilapia (Oreochromis niloticus). Environ. Pollut. 2018, 238, 1–9. [Google Scholar] [CrossRef]
  34. Torres-Ruiz, M.; de Alba González, M.; Morales, M.; Martin-Folgar, R.; González, M.C.; Cañas-Portilla, A.I.; De la Vieja, A. Neurotoxicity and Endocrine Disruption Caused by Polystyrene Nanoparticles in Zebrafish Embryo. Sci. Total Environ. 2023, 874, 162406. [Google Scholar] [CrossRef]
  35. Liu, X.; Zhao, Y.; Dou, J.; Hou, Q.; Cheng, J.; Jiang, X. Bioeffects of Inhaled Nanoplastics on Neurons and Alteration of Animal Behaviors through Deposition in the Brain. Nano Lett. 2022, 22, 1091–1099. [Google Scholar] [CrossRef] [PubMed]
  36. Jeong, C.-B.; Won, E.-J.; Kang, H.-M.; Lee, M.-C.; Hwang, D.-S.; Hwang, U.-K.; Zhou, B.; Souissi, S.; Lee, S.-J.; Lee, J.-S. Microplastic Size-Dependent Toxicity, Oxidative Stress Induction, and P-JNK and P-P38 Activation in the Monogonont Rotifer (Brachionus koreanus). Environ. Sci. Technol. 2016, 50, 8849–8857. [Google Scholar] [CrossRef]
  37. Trevisan, R.; Voy, C.; Chen, S.; Di, R.T. Nanoplastics Decrease the Toxicity of a Complex PAH Mixture but Impair Mitochondrial Energy Production in Developing Zebrafish. Environ. Sci. Technol. 2019, 53, 8405–8415. [Google Scholar] [CrossRef] [PubMed]
  38. Huang, Y.; Liang, B.; Li, Z.; Zhong, Y.; Wang, B.; Zhang, B.; Du, J.; Ye, R.; Xian, H.; Min, W.; et al. Polystyrene Nanoplastic Exposure Induces Excessive Mitophagy by Activating AMPK/ULK1 Pathway in Differentiated SH-SY5Y Cells and Dopaminergic Neurons in Vivo. Part. Fibre Toxicol. 2023, 20, 44. [Google Scholar] [CrossRef]
  39. Liang, B.; Huang, Y.; Zhong, Y.; Li, Z.; Ye, R.; Wang, B.; Zhang, B.; Meng, H.; Lin, X.; Du, J.; et al. Brain Single-Nucleus Transcriptomics Highlights That Polystyrene Nanoplastics Potentially Induce Parkinson’s Disease-like Neurodegeneration by Causing Energy Metabolism Disorders in Mice. J. Hazard. Mater. 2022, 430, 128459. [Google Scholar] [CrossRef]
  40. Zhang, C.; Li, Y.; Yu, H.; Ye, L.; Tian, L.; Zhang, X.; Wang, C.; Li, P.; Ji, H.; Gao, Q.; et al. Nanoplastics Promote Arsenic-Induced ROS Accumulation, Mitochondrial Damage and Disturbances in Neurotransmitter Metabolism of Zebrafish (Danio rerio). Sci. Total Environ. 2023, 863, 161005. [Google Scholar] [CrossRef]
  41. Kwon, W.; Kim, D.; Kim, H.-Y.; Jeong, S.W.; Lee, S.-G.; Kim, H.-C.; Lee, Y.-J.; Kwon, M.K.; Hwang, J.-S.; Han, J.E.; et al. Microglial Phagocytosis of Polystyrene Microplastics Results in Immune Alteration and Apoptosis in Vitro and in Vivo. Sci. Total Environ. 2022, 807, 150817. [Google Scholar] [CrossRef]
  42. Wang, G.; Lin, Y.; Shen, H. Exposure to Polystyrene Microplastics Promotes the Progression of Cognitive Impairment in Alzheimer’s Disease: Association with Induction of Microglial Pyroptosis. Mol. Neurobiol. 2023, 61, 900–907. [Google Scholar] [CrossRef] [PubMed]
  43. Wang, C.; Lin, K.; Zhang, Z.; Pan, Y.; Miao, Q.; Han, X.; Zhang, Z.; Zhu, P.; Yang, J.; Peng, Y.; et al. Adolescent Exposure to Micro/Nanoplastics Induces Cognitive Impairments in Mice with Neuronal Morphological Damage and Multi-Omic Alterations. Environ. Int. 2025, 197, 109323. [Google Scholar] [CrossRef]
  44. Zhang, R.; Quan, H.; Wang, Y.; Luo, F. Neurogenesis in Primates versus Rodents and the Value of Non-Human Primate Models. Natl. Sci. Rev. 2023, 10, nwad248. [Google Scholar] [CrossRef]
  45. Obernier, K.; Alvarez-Buylla, A. Neural Stem Cells: Origin, Heterogeneity and Regulation in the Adult Mammalian Brain. Development 2019, 146, dev156059. [Google Scholar] [CrossRef]
  46. Bonfanti, L.; La Rosa, C.; Ghibaudi, M.; Sherwood, C.C. Adult Neurogenesis and “Immature” Neurons in Mammals: An Evolutionary Trade-off in Plasticity? Brain Struct. Funct. 2024, 229, 1775–1793. [Google Scholar] [CrossRef]
  47. Cebrian-Silla, A.; Nascimento, M.A.; Mancia, W.; Gonzalez-Granero, S.; Romero-Rodriguez, R.; Obernier, K.; Steffen, D.M.; Lim, D.A.; Garcia-Verdugo, J.M.; Alvarez-Buylla, A. Neural Stem Cell Relay from B1 to B2 Cells in the Adult Mouse Ventricular-Subventricular Zone. Cell Rep. 2025, 44, 115264. [Google Scholar] [CrossRef]
  48. Rendell-Bhatti, F.; Paganos, P.; Pouch, A.; Mitchell, C.; D’Aniello, S.; Godley, B.J.; Pazdro, K.; Arnone, M.I.; Jimenez-Guri, E. Developmental Toxicity of Plastic Leachates on the Sea Urchin Paracentrotus Lividus. Environ. Pollut. 2020, 269, 115744. [Google Scholar] [CrossRef]
  49. Santos, D.; Luzio, A.; Félix, L.; Cabecinha, E.; Bellas, J.; Monteiro, S.M. Microplastics and Copper Induce Apoptosis, Alter Neurocircuits, and Cause Behavioral Changes in Zebrafish (Danio rerio) Brain. Ecotoxicol. Environ. Saf. 2022, 242, 113926. [Google Scholar] [CrossRef] [PubMed]
  50. Park, K.-Y.; Kim, M.S.; Oh, N. Cytotoxicity of Amine-Modified Polystyrene MPs and NPs on Neural Stem Cells Cultured from Mouse Subventricular Zone. Heliyon 2024, 10, e30518. [Google Scholar] [CrossRef] [PubMed]
  51. Prosperi, G.; Marchetti, N.; D’Elia, A.; Massari, R.; Giusto, M.; Pietrodangelo, A.; Rossi, T.; Nucara, A.; Scavizzi, F.; Strimpakos, G.; et al. Inhalation of Nanoplastics in the Mouse Model: Tissue Bio-Distribution and Effects on the Olfactory System. Sci. Total Environ. 2025, 968, 178853. [Google Scholar] [CrossRef]
  52. Prüst, M.; Meijer, J.; Westerink, R.H.S. The Plastic Brain: Neurotoxicity of Micro- and Nanoplastics. Part. Fibre Toxicol. 2020, 17, 24. [Google Scholar] [CrossRef]
  53. Winiarska, E.; Jutel, M.; Zemelka-Wiacek, M. The Potential Impact of Nano- and Microplastics on Human Health: Understanding Human Health Risks. Environ. Res. 2024, 251, 118535. [Google Scholar] [CrossRef]
  54. Santos, D.; Luzio, A.; Bellas, J.; Monteiro, S.M. Microplastics- and Copper-Induced Changes in Neurogenesis and DNA Methyltransferases in the Early Life Stages of Zebrafish. Chem.-Biol. Interact. 2022, 363, 110021. [Google Scholar] [CrossRef]
  55. Jung, B.-K.; Han, S.-W.; Park, S.-H.; Bae, J.-S.; Choi, J.; Ryu, K.-Y. Neurotoxic Potential of Polystyrene Nanoplastics in Primary Cells Originating from Mouse Brain. NeuroToxicology 2020, 81, 189–196. [Google Scholar] [CrossRef]
  56. So, Y.H.; Shin, H.S.; Lee, S.H.; Moon, H.J.; Jang, H.J.; Lee, E.-H.; Jung, E.-M. Maternal Exposure to Polystyrene Microplastics Impairs Social Behavior in Mouse Offspring with a Potential Neurotoxicity. NeuroToxicology 2023, 99, 206–216. [Google Scholar] [CrossRef]
  57. Yang, D.; Zhu, J.; Zhou, X.; Pan, D.; Nan, S.; Yin, R.; Lei, Q.; Ma, N.; Zhu, H.; Chen, J.; et al. Polystyrene Micro- and Nano-Particle Coexposure Injures Fetal Thalamus by Inducing ROS-Mediated Cell Apoptosis. Environ. Int. 2022, 166, 107362. [Google Scholar] [CrossRef] [PubMed]
  58. Ryoo, H.D.; Bergmann, A. The Role of Apoptosis-Induced Proliferation for Regeneration and Cancer. Cold Spring Harb. Perspect. Biol. 2012, 4, a008797. [Google Scholar] [CrossRef] [PubMed]
  59. Petrenko, V.; Mihhailova, J.; Salmon, P.; Kiss, J.Z. Apoptotic Neurons Induce Proliferative Responses of Progenitor Cells in the Postnatal Neocortex. Exp. Neurol. 2015, 273, 126–137. [Google Scholar] [CrossRef] [PubMed]
  60. Gratzner, H. Monoclonal Antibody to 5-Bromo- and 5-Iododeoxyuridine: A New Reagent for Detection of DNA Replication. Science 1982, 218, 474–475. [Google Scholar] [CrossRef] [PubMed]
  61. Nowakowski, R.S.; Lewin, S.B.; Miller, M.W. Bromodeoxyuridine Immunohistochemical Determination of the Lengths of the Cell Cycle and the DNA-Synthetic Phase for an Anatomically Defined Population. J. Neurocytol. 1989, 18, 311–318. [Google Scholar] [CrossRef] [PubMed]
  62. Altman, J.; Bayer, S.A. Development of the Cerebellar System in Relation to Its Evolution, Structure, and Functions; CRC Press: Boca Raton, FL, USA, 1997. [Google Scholar]
  63. Martí-Clúa, J. Developmental Timetables and Gradients of Neurogenesis in Cerebellar Purkinje Cells and Deep Glutamatergic Neurons: A Comparative Study between the Mouse and the Rat. Anat. Rec. 2021, 304, 2856–2864. [Google Scholar] [CrossRef] [PubMed]
  64. Martí-Clua, J. Times of Neuron Origin and Neurogenetic Gradients in Mice Purkinje Cells and Deep Cerebellar Nuclei Neurons during the Development of the Cerebellum. A Review. Tissue Cell 2022, 78, 101897. [Google Scholar] [CrossRef]
  65. Dori, I.; Bekiari, C.; Grivas, I.; Tsingotjidou, A.; Papadopoulos, G.C. Birth and Death of Neurons in the Developing and Mature Mammalian Brain. Int. J. Dev. Biol. 2022, 66, 9–22. [Google Scholar] [CrossRef]
  66. Svandova, E.; Lesot, H.; Sharpe, P.; Matalova, E. Making the Head: Caspases in Life and Death. Front. Cell Dev. Biol. 2023, 10, 1075751. [Google Scholar] [CrossRef]
  67. Ryu, J.R.; Hong, C.J.; Kim, J.Y.; Kim, E.-K.; Sun, W.; Yu, S.-W. Control of Adult Neurogenesis by Programmed Cell Death in the Mammalian Brain. Mol. Brain 2016, 9, 43. [Google Scholar] [CrossRef] [PubMed]
  68. Cameron, H.A.; Mckay, R.D.G. Adult Neurogenesis Produces a Large Pool of New Granule Cells in the Dentate Gyrus. J. Comp. Neurol. 2001, 435, 406–417. [Google Scholar] [CrossRef]
  69. Nandakumar, S.; Rozich, E.; Buttitta, L. Cell Cycle Re-Entry in the Nervous System: From Polyploidy to Neurodegeneration. Front. Cell Dev. Biol. 2021, 9, 698661. [Google Scholar] [CrossRef]
  70. Pavulraj, S.; Stout, R.W.; Paulsen, D.B.; Chowdhury, S.I. Live Triple Gene-Deleted Pseudorabies Virus-Vectored Subunit PCV2b and CSFV Vaccine Undergoes an Abortive Replication Cycle in the TG Neurons Following Latency Reactivation. Viruses 2023, 15, 473. [Google Scholar] [CrossRef]
  71. Ye, W.; Blain, S.W. S Phase Entry Causes Homocysteine-Induced Death While Ataxia Telangiectasia and Rad3 Related Protein Functions Anti-Apoptotically to Protect Neurons. Brain 2010, 133, 2295–2312. [Google Scholar] [CrossRef] [PubMed]
  72. Kuan, C.-Y. Hypoxia-Ischemia Induces DNA Synthesis without Cell Proliferation in Dying Neurons in Adult Rodent Brain. J. Neurosci. 2004, 24, 10763–10772. [Google Scholar] [CrossRef] [PubMed]
  73. Taupin, P. BrdU Immunohistochemistry for Studying Adult Neurogenesis: Paradigms, Pitfalls, Limitations, and Validation. Brain Res. Rev. 2007, 53, 198–214. [Google Scholar] [CrossRef]
  74. Rodríguez-Vázquez, L.; Martí, J. Administration of 5-Bromo-2′-Deoxyuridine Interferes with Neuroblast Proliferation and Promotes Apoptotic Cell Death in the Rat Cerebellar Neuroepithelium. J. Comp. Neurol. 2020, 529, 1081–1096. [Google Scholar] [CrossRef]
  75. Calcinotto, A.; Kohli, J.; Zagato, E.; Pellegrini, L.; Demaria, M.; Alimonti, A. Cellular Senescence: Aging, Cancer, and Injury. Physiol. Rev. 2019, 99, 1047–1078. [Google Scholar] [CrossRef]
  76. Rhinn, M.; Ritschka, B.; Keyes, W.M. Cellular Senescence in Development, Regeneration and Disease. Development 2019, 146, dev151837. [Google Scholar] [CrossRef]
  77. Wang, Y.; Kuca, K.; You, L.; Nepovimova, E.; Heger, Z.; Valko, M.; Adam, V.; Wu, Q.; Jomova, K. The Role of Cellular Senescence in Neurodegenerative Diseases. Arch. Toxicol. 2024, 98, 2393–2408. [Google Scholar] [CrossRef]
  78. Shiwakoti, S.; Ko, J.-Y.; Gong, D.-S.; Dhakal, B.; Lee, J.M.; Adhikari, R.; Gwak, Y.; Park, S.-H.; Choi, I.J.; Schini-Kerth, V.B.; et al. Effects of Polystyrene Nanoplastics on Endothelium Senescence and Its Underlying Mechanism. Environ. Int. 2022, 164, 107248. [Google Scholar] [CrossRef]
  79. Jin, W.; Zhang, W.; Tang, H.; Wang, P.; Yan, Z.; Liu, S.; Qiu, J.; Chen, H.; Wang, L.; Wang, R.; et al. Microplastics Exposure Causes the Senescence of Human Lung Epithelial Cells and Mouse Lungs by Inducing ROS Signaling. Environ. Int. 2024, 185, 108489. [Google Scholar] [CrossRef]
  80. Luo, H.; Xiao, T.; Sun, X.; Song, Y.; Shi, W.; Lu, K.; Chen, D.; Sun, C.; Bian, Q. The Regulation of CircRNA_kif26b on Alveolar Epithelial Cell Senescence via MiR-346-3p Is Involved in Microplastics-Induced Lung Injuries. Sci. Total Environ. 2023, 882, 163512. [Google Scholar] [CrossRef] [PubMed]
  81. Pan, C.; Wang, X.; Fan, Z.; Mao, W.; Shi, Y.; Wu, Y.; Liu, T.; Xu, Z.; Wang, H.; Chen, H. Polystyrene Microplastics Facilitate Renal Fibrosis through Accelerating Tubular Epithelial Cell Senescence. Food Chem. Toxicol. 2024, 191, 114888. [Google Scholar] [CrossRef] [PubMed]
  82. Eriko, M.; Nakabayashi, K.; Suzuki, T.; Kaul, S.C.; Ogino, H.; Fujii, M.; Mitsui, Y.; Ayusawa, D. 5-Bromodeoxyuridine Induces Senescence-like Phenomena in Mammalian Cells regardless of Cell Type or Species. J. Biochem. 1999, 126, 1052–1059. [Google Scholar] [CrossRef]
  83. Ross, H.H.; Levkoff, L.H.; Marshall, G.P.; Caldeira, M.; Steindler, D.A.; Reynolds, B.A.; Laywell, E.D. Bromodeoxyuridine Induces Senescence in Neural Stem and Progenitor Cells. STEM CELLS 2008, 26, 3218–3227. [Google Scholar] [CrossRef]
  84. Satou, W.; Suzuki, T.; Noguchi, T.; Ogino, H.; Fujii, M.; Ayusawa, D. AT-Hook Proteins Stimulate Induction of Senescence Markers Triggered by 5-Bromodeoxyuridine in Mammalian Cells. Exp. Gerontol. 2003, 39, 173–179. [Google Scholar] [CrossRef] [PubMed]
  85. Guerrero, A. Nucleosome Disruption by 5-Bromodeoxyuridine Leads to Senescence. FEBS J. 2022, 290, 684–687. [Google Scholar] [CrossRef]
  86. En, A.; Watanabe, K.; Ayusawa, D.; Fujii, M. The Key Role of a Basic Domain of Histone H2B N-Terminal Tail in the Action of 5-Bromodeoxyuridine to Induce Cellular Senescence. FEBS J. 2022, 290, 692–711. [Google Scholar] [CrossRef]
  87. Mattei, A.L.; Bailly, N.; Meissner, A. DNA Methylation: A Historical Perspective. Trends Genet. 2022, 38, 676–707. [Google Scholar] [CrossRef] [PubMed]
  88. Chera, A.; Stancu-Cretu, M.; Zabet, N.R.; Bucur, O. Shedding Light on DNA Methylation and Its Clinical Implications: The Impact of Long-Read-Based Nanopore Technology. Epigenetics Chromatin 2024, 17, 39. [Google Scholar] [CrossRef] [PubMed]
  89. Masiá, P.; Ardura, A.; García-Vázquez, E. Virgin Polystyrene Microparticles Exposure Leads to Changes in Gills DNA and Physical Condition in the Mediterranean Mussel Mytilus Galloprovincialis. Animals 2021, 11, 2317. [Google Scholar] [CrossRef]
  90. Menéndez, D.; Blanco-Fernandez, C.; Machado-Schiaffino, G.; Ardura, A.; Garcia-Vazquez, E. High Microplastics Concentration in Liver Is Negatively Associated with Condition Factor in the Benguela Hake Merluccius Polli. Ecotoxicol. Environ. Saf. 2023, 262, 115135. [Google Scholar] [CrossRef]
  91. Wright, S.L.; Thompson, R.C.; Galloway, T.S. The Physical Impacts of Microplastics on Marine Organisms: A Review. Environ. Pollut. 2013, 178, 483–492. [Google Scholar] [CrossRef] [PubMed]
  92. Im, J.; Eom, H.-J.; Choi, J. Effect of Early-Life Exposure of Polystyrene Microplastics on Behavior and DNA Methylation in Later Life Stage of Zebrafish. Arch. Environ. Contam. Toxicol. 2022, 82, 558–568. [Google Scholar] [CrossRef]
  93. Ortiz-Moriano, M.P.; Masiá, P.; Acle, S.; Ardura, A.; Garcia-Vazquez, E.; Machado-Schiaffino, G. Changes in Global Methylation Patterns of Mytilus Galloprovincialis Exposed to Microplastics. Aquat. Toxicol. 2024, 276, 107115. [Google Scholar] [CrossRef] [PubMed]
  94. Farag, A.A.; Youssef, H.S.; Sliem, R.E.; El, B.; Nabil, N.; Mouktar, M.M.; Marei, Y.M.; Ismail, N.S.; Radwaan, S.E.; Badr, A.M.; et al. Hematological Consequences of Polyethylene Microplastics Toxicity in Male Rats: Oxidative Stress, Genetic, and Epigenetic Links. Toxicology 2023, 492, 153545. [Google Scholar] [CrossRef] [PubMed]
  95. Jiang, Z.; Lai, Y.; Beaver, J.M.; Tsegay, P.S.; Zhao, M.-L.; Horton, J.K.; Zamora, M.; Rein, H.L.; Miralles, F.; Shaver, M.; et al. Oxidative DNA Damage Modulates DNA Methylation Pattern in Human Breast Cancer 1 (BRCA1) Gene via the Crosstalk between DNA Polymerase β and a de Novo DNA Methyltransferase. Cells 2020, 9, 225. [Google Scholar] [CrossRef] [PubMed]
  96. Schneider, L.; d’Adda di Fagagna, F. Neural Stem Cells Exposed to BrdU Lose Their Global DNA Methylation and Undergo Astrocytic Differentiation. Nucleic Acids Res. 2012, 40, 5332–5342. [Google Scholar] [CrossRef] [PubMed]
  97. Morris, S.M.; Domon, O.E.; McGarrity, L.J.; Kodelo, R.L.; Casciano, D.A. Effect of Bromodeoxyuridine on the Proliferation and Growth of Ethyl Methanesulfonate-Exposed P3 Cells: Relationship to the Induction of Sister-Chromatid Exchanges. Cell Biol. Toxicol. 1992, 8, 75–87. [Google Scholar] [CrossRef]
  98. Zhao, X.; van Praag, H. Steps towards Standardized Quantification of Adult Neurogenesis. Nat. Commun. 2020, 11, 4275. [Google Scholar] [CrossRef]
  99. Sorrells, S.F.; Paredes, M.F.; Zhang, Z.; Kang, G.; Pastor-Alonso, O.; Biagiotti, S.; Page, C.E.; Sandoval, K.; Knox, A.; Connolly, A.; et al. Positive Controls in Adults and Children Support That Very Few, If Any, New Neurons Are Born in the Adult Human Hippocampus. J. Neurosci. 2021, 41, 2554–2565. [Google Scholar] [CrossRef]
Figure 1. Plastic fragmentation generates microplastics and nanoplastics. These pollutants exert deleterious effects on both embryonic and adult neurogenesis. NSC: neural stem cells. INN: immature newborn neurons.
Figure 1. Plastic fragmentation generates microplastics and nanoplastics. These pollutants exert deleterious effects on both embryonic and adult neurogenesis. NSC: neural stem cells. INN: immature newborn neurons.
Ijms 26 07845 g001
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

Encinas, M.; Martí Clúa, J. Effects of Microplastics and Nanoplastics Exposure on Neurogenesis: Are Thymidine Analogs a Good Option to Study Such Effects? Int. J. Mol. Sci. 2025, 26, 7845. https://doi.org/10.3390/ijms26167845

AMA Style

Encinas M, Martí Clúa J. Effects of Microplastics and Nanoplastics Exposure on Neurogenesis: Are Thymidine Analogs a Good Option to Study Such Effects? International Journal of Molecular Sciences. 2025; 26(16):7845. https://doi.org/10.3390/ijms26167845

Chicago/Turabian Style

Encinas, Mercè, and Joaquin Martí Clúa. 2025. "Effects of Microplastics and Nanoplastics Exposure on Neurogenesis: Are Thymidine Analogs a Good Option to Study Such Effects?" International Journal of Molecular Sciences 26, no. 16: 7845. https://doi.org/10.3390/ijms26167845

APA Style

Encinas, M., & Martí Clúa, J. (2025). Effects of Microplastics and Nanoplastics Exposure on Neurogenesis: Are Thymidine Analogs a Good Option to Study Such Effects? International Journal of Molecular Sciences, 26(16), 7845. https://doi.org/10.3390/ijms26167845

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