Next Article in Journal
UVC Box: An Effective Way to Quickly Decontaminate Healthcare Facilities’ Wheelchairs
Previous Article in Journal
Oxytocin Modulation in Mindfulness-Based Pain Management for Chronic Pain
Previous Article in Special Issue
Postprandial Dysmetabolism and Its Medical Implications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Emerging Role of Micro- and Nanoplastics in Vascular Diseases

by
Seung Eun Lee
1,*,
Hyun Kyung Yoon
2,
Do Yun Kim
2,
Taek Seung Jeong
2 and
Yong Seek Park
1,*
1
Department of Microbiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
2
Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
*
Authors to whom correspondence should be addressed.
Life 2024, 14(2), 255; https://doi.org/10.3390/life14020255
Submission received: 30 December 2023 / Revised: 6 February 2024 / Accepted: 13 February 2024 / Published: 16 February 2024
(This article belongs to the Special Issue New Advances in Endocrinology and Metabolic Diseases: 2nd Edition)

Abstract

:
Vascular diseases are the leading causes of death worldwide, and they are attributable to multiple pathologies, such as atherosclerosis, diabetes, and chronic obstructive pulmonary disease. Exposure to various environmental contaminants is associated with the development of various diseases, including vascular diseases. Among environmental contaminants, micro- and nanoplastics have gained attention as global environmental risk factors that threaten human health. Recently, extensive research has been conducted on the effects of micro- and nanoplastics on various human diseases, including vascular diseases. In this review, we highlight the effects of micro- and nanoplastics on vascular diseases.

1. Introduction

Vascular diseases are the leading causes of death worldwide. These include any condition that affects the circulatory system or blood vessels, ranging from diseases of the veins, lymph vessels, and arteries to blood disorders that affect circulation. Typically, vascular diseases begin with an injury to the endothelial wall, and adaptive and maladaptive remodeling of the vascular wall represent key disease processes [1,2]. In several vascular diseases, the central pathophysiological events include the stimulation of pro-inflammatory signaling pathways, expression of cytokines/chemokines, and promotion of oxidative stress [3,4,5].
The most important risk factors for vascular diseases include hypertension, diabetes, obesity, hyperlipidemia, and smoking. Vascular diseases have also been associated with environmental contaminants, one of the most important causes of mortality and disability worldwide [6]. Chowdhury et al. reported that exposure to environmentally toxic metals is associated with an increased risk of cardiovascular and coronary heart diseases [7].
Ambient air pollution is the most important global environmental risk factor contributing to human death [8,9]. A recent study evaluated the contribution of long-term exposure to air pollutants (PM2.5, CO, and NO2) to the pathogenesis of peripheral arterial occlusive disease [10]. Specifically, accumulating evidence has revealed that exposure to environmental contaminants, particularly fine particulate matter, is a major contributor to cardiovascular mortality and morbidity [11,12]. Therefore, in this narrative review, we focus on vascular diseases related to micro- and nanoplastic (MNP) exposure.

2. MNPs as Environmental Contaminants

Plastics have infiltrated several aspects of modern life, and they have been utilized in packaging, health care, construction, aviation, logistics, and clothing [13]. The growth in plastic usage can be attributed to its beneficial properties, such as low cost, strength, lightweight, durability, corrosion resistance, electrical insulation, and high thermal properties [14]. Global plastic production doubled from 2000 to 2019 to 460 million tons, and this trend is expected to continue [15]. Supply chain systems encompassing the production, utilization, management, and disposal of fossil-based plastics pose serious environmental challenges [16,17]. Among plastic wastes, biodegradable plastics have garnered public attention as an encouraging substitute for conventional non-degradable plastic polymers that generate serious plastic pollution, given that biodegradable plastics are susceptible to microbial biodegradation and are thus environmentally harmless [18,19,20,21,22,23].

2.1. MNPs

In 2004, the risks associated with MNPs were introduced, and their potential adverse effects on living organisms have recently attracted considerable attention [24,25,26]. After deposition in the environment, plastic waste is degraded into small fragments by exposure to ultraviolet (UV) radiation, oxidation, biodegradation, disintegration, and mechanical erosion, eventually resulting in MNPs [27,28]. Microplastics (MPs) are small artificial polymer particles (<5 mm) that are not the end products of plastic waste, given that they degrade into nanoplastics (NPs) (<1000 nm) [29,30] (Figure 1).
Currently, two types of MPs are recognized. Primary MPs contain plastic fragments or particles ≥ 5.0 mm in size before entering the environment. They originate from plastic pellets in the pre-production stage of the manufacturing industries, including plastic powder, plastic resin flakes, scrubbers, commercial cleaning abrasives, or tow used to generate plastic products [31]. Meanwhile, secondary MPs are generated by the degradation of larger plastic objects through natural weathering processes, such as atmospheric deposition, aquaculture, land-based sources, air transportation, and textile production, after entering the environment [32,33]. MPs are potential contaminants of emerging concern owing to their abundance in global environments, including terrestrial, atmospheric, and aquatic environments [34,35] (Figure 2).

2.2. Environmental Pollution with MNPs

Despite widespread awareness that the excessive usage and mismanagement of plastics are responsible for global environmental problems, global plastic waste production has more than doubled from 2000 to 2019 to 359 million tons. Furthermore, approximately 6.1 million tons of plastic waste is dumped in aquatic environments, with approximately 1.7 million tons presumed to have entered the oceans [36]. Under this ambitious scenario, Borrelle et al. have predicted that 20–53 million metric tons of plastic emissions/year will enter aquatic ecosystems by 2030 [37]. The presence of MPs in aquatic environments has been reported elsewhere as a global concern [38]. MPs have been found in abiotic and biotic areas of the ocean, including beaches [39,40], surface waters [41,42], sediment [43,44,45,46], fish [47,48,49], and shellfish [50,51].
Soil (such as home gardens, greenhouses, farmlands, and floodplain soils) is an enormous reservoir of accumulated MPs. Sewage sludge is among the several sources of MPs in the soil. Sewage-irrigated farmlands contain a significantly higher distribution of MPs compared with non-sewage-irrigated agricultural fields (5190 particles/kg vs. 2030 particles/kg) [52].
Furthermore, atmospheric pollution caused by airborne MPs is a growing concern worldwide. In recent years, airborne MPs have been discovered worldwide [53,54,55,56,57,58], with concentrations ranging from 0.01 MP m−3 (West Pacific Ocean) [59] to 5650 MP m−3 (Beijing, China) [60]. Airborne MPs primarily originate from textile fragmentation and decomposition processes, whereas nonfibrous particles predominantly arise from the degradation of packaging materials.

2.3. MNPs in the Food Chains and Food

The notable properties of plastics, such as strength, variability, flexibility, lightness, and persistence, make them suitable for packaging food and other goods [61]. As the use of plastics pervades various segments of the food chain, the instances of consumer exposure to MPs have increased considerably. MPs enter the human food chain mainly through contaminated foods and can affect human health [62]. Recent studies have reported an abundance of MPs in terrestrial food chains, including chicken stomachs (5.1 particles/g) and feces (105 particles/g) [26,63] and sheep feces (1000 particles/g) [64]. In addition, the presence of MPs has been documented in water, honey, beer, seafood, and sugar [65]. Overall, MNPs have the potential to be absorbed into the body through various stages of the food chain, circulate, and accumulate, and are thereby expected to have an adverse effect on the circulatory system. For instance, in vivo murine models exhibited that intestinal-delivered MPs reach the bloodstream within 15 min after ingestion, mainly accumulating in the liver [66,67].

3. MNP Toxicity in the Vascular System

3.1. Toxicities in Other Animal Cells

Florance et al. showed that anionic surfactant-stabilized NPs potently stimulate dysfunctional lipid homeostasis in brain- and tissue-specific macrophages in mice. The authors suggested that NPs were potent stimuli for foam cell formation and lipotoxicity, leading to the pathogenesis of atherosclerosis and posing a major risk to animal and human health [68].
A recent study examined the effects of MPs on endothelial and immune cells associated with vascular inflammation in mice. MP administration augmented the expression of adhesion molecules and cytokines in the aorta, indicating that MPs are a new environmental risk factor attributable to endothelial inflammation [69].
Furthermore, the in vivo effects of long-term MP exposure and potential exposure-induced kidney, liver, and cardiovascular toxicities have been investigated. Exposure to MPs aggravates vascular lesions and organ injuries, particularly in the heart, liver, and kidney. Based on RNA sequencing data, long-term exposure to MPs may be related to alterations in the biological features and global gene expression patterns in mice, providing novel insights into the biological toxicity of MPs [70]. N6-methyladenosine (m6A) modifications in ncRNAs have been identified as manipulating factors in cardiovascular diseases [71,72]. Zhang et al. demonstrated the N6-methyladenosine (m6A) modification profile of non-coding RNA (ncRNAs) in myocardial tissues exposed to MP using RNA sequencing (RNA-seq) and methylated RNA immunoprecipitation sequencing (MeRIP-seq). Differentially expressed ncRNAs were altered by MP exposure and were enriched in several cardiotoxicity-related signaling pathways. This study suggests that m6A modification of ncRNAs is involved in MP-induced myocardial damage [73].
Shiwakoti et al. have demonstrated the potential effects of NPs on the cardiovascular system at the cellular level. NP-exposed endothelial cells (ECs) could promote the acquisition of senescence markers; senescence-associated β-galactosidase activity; and p16, p21, and p53 protein expression, resulting in suppressed proliferation. Their findings support the effects and underlying mechanisms of NPs on the cardiovascular system and may provide pharmacological targets for the prevention of cardiovascular diseases [74]. Dhakal et al. demonstrated the role of sodium glucose co-transporter (SGLT2) in NP-induced endothelial senescence and dysfunction of porcine coronary arteries. NPs significantly upregulated SGLT2 and senescence-associated-β-galactosidase (SA-β-gal) activity, whereas suppression of SGLT2 significantly diminished SA-β-gal activity in porcine coronary artery endothelial cells (PCAEC). This study highlights the association of premature endothelial senescence by NPs with SGLT2 and provides a potential therapeutic target to prevent environmental pollutant-elicited cardiovascular diseases mediated by endothelial senescence and dysfunction [75]. Basini et al. revealed the potential effects of NPs on endothelial aortic cells (AOCs) by evaluating the ability of NPs to specifically interact with endothelial cells using fluorescence microscopy. NPs co-localize with AOC, increase AOC metabolic activity and vascular endothelial growth factor (VEGF) production, and disrupt the redox status [76].
Exposure to MPs can induce cerebral hemorrhage and promote the production of microthrombi and loss of Purkinje cells in chicken brain tissue. Moreover, a previous study elucidated the detailed mechanism underlying subsequent brain injury from the perspective of MP-induced physical injury (cerebral hemorrhage), thereby comprehensively clarifying MP exposure-induced neurotoxicity [77]. According to a study by Zhang et al., MPs can induce cardiotoxicity via ROS-driven NF-κB-NLRP3-GSDMD and the AMPK-PGC-1α axis in chickens. MP exposure triggered myocardial inflammation and pyroptosis through NF-κB-NLRP3-GSDMD axis- and AMPK-PGC-1α axis-mediated mitochondrial and energy metabolism dysfunction. The authors provided a better understanding of the potential role of MPs in MP exposure-induced cardiotoxicity [78].
Zhang et al. observed the effect of MPs on myocardial development in the primary cardiomyocytes of chicken embryos. Histopathological observations showed that MPs had a loose and irregular myocardial arrangement, large cell gaps, and broken myocardial fiber bundles. These data suggest that MP-induced myocardial dysplasia in birds is principally attributed to the ER stress-mediated autophagic pathway [79].
Moreover, the effects of MPs on cardiac tissues and the mechanisms of pyroptosis and oxidative stress during cardiomyocyte injury have been reported in Wistar rats. Administration of MPs can lead to apoptosis and pyroptosis, as evidenced by the stimulation of interleukin (IL)-1β and IL-18 expression. These findings illustrate that pyroptosis plays a crucial role in MP-induced cardiotoxicity, which may help clarify the mechanism underlying MP exposure-induced cardiac dysfunction [80]. Roshanzadeh et al. showed that MPs can reduce the contractility of cardiomyocytes by influencing intracellular Ca2+ level, electrophysiological activity, mitochondrial membrane potential, and cell metabolism of neonatal rat ventricular cardiomyocytes (NRVMs) [81].
Zhao et al. revealed that consumption of MPs promotes adiposity, hyperglycemia, and changes in the gut microbiome in mice. As the major finding, the authors noted that MP ingestion could promote increased obesity in mice in a dose- and size-dependent manner. These results suggest that the ingestion of MPs stimulates a cardiometabolic disease phenotype and thus may be considered a new risk factor for cardiovascular disease [82]. Wang et al. demonstrated that NPs in the blood and aorta of ApoE−/− mice aggravate the artery stiffness and enhance atherosclerotic plaque formation. NP exposure stimulates phagocytosis by M1 macrophages, interrupts lipid metabolism, and increases long-chain acyl carnitines (LCACs). This study provides new insights into the mechanisms underlying MNP-induced cardiovascular toxicity [83]. In vivo studies by Wang et al. demonstrated that NPs can induce vascular injury, structural damage to vascular endothelial cells, and inflammatory responses. Structural damage and dysfunction of vascular endothelial cells upon exposure to NPs occur through the activation of the Janus kinase 1/signal transducer and activator of the transcription-3/tissue factor (JAK1/STAT3/TF) signaling pathway [84]. Zhang et al. evaluated the cardiotoxicity induced by respiratory exposure to NPs, including the accumulation of NPs, histological observation, assessment of cardiac function, detection of biomarkers, and transcriptomic studies, and showed that NP exposure induced acute cardiotoxicity in a dose- and time-dependent manner [85]. Shan et al. found that NPs could accumulate in the brains of mice, promptly activate microglia, and cause damage to neurons. Moreover, NPs could accumulate in human cerebral microvascular endothelial cell (hCMEC/D3) cells and stimulate nuclear factor kappa-B (NF-κB) activation, ROS generation, necroptosis, and inhibition of transendothelial electrical resistance (TEER) and occludin expression. These findings suggest that NPs stimulate the inflammatory response, oxidative stress, and necroptosis in hCMEC/D3 cells, which may also contribute to the impairment of tight junctions (TJs) in the blood–brain barrier (BBB) [86].
A recent study has provided insights into the role of MPs as potential mediators of unbridled oxidative imbalance and dyslipidemia in mammals. Oral administration of MPs lowered high-density lipoprotein cholesterol and elevated low-density lipoprotein cholesterol levels. In addition, the authors observed that MP-exposed rats exhibited sex-based differences in atherogenic indices. These data indicate that dietary MPs may lead to dyslipidemia, and male rats have a high cardiovascular risk [87].
The effects of inhaled MNPs on the cardiovascular system have been previously reported in rats. Cardiovascular function was assessed by measuring mean arterial pressure (MAP), wire myography of the aorta and uterine artery, and pressure myography of the radial artery. MNP inhalation induces various adverse cardiovascular dysfunctions such as increased blood pressure and diminished uterine vascular dilation. These data suggest that the inhalation of MNP aerosols leads to alteration of inflammatory, cardiovascular, and endocrine activities [88].
Li et al. revealed that MPs could induce cardiomyocyte apoptosis and activate Wnt/β-catenin signaling via oxidative stress induction, leading to heart fibrosis and cardiac dysfunction in rats. These findings provide novel evidence for a potential mechanism of MP-induced cardiovascular toxicity [89]. Yan et al. demonstrated that MP exposure elicited mild vascular calcification (VC) in normal murine and aggravated VC in vitamin D3 plus nicotine (VDN)-treated murine. Their work revealed that the mechanism of vascular injury by MPs may involve the induction of intestinal mucosal barrier dysfunction and an inflammatory response in the body. These observations present a theoretical basis for the risk assessment of MP-induced cardiovascular diseases [90].
The potential toxic effects of MNP exposure were estimated in zebrafish embryos at various developmental stages. Exposure to MNPs impairs intact development of the caudal vein plexus, leading to prematurity via pathological angiogenesis. These findings indicate that exposure to MNPs can lead to developmental disorders with substantial growth impairment and peripheral microcirculation dysfunction, and thus provide new insights into the use of plastics [91]. Sun et al. found that NPs could induce pericardial edema, restrain sub-intestinal angiogenesis, lead to endothelial cell dysfunction, and simultaneously obstruct CO and blood flow velocity in zebrafish embryos. These findings suggest that NPs can elicit cardiovascular toxicity in zebrafish embryos, thereby providing essential information for the safety assessment of NPs [92].
Pitt et al. investigated the potential toxicity of NPs during the development of zebrafish (Danio rerio). The authors observed that NPs accumulated in the yolk sac as early as 24 h post-fertilization (hpf), migrated to the gallbladder, gastrointestinal tract, pancreas, liver, heart, and brain throughout development (48–120 hpf), and diminished during the depuration phase (120–168 hpf) in all organs. Notably, NPs were localized in the pericardium only at 1 and 10 ppm; however, a significant decrease in heart rate was observed even when larvae were exposed to 0.1 ppm NP, indicating that NPs can induce cardiotoxicity even at a low concentration. Collectively, these data demonstrated that NPs may produce organ toxicity specific to their developmental distribution patterns [93]. A recent study demonstrated that exposure to NPs can prompt size- and dose-dependent effects on neurobehavior and potential hepatic, cardiovascular, gastrointestinal, epigenetic, and metabolic dysfunction, as determined by transcriptomic analysis of NP-exposed zebrafish [94]. Dai et al. evaluated the significant developmental toxicity of NPs in zebrafish embryos. Developmental impairments were observed in zebrafish embryos following exposure to various NP concentrations. Additionally, NP exposure led to vascular malformation by modulating VEGFA/VEGFR pathway-correlated gene expression during the early developmental stages of zebrafish. Thus, their findings demonstrated the toxicological effects of NPs on the development of the cardiovascular system [95]. Santos et al. investigated vasotoxicity, developmental toxicity, cytotoxicity, and behavioral impairments in zebrafish induced by NPs exposure. The authors assessed the cardiotoxicity, neurotoxicity, teratogenic effects, and larval morphological changes caused by NPs using the zebrafish embryo–larval toxicity test (ZELT). This study showed the harmful effects of NPs on the early developmental stages of fish, highlighting their environmental risk [96].
Wu et al. demonstrated the adverse effects of NPs on cardiac apoptosis and inflammation in carp, mediated by an oxidative stress-induced Th1/Th2 imbalance and activation of the IGFBP3/p53/ACHE signaling pathway. The authors provided a theoretical basis for toxicological research on MNPs in aquatic animal hearts [97].

3.2. Toxicities in Human Cells

In humans, the routes of MNP exposure include ingestion of plastic-contaminated food and water or inhalation of plastic-contaminated air [54,55]. Consequently, the digestive and respiratory systems are the primary sites of contact for MNPs. MNPs produce systemic toxicity by penetrating the cell membranes and causing cellular internalization.
Rotchell et al. examined digested human saphenous vein tissue samples using μFTIR spectroscopy and reported the first evidence of MP contamination in human vascular tissues [98]. Sun et al. qualitatively and quantitatively identified the presence of various types of MPs in the human endometrium using laser direct infrared (LDIR) spectroscopy [99]. These studies provide a prospective direction for future biological and environmental studies.
Lee et al. reported that MP exposure is a noteworthy risk factor for endothelial dysfunction and vascular malformations. Prolonged MP exposure can lead to reduced cell viability associated with the stimulation of autophagy/necrosis. Thus, the authors provided considerable evidence regarding the possible threat posed by MPs to human vascular system homeostasis [100].
A recent study has explored the interaction between low levels of MNPs and human umbilical vein endothelial cells (HUVECs) as a model of vascular endothelial cells. In this study, no remarkable changes in inflammation, autophagy, reactive oxygen species (ROS) levels, lactate dehydrogenase release, or adhesion molecule expression at exposure concentrations ranging from 5 to 25 μg/mL were detected. However, cell viability declined at extremely high exposure (100 μg/mL). These findings contribute to further risk assessments of MNPs in human health, including the vascular endothelium [101]. Moreover, NP-stimulated vasculature permeability is primarily biophysical/biochemical in nature and is not correlated with cytotoxic events, including ROS production, apoptosis, and autophagy [102]. Lu et al. demonstrated that smaller NPs (100 nm) result in cell membrane damage, autophagosome formation, and autophagic flux blockage in HUVECs. This study provided new insights into the potential effects of NPs on human vascular endothelial cells, which is a crucial contribution to the health risk assessment of NPs in the vascular system [103].
MPs can induce endothelial cell (EC) dysfunction by impairing mitochondrial function and upregulating pro-inflammatory cytokines, thereby triggering apoptosis. In addition, MPs induce atherosclerotic plaque formation and vascular inflammatory responses in vivo. Moreover, correlation analysis between transcriptomics/metabolites and cell injury has been reported. These findings indicate that MPs prompt abnormal transcriptomic and metabolomic responses in ECs both in vitro and in vivo, and these results may be utilized in the development of effective preventive strategies against MP damage [104]. Additionally, Fu et al. revealed that NPs induce oxidative stress and damage the mitochondria membrane potential in HUVECs. NPs dysregulate mitochondrial dynamics, function-related gene expression, and replication. Results showed that NPs were cytotoxic in HUVECs, as evidenced by reduced cell viability, diminished mitochondrial membrane potential, and augmented ROS generation, which provides direct evidence of the potential risks of NPs [105].
Chen et al. introduced an optically assisted thrombus platform to reveal interactions between MPs and the vascular system. According to the on-chip results, MP invasion decreased fibrin binding to platelets (p-value < 0.0001) and showed a high risk of thrombus shedding into the circulation compared to that in normal thrombus. Accordingly, they demonstrated the mechanism of thrombus invasion mediated by MPs using a microfluidic platform, suggesting a potential method for further exploration of MP-related health concerns [106].
Zhou et al. utilized a human pluripotent stem cell-derived three-dimensional cardiac organoid (CO) model to explore the adverse effects of MP exposure on the human heart in vitro. MPs augmented the inflammatory response, oxidative stress, collagen accumulation, and apoptosis, which are associated with increased expression of the cardiac-specific markers MYL2, MYL4, and CX43 in COs [107].
MPs have been shown to induce oxidative stress by reducing antioxidant expression in human EA.hy926 vascular endothelial cells. Exposure to MPs resulted in vascular barrier dysfunction and altered apoptotic cytotoxicity. These data provide insights into the potential toxicity risks of MPs in the human circulatory system [108].
Florance et al. reported that NPs induce lipid droplet accumulation and acute mitochondrial oxidative stress in humans. Accumulated lipid droplets are further delivered and accumulate in lysosomes, leading to impaired lysosomal clearance. The authors suggested that exposure to NPs is a potent stimulus for dysregulated macrophage foam cell formation and lipid metabolism, a characteristic feature observed during atherosclerosis, thereby posing a severe risk to human health [109].
Plastic particles such as MNPs enter the body through multiple pathways involving dermal areas, inhalation, and ingestion, and are transported via the bloodstream. Therefore, understanding the correlation between pollutants such as MNPs and blood cells might have significant implications for the pathophysiology of various human diseases. Recent studies revealed that aggregation and distribution of MNPs in blood are associated with the bioavailability, inflammatory, and immune responses of multiple cells such as monocytes, leukocytes, erythrocytes, and endothelial cells in the human body [110,111,112,113,114].
Recently, MNPs have emerged as a serious hazard in the environmental and food safety fields, causing cellular damage and affecting cell survival, death, apoptosis, inflammation, and various immune responses. Overall, this review focused on discussing the association between MNP exposure and vascular diseases (summarized in Table 1 and Table 2 and Figure 3).

4. Conclusions

Plastic production worldwide has increased 20-fold compared to its initial value, owing to its ease of use, cost-effectiveness, and durability. The most commonly distributed plastics in the environment are polyvinyl chloride, polyethylene, polypropylene, polyethylene terephthalate, and polystyrene [115]. Global concerns about plastic-induced environmental pollution have increased substantially. Plastic waste is exposed to the environment and forms MPs through mechanical abrasion, ultraviolet (UV) irradiation, and biological weathering. MPs have been found in every component of nature, including rivers, lakes, oceans, soil, atmosphere, and the Antarctic Ocean [116]. MPs can be attributed to multiple sources, including primary (synthetic materials) and secondary sources (derived from the breakdown of larger plastic particles). MNPs have been shown to negatively affect animal growth, reproduction, and metabolism through several mechanisms, including ingestion and bioaccumulation. Furthermore, MPs can induce oxidative stress, DNA damage, histopathological damage, intestinal microflora dysbiosis, neurotoxicity, genotoxicity, reproductive toxicity, and metabolic disorders, which are primarily governed by the MP type, size, shape, and dosage.
Overall, this review provides a comprehensive overview of the current knowledge regarding the effects of MNPs on the structure and function of the vascular system and their impact on the pathophysiology of vascular disorders. A better understanding of the roles of MNPs will broaden our understanding of cellular systems.

Author Contributions

Conceptualization, S.E.L. and Y.S.P.; writing—original draft preparation, S.E.L.; writing—review and editing, S.E.L., H.K.Y., D.Y.K., T.S.J. and Y.S.P.; funding acquisition, Y.S.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Research Foundation of Korea (NRF) (grant number NRF-2020R1A2C1011704) and the Ministry of Health & Welfare, Republic of Korea (grant number: HV22C0160).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This study was supported by the National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIP) (Grant No.: NRF-2020R1A2C1011704) and the Korean Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI) funded by the Ministry of Health & Welfare, Republic of Korea (grant number: HV22C0160).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Lyle, A.N.; Taylor, W.R. The pathophysiological basis of vascular disease. Lab. Invest. 2019, 99, 284–289. [Google Scholar] [CrossRef] [PubMed]
  2. Tesfamariam, B.; DeFelice, A.F. Endothelial injury in the initiation and progression of vascular disorders. Vascul. Pharmacol. 2007, 46, 229–237. [Google Scholar] [CrossRef] [PubMed]
  3. Abdellatif, M.; Bugger, H.; Kroemer, G.; Sedej, S. NAD(+) and Vascular Dysfunction: From Mechanisms to Therapeutic Opportunities. J. Lipid Atheroscler. 2022, 11, 111–132. [Google Scholar] [CrossRef] [PubMed]
  4. Jeong, S.J.; Oh, G.T. Unbalanced Redox With Autophagy in Cardiovascular Disease. J. Lipid Atheroscler. 2023, 12, 132–151. [Google Scholar] [CrossRef] [PubMed]
  5. Woo, S.H.; Kim, D.Y.; Choi, J.H. Roles of Vascular Smooth Muscle Cells in Atherosclerotic Calcification. J. Lipid Atheroscler. 2023, 12, 106–118. [Google Scholar] [CrossRef] [PubMed]
  6. Serra, R.; Abramo, A.; Ielapi, N.; Procopio, S.; Marino, P. Environmental Pollution and Peripheral Artery Disease. Risk Manag. Healthc. Policy 2021, 14, 2181–2190. [Google Scholar] [CrossRef] [PubMed]
  7. Chowdhury, R.; Ramond, A.; O’Keeffe, L.M.; Shahzad, S.; Kunutsor, S.K.; Muka, T.; Gregson, J.; Willeit, P.; Warnakula, S.; Khan, H.; et al. Environmental toxic metal contaminants and risk of cardiovascular disease: Systematic review and meta-analysis. BMJ 2018, 362, k3310. [Google Scholar] [CrossRef]
  8. Landrigan, P.J.; Fuller, R.; Acosta, N.J.; Adeyi, O.; Arnold, R.; Baldé, A.B.; Bertollini, R.; Bose-O’Reilly, S.; Boufford, J.I.; Breysse, P.N. The Lancet Commission on pollution and health. Lancet 2018, 391, 462–512. [Google Scholar] [CrossRef]
  9. Burnett, R.; Chen, H.; Szyszkowicz, M.; Fann, N.; Hubbell, B.; Pope Iii, C.A.; Apte, J.S.; Brauer, M.; Cohen, A.; Weichenthal, S. Global estimates of mortality associated with long-term exposure to outdoor fine particulate matter. Proc. Natl. Acad. Sci. USA 2018, 115, 9592–9597. [Google Scholar] [CrossRef]
  10. Liao, S.H.; Chiu, C.S.; Jang, L.; Hu, S.Y.; How, C.K.; Hsieh, V.C.R.; Hsieh, M.S. Long-Term Exposures to Air Pollutants and Risk of Peripheral Arterial Occlusive Disease: A Nationwide Cohort Study in Taiwan. Front. Cardiovasc. Med. 2022, 9, 796423. [Google Scholar] [CrossRef]
  11. Konduracka, E.; Rostoff, P. Links between chronic exposure to outdoor air pollution and cardiovascular diseases: A review. Environ. Chem. Lett. 2022, 20, 2971–2988. [Google Scholar] [CrossRef]
  12. Zhu, X.; Wang, C.; Duan, X.; Liang, B.; Genbo Xu, E.; Huang, Z. Micro- and nanoplastics: A new cardiovascular risk factor? Environ. Int. 2023, 171, 107662. [Google Scholar] [CrossRef] [PubMed]
  13. Horton, A.A. Plastic pollution: When do we know enough? J. Hazard. Mater. 2022, 422, 126885. [Google Scholar] [CrossRef] [PubMed]
  14. Thompson, R.C.; Moore, C.J.; vom Saal, F.S.; Swan, S.H. Plastics, the environment and human health: Current consensus and future trends. Philos. Trans. R. Soc. Lond. B. Biol. Sci. 2009, 364, 2153–2166. [Google Scholar] [CrossRef] [PubMed]
  15. Ritchie, H.; Samborska, V.; Roser, M. Plastic Pollution. Our World Data 2023. Available online: https://ourworldindata.org/plastic-pollution (accessed on 1 December 2023).
  16. Batel, A.; Linti, F.; Scherer, M.; Erdinger, L.; Braunbeck, T. Transfer of benzo[a]pyrene from microplastics to Artemia nauplii and further to zebrafish via a trophic food web experiment: CYP1A induction and visual tracking of persistent organic pollutants. Environ. Toxicol. Chem. 2016, 35, 1656–1666. [Google Scholar] [CrossRef]
  17. Schirinzi, G.F.; Perez-Pomeda, I.; Sanchis, J.; Rossini, C.; Farre, M.; Barcelo, D. Cytotoxic effects of commonly used nanomaterials and microplastics on cerebral and epithelial human cells. Environ. Res. 2017, 159, 579–587. [Google Scholar] [CrossRef] [PubMed]
  18. Rujnic-Sokele, M.; Pilipovic, A. Challenges and opportunities of biodegradable plastics: A mini review. Waste Manag. Res. 2017, 35, 132–140. [Google Scholar] [CrossRef] [PubMed]
  19. Qin, M.; Chen, C.Y.; Song, B.; Shen, M.C.; Cao, W.C.; Yang, H.L.; Zeng, G.M.; Gong, J.L. A review of biodegradable plastics to biodegradable microplastics: Another ecological threat to soil environments? J. Clean. Prod. 2021, 312, 127816. [Google Scholar] [CrossRef]
  20. Shen, M.C.; Song, B.; Zeng, G.M.; Zhang, Y.X.; Huang, W.; Wen, X.F.; Tang, W.W. Are biodegradable plastics a promising solution to solve the global plastic pollution? Environ. Pollut. 2020, 263, 114469. [Google Scholar] [CrossRef]
  21. Lee, H.; Shim, J.E.; Park, I.H.; Choo, K.S.; Yeo, M.K. Physical and biomimetic treatment methods to reduce microplastic waste accumulation. Mol. Cell Toxicol. 2023, 19, 13–25. [Google Scholar] [CrossRef]
  22. Kim, J.; Rhee, J.S. Biochemical and physiological responses of the water flea Moina macrocopa to microplastics: A multigenerational study. Mol. Cell. Toxicol. 2021, 17, 523–532. [Google Scholar] [CrossRef]
  23. Lee, S.E.; Yi, Y.; Moon, S.; Yoon, H.; Park, Y.S. Impact of Micro- and Nanoplastics on Mitochondria. Metabolites 2022, 12, 897. [Google Scholar] [CrossRef] [PubMed]
  24. Lebordais, M.; Gutierrez-Villagomez, J.M.; Gigault, J.; Baudrimont, M.; Langlois, V.S. Molecular impacts of dietary exposure to nanoplastics combined with arsenic in Canadian oysters (Crassostrea virginica) and bioaccumulation comparison with Caribbean oysters (Isognomon alatus). Chemosphere 2021, 277, 130331. [Google Scholar] [CrossRef] [PubMed]
  25. Ricciardi, M.; Pironti, C.; Motta, O.; Miele, Y.; Proto, A.; Montano, L. Microplastics in the Aquatic Environment: Occurrence, Persistence, Analysis, and Human Exposure. Water 2021, 13, 973. [Google Scholar] [CrossRef]
  26. Zolotova, N.; Kosyreva, A.; Dzhalilova, D.; Fokichev, N.; Makarova, O. Harmful effects of the microplastic pollution on animal health: A literature review. PeerJ 2022, 10, e13503. [Google Scholar] [CrossRef] [PubMed]
  27. Uchida, K.; Hagita, R.; Hayashi, T.; Tokai, T. Distribution of small plastic fragments floating in the western Pacific Ocean from 2000 to 2001. Fish. Sci. 2016, 82, 969–974. [Google Scholar] [CrossRef]
  28. MacLeo, M.; Arp, H.P.H.; Tekman, M.B.; Jahnke, A. The global threat from plastic pollution. Science 2021, 373, 61–65. [Google Scholar] [CrossRef]
  29. Karlsson, T.M.; Arneborg, L.; Brostrom, G.; Almroth, B.C.; Gipperth, L.; Hassellov, M. The unaccountability case of plastic pellet pollution. Mar. Pollut. Bull. 2018, 129, 52–60. [Google Scholar] [CrossRef]
  30. 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]
  31. Lambert, S.; Sinclair, C.; Boxall, A. Occurrence, degradation, and effect of polymer-based materials in the environment. Rev. Environ. Contam. Toxicol. 2014, 227, 1–53. [Google Scholar] [CrossRef]
  32. Lamichhane, G.; Acharya, A.; Marahatha, R.; Modi, B.; Paudel, R.; Adhikari, A.; Raut, B.K.; Aryal, S.; Parajuli, N. Microplastics in environment: Global concern, challenges, and controlling measures. Int. J. Environ. Sci. Technol. 2023, 20, 4673–4694. [Google Scholar] [CrossRef]
  33. Akdogan, Z.; Guven, B. Microplastics in the environment: A critical review of current understanding and identification of future research needs. Environ. Pollut. 2019, 254, 113011. [Google Scholar] [CrossRef]
  34. Lambert, S.; Wagner, M. Freshwater Microplastics: Emerging Environmental Contaminants? In The Handbook of Environmental Chemistry; 1 online resource (XIV, 303 pages 330 illustrations, 321 illustrations in color; Springer: Berlin/Heidelberg, Germany, 2018. [Google Scholar] [CrossRef]
  35. Miri, S.; Saini, R.; Davoodi, S.M.; Pulicharla, R.; Brar, S.K.; Magdouli, S. Biodegradation of microplastics: Better late than never. Chemosphere 2022, 286, 131670. [Google Scholar] [CrossRef]
  36. OECD. Plastic Waste in 2019; OECD Environment Statistics (Database) 2022. Available online: https://www.oecd-ilibrary.org/environment/data/global-plastic-outlook/plastic-waste-in-2019_a92f5ea3-en (accessed on 1 December 2023).
  37. Borrelle, S.B.; Ringma, J.; Law, K.L.; Monnahan, C.C.; Lebreton, L.; McGivern, A.; Murphy, E.; Jambeck, J.; Leonard, G.H.; Hilleary, M.A.; et al. Predicted growth in plastic waste exceeds efforts to mitigate plastic pollution. Science 2020, 369, 1515–1518. [Google Scholar] [CrossRef]
  38. Hidalgo-Ruz, V.; Gutow, L.; Thompson, R.C.; Thiel, M. Microplastics in the Marine Environment: A Review of the Methods Used for Identification and Quantification. Environ. Sci. Technol. 2012, 46, 3060–3075. [Google Scholar] [CrossRef] [PubMed]
  39. Gao, Z.; Wontor, K.; Cizdziel, J.V.; Lu, H. Distribution and characteristics of microplastics in beach sand near the outlet of a major reservoir in north Mississippi, USA. Microplast. Nanoplast. 2022, 2, 10. [Google Scholar] [CrossRef]
  40. Munari, C.; Scoponi, M.; Mistri, M. Plastic debris in the Mediterranean Sea: Types, occurrence and distribution along Adriatic shorelines. Waste Manag. 2017, 67, 385–391. [Google Scholar] [CrossRef] [PubMed]
  41. Osorio, E.D.; Tanchuling, M.A.N.; Diola, M.B.L.D. Microplastics Occurrence in Surface Waters and Sediments in Five River Mouths of Manila Bay. Front. Environ. Sci. 2021, 9, 719274. [Google Scholar] [CrossRef]
  42. Vianello, A.; Da Ros, L.; Boldrin, A.; Marceta, T.; Moschino, V. First evaluation of floating microplastics in the Northwestern Adriatic Sea. Environ. Sci. Pollut. R. 2018, 25, 28546–28561. [Google Scholar] [CrossRef] [PubMed]
  43. Imhof, H.K.; Sigl, R.; Brauer, E.; Feyl, S.; Giesemann, P.; Klink, S.; Leupolz, K.; Loder, M.G.J.; Loschel, L.A.; Missun, J.; et al. Spatial and temporal variation of macro-, meso- and microplastic abundance on a remote coral island of the Maldives, Indian Ocean. Mar. Pollut. Bull. 2017, 116, 340–347. [Google Scholar] [CrossRef]
  44. Wang, J.D.; Peng, J.P.; Tan, Z.; Gao, Y.F.; Zhan, Z.W.; Chen, Q.Q.; Cai, L.Q. Microplastics in the surface sediments from the Beijiang River littoral zone: Composition, abundance, surface textures and interaction with heavy metals. Chemosphere 2017, 171, 248–258. [Google Scholar] [CrossRef]
  45. Boskovic, N.; Joksimovic, D.; Perosevic-Bajceta, A.; Pekovic, M.; Bajt, O. Distribution and characterization of microplastics in marine sediments from the Montenegrin coast. J. Soils Sediments 2022, 22, 2958–2967. [Google Scholar] [CrossRef]
  46. Boskovic, N.; Joksimovic, D.; Pekovic, M.; Perosevic-Bajceta, A.; Bajt, O. Microplastics in Surface Sediments along the Montenegrin Coast, Adriatic Sea: Types, Occurrence, and Distribution. J. Mar. Sci. Eng. 2021, 9, 841. [Google Scholar] [CrossRef]
  47. Wootton, N.; Ferreira, M.; Reis-Santos, P.; Gillanders, B.M. A Comparison of Microplastic in Fish From Australia and Fiji. Front. Mar. Sci. 2021, 8, 690991. [Google Scholar] [CrossRef]
  48. Giani, D.; Baini, M.; Galli, M.; Casini, S.; Fossi, M.C. Microplastics occurrence in edible fish species (Mullus barbatus and Merluccius merluccius) collected in three different geographical sub-areas of the Mediterranean Sea. Mar. Pollut. Bull. 2019, 140, 129–137. [Google Scholar] [CrossRef]
  49. Anastasopoulou, A.; Viršek, M.K.; Varezić, D.B.; Digka, N.; Fortibuoni, T.; Koren, Š.; Mandić, M.; Mytilineou, C.; Pešić, A.; Ronchi, F. Assessment on marine litter ingested by fish in the Adriatic and NE Ionian Sea macro-region (Mediterranean). Mar. Pollut. Bull. 2018, 133, 841–851. [Google Scholar] [CrossRef] [PubMed]
  50. De Simone, S.; Perošević-Bajčeta, A.; Joksimović, D.; Beccherelli, R.; Zografopoulos, D.C.; Mussi, V. Study of microplastics and inorganic contaminants in mussels from the Montenegrin Coast, Adriatic Sea. J. Mar. Sci. Eng. 2021, 9, 544. [Google Scholar] [CrossRef]
  51. Gomiero, A.; Strafella, P.; Øysæd, K.B.; Fabi, G. First occurrence and composition assessment of microplastics in native mussels collected from coastal and offshore areas of the northern and central Adriatic Sea. Environ. Sci. Pollut. R. 2019, 26, 24407–24416. [Google Scholar] [CrossRef] [PubMed]
  52. van den Berg, P.; Huerta-Lwanga, E.; Corradini, F.; Geissen, V. Sewage sludge application as a vehicle for microplastics in eastern Spanish agricultural soils. Environ. Pollut. 2020, 261, 114198. [Google Scholar] [CrossRef] [PubMed]
  53. Akhbarizadeh, R.; Dobaradaran, S.; Torkmahalleh, M.A.; Saeedi, R.; Aibaghi, R.; Ghasemi, F.F. Suspended fine particulate matter (PM2. 5), microplastics (MPs), and polycyclic aromatic hydrocarbons (PAHs) in air: Their possible relationships and health implications. Environ. Res. 2021, 192, 110339. [Google Scholar] [CrossRef]
  54. Allen, S.; Allen, D.; Phoenix, V.R.; Le Roux, G.; Durántez Jiménez, P.; Simonneau, A.; Binet, S.; Galop, D. Atmospheric transport and deposition of microplastics in a remote mountain catchment. Nat. Geosci. 2019, 12, 339–344. [Google Scholar] [CrossRef]
  55. Allen, S.; Allen, D.; Moss, K.; Le Roux, G.; Phoenix, V.R.; Sonke, J.E. Examination of the ocean as a source for atmospheric microplastics. PLoS ONE 2020, 15, e0232746. [Google Scholar] [CrossRef] [PubMed]
  56. Brahney, J.; Hallerud, M.; Heim, E.; Hahnenberger, M.; Sukumaran, S. Plastic rain in protected areas of the United States. Science 2020, 368, 1257–1260. [Google Scholar] [CrossRef] [PubMed]
  57. Gaston, E.; Woo, M.; Steele, C.; Sukumaran, S.; Anderson, S. Microplastics differ between indoor and outdoor air masses: Insights from multiple microscopy methodologies. Appl. Spectrosc. 2020, 74, 1079–1098. [Google Scholar] [CrossRef] [PubMed]
  58. Liu, K.; Wang, X.; Song, Z.; Wei, N.; Ye, H.; Cong, X.; Zhao, L.; Li, Y.; Qu, L.; Zhu, L. Global inventory of atmospheric fibrous microplastics input into the ocean: An implication from the indoor origin. J. Hazard. Mater. 2020, 400, 123223. [Google Scholar] [CrossRef] [PubMed]
  59. Liu, K.; Wu, T.; Wang, X.; Song, Z.; Zong, C.; Wei, N.; Li, D. Consistent transport of terrestrial microplastics to the ocean through atmosphere. Environ. Sci. Technol. 2019, 53, 10612–10619. [Google Scholar] [CrossRef] [PubMed]
  60. Li, Y.; Shao, L.; Wang, W.; Zhang, M.; Feng, X.; Li, W.; Zhang, D. Airborne fiber particles: Types, size and concentration observed in Beijing. Sci. Total Environ. 2020, 705, 135967. [Google Scholar] [CrossRef] [PubMed]
  61. Beswick, R.; Dunn, D. Plastics in Packaging: Western Europe and North America; iSmithers Rapra Publishing: Shawbury, UK, 2002. [Google Scholar]
  62. Cverenkarova, K.; Valachovicova, M.; Mackulak, T.; Zemlicka, L.; Birosova, L. Microplastics in the Food Chain. Life 2021, 11, 1349. [Google Scholar] [CrossRef]
  63. Lwanga, E.H.; Vega, J.M.; Quej, V.K.; Chi, J.D.; del Cid, L.S.; Chi, C.; Segura, G.E.; Gertsen, H.; Salanki, T.; van der Ploeg, M.; et al. Field evidence for transfer of plastic debris along a terrestrial food chain. Sci. Rep. 2017, 7, 14071. [Google Scholar] [CrossRef]
  64. Beriot, N.; Peek, J.; Zornoza, R.; Geissen, V.; Huerta Lwanga, E. Low density-microplastics detected in sheep faeces and soil: A case study from the intensive vegetable farming in Southeast Spain. Sci. Total Environ. 2021, 755, 142653. [Google Scholar] [CrossRef]
  65. Karbalaei, S.; Hanachi, P.; Walker, T.R.; Cole, M. Occurrence, sources, human health impacts and mitigation of microplastic pollution. Environ. Sci. Pollut. Res. Int. 2018, 25, 36046–36063. [Google Scholar] [CrossRef] [PubMed]
  66. Eyles, J.; Alpar, O.; Field, W.N.; Lewis, D.A.; Keswick, M. The transfer of polystyrene microspheres from the gastrointestinal tract to the circulation after oral administration in the rat. J. Pharm. Pharmacol. 1995, 47, 561–565. [Google Scholar] [CrossRef] [PubMed]
  67. Jani, P.; Halbert, G.W.; Langridge, J.; Florence, A.T. Nanoparticle uptake by the rat gastrointestinal mucosa: Quantitation and particle size dependency. J. Pharm. Pharmacol. 1990, 42, 821–826. [Google Scholar] [CrossRef] [PubMed]
  68. Florance, I.; Ramasubbu, S.; Mukherjee, A.; Chandrasekaran, N. Polystyrene nanoplastics dysregulate lipid metabolism in murine macrophages in vitro. Toxicology 2021, 458, 152850. [Google Scholar] [CrossRef] [PubMed]
  69. Vlacil, A.K.; Banfer, S.; Jacob, R.; Trippel, N.; Kuzu, I.; Schieffer, B.; Grote, K. Polystyrene microplastic particles induce endothelial activation. PLoS ONE 2021, 16, e0260181. [Google Scholar] [CrossRef]
  70. Shi, J.; Deng, H.P.; Zhang, M. Whole transcriptome sequencing analysis revealed key RNA profiles and toxicity in mice after chronic exposure to microplastics. Chemosphere 2022, 304, 135321. [Google Scholar] [CrossRef]
  71. Choy, M.; Xue, R.; Wu, Y.; Fan, W.; Dong, Y.; Liu, C. Role of N6-methyladenosine Modification in Cardiac Remodeling. Front. Cardiovasc. Med. 2022, 9, 774627. [Google Scholar] [CrossRef] [PubMed]
  72. Zhang, M.; Shi, J.; Deng, H.P. Transcriptome-wide m6A modification mediates cardiotoxicity in mice after chronic exposure to microplastics. Chemosphere 2023, 317, 137877. [Google Scholar] [CrossRef]
  73. Zhang, M.; Shi, J.; Zhou, J.; Song, L.; Ding, J.; Deng, H.P.; Weng, L.; Zhu, Y.; Xu, Z. N6-methyladenosine methylation mediates non-coding RNAs modification in microplastic-induced cardiac injury. Ecotoxicol. Environ. Saf. 2023, 262, 115174. [Google Scholar] [CrossRef]
  74. Shiwakoti, S.; Ko, J.Y.; Gong, D.; Dhakal, B.; Lee, J.H.; 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]
  75. Dhakal, B.; Shiwakoti, S.; Park, E.Y.; Kang, K.W.; Schini-Kerth, V.B.; Park, S.H.; Ji, H.Y.; Park, J.S.; Ko, J.Y.; Oak, M.H. SGLT2 inhibition ameliorates nano plastics-induced premature endothelial senescence and dysfunction. Sci. Rep. 2023, 13, 6256. [Google Scholar] [CrossRef] [PubMed]
  76. Basini, G.; Grolli, S.; Bertini, S.; Bussolati, S.; Berni, M.; Berni, P.; Ramoni, R.; Scaltriti, E.; Quintavalla, F.; Grasselli, F. Nanoplastics induced oxidative stress and VEGF production in aortic endothelial cells. Environ. Toxicol. Pharmacol. 2023, 104, 104294. [Google Scholar] [CrossRef] [PubMed]
  77. Yin, K.; Lu, H.M.; Zhang, Y.; Hou, L.L.; Meng, X.; Li, J.B.; Zhao, H.J.; Xing, M.W. Secondary brain injury after polystyrene microplastic-induced intracerebral hemorrhage is associated with inflammation and pyroptosis. Chem. Biol. Interact. 2022, 367, 110180. [Google Scholar] [CrossRef] [PubMed]
  78. Zhang, Y.; Yin, K.; Wang, D.X.; Wang, Y.; Lu, H.M.; Zhao, H.J.; Xing, M.W. Polystyrene microplastics-induced cardiotoxicity in chickens via the ROS-driven NF-?B-NLRP3-GSDMD and AMPK-PGC-1a axes. Sci. Total Environ. 2022, 840, 156727. [Google Scholar] [CrossRef] [PubMed]
  79. Zhang, Y.; Wang, D.X.; Yin, K.; Zhao, H.J.; Lu, H.M.; Meng, X.; Hou, L.L.; Li, J.B.; Xing, M.W. Endoplasmic reticulum stress-controlled autophagic pathway promotes polystyrene microplastics-induced myocardial dysplasia in birds. Environ. Pollut. 2022, 311, 119963. [Google Scholar] [CrossRef] [PubMed]
  80. Wei, J.L.; Wang, X.F.; Liu, Q.; Zhou, N.; Zhu, S.X.; Li, Z.K.; Li, X.L.; Yao, J.P.; Zhang, L.S. The impact of polystyrene microplastics on cardiomyocytes pyroptosis through NLRP3/Caspase-1 signaling pathway and oxidative stress in Wistar rats. Environ. Toxicol. 2021, 36, 935–944. [Google Scholar] [CrossRef] [PubMed]
  81. Roshanzadeh, A.; Oyunbaatar, N.E.; Ganjbakhsh, S.E.; Park, S.; Kim, D.S.; Kanade, P.P.; Lee, S.; Lee, D.W.; Kim, E.S. Exposure to nanoplastics impairs collective contractility of neonatal cardiomyocytes under electrical synchronization. Biomaterials 2021, 278, 121175. [Google Scholar] [CrossRef]
  82. Zhao, J.J.; Gomes, D.; Jin, L.X.; Mathis, S.P.; Li, X.H.; Rouchka, E.C.; Bodduluri, H.; Conklin, D.J.; O’Toole, T.E. Polystyrene bead ingestion promotes adiposity and cardiometabolic disease in mice. Ecotoxicol. Environ. Saf. 2022, 232, 113239. [Google Scholar] [CrossRef]
  83. Wang, B.; Liang, B.; Huang, Y.; Li, Z.; Zhang, B.; Du, J.; Ye, R.; Xian, H.; Deng, Y.; Xiu, J.; et al. Long-Chain Acyl Carnitines Aggravate Polystyrene Nanoplastics-Induced Atherosclerosis by Upregulating MARCO. Adv. Sci. 2023, 10, e2205876. [Google Scholar] [CrossRef]
  84. Wang, X.X.; Jia, Z.Z.; Zhou, X.R.; Su, L.; Wang, M.L.; Wang, T.; Zhang, H.Y. Nanoplastic-induced vascular endothelial injury and coagulation dysfunction in mice. Sci. Total Environ. 2023, 865, 161271. [Google Scholar] [CrossRef]
  85. Zhang, T.Y.; Yang, S.; Ge, Y.L.; Wan, X.; Zhu, Y.X.; Yang, F.; Li, J.; Gong, S.S.; Cheng, Y.P.; Hu, C.Y.; et al. Multi-dimensional evaluation of cardiotoxicity in mice following respiratory exposure to polystyrene nanoplastics. Part. Fibre Toxicol. 2023, 20, 46. [Google Scholar] [CrossRef]
  86. 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]
  87. Nnoruka, U.C.; Okonkwo, C.J.; Ilechukwu, I.; Okonkwo, C.J.; Belonwu, D.C. Impact of polystyrene microplastic exposure on lipid profile and oxidative stress status of male and female Wistar rats. Environ. Anal. Health Toxicol. 2022, 37, e2022024. [Google Scholar] [CrossRef]
  88. Cary, C.M.; Seymore, T.N.; Singh, D.; Vayas, K.N.; Goedken, M.J.; Adams, S.; Polunas, M.; Sunil, V.R.; Laskin, D.L.; Demokritou, P.; et al. Single inhalation exposure to polyamide micro and nanoplastic particles impairs vascular dilation without generating pulmonary inflammation in virgin female Sprague Dawley rats. Part. Fibre Toxicol. 2023, 20, 16. [Google Scholar] [CrossRef] [PubMed]
  89. Li, Z.K.; Zhu, S.X.; Liu, Q.; Wei, J.L.; Jin, Y.C.; Wang, X.F.; Zhang, L.S. Polystyrene microplastics cause cardiac fibrosis by activating Wnt/beta-catenin signaling pathway and promoting cardiomyocyte apoptosis in rats. Environ. Pollut. 2020, 265, 115025. [Google Scholar] [CrossRef] [PubMed]
  90. Yan, J.; Pan, Y.; He, J.; Pang, X.; Shao, W.; Wang, C.; Wang, R.; He, Y.; Zhang, M.; Ye, J.; et al. Toxic vascular effects of polystyrene microplastic exposure. Sci. Total Environ. 2023, 905, 167215. [Google Scholar] [CrossRef] [PubMed]
  91. Park, S.H.; Kim, K. Microplastics induced developmental toxicity with microcirculation dysfunction in zebrafish embryos. Chemosphere 2022, 286, 131868. [Google Scholar] [CrossRef] [PubMed]
  92. Sun, M.Q.; Ding, R.Y.; Ma, Y.M.; Sun, Q.L.; Ren, X.K.; Sun, Z.W.; Duan, J.C. Cardiovascular toxicity assessment of polyethylene nanoplastics on developing zebrafish embryos. Chemosphere 2021, 282, 131124. [Google Scholar] [CrossRef] [PubMed]
  93. Pitt, J.A.; Kozal, J.S.; Jayasundara, N.; Massarsky, A.; Trevisan, R.; Geitner, N.; Wiesner, M.; Levin, E.D.; Di Giulio, R.T. Uptake, tissue distribution, and toxicity of polystyrene nanoparticles in developing zebrafish (Danio rerio). Aquat. Toxicol. 2018, 194, 185–194. [Google Scholar] [CrossRef] [PubMed]
  94. Pedersen, A.F.; Meyer, D.N.; Petriv, A.M.V.; Soto, A.L.; Shields, J.N.; Akemann, C.; Baker, B.B.; Tsou, W.L.; Zhang, Y.L.; Baker, T.R. Nanoplastics impact the zebrafish (Danio rerio) transcriptome: Associated developmental and neurobehavioral consequences. Environ. Pollut. 2020, 266, 115090. [Google Scholar] [CrossRef]
  95. Dai, L.; Luo, J.; Feng, M.; Wang, M.; Zhang, J.; Cao, X.; Yang, X.; Li, J. Nanoplastics exposure induces vascular malformation by interfering with the VEGFA/VEGFR pathway in zebrafish (Danio rerio). Chemosphere 2023, 312, 137360. [Google Scholar] [CrossRef] [PubMed]
  96. Santos, A.L.; Rodrigues, L.C.; Rodrigues, C.C.; Cirqueira, F.; Malafaia, G.; Rocha, T.L. Polystyrene nanoplastics induce developmental impairments and vasotoxicity in zebrafish (Danio rerio). J. Hazard. Mater. 2024, 464, 132880. [Google Scholar] [CrossRef] [PubMed]
  97. Wu, H.; Guo, J.; Yao, Y.; Xu, S. Polystyrene nanoplastics induced cardiomyocyte apoptosis and myocardial inflammation in carp by promoting ROS production. Fish. Shellfish Immunol. 2022, 125, 1–8. [Google Scholar] [CrossRef] [PubMed]
  98. Rotchell, J.M.; Jenner, L.C.; Chapman, E.; Bennett, R.T.; Bolanle, I.O.; Loubani, M.; Sadofsky, L.; Palmer, T.M. Detection of microplastics in human saphenous vein tissue using μFTIR: A pilot study. PLoS ONE 2023, 18, e0280594. [Google Scholar] [CrossRef] [PubMed]
  99. Sun, J.N.; Sui, M.S.; Wang, T.; Teng, X.M.; Sun, J.; Chen, M.X. Detection and quantification of various microplastics in human endometrium based on laser direct infrared spectroscopy. Sci. Total Environ. 2024, 906, 167760. [Google Scholar] [CrossRef] [PubMed]
  100. Lee, H.S.; Amarakoon, D.; Wei, C.I.; Choi, K.Y.; Smolensky, D.; Lee, S.H. Adverse effect of polystyrene microplastics (PS-MPs) on tube formation and viability of human umbilical vein endothelial cells. Food Chem. Toxicol. 2021, 154, 112356. [Google Scholar] [CrossRef] [PubMed]
  101. Lu, Y.Y.; Cao, M.Y.; Tian, M.P.; Huang, Q.Y. Internalization and cytotoxicity of polystyrene microplastics in human umbilical vein endothelial cells. J. Appl. Toxicol. 2023, 43, 262–271. [Google Scholar] [CrossRef] [PubMed]
  102. Wei, W.; Li, Y.H.; Lee, M.; Andrikopoulos, N.; Lin, S.J.; Chen, C.Y.; Leong, D.T.; Ding, F.; Song, Y.; Ke, P.C. Anionic nanoplastic exposure induces endothelial leakiness. Nat. Commun. 2022, 13, 4757. [Google Scholar] [CrossRef] [PubMed]
  103. Lu, Y.Y.; Li, H.; Ren, H.; Zhang, X.; Huang, F.; Zhang, D.; Huang, Q.; Zhang, X. Size-dependent effects of polystyrene nanoplastics on autophagy response in human umbilical vein endothelial cells. J. Hazard. Mater. 2022, 421, 126770. [Google Scholar] [CrossRef]
  104. Zhang, M.; Shi, J.; Huang, Q.; Xie, Y.; Wu, R.H.; Zhong, J.Y.; Deng, H.P. Multi-omics analysis reveals size-dependent toxicity and vascular endothelial cell injury induced by microplastic exposure in vivo and in vitro. Environ. Sci.-Nano 2022, 9, 663–683. [Google Scholar] [CrossRef]
  105. Fu, Y.; Fan, M.; Xu, L.; Wang, H.; Hu, Q.; Jin, Y. Amino-Functionalized Polystyrene Nano-Plastics Induce Mitochondria Damage in Human Umbilical Vein Endothelial Cells. Toxics 2022, 10, 215. [Google Scholar] [CrossRef] [PubMed]
  106. Chen, L.F.; Zheng, Y.J.; Liu, Y.T.; Tian, P.F.; Yu, L.; Bai, L.; Zhou, F.L.; Yang, Y.; Cheng, Y.X.; Wang, F.B.; et al. Microfluidic-based in vitro thrombosis model for studying microplastics toxicity. Lab A Chip 2022, 22, 1344–1353. [Google Scholar] [CrossRef] [PubMed]
  107. Zhou, Y.; Wu, Q.; Li, Y.; Feng, Y.; Wang, Y.; Cheng, W. Low-dose of polystyrene microplastics induce cardiotoxicity in mice and human-originated cardiac organoids. Environ. Int. 2023, 179, 108171. [Google Scholar] [CrossRef]
  108. Chen, Y.C.; Chen, K.F.; Andrew Lin, K.Y.; Su, H.P.; Wu, D.N.; Lin, C.H. Evaluation of toxicity of polystyrene microplastics under realistic exposure levels in human vascular endothelial EA.hy926 cells. Chemosphere 2023, 313, 137582. [Google Scholar] [CrossRef] [PubMed]
  109. Florance, I.; Chandrasekaran, N.; Gopinath, P.M.; Mukherjee, A. Exposure to polystyrene nanoplastics impairs lipid metabolism in human and murine macrophages in vitro. Ecotoxicol. Environ. Saf. 2022, 238, 113612. [Google Scholar] [CrossRef] [PubMed]
  110. Mohr, K.; Sommer, M.; Baier, G.; Schöttler, S.; Okwieka, P.; Tenzer, S.; Landfester, K.; Mailänder, V.; Schmidt, M.; Meyer, R.G. Aggregation behavior of polystyrene-nanoparticles in human blood serum and its impact on the in vivo distribution in mice. J. Nanomed. Nanotechnol. 2014, 5, 1–10. [Google Scholar] [CrossRef]
  111. Ballesteros, S.; Domenech, J.; Barguilla, I.; Cortés, C.; Marcos, R.; Hernández, A. Genotoxic and immunomodulatory effects in human white blood cells after ex vivo exposure to polystyrene nanoplastics. Environ. Sci. Nano 2020, 7, 3431–3446. [Google Scholar] [CrossRef]
  112. Prietl, B.; Meindl, C.; Roblegg, E.; Pieber, T.; Lanzer, G.; Fröhlich, E. Nano-sized and micro-sized polystyrene particles affect phagocyte function. Cell Biol. Toxicol. 2014, 30, 1–16. [Google Scholar] [CrossRef]
  113. Gopinath, P.M.; Saranya, V.; Vijayakumar, S.; Mythili Meera, M.; Ruprekha, S.; Kunal, R.; Pranay, A.; Thomas, J.; Mukherjee, A.; Chandrasekaran, N. Assessment on interactive prospectives of nanoplastics with plasma proteins and the toxicological impacts of virgin, coronated and environmentally released-nanoplastics. Sci. Rep. 2019, 9, 8860. [Google Scholar] [CrossRef]
  114. Barshtein, G.; Livshits, L.; Shvartsman, L.D.; Shlomai, N.O.; Yedgar, S.; Arbell, D. Polystyrene nanoparticles activate erythrocyte aggregation and adhesion to endothelial cells. Cell Biochem. Biophys. 2016, 74, 19–27. [Google Scholar] [CrossRef] [PubMed]
  115. Rochman, C.M.; Hoh, E.; Hentschel, B.T.; Kaye, S. Long-Term Field Measurement of Sorption of Organic Contaminants to Five Types of Plastic Pellets: Implications for Plastic Marine Debris. Environ. Sci. Technol. 2013, 47, 1646–1654. [Google Scholar] [CrossRef] [PubMed]
  116. Kefer, S.; Miesbauer, O.; Langowski, H.C. Environmental Microplastic Particles vs. Engineered Plastic Microparticles-A Comparative Review. Polymers 2021, 13, 2881. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The fate of micro- and nanoplastics in mammalian bodies.
Figure 1. The fate of micro- and nanoplastics in mammalian bodies.
Life 14 00255 g001
Figure 2. Sources of micro- and nanoplastics in the environment.
Figure 2. Sources of micro- and nanoplastics in the environment.
Life 14 00255 g002
Figure 3. The possible influence of micro- and nanoplastics on the vascular system.
Figure 3. The possible influence of micro- and nanoplastics on the vascular system.
Life 14 00255 g003
Table 1. Summary of micro- and nanoplastic toxicity on the vascular system.
Table 1. Summary of micro- and nanoplastic toxicity on the vascular system.
ModelsType and Size of ParticlesConc.Modes of ActionReferences
RAW 264.7 and BV2 microglial cellsPolystyrene (PS) NPs
0.20 μm
50, 100, and 200 μg/mLDisruption of lipid metabolismFlorance et al., 2021 [68]
Murine myocardial endothelial cellsPS MPs
1 μm
0.54 ng/mL, 54 ng/mL, and 5.4 μg/mLInduction of endothelial activation and monocyte adhesionVlacil et al., 2021 [69]
Porcine coronary artery endothelial cells (PCAECs)PS NPs
25 nm
0.1, 1, and 10 μg/mLInduction of premature EC senescenceShiwakoti et al., 2022 [74]
PS NPs5 and 10 μg/mLInduction of premature EC senescence and dysfunctionDhakal et al., 2023 [75]
Porcine aortic endothelial cell line (AOC)PS NPs
100 nm
5, 25, and 75 μg/mLAlteration of AOC metabolic activity, redox status, and VEGF productionBasini et al., 2023 [76]
ChickenPS MPs
5 μm
1, 10, and 100 mg/L
in drinking water
Alteration of intracerebral hemorrhage and mitochondrial dysfunctionYin et al., 2022 [77]
PS MPs
5 μm
1, 10, and 100 mg/L
in drinking water
Induction of myocardial inflammation and pyroptosisZhang et al., 2022 [78]
BirdPS MPs
5 μm
1, 10, and 100 mg/L
in drinking water
Induction of endoplasmic reticulum (ER) stress in the myocardiumZhang et al., 2022 [79]
MouseMPs
5 μm
1000 μg/L
in drinking water
Alterations in biological features and global gene expression patternsShi et al., 2022 [70]
PS MPs
10 μm
1000 μg/L
in drinking water
Induction of m6A modifications of ncRNAsZhang et al., 2023 [73]
PS NPs
0.5 and 5 μm
0.1 and 1.0 μg/mL
in drinking water
Induction of adiposity and hyperglycemiaZhao et al., 2022 [82]
PS NPs
50 nm
2.5, 25, and 250 mg/kgInduction of MARCO upregulation, macrophage activation, and lipid metabolism disruptionWang et al., 2023 [83]
PS, PS-NH2, and PS-COOH NPs
50 nm
0.05, 0.5, 5, 10, and 20 mg/kgInduction of injury and dysfunction through the activation of the JAK1/STAT3/TF pathwayWang et al., 2023 [84]
PS NPs
40 nm
7.9 × 1012, 1.85 × 1013, and 4.6 × 1013 items/m3Induction of acute cardiotoxicityZhang et al., 2023 [85]
PS NPs
42 nm
0.5, 2.5, 10, and 50 mg/kgInduction of microglia activation and neuron damageShan et al., 2022 [86]
RatPS MPs
0.5 μm
0.5, 5, and 50 mg/L
in drinking water
Induction of pyroptosis of cardiomyocytes and oxidative stress/inflammationWei et al., 2021 [80]
Amine-modified PS NPs
50 nm
25 μg/mLReduction of collective contractilityRoshanzadeh et al., 2021 [81]
PS MPs
5 mm
Feed containing
1, 5, and 10%
Alteration of dyslipidemia and oxidative imbalanceNnoruka et al., 2022 [87]
Polyamide MNPs
with a median diameter of 2.81 μm
Exposure concentration of near 10 mg/m3Alteration of inflammatory, cardiovascular, and endocrine activityCary et al., 2023 [88]
PS MPs
0.5 μm
0.5, 5, and 50 mg/L Alteration of cardiac fibrosis and dysfunctionLi et al., 2020 [89]
PS MPs
5 μm
0.5 mg/LInduction of mild vascular calcification (VC)Yan et al., 2023 [90]
Zebrafish embryosPS NPs, PS MPs
0.4 and 1 μm
0.00075–0.006%Induction of developmental toxicity and microcirculation dysfunctionPark et al., 2022 [91]
Polyethylene NPs
191.10 ± 3.13 nm
25, 50, 100, 200, 400, 600, 800, and 1000 μg/mLAlteration of endothelial damage and hemodynamics changesSun et al., 2021 [92]
PS NPs
20 nm
2, 5, and 8 mg/LInduction of various vascular malformationsDai et al., 2023 [95]
PS NPs
23.03 ± 0.266 nm
0.04 and 34 ng/L,
34 μg/L
Induction of neurotoxicity and cardiotoxicitySantos et al., 2024 [96]
Zebrafish (Danio rerio)PS NPs
51 nm
0, 0.1, 1, and 10 ppmInduction of cardiotoxicityPitt et al., 2018 [93]
PS NPs
50 and 200 nm
0, 10, 100, 1000, and 10,000 ppbAlteration of behavioral response (swimming hyperactivity)Pedersen et al., 2020 [94]
CarpPS NPs
50, 100, and 400 nm
1000 μg/LInduction of cardiomyocyte apoptosis and myocardial inflammationWu et al., 2022 [97]
Human umbilical vein endothelial cells (HUVECs)PS MPs
0.5, 1, and 5 μm
0, 20, 40, 60, 80, and 100 μg/mLReduction of viability and stimulation of autophagy/necrosisLee et al., 2021 [100]
PS MNPs
1 μm
0, 5, 10, and 25 μg/mLReduction of cell viabilityLu et al., 2023 [101]
PS, NH2-PS, PMMA NPs
30 and 50 nm
0.05 and 0.5 mg/mLInduction of endothelial leakinessWei et al., 2022 [102]
PS NPs
100 and 500 nm
0, 5, 10, 25, 50, and 100 μg/mLInduction of autophagosome formation and autophagic flux blockageLu et al., 2022 [103]
PS MPs
20, 50, 100, and 500 nm, 5 and 10 μm
1000 μg/mLAlteration of endothelial dysfunctionZhang et al., 2022 [104]
PS NPs and NH2-PS NPs
50 nm
5, 10, 15, 20, and 25 μg/mLInduction of oxidative stressFu et al., 2022 [105]
HUVECs and blood (human/mouse) PS MPs
1 μm
0.1 and 1 μg/mLIncreased risk of thrombosisChen et al., 2022 [106]
Human embryonic stem cell line H1 (H1 ES)PS MPs
1 μm
0, 0.025, 0.25, and 2.5 μg/mLIncreased the expression levels of cardiac-specific markers MYL2, MYL4, and CX43Zhou et al., 2023 [107]
Human vascular endothelial EA.hy926 cellsPS MPs
2.2–6.5 μm
4 × 10−6–40 μg/mLInduction of oxidative vascular cytotoxicityChen et al., 2023 [108]
THP-1 (human monocytes) and RAW 264.7 (murine macrophages)PS NPs
0.2 μm
100 µg/mLDysregulation of lipid metabolism homeostasisFlorance et al., 2022 [109]
Table 2. Summary of micro- and nanoplastic effects on human vascular system.
Table 2. Summary of micro- and nanoplastic effects on human vascular system.
SpecimenType and Size of ParticlesEffects on Vascular SystemReference
Human serum PS NPs
80–170 nm
Internalization into granulocytes, monocytes, and myeloid dendritic cells Mohr et al., 2014 [110]
Human whole bloodPS NPs
0.05–0.1 μm
Internalization into monocytes and white cells, genotoxic effects on monocytes and polymorphonuclear leukocytes Ballesteros et al., 2020 [111]
Human peripheral blood mononuclear cellsPS NPs, PS MPs
20, 100, 200, 500, and 1000 nm
Internalization into phagocytes and macrophages, IL-6 secretionPrietl et al., 2014 [112]
Human plasmaPS NPs
100 nm
Erythrocytes and lymphocytes inducing cytotoxicity, hemolysis, and genotoxicity Gopinath et al., 2019 [113]
Human red blood cellsPS NPs
49.9 ± 6.3, 107.9 ± 1.4, and 243 ± 3.0 nm
Adhesion and aggregation to
endothelial cells
Barshtein et al., 2016 [114]
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

Lee, S.E.; Yoon, H.K.; Kim, D.Y.; Jeong, T.S.; Park, Y.S. An Emerging Role of Micro- and Nanoplastics in Vascular Diseases. Life 2024, 14, 255. https://doi.org/10.3390/life14020255

AMA Style

Lee SE, Yoon HK, Kim DY, Jeong TS, Park YS. An Emerging Role of Micro- and Nanoplastics in Vascular Diseases. Life. 2024; 14(2):255. https://doi.org/10.3390/life14020255

Chicago/Turabian Style

Lee, Seung Eun, Hyun Kyung Yoon, Do Yun Kim, Taek Seung Jeong, and Yong Seek Park. 2024. "An Emerging Role of Micro- and Nanoplastics in Vascular Diseases" Life 14, no. 2: 255. https://doi.org/10.3390/life14020255

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