Abstract
The cardiovascular system is pivotal in the systemic distribution and toxicity of nanoparticles (NPs) upon their entry into circulation. Therefore, it is crucial to extensively investigate the adverse cardiovascular effects of NPs. This review presents the advancements in understanding the cardiovascular distribution of NPs and their potential adverse effects, particularly in humans. We first discussed the uptake and distribution of NPs in cardiovascular tissues and cells, which result in adverse outcomes such as hemolysis, blood coagulation dysfunction, vascular endothelial damage, and cardiac pathological damage and function impairment. Additionally, we discuss the current understanding of the mechanisms underlying NP-induced cardiovascular toxicity, including oxidative stress, inflammation, mitochondrial damage, and autophagy. Moreover, we systematically reviewed the factors influencing NPs circulation, retention, clearance, and toxicity within the cardiovascular system, which include the primary physicochemical properties of NPs, modifications of NPs with biological molecules, physiological and pathological conditions of the body, and their interaction with other environmental chemicals. Finally, we proposed the challenges associated with NP cardiovascular toxicity, with the aim of providing insights into the interactions between NPs and cardiovascular components and offering valuable perspectives for the development of safer NP-based therapies and biomedical applications.
Graphical abstract

1 Introduction
Nanotechnology, defined as the manipulation of matter on an atomic or molecular scale, has revolutionized numerous fields [1,2,3,4]. However, the rapid advancement and widespread application of nanotechnology inevitably lead to environmental release and human exposure to nanoparticles (NPs) [5]. Whether encountered environmentally, occupationally, or medically, NPs can interact uniquely with biological systems due to their extremely small size and high surface area-to-volume ratio, raising significant concerns about their health impact [6,7,8].
Humans may be exposed to NPs through various ways, including skin contact, inhalation, and ingestion [5,9]. After crossing primary biological barriers such as the skin, the air–blood barrier, and the intestinal barrier, NPs initially enter the cardiovascular system before being transported and accumulated in vital organs [10,11]. For biomedical applications, NPs are designed to have a long-circulating effect to facilitate drug delivery [12,13]. NP-based drug delivery systems have shown great potential in overcoming the limitations of free drugs and offering multiple therapeutic effects in treating several diseases through improving bioavailability, targeting ability, efficacy, and safety [14,15,16]. However, nanomedicines still face challenges that limit their clinical translation due to insufficient information on the mechanisms influencing blood circulation, biodistribution, and fate, as well as the biocompatibility of nanomedicines [7,17]. Consequently, the cardiovascular system is at the forefront of nano bio-distribution, making the potential adverse effects of NPs on the cardiovascular system, a critical area of concern that requires extensive attention. Mounting evidence has shown that NPs can damage the cardiovascular system through interactions with various blood cells [18,19], vascular endothelial cells (ECs) [20,21], and myocardial cells [22,23]. Recently, potential adverse effects of NPs on the cardiovascular system, particularly focusing on specific types of NPs [24,25,26], or special cells and animal models [27,28], have been reviewed extensively. However, a comprehensive review providing a complete and up-to-date overview of the cardiovascular distribution and toxicity of NPs is still lacking.
Using “‘cardiovascular toxicity’ AND ‘nanoparticles’” as search terms, we screened and analyzed the 1,569 and 2,540 research papers obtained in the Web of Science and Google Scholar databases. The top five concepts of the obtained papers include biomaterials, pharmacology, toxicology, biochemistry and molecular biophysics, and transport and circulation. Based on the literature on the reported adverse cardiovascular effects of NPs, we comprehensively reviewed recent advancements in the distribution of NPs within the cardiovascular system, their potential adverse effects, and the underlying mechanisms governing NP interactions with cardiovascular tissues, particularly in humans. Additionally, detailed scrutiny was conducted on the factors influencing NPs circulation, retention, clearance, and toxicity within the cardiovascular system. We also addressed the challenges associated with NP-induced cardiovascular toxicity, aiming to illuminate NP interactions with cardiovascular components and provide valuable perspectives for the development of safer NP-based therapies and biomedical applications.
2 Distribution of NPs in cardiovascular system
The reticuloendothelial system generally exhibits higher NP accumulation compared to the heart [29]. Nonetheless, the cardiovascular system is recognized as a major site for NP distribution [24]. Accumulation of various types of NPs in the heart has been documented across different animals, irrespective of exposure route-whether through inhalation or ingestion. Polymer NPs, including nanoplastics [30,31], metal NPs [32,33], carbon-based NPs [34,35], and other inorganic NPs [36,37], all exhibited heart accumulation. Experimental evidence directly confirms the presence of NPs in myocardial cells of Wistar rats orally exposed to polystyrene microplastics (PS, diameter = 0.5 μm) at doses of 5 or 50 mg/L for 90 days [38]. Although NP uptake by cardiomyocytes has been observed in various cell lines in vitro, such as primary cardiomyocytes from Wistar rats [39,40], cardiomyocyte-derived HL-1 cells [41], human cardiomyocytes (AC16) [42], and H9c2 rat cardiomyoblasts [39,43], detailed studies on the mechanism of NP-cardiomyocyte interactions remain limited. For example, it was found that the titanate nanotubes (TiONts) were taken up by neonatal rat cardiomyocytes through two distinct pathways, which include endocytosis, specifically the macropinocytosis (in the case of nanohybrids larger than 500 nm) and diffusion through the membrane, while the spherical TiO2 was considered to be exclusively taken up through endocytosis, indicating that the shape and probably their specific surface have a predominant impact in the internalization step [40]. Consistently, macropinocytosis was found to be involved in the cellular uptake of hydroxyapatite (HAp) by cardiomyocyte-derived HL-1 cells [41]. In contrast, CsPbBr3 NPs were taken up by human cardiomyocytes AC16 via macropinocytosis and phagocytosis, followed by vesicle delivery into lysosomes [42]. However, the atrial natriuretic peptide-modified, heart-targeted porous silicon NPs were internalized by primary cardiomyocytes partly via the guanylate cyclase-A receptor expressed at the cell membrane [39].
Besides the interaction with myocardial cells, NPs entering circulation can interact directly with various types of blood cells. For instance, it was demonstrated that the interaction between silver NPs (Ag NPs) and fish red blood cells (RBCs) was regulated by the size of Ag NPs, with more particles of middle size (50 nm) being adsorbed to the cell surface and internalized by RBCs, compared with smaller (15 nm) or larger (100 nm) ones [18]. In another investigation, gold nanorods (Au NRs), coated with hexadecyltrimethylammonium bromide but not poly(sodium-p-styrenesulfonate) were internalized by mouse RBCs, leading to induced hemolysis [44]. Circulating NPs have also been observed to be actively internalized into human granulocytes and monocytes, with the specific endocytic mechanism yet to be fully elucidated [45]. In addition to the physicochemical properties of NPs, the interaction of NPs with immune cells in human blood is significantly influenced by the adsorption of various plasma components in a person-specific manner, contributing to the complexity of nano-blood cell interaction [46]. Although specific plasma proteins such as hemoglobin [46] and coagulation factor XII [47] are known to play crucial roles in NP-induced hematological responses, further investigation is required to understand how biological molecules modulate NP characteristics and subsequently regulate their hematological behaviors.
Furthermore, the vascular endothelium, a monolayer of ECs, is in direct contact with circulating NPs, and ECs have demonstrated the capacity to uptake NPs [48]. Four distinct pathways of endocytosis, including caveolin-mediated, clathrin-mediated, phagocytosis, and micropinocytosis, are well-recognized for their involvement in NP uptake of ECs [49]. Although the endothelial cellular uptake of NPs depends mainly on the characteristics of the NPs, it showed a negative correlation with the flow rate of the bloodstream [50,51]. However, the uptake of rod-like NPs is less affected by increased flow rate compared to spherical NPs [51]. Additionally, shear stress is an important factor contributing to mechanical damage of ECs induced by rod-like NPs, but not by the spherical NPs.
As has been shown above, the cardiovascular system is highly complex with multiple functional cells and various biological molecules. To date, research studies have revealed interactions between NPs and different cell types within the cardiovascular system, highlighting the crucial role of biomolecules in the behaviors and distribution of NPs (Figure 1). Given the critical and multifaceted role of the cardiovascular system in the distribution and toxicity of NPs, further study should focus on elucidating the detailed molecular mechanisms underlying these interactions. Specifically, studies should target cardiomyocytes, blood cells, and vascular ECs, aiming to understand the endocytic pathways and the role of specific receptors and plasma proteins in mediating NP uptake and effects.

Routes of exposure, distribution of NPs, and interactions between NPs and different cell types within the cardiovascular system.
3 Toxicity of NPs to the cardiovascular system and the underlying mechanism
3.1 Effect on blood components
Whole blood comprises essential components such as RBCs, white blood cells, platelets, and plasma, which are responsible for transporting oxygen and nutrients to tissues (RBCs), defending against external xenobiotic agents (white blood cells), and promoting blood clots formation in wounds (platelets). Plasma, on the other hand, creates a balanced acid–base environment and osmotic pressure, enabling the circulation of blood cells and critical proteins throughout the body. The toxicity of NPs to the cardiovascular system is initiated through their interaction with the diverse blood components.
In male BALB/c mice, administration of PS NPs via caudal vein injection led to coagulation dysfunction. This was evidenced by decreased prothrombin time and activated partial thromboplastin time, along with the increased plasma fibrinogen and elevated levels of tissue factor (TF), protease-activated receptor-1, and fibrinogen in the aortic arch [52]. The activation of the Janus kinase 1/signal transducers and activators of transcription-3/TF pathway was proposed as the main molecular mechanism underlying the nanoplastic-induced coagulation dysfunctions and prethrombotic state. Similarly, pulmonary exposure to Ag NPs triggered prothrombotic events and alterations in plasma coagulation markers, including fibrinogen and plasminogen activator inhibitor-1, in BALB/c mice [53]. Consistently, activation of various hematologic parameters in human blood was observed after exposure to Ag NPs (12 nm, 30 mg/L). These included hemolysis of RBCs, α-granule secretion in platelets, approximately 4-fold increase of CD11b expression on granulocytes, elevated coagulation markers such as thrombin–antithrombin-III complex (about 1.6-fold increase), kallikrein-like and FXIIa-like activities, and activation of complement cascade [54]. In contrast, commercially available carboxylated PS NPs (hydrodynamic diameters of 49 nm in water) exhibited minimal hemolytic potential to RBCs but upregulated the expression of P-selection, a marker of platelets activation, and promoted platelet–monocyte aggregation at a concentration of 300 mg/L [55]. Furthermore, NPs such as graphene oxide NPs [56] and poly(n-butylcyano-acrylate) NPs were found to elicit an inflammatory reaction in blood [57]. When RBCs, mononuclear cells, and platelets were isolated and used to investigate the direct interaction between NPs and specific blood cell types. The RBCs displayed morphological changes [58,59] and hemolysis response [19,60] upon exposure to various types of NPs. The hemolytic activity of NPs is primarily attributed to oxidative stress and direct interactions with cell membranes [18,61,62]. Human peripheral blood lymphocytes exposed to NPs exhibited inflammatory response [63], G0/G1 cell cycle arrest [64], apoptosis [65], and DNA damage [66], dependent on the NPs’ chemical composition, particle size, and surface modifications. Similar effects were also observed in lymphocytes from rodents [67] and cell lines such as the THP-1 human leukemia monocytic cell [68] and RAW 264.7 mouse monocyte/macrophage-like cell [56]. The toxicity of NPs to lymphocytes can be partly attributed to intracellular reactive oxygen species (ROS) imbalance [69] and dysfunction of mitochondria and lysosomes [70]. Additionally, various studies have shown that NPs affect platelets function through either procoagulant or anticoagulant activities, which were dictated by their physicochemical characteristics, like particle size and surface area [71,72]. Understanding NP–platelet interactions is crucial not only for developing safe nanomedicines to mitigate adverse biological effects but also for creating targeted therapeutics for critical clinical scenarios.
It is well-known that NPs entering the circulatory system can interact with proteins circulating in the blood, thereby altering their primary properties, biological behaviors, and toxicity [73,74,75]. However, much less is known regarding the pathological outcomes of NP-altered plasma protein components on individuals. A recent work reported that a primary lipid-based corona formed on the surface of silica (SiO2) NPs following their entry into the alveoli of mice after intranasal instillation, which enhanced the adsorption of apolipoprotein A-I (Apo A-I) in the blood [76]. Although protein adsorption reduced the cellular uptake and cytotoxicity of SiO2 NPs, the rapid elimination of the NPs decreased the Apo A-I level in the blood, resulting in atherosclerotic lesions, indicating the correlation between plasma protein adsorption and cardiovascular damage induced by NPs exposure. In contrast, polyvinylpyrrolidone-stabilized gold NP (Au NP) coatings could inhibit blood protein adsorption, indicating the potential use in blood-contact medical applications [77].
3.2 Damage to vascular structures
The vascular endothelium, comprising ECs, constitutes the inner layer of blood vessels and plays a pivotal role in regulating vascular functions such as blood flow, blood pressure, and angiogenesis [78]. ECs secrete various biologically active mediators, including nitric oxide (NO) and arachidonic acid derivatives, which contribute to vascular homeostasis [79]. The endothelial layer forms a non-thrombogenic and non-adhesive surface that maintains blood fluidity and acts as a selectively permeable barrier for molecules [80]. The interaction between NPs and ECs occurs as NPs pass through biological barriers via non-injected routes, circulate in the bloodstream, and undergo clearance from the blood. So far, NP-induced damage to the vascular structure has been confirmed in vitro [81], ex vivo [82], and in vivo [83]. Its exposure triggers endothelial leakiness and impairs angiogenesis by disrupting cytoskeleton organization, reducing the expression of cellular adhesion molecules, and inducing apoptosis in ECs [84,85,86]. Furthermore, NPs cause endothelial nitric oxide synthases (NOS) system disorder [87], oxidative stress [88], and endoplasmic reticulum (ER) stress [89], promoting vascular endothelial dysfunction. It is worth noting that non-coding RNAs also play key roles in NP-induced toxicity. For example, a cluster of RNA transcripts involved in the phagosome, gap junction, TNF, and chemokine signaling pathways is dysregulated in EA.hy926 human ECs after exposure to PM2.5 [90]. miR-451a negatively regulates the interleukin-6 receptor (IL6R)/STAT/TF signaling pathway, which accelerates SiO2 NP-induced vascular endothelial dysfunction and prethrombotic state [91]. Additionally, autophagy perturbation is crucial in NP-induced EC dysfunction. For instance, various classical autophagy signaling pathways, generally mediated by ROS or NO/NOS system disorder, such as phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (mTOR) [92], mitogen-activated protein kinases (MAPK)/B-cell leukemia/lymphoma 2 (Bcl-2) [93], and vascular endothelial growth factor receptor 2 (VEGFR2)/MAPK/extracellular signal-regulated kinase 1/2 (Erk1/2)/mTOR pathways [84], are involved in SiO2 NP-induced endothelial dysfunction. Similarly, ROS-mediated mitochondria-lysosome damage [94,95] and NO/NOS system disorder-mediated autophagy [87] are responsible for Ag NP- and magnetic ferroferric oxide NP (Fe3O4 NP)-induced EC dysfunction, respectively. Recently, it was found that SiO2 NPs (50 nm) induced cardiovascular toxicity in primary human umbilical vein endothelial cells (HUVECs) by triggering nuclear receptor coactivator 4 (NCOA4)-mediated autophagy, leading to ferroptosis [96]. Interestingly, NPs may also induce ROS- and endocytosis-independent endothelial leakiness. For example, anionic PS and poly(methyl methacrylate) NPs induced endothelial leakiness by disrupting vascular endothelial cadherin junctions in ECs, independent of ROS production, autophagy, and apoptosis [82].
3.3 Injury to the heart
The heart is a muscular organ that pumps blood throughout the body. According to a 2022 Statistical Update, heart failure impacts approximately 64 million people worldwide [97]. The incidence and mortality of heart failure are strongly associated with environmental pollution [98,99,100]. Studies have shown that exposure to NPs triggers cardiac dysfunction in rodents. For example, multi-walled carbon nanotubes exposed via oropharyngeal aspiration (4 mg/kg body weight) exacerbate myocardial ischemia–reperfusion (I/R) injury by activating mast cell and the IL-33/IL-1-like receptor ST2 axis [101]. In an ex vivo heart model, amorphous SiO2 NPs disturbed cardiac function by increasing mitochondrial permeability [102]. In a mouse embryo model, zinc oxide NPs (ZnO NPs) increased the risk of heart tube malformation during early cardiogenesis by inducing apoptosis, programmed necrosis, and ferroptosis-mediated precardiac cell death [103].
In addition to mammalian models, the zebrafish (Danio rerio) model is a viable alternative for the cardiovascular toxicity study of hazardous chemicals [104]. A recent study proposed that zebrafish embryos can be utilized to predict the circulation times of NPs in mice and to facilitate the quantification of NP–cell interactions [105]. Potential adverse cardiac effects of various NPs, including neodymium oxide (Nd2O3) NPs [106], copper NPs [107], Au NRs [108], and carbon NPs [109], have been identified in zebrafish embryos. The primary adverse outcomes include pericardial edema, cardiac arrhythmia, and reductions in cardiac parameters such as stroke volume, ejection fraction, and cardiac output [86,107,110,111]. Interestingly, raising the environmental temperature from 27 to 30°C increased lethality but reduced cardiovascular toxicity of PS NPs by enhancing the myocardial contractility of larvae [111].
Recently, some non-animal models, including 3D cell co-cultures, organoids, and organ-on-a-chip technologies, have been developed for evaluating the health risks of environmental toxicants [27,112]. For high-throughput determination of the cardiotoxicity of NPs, a heart-on-a-chip model based on neonatal rat ventricular myocytes was constructed using a mussel-inspired 3D fiber scaffold [113]. Using this platform, researchers found that TiO2 and Ag NPs impaired the contractile function of cardiac tissues by causing structural damage. In another study, a heart-on-a-chip platform was developed by seeding ECs and iPSC-derived cardiomyocytes onto a 3D perfusable and vascularized microfluidic system to evaluate the toxicity of air pollution-related NPs including CuO and SiO2 NPs [114]. Results showed that CuO NPs are highly toxic to cardiac tissue, causing ROS-mediated electrical and contractile dysfunction through penetrating, which further disrupted cardiac troponin T and led to the release of cardiac injury biomarkers like B-type natriuretic peptide, N-terminated pro-hormone BNP, and Troponin I. In contrast, SiO2 NPs primarily induced the secretion of pro-inflammatory cytokines and modulated the intracellular Ca2+ handling.
In addition to the cardiotoxicity induced by NPs, some studies have demonstrated that certain NPs may have protective effects against cardiotoxicity. For instance, curcumin NPs protected male Wistar albino rats from doxorubicin- and cisplatin-induced cardiotoxicity by preventing the chemical-induced oxidative stress and restoring the altered levels and activities of various cardiac parameters [115,116]. Similarly, zero-valent iron NPs functionalized with the biocompatible polymer sodium carboxymethylcellulose significantly protected male BALB/c mice from cecal ligation and puncture-induced septic myocardial injury. This protection was primarily achieved through attenuation of inflammation, inhibition of oxidative stress and apoptosis, improvement of mitochondrial function, regulation of ER stress, and activation of the AMP-activated protein kinase (AMPK) pathway [117]. These findings highlight the complexity of nano-heart interaction, underscoring the need for a more comprehensive and in-depth exploration of NP-disturbed cardiac function.
On the whole, NPs can interact with blood components and various cells within the cardiovascular system, causing toxicity at multiple levels. The induction of cellular stress and inflammation is a common mechanism underlying NP-induced cardiovascular toxicity, although the specific signaling pathways involved can vary significantly. Under real exposure conditions, NP-induced alterations in blood components, damage to vascular structures, and injury to the heart collectively contribute to adverse cardiovascular outcomes, such as arrhythmia, atherosclerosis, myocardial infarction, and other cardiovascular diseases (Figure 2).

Key mechanism involved in NP-induced adverse cardiovascular effects.
4 Critical factors affecting the cardiovascular toxicity of NPs
4.1 Primary physicochemical characteristics of NPs
It is well recognized that the physicochemical properties are key determinants of NP-induced toxicity [118,119]. Composition, particle size, shape, and surface functionalization are the four fundamental characteristics of NPs. This section discusses how these factors affect the cardiovascular toxicity of NPs and how to minimize the NP toxicity based on these four properties. Additionally, the distinct contribution of free metal ions released from metal-based particles is included (Figure 3a).

Critical factors affecting the cardiovascular toxicity of NPs. (a) The primary physicochemical characteristics of NPs. (b) Formation of biomolecular corona on the surface of NPs. (c) Different physiological and pathological conditions of the body. (d) Combined exposure to environmental chemicals.
4.1.1 Composition
A pilot study comparing the differential toxicity of noble metal NPs indicated that Ag NPs are the most toxic, while Au NPs are non-toxic. Both Ag NPs and Au NPs exhibit similar uptake and accumulation in zebrafish embryos, which is higher than platinum (Pt) NPs [120]. Under identical exposure conditions, Ag NPs induce hatching delays, decreased heart rate, impaired touch response, axis curvatures, pericardial effusion, abnormal cardiac morphology, circulatory defects, and eye malformation. Pt NPs, on the other hand, cause hatching delays, decreased heart rate, impaired touch response, and axis curvatures. Furthermore, distinct toxicity to cardiovascular cells is observed among various metal-based NPs including ZnO, Ag, TiO2, Fe2O3, Fe3O4, MgO, and Y2O3 NPs, as summarized previously [121]. For cross-comparison of results from different studies, most types of NPs exhibited procoagulant activity on platelets [122,123,124,125], whereas Ag NPs have been reported to have an anticoagulant effect in several studies [126,127]. Collectively, these studies suggest that composition is crucial in determining the cardiovascular toxicity of NPs.
4.1.2 Particle size
Several studies have shown that smaller NPs tend to be more toxic to the cardiovascular system than larger ones. For instance, 20 nm Fe3O4 NPs exhibit a greater capability to reduce lesion plaque stability and accelerate the progression of atherosclerosis compared to 200 nm Fe3O4 NPs in male C57BL/6J background apolipoprotein E-deficient (ApoE−/−) mice, with the polarization of M1 macrophages associated with the unstable plaques [128]. Similarly, for SiO2 NPs in the size range of 30–600 nm, smaller NPs show higher cardiovascular toxicity in male Wistar rats, and the endothelial NO/NOS system disorder was considered one of the mechanisms involved in SiO2 NP-induced endothelial dysfunction [83]. However, this tendency is not always consistent. A study comparing the cardiovascular toxicity of Ag NPs of varying sizes with capping agents found that 20 nm Ag NPs induced greater cardiac I/R injury in male Sprague–Dawley rats than the 110 nm NPs 1 day following intratracheal instillation, independent of the capping agent [129]. However, the greatest injury occurred in rats exposed to 110 nm polyvinylprryolidone-capped Ag NPs 7 days after intratracheal instillation. Particle size showed minimal effect on NP-triggered toxicity to vascular ECs for ZnO NPs of 20 and 90 nm [130], and nitrodopamide–poly(ethylene glycol)-grafted iron oxide NPs of 3.3, 6.7, and 8.0 nm [131]. While Au NPs (50 nm) showed the highest surface adsorption and internalization by fish RBCs compared to smaller (15 nm) or larger (100 nm) ones [18], Au NPs in the size range of 10–30 nm were considered effective in inducing endothelial leakiness [132]. Hence, there might be a cut-off size for NPs to induce cardiovascular toxicity, which may depend on the properties of NPs and the adverse outcomes measured.
4.1.3 Shape
Shape regulates the cardiovascular fate and toxicity of NPs, particularly during the interaction with ECs in the cardiovascular system. More spherical and rod-like Au NPs are taken up by HUVECs than hollow NPs and core/shell silica@gold nanocrystals with the same coating [133]. Rod-like NPs generally interact more with vascular ECs than spherical NPs under both static and flow conditions. For example, elongated rod-shaped PS NPs demonstrate greater binding ability with cerebral microvasculature cells than the PS spheres [134]. The uptake and elimination of rod-like SiO2 NPs are faster than those of spherical NPs, although their peak accumulations in HUVECs are comparable [51]. Interestingly, larger rod- and disk-shaped PEG NPs show higher uptake than smaller ones, which is opposite to the size effect observed for spherical particles under flow conditions [135]. Utilizing this shape-induced enhancement of endothelial targeting, a pilot study confirmed the possibility of applying antibody-coated rod-shaped PS structures for targeting lung and brain endothelium [136].
4.1.4 Surface coating
Modifying the NP surface can easily alter their physicochemical properties, including electronegativity, hydrophobicity, and even targeting specific proteins. Consequently, surface functionalization has become a popular strategy for designing and synthesizing NPs. Surface chemistry is critical for NP-induced toxicity in the cardiovascular system. For example, in H9c2 cells, cerium oxide NPs showed surface-charge-dependent cell localization and cytotoxicity. Positively charged NPs were the most toxic, although neutral NPs had the highest cellular internalization [137]. While PEG-modified Au nanorods exhibited negligible toxicity to both vascular endothelial and smooth muscle cells because of the lower cellular uptake, rod-like Au NPs coated with polyelectrolyte could be internalized by vascular endothelium, causing cytotoxicity and impairing the relaxation function of aortic rings [138]. In another study comparing the differential adverse effects of SiO2 NPs with various functionalization-amino (NH2-SiO2), polyethyleneimine (PEI-SiO2), succinic acid (SUCC-SiO2), and PEG functionalization (PEG-SiO2). PEI-SiO2 had the most disruptive effect on blood circulation due to its efficient penetration into the zebrafish embryo, followed by SUCC-SiO2 and PEG-SiO2, depending on surface-functionalization-controlled penetrance of biological membranes [139]. Drug coating (e.g., antibodies, RNA) can also impact the toxicity of NPs by altering their surface charge, size, or biocompatibility of NPs, which may, in turn, affect their toxicity to the cardiovascular system. For instance, a noncationic NP coated with a shell of microRNA-146a oligonucleotides, which regulate the NF-κB pathway, can specifically target class A scavenger receptors on plaque macrophages and ECs, offering potential benefits for cardiovascular disease [140]. NPs conjugated with antibodies have been considered as an effective tool for targeted therapy of cardiovascular diseases [14]. However, it has been found recently that antibody-targeted NPs prepared by conjugation chemistries activated the complement cascade of plasma proteins, leading to large changes in the biodistribution of NPs and multiple toxicities, which included a 50% drop in platelet count [141].
4.1.5 Ion release
Metal-based NPs release metal ions into the solution, potentially contributing to cardiovascular toxicity following NP exposure. In a recent study, Ag NPs were found to induce oxidative stress, DNA damage, and apoptosis in the heart and trigger prothrombotic events and alterations in coagulation markers in BALB/c mice [53]. Variations in effects were observed between the particles and Ag+. Specifically, Ag+ increased thrombogenicity and oxidative stress marker without causing DNA damage or apoptosis in the heart, unlike Ag NPs. Some studies attribute observed toxic effects solely to either particles or metal ions. For instance, selenium NPs, but not sodium selenite, were reported to increase mortality, pericardial edema, and cardiac arrhythmia in zebrafish (D. rerio) embryos in a dose-dependent manner [110]. Conversely, Zn2+ not ZnO NPs, contributed to cytotoxicity in H9c2 cells, evidenced by morphological damage, decreased intracellular expression of cardiac protein (troponin I and atrial natriuretic peptide), alterations in mitochondrial membrane potential, and cell death [23]. In a word, distinguishing particle effects from those of the released ions heavily relies on the advancing analytical methods for in situ characterization of the NP state, necessitating future research efforts.
4.2 Secondary modification by biological molecules
Although NPs with varied physicochemical properties are used to evaluate their adverse effects on cells or animals, they are not what the cells exactly “see.” Upon entering the complex physiological environments, NPs adsorb a variety of biomolecules to form biomolecule corona, such as protein corona, which gives the NPs a new identity that determines the nano-bio interactions [142,143]. As a result, secondary modification by biological molecules is crucial in cardiovascular nanotoxicity (Figure 3b).
The formation of protein corona generally reduces the cardiovascular toxicity of NPs. For instance, the pre-formation of a serum protein corona around citrate-capped Au NRs eliminated nanorod-induced hemolysis and morphology change in mouse RBCs, reduced the uptake of nanorods by phagocytic U937 monocytes, and mitigated complement activation induced by citrate-capped Au NRs [144]. The protein corona also mitigated the cytotoxicity and inflammatory response induced by NPs in various types of cardiovascular cells, such as HUVECs [145,146], THP-1 [147,148], and RAW264.7 cells [149,150], by preventing direct interaction of NPs with cell membranes, aggregation and/or degradation of NPs, reducing cellular uptake of NPs, and subsequently decreasing ROS production [143]. In BALB/c mice, the specific adsorption of blood coagulation factor XII prevented the abnormal activation of the coagulation cascade induced by amorphous silica NPs with diameters of 70 nm [151]. In contrast, some studies have suggested that protein adsorption enhances the cardiovascular toxicity of NPs. On the one hand, an inflammatory response was promoted in various circulation-related cells like neutrophils [152], HUVECs [153], THP-1 [154,155], and RAW264.7 cells [153], with the induction of oxidative stress, unfolded protein response, and activation of inflammatory signaling pathways. On the other hand, the enhancement of the NP-activated complement system was confirmed both in vivo [156] and in vitro [157]. This disagreement is understandable due to the extremely complex protein components in blood, which can regulate the properties and bio-effects of NPs in diverse and person-specific manner [46,158].
In addition to governing whole-body circulation, the cardiovascular system serves as a crucial conduit linking various internal organs while biological barriers demarcate them. Limited research has indicated that the protein corona formed on NPs governs barrier crossing, tissue targeting, and NP accumulation [159,160]. Conversely, barrier crossing has been observed to alter the components of the protein corona [161]. Nevertheless, the intricate interplay between barrier crossing and protein corona structure remains insufficiently understood.
4.3 Physiological and pathological conditions of the body
The biological functions of the body vary under different physiological and pathological conditions, leading to distinct responses to environmental contaminant exposures. Consequently, the health risks of environmental contaminants, including NPs, to susceptible population groups particularly vulnerable to pollutants or other environmental hazards [162], have been evaluated recently [163]. For example, in atherosclerosis-prone ApoE−/− mice, exposure to NPs such as nickel NPs [164] and single-walled carbon nanotubes (SWCNTs) [165] resulted in increased plaque formation and accelerated atherosclerosis progression. Inhalation of Zr-doped CeO2 NPs for 4 weeks via nose-only exposure (4 mg/m3 NP for 3 h/day, 5 days/week, and recovery for 4 weeks post the final exposure) triggered lung inflammation in ApoE−/− mice exclusively, without altering the size of atherosclerotic plaques [166]. In perfused hearts isolated from spontaneously diabetic Wistar rats or hypertensive rats, Ag NPs induce sustained vasoconstriction and enhanced cardiac contractility, with or without impacting the NO/NOS system functionality (Figure 4). Both diabetes and hypertension conditions heightened the cardiotoxic effects of Ag NPs [167,168]. A similar accelerated cardiovascular response was observed in a spontaneously hypertensive rat model, demonstrating that SWCNTs induced endothelial dysfunction in the pulmonary circulation and peripheral vascular thrombosis [169]. These results suggest that individuals with preexisting cardiovascular diseases are more vulnerable to NP exposures (Figure 3c).
![Figure 4
Diabetes and hypertension intensified cardiotoxicity because of Ag NPs administration in isolated perfused hearts. (a) Ag NP cardiac effects in diabetes were evaluated using perfused hearts from streptozotocin (STZ)-induced diabetic rats using the Langendorff preparation. STZ-induced diabetic rats showed low basal levels of NO. Ag NPs promoted persistent vasoconstriction, increased cardiac contractility, ROS release, and catalase (CAT) expression. Furthermore, Ag NPs did not alter NO production nor endothelial NO-synthase (eNOS) expression. Reprinted from Ramirez-Lee et al. [167]. Copyright © 2017 Elsevier Inc. (b) Ag NP cardiac effects in hypertension were evaluated using perfused hearts from spontaneously hypertensive rats according to the Langendorff preparation. Ag NPs promoted persistent vasoconstriction and increased cardiac contractility. Furthermore, Ag NPs reduced NO generation by reducing endothelial/inducible NO-synthase expressions. Ag NPs increased ROS release and reduced antioxidant enzyme expression such as CAT and superoxide anion dismutase (SOD). Reprinted from Ramirez-Lee et al. [168]. Copyright © 2017 Elsevier Inc.](/document/doi/10.1515/ntrev-2025-0161/asset/graphic/j_ntrev-2025-0161_fig_004.jpg)
Diabetes and hypertension intensified cardiotoxicity because of Ag NPs administration in isolated perfused hearts. (a) Ag NP cardiac effects in diabetes were evaluated using perfused hearts from streptozotocin (STZ)-induced diabetic rats using the Langendorff preparation. STZ-induced diabetic rats showed low basal levels of NO. Ag NPs promoted persistent vasoconstriction, increased cardiac contractility, ROS release, and catalase (CAT) expression. Furthermore, Ag NPs did not alter NO production nor endothelial NO-synthase (eNOS) expression. Reprinted from Ramirez-Lee et al. [167]. Copyright © 2017 Elsevier Inc. (b) Ag NP cardiac effects in hypertension were evaluated using perfused hearts from spontaneously hypertensive rats according to the Langendorff preparation. Ag NPs promoted persistent vasoconstriction and increased cardiac contractility. Furthermore, Ag NPs reduced NO generation by reducing endothelial/inducible NO-synthase expressions. Ag NPs increased ROS release and reduced antioxidant enzyme expression such as CAT and superoxide anion dismutase (SOD). Reprinted from Ramirez-Lee et al. [168]. Copyright © 2017 Elsevier Inc.
NPs also exhibit increased toxicity to the cardiovascular system in other susceptible populations. For instance, carbon black [170], carbon nanotubes [171], and TiO2 NPs [172] impaired the coagulatory functions of mice with pulmonary inflammation, likely due to enhanced permeability in the pulmonary vessel. RBCs isolated from cancer patients were more sensitive to the procoagulant effects of Ag NPs [173]. In a study comparing the differential toxicity of SiO2 NPs in male Sprague–Dawley rats at ages 3 weeks, 8 weeks, and 20 months – representing young, adult, and old age groups – myocardial ischemic damage, atrioventricular blockages, and increased fibrinogen concentration and blood viscosity were observed exclusively in old age rats [174].
4.4 Combination with other environmental chemicals
In general, NPs do not exist independently but are combined with other organic or inorganic chemicals in the natural environment. It is well-known that the surface properties and biological performance of NPs can be modified by co-existing environmental chemicals [175]. However, the combined effect of NPs and other environmental chemicals is often overlooked [176]. Understanding these interactions is crucial for assessing the real-world risks associated with NP exposure in environments where multiple chemicals coexist. It underscores the importance of evaluating not only individual NP toxicity but also considering their interactions with other environmental stressors for comprehensive cardiovascular risk assessment and mitigation strategies (Figure 3d).
To date, various studies have shown that NPs and their bound chemicals can independently induce oxidative stress and inflammation in cardiovascular tissues. Co-exposure may exacerbate these effects through synergistic interactions, intensifying cellular damage and impairing cardiovascular function. For example, co-exposure with methylmercury further aggravates SiO2 NP-induced bradycardia, vascular endothelial damage, and neutrophil-mediated inflammation in zebrafish (D. rerio) embryos. Oxidative stress and genes including stxbp1a, celf4, ahr1b, and bai2 are associated with co-exposure-induced cardiovascular toxicity, and cardiac muscle contraction is targeted by dre-miR-7147, dre-miR-26a, and dre-miR-375 [177,178]. In another study, combined exposure to SiO2 NPs and CdCl2 caused more severe cardiovascular damage, indicated by altered heart morphology, reduced heart rate, and damaged vascular ECs through oxidative stress, cellular apoptosis, and inflammation [179]. In male Sprague–Dawley rats, co-administration of SiO2 NPs with lead acetate or methylmercury also led to additive or synergistic cardiovascular toxicity, evidenced by endothelial damage, hypercoagulation, myocardial interstitial edema, and ultrastructural changes such as myofibril disorder, myocardial gap expansion, and mitochondrial damage [180,181]. Similar findings were observed when NPs were co-exposed with 2,2′,4,4′,5,5′-hexachlorobiphenyl (PCB153), a typical organic environmental contaminant, exhibiting increased stroke volume and potentiated brain damage in C57BL/6 mice subjected to I/R [182].
Despite the enhanced cardiotoxicity of NPs and environmental contaminants, antagonistic effects have also been reported. A recent study showed that PS NPs could adsorb the polycyclic aromatic hydrocarbons (PAHs) from the exposure medium, decreasing the bioavailability and bioaccumulation of PAHs, and consequently diminishing PAH-induced cardiovascular toxicity in developing zebrafish [183]. Taken together, the complex interactions between NPs and other environmental chemicals require more comprehensive investigation in future research. Specifically, the effects of co-existing natural organic matter and inorganic ligands, beyond conventional contaminants, on NP-induced cardiovascular toxicity should be carefully evaluated.
5 Gaps and insight
Besides the extensive toxicity/safety evaluation of NPs in relation to human health and environmental safety, so far, whether therapeutic NPs will provide more benefits or pose greater toxicity is a critical issue in the field of nanomedicine. On the one hand, therapeutic NPs can improve the efficacy and bioavailability of free drugs and minimize systemic side effects, which might result in better therapy outcomes. However, concerns remain regarding the toxicity of the NPs themselves and their interaction with the body. For example, some NPs may accumulate in certain organs (like the liver or spleen) over time leading to adverse effects. Furthermore, the drug release profile from NPs may not always be as predictable or controllable as expected, which could result in off-target effects. As mentioned above, the properties of the NPs, such as size, surface charge, and material composition, influence their biocompatibility, toxicity, and other nano-bio interactions. Therefore, specific design strategies can be employed to increase the efficacy/toxicity ratio of NPs. For instance, surface modification is the most commonly used strategy for minimizing NP toxicity. Functionalizing NPs with biocompatible polymers (e.g., PEGylation) or preparation biomimetic NPs with natural molecules (e.g., albumin, chitosan, cell membrane, and extracellular vesicles) can evade the immune system, extend the circulation time, and increase therapeutic efficacy.
Despite these research advances, several specific gaps in current knowledge warrant immediate attention, such as the influence on cardiac electrophysiology, especially their effects on arrhythmogenic potential and conduction system. Understanding and evaluating how NP can affect cardiac electrical activity, cardiac conduction system (e.g., atrioventricular node and Purkinje fibers) and impulse propagation is crucial for assessing potential cardiovascular toxicity. Another special gap is the NP-bio interactions with pre-existing cardiovascular conditions. Studying how NPs interact with cardiovascular tissues already compromised by diseases (e.g., hypertension and atherosclerosis) is vital. Such studies can determine whether NPs exacerbate existing conditions or induce novel pathophysiological responses.
Moreover, knowledge gaps regarding the fate and adverse impacts of NPs on the human cardiovascular system remain to be elucidated, primarily due to limited direct evidence on the health risks of NPs exposure in humans. Addressing these gaps requires interdisciplinary research efforts integrating nanotechnology, cardiovascular biology, toxicology, and clinical medicine. Robust preclinical models, advanced imaging techniques, and biomarker assessments are essential tools to advance our understanding of NP cardiovascular distribution and toxicity, ultimately facilitating safer clinical translation of NP-based therapies and diagnostics.
6 Conclusion
The increased applications of nanotechnology have led to innovations in various fields; however, serious concerns are rapidly arising regarding NP-induced potential toxicity to public health. NPs entering the circulation can interact with various components of the cardiovascular system at multiple levels, leading to cardiovascular toxicity. Both the synthetic identity including composition, size, shape, surface coating, Ion release, and biological identity (after absorption with biological molecules) of NPs, as well as the exposure dose and time affect the NP-induced cardiovascular toxicity. However, it is worth noting that the real-world NP exposure scenarios differ significantly from controlled laboratory conditions. In the real world, organisms are exposed to a complex mixture of NPs, which can interact differently with biological systems compared to simplified NPs used in controlled experiments; Low-dose and chronic exposure would contribute to the potential accumulation of NPs in tissues and organs over time. Moreover, the combined effects of NPs and other environmental pollutants or stressors (e.g., heavy metals and organic pollutants) on human health are often overlooked. On the other hand, specific design strategies can be employed to mitigate adverse biological effects while preserving the intended therapeutic and diagnostic benefits of NPs. For instance, modulating surface charge and hydrophilicity can influence NP biodistribution and cellular uptake, potentially minimizing toxicity by reducing off-target interactions and enhancing biocompatibility. Optimizing NP size and shape, and choosing biocompatible core materials are also crucial to avoid harmful effects. Collectively, these strategies advance the development of safer and more effective nanomedicines and diagnostic tools, contributing to improved outcomes in biomedical applications.
-
Funding information: The work was supported by the National Key R&D Program of China (2023YFA0915103) and the Shandong Provincial Key Research and Development Plan (2021CXGC011306).
-
Author contributions: All authors have accepted responsibility for the entire content of this manuscript and approved its submission.
-
Conflict of interest: The authors state no conflict of interest.
-
Data availability statement: Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.
References
[1] Osherov A, Prasad R, Chrzanowski W, New EJ, Brazaca L, Sadik O, et al. Responsible nanotechnology for a sustainable future. One Earth. 2023;6:763–66.10.1016/j.oneear.2023.06.010Suche in Google Scholar
[2] Squire HJ, Tomatz S, Voke E, González-Grandío E, Landry M. The emerging role of nanotechnology in plant genetic engineering. Nat Rev Bioeng. 2023;1:314–28.10.1038/s44222-023-00037-5Suche in Google Scholar
[3] Ren H, Jia W, Xie Y, Yu M, Chen Y. Adjuvant physiochemistry and advanced nanotechnology for vaccine development. Chem Soc Rev. 2023;52:5172–254.10.1039/D2CS00848CSuche in Google Scholar
[4] Liu J, Yuan S, Bremmer A, Hu Q. Convergence of nanotechnology and bacteriotherapy for biomedical applications. Adv Sci. 2024;11:2309295.10.1002/advs.202309295Suche in Google Scholar PubMed PubMed Central
[5] Malakar A, Kanel SR, Ray C, Snow DD, Nadagouda MN. Nanomaterials in the environment, human exposure pathway, and health effects: A review. Sci Total Environ. 2021;759:143470.10.1016/j.scitotenv.2020.143470Suche in Google Scholar PubMed
[6] Fadeel B, Keller AA. Nanosafety: A perspective on nano-bio interactions. Small. 2024;20:2310540.10.1002/smll.202310540Suche in Google Scholar PubMed
[7] Bondarenko O, Mortimer M, Kahru A, Feliu N, Javed I, Kakinen A, et al. Nanotoxicology and nanomedicine: The Yin and Yang of nano-bio interactions for the new decade. Nano Today. 2021;39:101184.10.1016/j.nantod.2021.101184Suche in Google Scholar PubMed PubMed Central
[8] Elsaesser A, Howard CV. Toxicology of nanoparticles. Adv Drug Delivery Rev. 2012;64:129–37.10.1016/j.addr.2011.09.001Suche in Google Scholar PubMed
[9] Karimi M, Sadeghi R, Kokini J. Human exposure to nanoparticles through trophic transfer and the biosafety concerns that nanoparticle-contaminated foods pose to consumers. Trends Food Sci Technol. 2018;75:129–45.10.1016/j.tifs.2018.03.012Suche in Google Scholar
[10] Jia J, Wang Z, Yue T, Su G, Teng C, Yan B. Crossing biological barriers by engineered nanoparticles. Chem Res Toxicol. 2020;33:1055–60.10.1021/acs.chemrestox.9b00483Suche in Google Scholar PubMed
[11] Meng H, Leong W, Leong KW, Chen C, Zhao Y. Walking the line: The fate of nanomaterials at biological barriers. Biomaterials. 2018;174:41–53.10.1016/j.biomaterials.2018.04.056Suche in Google Scholar PubMed PubMed Central
[12] Wang R, Zhang Z, Liu B, Xue J, Liu F, Tang T, et al. Strategies for the design of nanoparticles: starting with long-circulating nanoparticles, from lab to clinic. Biomater Sci. 2021;9:3621–37.10.1039/D0BM02221GSuche in Google Scholar PubMed
[13] Haroon HB, Hunter AC, Farhangrazi ZS, Moghimi SM. A brief history of long circulating nanoparticles. Adv Drug Delivery Rev. 2022;188:114396.10.1016/j.addr.2022.114396Suche in Google Scholar PubMed
[14] Gao W, Sun YH, Cai M, Zhao YJ, Cao WH, Liu ZH, et al. Copper sulfide nanoparticles as a photothermal switch for TRPV1 signaling to attenuate atherosclerosis. Nat Commun. 2018;9:231.10.1038/s41467-017-02657-zSuche in Google Scholar PubMed PubMed Central
[15] Etemad L, Salmasi Z, Kalat SAM, Moshiri M, Zamanian J, Kesharwani P, et al. An overview on nanoplatforms for statins delivery: Perspectives for safe and effective therapy. Environ Res. 2023;234:116572.10.1016/j.envres.2023.116572Suche in Google Scholar PubMed
[16] Chen W, Schilperoort M, Cao YH, Shi JJ, Tabas I, Tao W. Macrophage-targeted nanomedicine for the diagnosis and treatment of atherosclerosis. Nat Rev Cardiol. 2022;19:228–49.10.1038/s41569-021-00629-xSuche in Google Scholar PubMed PubMed Central
[17] Yang Y, Qina Z, Zeng W, Yang T, Cao YB, Mei CR, et al. Toxicity assessment of nanoparticles in various systems and organs. Nanotechnol Rev. 2017;6:279–89.10.1515/ntrev-2016-0047Suche in Google Scholar
[18] Chen LQ, Fang L, Ling J, Ding CZ, Kang B, Huang CZ. Nanotoxicity of silver nanoparticles to red blood cells: Size dependent adsorption, uptake, and hemolytic activity. Chem Res Toxicol. 2015;28:501–9.10.1021/tx500479mSuche in Google Scholar PubMed
[19] Zhao Y, Sun X, Zhang G, Trewyn BG, Slowing II, Lin VSY. Interaction of mesoporous silica nanoparticles with human red blood cell membranes: Size and surface effects. ACS Nano. 2011;5:1366–75.10.1021/nn103077kSuche in Google Scholar PubMed
[20] Su L, Han L, Ge F, Zhang SL, Zhang Y, Zhao BX, et al. The effect of novel magnetic nanoparticles on vascular endothelial cell function in vitro and in vivo. J Hazard Mater. 2012;235-236:316–25.10.1016/j.jhazmat.2012.08.003Suche in Google Scholar PubMed
[21] Qin X, Zhang J, Wang B, Xu G, Yang X, Zou Z, et al. Ferritinophagy is involved in the zinc oxide nanoparticles-induced ferroptosis of vascular endothelial cells. Autophagy. 2021;17:4266–85.10.1080/15548627.2021.1911016Suche in Google Scholar PubMed PubMed Central
[22] Cui G, Li Z, Cao F, Li P, Jin M, Hou S, et al. Activation of Nrf2/HO-1 signaling pathway attenuates ROS-mediated autophagy induced by silica nanoparticles in H9c2 cells. Environ Toxicol. 2021;36:1389–401.10.1002/tox.23134Suche in Google Scholar PubMed
[23] Mendoza-Milla C, Macías Macías FI, Velázquez Delgado KA, Herrera Rodríguez MA, Colín-Val Z, Ramos-Godinez MdP, et al. Zinc oxide nanoparticles induce toxicity in H9c2 rat cardiomyoblasts. Int J Mol Sci. 2022;23:12940.10.3390/ijms232112940Suche in Google Scholar PubMed PubMed Central
[24] Guo C, Liu Y, Li Y. Adverse effects of amorphous silica nanoparticles: Focus on human cardiovascular health. J Hazard Mater. 2021;406:124626.10.1016/j.jhazmat.2020.124626Suche in Google Scholar PubMed
[25] Gonzalez C, Rosas-Hernandez H, Ramirez-Lee MA, Salazar-García S, Ali SF. Role of silver nanoparticles (AgNPs) on the cardiovascular system. Arch Toxicol. 2016;90:493–511.10.1007/s00204-014-1447-8Suche in Google Scholar PubMed
[26] Huang CF, Liu XT, Wu QC, Cao JZ, Zhu XY, Wang XY, et al. Cardiovascular toxic effects of nanoparticles and corresponding molecular mechanisms. Environ Pollut. 2024;356:124360.10.1016/j.envpol.2024.124360Suche in Google Scholar PubMed
[27] Cheng Y, Chen Z, Yang S, Liu T, Yin L, Pu Y, et al. Nanomaterials-induced toxicity on cardiac myocytes and tissues, and emerging toxicity assessment techniques. Sci Total Environ. 2021;800:149584.10.1016/j.scitotenv.2021.149584Suche in Google Scholar PubMed
[28] Wang Z, Tang M. Research progress on toxicity, function, and mechanism of metal oxide nanoparticles on vascular endothelial cells. J Appl Toxicol. 2021;41:683–700.10.1002/jat.4121Suche in Google Scholar PubMed
[29] Wei Y, Quan L, Zhou C, Zhan Q. Factors relating to the biodistribution & clearance of nanoparticles & their effects on in vivo application. Nanomedicine. 2018;13:1495–512.10.2217/nnm-2018-0040Suche in Google Scholar PubMed
[30] Kaga S, Truong NP, Esser L, Senyschyn D, Sanyal A, Sanyal R, et al. Influence of size and shape on the biodistribution of nanoparticles prepared by polymerization-induced self-assembly. Biomacromolecules. 2017;18:3963–70.10.1021/acs.biomac.7b00995Suche in Google Scholar PubMed
[31] Zhu X, Wang C, Duan X, Liang B, Genbo Xu E, Huang Z. Micro- and nanoplastics: A new cardiovascular risk factor? Environ Int. 2023;171:107662.10.1016/j.envint.2022.107662Suche in Google Scholar PubMed
[32] Khlebtsov N, Dykman L. Biodistribution and toxicity of engineered gold nanoparticles: a review of in vitro and in vivo studies. Chem Soc Rev. 2011;40:1647–71.10.1039/C0CS00018CSuche in Google Scholar
[33] Jia J, Li F, Zhou H, Bai Y, Liu S, Jiang Y, et al. Oral exposure to silver nanoparticles or silver ions may aggravate fatty liver disease in overweight mice. Environ Sci Technol. 2017;51:9334–43.10.1021/acs.est.7b02752Suche in Google Scholar PubMed
[34] Yang K, Gong H, Shi X, Wan J, Zhang Y, Liu Z. In vivo biodistribution and toxicology of functionalized nano-graphene oxide in mice after oral and intraperitoneal administration. Biomaterials. 2013;34:2787–95.10.1016/j.biomaterials.2013.01.001Suche in Google Scholar PubMed
[35] Czarny B, Georgin D, Berthon F, Plastow G, Pinault M, Patriarche G, et al. Carbon nanotube translocation to distant organs after pulmonary exposure: Insights from in Situ 14C-radiolabeling and tissue radioimaging. ACS Nano. 2014;8:5715–24.10.1021/nn500475uSuche in Google Scholar PubMed
[36] Xie G, Sun J, Zhong G, Shi L, Zhang D. Biodistribution and toxicity of intravenously administered silica nanoparticles in mice. Arch Toxicol. 2010;84:183–90.10.1007/s00204-009-0488-xSuche in Google Scholar PubMed
[37] Yu T, Hubbard D, Ray A, Ghandehari H. In vivo biodistribution and pharmacokinetics of silica nanoparticles as a function of geometry, porosity and surface characteristics. J Controlled Rel. 2012;163:46–54.10.1016/j.jconrel.2012.05.046Suche in Google Scholar PubMed PubMed Central
[38] Li Z, Zhu S, Liu Q, Wei J, Jin Y, Wang X, et al. Polystyrene microplastics cause cardiac fibrosis by activating Wnt/β-catenin signaling pathway and promoting cardiomyocyte apoptosis in rats. Environ Pollut. 2020;265:115025.10.1016/j.envpol.2020.115025Suche in Google Scholar PubMed
[39] Ferreira MPA, Ranjan S, Correia AMR, Mäkilä EM, Kinnunen SM, Zhang H, et al. In vitro and in vivo assessment of heart-homing porous silicon nanoparticles. Biomaterials. 2016;94:93–104.10.1016/j.biomaterials.2016.03.046Suche in Google Scholar PubMed
[40] Papa AL, Dumont L, Vandroux D, Millot N. Titanate nanotubes: Towards a novel and safer nanovector for cardiomyocytes. Nanotoxicology. 2013;7:1131–42.10.3109/17435390.2012.710661Suche in Google Scholar PubMed
[41] Komuro H, Yamazoe M, Nozaki K, Nagai A, Sasano T. Cardiomyocyte uptake mechanism of a hydroxyapatite nanoparticle mediated gene delivery system. Beilstein J Nanotechnol. 2020;11:1685–92.10.3762/bjnano.11.150Suche in Google Scholar PubMed PubMed Central
[42] He R, Ding X, Zhang T, Mei L, Zhu S, Wang C, et al. Study on myocardial toxicity induced by lead halide perovskites nanoparticles. Nanotoxicology. 2023;17:449–70.10.1080/17435390.2023.2255269Suche in Google Scholar PubMed
[43] Huerta-García E, Zepeda-Quiroz I, Sánchez-Barrera H, Colín-Val Z, Alfaro-Moreno E, Ramos-Godinez MDP, et al. Internalization of titanium dioxide nanoparticles is cytotoxic for H9c2 rat cardiomyoblasts. Molecules. 2018;23:1955.10.3390/molecules23081955Suche in Google Scholar PubMed PubMed Central
[44] Zhao X, Lu D, Liu QS, Li Y, Feng R, Hao F, et al. Hematological effects of gold nanorods on erythrocytes: Hemolysis and hemoglobin conformational and functional changes. Adv Sci. 2017;4:1700296.10.1002/advs.201700296Suche in Google Scholar PubMed PubMed Central
[45] Maitz MF, Sperling C, Wongpinyochit T, Herklotz M, Werner C, Seib FP. Biocompatibility assessment of silk nanoparticles: Hemocompatibility and internalization by human blood cells. Nanomed: Nanotechnol Biol Med. 2017;13:2633–42.10.1016/j.nano.2017.07.012Suche in Google Scholar PubMed
[46] Ju Y, Kelly HG, Dagley LF, Reynaldi A, Schlub TE, Spall SK, et al. Person-specific biomolecular coronas modulate nanoparticle interactions with immune cells in human blood. ACS Nano. 2020;14:15723–37.10.1021/acsnano.0c06679Suche in Google Scholar PubMed
[47] Hao F, Liu QS, Chen X, Zhao X, Zhou Q, Liao C, et al. Exploring the heterogeneity of nanoparticles in their interactions with plasma coagulation factor XII. ACS Nano. 2019;13:1990–2003.10.1021/acsnano.8b08471Suche in Google Scholar PubMed
[48] Gomez-Garcia MJ, Doiron AL, Steele RRM, Labouta HI, Vafadar B, Shepherd RD, et al. Nanoparticle localization in blood vessels: Dependence on fluid shear stress, flow disturbances, and flow-induced changes in endothelial physiology. Nanoscale. 2018;10:15249–61.10.1039/C8NR03440KSuche in Google Scholar PubMed
[49] Setyawati MI, Tay CY, Docter D, Stauber RH, Leong DT. Understanding and exploiting nanoparticles’ intimacy with the blood vessel and blood. Chem Soc Rev. 2015;44:8174–99.10.1039/C5CS00499CSuche in Google Scholar PubMed
[50] Chen YY, Syed AM, MacMillan P, Rocheleau JV, Chan WCW. Flow rate affects nanoparticle uptake into endothelial cells. Adv Mater. 2020;32:1906274.10.1002/adma.201906274Suche in Google Scholar PubMed
[51] Cheng Y, Tao J, Zhang Y, Xi L, Han R, Xu M, et al. Shape and shear stress impact on the toxicity of mesoporous silica nanoparticles: In vitro and in vivo evidence. Mol Pharm. 2023;20:3187–201.10.1021/acs.molpharmaceut.3c00180Suche in Google Scholar PubMed
[52] Qi M, Wang X, Chen J, Liu Y, Liu Y, Jia J, et al. Transformation, absorption and toxicological mechanisms of silver nanoparticles in the gastrointestinal tract following oral exposure. ACS Nano. 2023;17:8851–65.10.1021/acsnano.3c00024Suche in Google Scholar PubMed
[53] Ferdous Z, Al-Salam S, Greish YE, Ali BH, Nemmar A. Pulmonary exposure to silver nanoparticles impairs cardiovascular homeostasis: Effects of coating, dose and time. Toxicol Appl Pharmacol. 2019;367:36–50.10.1016/j.taap.2019.01.006Suche in Google Scholar PubMed
[54] Krajewski S, Prucek R, Panacek A, Avci-Adali M, Nolte A, Straub A, et al. Hemocompatibility evaluation of different silver nanoparticle concentrations employing a modified Chandler-loop in vitro assay on human blood. Acta Biomater. 2013;9:7460–68.10.1016/j.actbio.2013.03.016Suche in Google Scholar PubMed
[55] Khanbeigi RA, Hashim Z, Abelha TF, Pitchford S, Collins H, Green M, et al. Interactions of stealth conjugated polymer nanoparticles with human whole blood. J Mater Chem B. 2015;3:2463–71.10.1039/C4TB01822BSuche in Google Scholar PubMed
[56] Lategan K, Alghadi H, Bayati M, De Cortalezzi MF, Pool E. Effects of graphene oxide nanoparticles on the immune system biomarkers produced by RAW 264.7 and human whole blood cell cultures. Nanomaterials. 2018;8:125.10.3390/nano8020125Suche in Google Scholar PubMed PubMed Central
[57] Kolter M, Ott M, Hauer C, Reimold I, Fricker G. Nanotoxicity of poly(n-butylcyano-acrylate) nanoparticles at the blood–brain barrier, in human whole blood and in vivo. J Controlled Rel. 2015;197:165–79.10.1016/j.jconrel.2014.11.005Suche in Google Scholar PubMed
[58] Kozelskaya AI, Panin AV, Khlusov IA, Mokrushnikov PV, Zaitsev BN, Kuzmenko DI, et al. Morphological changes of the red blood cells treated with metal oxide nanoparticles. Toxicol Vitro. 2016;37:34–40.10.1016/j.tiv.2016.08.012Suche in Google Scholar PubMed
[59] Ghosh M, Chakraborty A, Mukherjee A. Cytotoxic, genotoxic and the hemolytic effect of titanium dioxide (TiO2) nanoparticles on human erythrocyte and lymphocyte cells in vitro. J Appl Toxicol. 2013;33:1097–110.10.1002/jat.2863Suche in Google Scholar PubMed
[60] Maurer-Jones MA, Lin Y-S, Haynes CL. Functional assessment of metal oxide nanoparticle toxicity in immune cells. ACS Nano. 2010;4:3363–73.10.1021/nn9018834Suche in Google Scholar PubMed
[61] Chen LQ, Kang B, Ling J. Cytotoxicity of cuprous oxide nanoparticles to fish blood cells: Hemolysis and internalization. J Nanopart Res. 2013;15:1507.10.1007/s11051-013-1507-7Suche in Google Scholar
[62] Li S-Q, Zhu R-R, Zhu H, Xue M, Sun X-Y, Yao S-D, et al. Nanotoxicity of TiO2 nanoparticles to erythrocyte in vitro. Food Chem Toxicol. 2008;46:3626–31.10.1016/j.fct.2008.09.012Suche in Google Scholar PubMed
[63] Andersson-Willman B, Gehrmann U, Cansu Z, Buerki-Thurnherr T, Krug HF, Gabrielsson S, et al. Effects of subtoxic concentrations of TiO2 and ZnO nanoparticles on human lymphocytes, dendritic cells and exosome production. Toxicol Appl Pharmacol. 2012;264:94–103.10.1016/j.taap.2012.07.021Suche in Google Scholar PubMed
[64] Lankoff A, Arabski M, Wegierek-Ciuk A, Kruszewski M, Lisowska H, Banasik-Nowak A, et al. Effect of surface modification of silica nanoparticles on toxicity and cellular uptake by human peripheral blood lymphocytes in vitro. Nanotoxicology. 2013;7:235–50.10.3109/17435390.2011.649796Suche in Google Scholar PubMed
[65] Ghosh M, Sinha S, Jothiramajayam M, Jana A, Nag A, Mukherjee A. Cyto-genotoxicity and oxidative stress induced by zinc oxide nanoparticle in human lymphocyte cells in vitro and Swiss albino male mice in vivo. Food Chem Toxicol. 2016;97:286–96.10.1016/j.fct.2016.09.025Suche in Google Scholar PubMed
[66] Dworak N, Wnuk M, Zebrowski J, Bartosz G, Lewinska A. Genotoxic and mutagenic activity of diamond nanoparticles in human peripheral lymphocytes in vitro. Carbon. 2014;68:763–76.10.1016/j.carbon.2013.11.067Suche in Google Scholar
[67] Gaharwar US, Meena R, Rajamani P. Iron oxide nanoparticles induced cytotoxicity, oxidative stress and DNA damage in lymphocytes. J Appl Toxicol. 2017;37:1232–44.10.1002/jat.3485Suche in Google Scholar PubMed
[68] Senapati VA, Kumar A, Gupta GS, Pandey AK, Dhawan A. ZnO nanoparticles induced inflammatory response and genotoxicity in human blood cells: A mechanistic approach. Food Chem Toxicol. 2015;85:61–70.10.1016/j.fct.2015.06.018Suche in Google Scholar PubMed
[69] Ding Z, Zhang Z, Ma H, Chen Y. In vitro hemocompatibility and toxic mechanism of graphene oxide on human peripheral blood T lymphocytes and serum albumin. ACS Appl Mater Interfaces. 2014;6:19797–807.10.1021/am505084sSuche in Google Scholar PubMed
[70] Assadian E, Zarei MH, Gilani AG, Farshin M, Degampanah H, Pourahmad J. Toxicity of copper oxide (CuO) nanoparticles on human blood lymphocytes. Biol Trace Elem Res. 2018;184:350–57.10.1007/s12011-017-1170-4Suche in Google Scholar PubMed
[71] Matus MF, Vilos C, Cisterna BA, Fuentes E, Palomo I. Nanotechnology and primary hemostasis: Differential effects of nanoparticles on platelet responses. Vasc Pharmacol. 2018;101:1–8.10.1016/j.vph.2017.11.004Suche in Google Scholar PubMed
[72] Asaad Y, Nemcovsky-Amar D, Sznitman J, Mangin PH, Korin N. A double-edged sword: The complex interplay between engineered nanoparticles and platelets. Bioeng Transl Med. 2024;9:e10669.10.1002/btm2.10669Suche in Google Scholar PubMed PubMed Central
[73] Docter D, Westmeier D, Markiewicz M, Stolte S, Knauer SK, Stauber RH. The nanoparticle biomolecule corona: Lessons learned – challenge accepted? Chem Soc Rev. 2015;44:6094–121.10.1039/C5CS00217FSuche in Google Scholar
[74] Liam-Or R, Faruqu FN, Walters A, Han S, Xu L, Wang JT-W, et al. Cellular uptake and in vivo distribution of mesenchymal-stem-cell-derived extracellular vesicles are protein corona dependent. Nat Nanotechnol. 2024;19:846–55.10.1038/s41565-023-01585-ySuche in Google Scholar PubMed PubMed Central
[75] Monopoli MP, Åberg C, Salvati A, Dawson KA. Biomolecular coronas provide the biological identity of nanosized materials. Nat Nanotechnol. 2012;7:779–86.10.1038/nnano.2012.207Suche in Google Scholar PubMed
[76] Liu X, Wei W, Liu Z, Song E, Lou J, Feng L, et al. Serum apolipoprotein A-I depletion is causative to silica nanoparticles–induced cardiovascular damage. Proc Natl Acad Sci. 2021;118:e2108131118.10.1073/pnas.2108131118Suche in Google Scholar PubMed PubMed Central
[77] Tiyyagura HR, Rudolf R, Bracic M. Polyvinylpyrrolidone-stabilised gold nanoparticle coatings inhibit blood protein adsorption. Nanotechnol Rev. 2024;13:20230176.10.1515/ntrev-2023-0176Suche in Google Scholar
[78] Michiels C. Endothelial cell functions. J Cell Physiol. 2003;196:430–43.10.1002/jcp.10333Suche in Google Scholar PubMed
[79] Pate M, Damarla V, Chi DS, Negi S, Krishnaswamy G. Chapter 5 - Endothelial cell biology: Role in the inflammatory response. In: Makowski GS, editor. Advances in clinical chemistry. Amsterdam: Elsevier; 2010. p. 109–30.10.1016/S0065-2423(10)52004-3Suche in Google Scholar
[80] Krüger-Genge A, Blocki A, Franke R-P, Jung F. Vascular endothelial cell biology: An update. Int J Mol Sci. 2019;20:4411.10.3390/ijms20184411Suche in Google Scholar PubMed PubMed Central
[81] Wierzbicki M, Zawadzka K, Wójcik B, Jaworski S, Strojny B, Ostrowska A, et al. Differences in the cell type-specific toxicity of diamond nanoparticles to endothelial cells depending on the exposure of the cells to nanoparticles. Int J Nanomed. 2023;18:2821–38.10.2147/IJN.S411424Suche in Google Scholar PubMed PubMed Central
[82] Wei W, Li Y, Lee M, Andrikopoulos N, Lin S, Chen C, et al. Anionic nanoplastic exposure induces endothelial leakiness. Nat Commun. 2022;13:4757.10.1038/s41467-022-32532-5Suche in Google Scholar PubMed PubMed Central
[83] Du Z, Zhao D, Jing L, Cui G, Jin M, Li Y, et al. Cardiovascular toxicity of different sizes amorphous silica nanoparticles in rats after intratracheal instillation. Cardiovasc Toxicol. 2013;13:194–207.10.1007/s12012-013-9198-ySuche in Google Scholar PubMed
[84] Duan J, Yu Y, Yu Y, Li Y, Huang P, Zhou X, et al. Silica nanoparticles enhance autophagic activity, disturb endothelial cell homeostasis and impair angiogenesis. Part Fibre Toxicol. 2014;11:50.10.1186/s12989-014-0050-8Suche in Google Scholar PubMed PubMed Central
[85] Duan J, Yu Y, Li Y, Yu Y, Sun Z. Cardiovascular toxicity evaluation of silica nanoparticles in endothelial cells and zebrafish model. Biomaterials. 2013;34:5853–62.10.1016/j.biomaterials.2013.04.032Suche in Google Scholar PubMed
[86] Bangeppagari M, Park SH, Kundapur RR, Lee SJ. Graphene oxide induces cardiovascular defects in developing zebrafish (Danio rerio) embryo model: In-vivo toxicity assessment. Sci Total Environ. 2019;673:810–20.10.1016/j.scitotenv.2019.04.082Suche in Google Scholar PubMed
[87] Zhang L, Wang X, Miao Y, Chen Z, Qiang P, Cui L, et al. Magnetic ferroferric oxide nanoparticles induce vascular endothelial cell dysfunction and inflammation by disturbing autophagy. J Hazard Mater. 2016;304:186–95.10.1016/j.jhazmat.2015.10.041Suche in Google Scholar PubMed
[88] Guo H, Zhang J, Boudreau M, Meng J, Yin J-J, Liu J, et al. Intravenous administration of silver nanoparticles causes organ toxicity through intracellular ROS-related loss of inter-endothelial junction. Part Fibre Toxicol. 2016;13:21.10.1186/s12989-016-0133-9Suche in Google Scholar PubMed PubMed Central
[89] Li Y, Ma R, Liu X, Qi Y, Abulikemu A, Zhao X, et al. Endoplasmic reticulum stress-dependent oxidative stress mediated vascular injury induced by silica nanoparticles in vivo and in vitro. NanoImpact. 2019;14:100169.10.1016/j.impact.2019.100169Suche in Google Scholar
[90] Wang Y, Wu T, Zou L, Xiong L, Zhang T, Kong L, et al. Genome-wide identification and functional analysis of long non-coding RNAs in human endothelial cell line after incubation with PM2.5. Chemosphere. 2019;216:396–403.10.1016/j.chemosphere.2018.10.101Suche in Google Scholar PubMed
[91] Feng L, Yang X, Liang S, Xu Q, Miller MR, Duan J, et al. Silica nanoparticles trigger the vascular endothelial dysfunction and prethrombotic state via miR-451 directly regulating the IL6R signaling pathway. Part Fibre Toxicol. 2019;16:16.10.1186/s12989-019-0300-xSuche in Google Scholar PubMed PubMed Central
[92] Duan J, Yu Y, Yu Y, Li Y, Wang J, Geng W, et al. Silica nanoparticles induce autophagy and endothelial dysfunction via the PI3K/Akt/mTOR signaling pathway. Int J Nanomed. 2014;9:5131–41.10.2147/IJN.S71074Suche in Google Scholar PubMed PubMed Central
[93] Guo C, Yang M, Jing L, Wang J, Yu Y, Li Y, et al. Amorphous silica nanoparticles trigger vascular endothelial cell injury through apoptosis and autophagy via reactive oxygen species-mediated MAPK/Bcl-2 and PI3K/Akt/mTOR signaling. Int J Nanomed. 2016;11:5257–76.10.2147/IJN.S112030Suche in Google Scholar PubMed PubMed Central
[94] He J, Ma Y, Niu X, Pei J, Yan R, Xu F, et al. Silver nanoparticles induce endothelial cytotoxicity through ROS-mediated mitochondria-lysosome damage and autophagy perturbation: The protective role of N-acetylcysteine. Toxicology. 2024;502:153734.10.1016/j.tox.2024.153734Suche in Google Scholar PubMed
[95] Abulikemu A, Zhao X, Qi Y, Liu Y, Wang J, Zhou W, et al. Lysosomal impairment-mediated autophagy dysfunction responsible for the vascular endothelial apoptosis caused by silica nanoparticle via ROS/PARP1/AIF signaling pathway. Environ Pollut. 2022;304:119202.10.1016/j.envpol.2022.119202Suche in Google Scholar PubMed
[96] Li Z, Wang Y, Xu J, Sun J, Zhang W, Liu Z, et al. Silica nanoparticles induce ferroptosis of HUVECs by triggering NCOA4-mediated ferritinophagy. Ecotoxicol Environ Saf. 2024;270:115889.10.1016/j.ecoenv.2023.115889Suche in Google Scholar PubMed
[97] Tsao CW, Aday AW, Almarzooq ZI, Alonso A, Beaton AZ, Bittencourt MS, et al. Heart disease and stroke statistics—2022 update: A report from the American Heart Association. Circulation. 2022;145:e153–639.Suche in Google Scholar
[98] Shah ASV, Langrish JP, Nair H, McAllister DA, Hunter AL, Donaldson K, et al. Global association of air pollution and heart failure: A systematic review and meta-analysis. Lancet. 2013;382:1039–48.10.1016/S0140-6736(13)60898-3Suche in Google Scholar PubMed PubMed Central
[99] Jia Y, Lin Z, He Z, Li C, Zhang Y, Wang J, et al. Effect of air pollution on heart failure: Systematic review and meta-analysis. Environ Health Perspect. 2023;131:076001.10.1289/EHP11506Suche in Google Scholar PubMed PubMed Central
[100] Xu H, Jia Y, Sun Z, Su J, Liu QS, Zhou Q, et al. Environmental pollution, a hidden culprit for health issues. Eco-Environ Health. 2022;1:31–45.10.1016/j.eehl.2022.04.003Suche in Google Scholar PubMed PubMed Central
[101] Katwa P, Wang X, Urankar RN, Podila R, Hilderbrand SC, Fick RB, et al. A carbon nanotube toxicity paradigm driven by mast cells and the IL-33/ST2 axis. Small. 2012;8:2904–12.10.1002/smll.201200873Suche in Google Scholar PubMed PubMed Central
[102] Lozano O, Silva-Platas C, Chapoy-Villanueva H, Pérez BE, Lees JG, Ramachandra CJA, et al. Amorphous SiO2 nanoparticles promote cardiac dysfunction via the opening of the mitochondrial permeability transition pore in rat heart and human cardiomyocytes. Part Fibre Toxicol. 2020;17:15.10.1186/s12989-020-00346-2Suche in Google Scholar PubMed PubMed Central
[103] Wang M, Zhang P, Li Z, Yan Y, Cheng X, Wang G, et al. Different cellular mechanisms from low- and high-dose zinc oxide nanoparticles-induced heart tube malformation during embryogenesis. Nanotoxicology. 2022;16:580–96.10.1080/17435390.2022.2124130Suche in Google Scholar PubMed
[104] Shen C, Zuo Z. Zebrafish (Danio rerio) as an excellent vertebrate model for the development, reproductive, cardiovascular, and neural and ocular development toxicity study of hazardous chemicals. Environ Sci Pollut Res. 2020;27:43599–614.10.1007/s11356-020-10800-5Suche in Google Scholar PubMed
[105] Dal N-JK, Kocere A, Wohlmann J, Van Herck S, Bauer TA, Resseguier J, et al. Zebrafish embryos allow prediction of nanoparticle circulation times in mice and facilitate quantification of nanoparticle–cell interactions. Small. 2020;16:1906719.10.1002/smll.201906719Suche in Google Scholar PubMed
[106] Chen Y, Zhu W, Shu F, Fan Y, Yang N, Wu T, et al. Nd2O3 nanoparticles induce toxicity and cardiac/cerebrovascular abnormality in zebrafish embryos via the apoptosis pathway. Int J Nanomed. 2020;15:387–400.10.2147/IJN.S220785Suche in Google Scholar PubMed PubMed Central
[107] Hsiao B-Y, Horng J-L, Yu C-H, Lin W-T, Wang Y-H, Lin L-Y. Assessing cardiovascular toxicity in zebrafish embryos exposed to copper nanoparticles. Comp Biochem Physiol Part C: Toxicol Pharmacol. 2024;277:109838.10.1016/j.cbpc.2024.109838Suche in Google Scholar PubMed
[108] Zakaria ZZ, Mahmoud NN, Benslimane FM, Yalcin HC, Al Moustafa A-E, Al-Asmakh M. Developmental toxicity of surface-modified gold nanorods in the zebrafish model. ACS Omega. 2022;7:29598–611.10.1021/acsomega.2c01313Suche in Google Scholar PubMed PubMed Central
[109] Shi L, Qian Y, Shen Q, He Y, Jia Y, Wang F. The developmental toxicity and transcriptome analyses of zebrafish (Danio rerio) embryos exposed to carbon nanoparticles. Ecotoxicol Environ Saf. 2022;234:113417.10.1016/j.ecoenv.2022.113417Suche in Google Scholar PubMed
[110] Kalishwaralal K, Jeyabharathi S, Sundar K, Muthukumaran A. Comparative analysis of cardiovascular effects of selenium nanoparticles and sodium selenite in zebrafish embryos. Artif Cells Nanomed Biotechnol. 2016;44:990–96.10.3109/21691401.2015.1008507Suche in Google Scholar PubMed
[111] Duan Z, Wang J, Zhang H, Wang Y, Chen Y, Cong J, et al. Elevated temperature decreases cardiovascular toxicity of nanoplastics but adds to their lethality: A case study during zebrafish (Danio rerio) development. J Hazard Mater. 2023;458:131679.10.1016/j.jhazmat.2023.131679Suche in Google Scholar PubMed
[112] Yang S, Chen Z, Cheng Y, Liu T, Lihong Y, Pu Y, et al. Environmental toxicology wars: Organ-on-a-chip for assessing the toxicity of environmental pollutants. Environ Pollut. 2021;268:115861.10.1016/j.envpol.2020.115861Suche in Google Scholar PubMed
[113] Ahn S, Ardoña HAM, Lind JU, Eweje F, Kim SL, Gonzalez GM, et al. Mussel-inspired 3D fiber scaffolds for heart-on-a-chip toxicity studies of engineered nanomaterials. Anal Bioanal Chem. 2018;410:6141–54.10.1007/s00216-018-1106-7Suche in Google Scholar PubMed PubMed Central
[114] Lu RXZ, Lai BFL, Benge T, Wang EY, Davenport Huyer L, Rafatian N, et al. Heart-on-a-chip platform for assessing toxicity of air pollution related nanoparticles. Adv Mater Technol. 2021;6:2000726.10.1002/admt.202000726Suche in Google Scholar
[115] Mohammed HS, Hosny EN, Khadrawy YA, Magdy M, Attia YS, Sayed OA, et al. Protective effect of curcumin nanoparticles against cardiotoxicity induced by doxorubicin in rat. Biochim Biophys Acta (BBA) - Mol Basis Dis. 2020;1866:165665.10.1016/j.bbadis.2020.165665Suche in Google Scholar PubMed
[116] Khadrawy YA, Hosny EN, El-Gizawy MM, Sawie HG, Aboul Ezz HS. The effect of curcumin nanoparticles on cisplatin-induced cardiotoxicity in male wistar albino rats. Cardiovasc Toxicol. 2021;21:433–43.10.1007/s12012-021-09636-3Suche in Google Scholar PubMed
[117] Wang D, Wang C, Liang Z, Lei W, Deng C, Liu X, et al. Protection of zero-valent iron nanoparticles against sepsis and septic heart failure. J Nanobiotechnol. 2022;20:405.10.1186/s12951-022-01589-1Suche in Google Scholar PubMed PubMed Central
[118] Breznan D, Das DD, MacKinnon-Roy C, Bernatchez S, Sayari A, Hill M, et al. Physicochemical properties can be key determinants of mesoporous silica nanoparticle potency in vitro. ACS Nano. 2018;12:12062–79.10.1021/acsnano.8b04910Suche in Google Scholar PubMed
[119] Xu M, Soliman MG, Sun X, Pelaz B, Feliu N, Parak WJ, et al. How entanglement of different physicochemical properties complicates the prediction of in vitro and in vivo interactions of gold nanoparticles. ACS Nano. 2018;12:10104–13.10.1021/acsnano.8b04906Suche in Google Scholar PubMed
[120] Asharani PV, lianwu Y, Gong Z, Valiyaveettil S. Comparison of the toxicity of silver, gold and platinum nanoparticles in developing zebrafish embryos. Nanotoxicology. 2011;5:43–54.10.3109/17435390.2010.489207Suche in Google Scholar PubMed
[121] Cao Y, Gong Y, Liao W, Luo Y, Wu C, Wang M, et al. A review of cardiovascular toxicity of TiO2, ZnO and Ag nanoparticles (NPs). BioMetals. 2018;31:457–76.10.1007/s10534-018-0113-7Suche in Google Scholar PubMed
[122] Smyth E, Solomon A, Vydyanath A, Luther PK, Pitchford S, Tetley TD, et al. Induction and enhancement of platelet aggregation in vitro and in vivo by model polystyrene nanoparticles. Nanotoxicology. 2015;9:356–64.10.3109/17435390.2014.933902Suche in Google Scholar PubMed
[123] McGuinnes C, Duffin R, Brown S, Mills NL, Megson IL, MacNee W, et al. Surface derivatization state of polystyrene latex nanoparticles determines both their potency and their mechanism of causing human platelet aggregation in vitro. Toxicol Sci. 2010;119:359–68.10.1093/toxsci/kfq349Suche in Google Scholar PubMed
[124] Corbalan JJ, Medina C, Jacoby A, Malinski T, Radomski MW. Amorphous silica nanoparticles aggregate human platelets: potential implications for vascular homeostasis. Int J Nanomed. 2012;7:631–39.10.2147/IJN.S28293Suche in Google Scholar PubMed PubMed Central
[125] Geys J, Nemmar A, Verbeken E, Smolders E, Ratoi M, Hoylaerts MF, et al. Acute toxicity and prothrombotic effects of quantum dots: Impact of surface charge. Environ Health Perspect. 2008;116:1607–13.10.1289/ehp.11566Suche in Google Scholar PubMed PubMed Central
[126] Shrivastava S, Bera T, Singh SK, Singh G, Ramachandrarao P, Dash D. Characterization of antiplatelet properties of silver nanoparticles. ACS Nano. 2009;3:1357–64.10.1021/nn900277tSuche in Google Scholar PubMed
[127] Hajtuch J, Hante N, Tomczyk E, Wojcik M, Radomski MW, Santos-Martinez MJ, et al. Effects of functionalized silver nanoparticles on aggregation of human blood platelets. Int J Nanomed. 2019;14:7399–417.10.2147/IJN.S213499Suche in Google Scholar PubMed PubMed Central
[128] Yan Z, Liu Z, Yang B, Zhu X, Song E, Song Y. Long-term pulmonary iron oxide nanoparticles exposure disrupts hepatic iron-lipid homeostasis and increases plaque vulnerability in ApoE−/− mice. Environ Pollut. 2024;341:122905.10.1016/j.envpol.2023.122905Suche in Google Scholar PubMed
[129] Holland NA, Thompson LC, Vidanapathirana AK, Urankar RN, Lust RM, Fennell TR, et al. Impact of pulmonary exposure to gold core silver nanoparticles of different size and capping agents on cardiovascular injury. Part Fibre Toxicol. 2016;13:48.10.1186/s12989-016-0159-zSuche in Google Scholar PubMed PubMed Central
[130] Chuang K-J, Lee K-Y, Pan C-H, Lai C-H, Lin L-Y, Ho S-C, et al. Effects of zinc oxide nanoparticles on human coronary artery endothelial cells. Food Chem Toxicol. 2016;93:138–44.10.1016/j.fct.2016.05.008Suche in Google Scholar PubMed
[131] Gal N, Charwat V, Städler B, Reimhult E. Poly(ethylene glycol) grafting of nanoparticles prevents uptake by cells and transport through cell barrier layers regardless of shear flow and particle size. ACS Biomater Sci Eng. 2019;5:4355–65.10.1021/acsbiomaterials.9b00611Suche in Google Scholar PubMed
[132] Setyawati MI, Tay CY, Bay BH, Leong DT. Gold nanoparticles induced endothelial leakiness depends on particle size and endothelial cell origin. ACS Nano. 2017;11:5020–30.10.1021/acsnano.7b01744Suche in Google Scholar PubMed
[133] Bartczak D, Muskens OL, Nitti S, Sanchez-Elsner T, Millar TM, Kanaras AG. Interactions of human endothelial cells with gold nanoparticles of different morphologies. Small. 2012;8:122–30.10.1002/smll.201101422Suche in Google Scholar PubMed
[134] Da Silva-Candal A, Brown T, Krishnan V, Lopez-Loureiro I, Ávila-Gómez P, Pusuluri A, et al. Shape effect in active targeting of nanoparticles to inflamed cerebral endothelium under static and flow conditions. J Controlled Rel. 2019;309:94–105.10.1016/j.jconrel.2019.07.026Suche in Google Scholar PubMed
[135] Jurney P, Agarwal R, Singh V, Choi D, Roy K, Sreenivasan SV, et al. Unique size and shape-dependent uptake behaviors of non-spherical nanoparticles by endothelial cells due to a shearing flow. J Controlled Rel. 2017;245:170–76.10.1016/j.jconrel.2016.11.033Suche in Google Scholar PubMed
[136] Kolhar P, Anselmo AC, Gupta V, Pant K, Prabhakarpandian B, Ruoslahti E, et al. Using shape effects to target antibody-coated nanoparticles to lung and brain endothelium. Proc Natl Acad Sci. 2013;110:10753–8.10.1073/pnas.1308345110Suche in Google Scholar PubMed PubMed Central
[137] Asati A, Santra S, Kaittanis C, Perez JM. Surface-charge-dependent cell localization and cytotoxicity of cerium oxide nanoparticles. ACS Nano. 2010;4:5321–31.10.1021/nn100816sSuche in Google Scholar PubMed PubMed Central
[138] Alkilany AM, Shatanawi A, Kurtz T, Caldwell RB, Caldwell RW. Toxicity and cellular uptake of gold nanorods in vascular endothelium and smooth muscles of isolated rat blood vessel: Importance of surface modification. Small. 2012;8:1270–8.10.1002/smll.201101948Suche in Google Scholar PubMed PubMed Central
[139] Paatero I, Casals E, Niemi R, Özliseli E, Rosenholm JM, Sahlgren C. Analyses in zebrafish embryos reveal that nanotoxicity profiles are dependent on surface-functionalization controlled penetrance of biological membranes. Sci Rep. 2017;7:8423.10.1038/s41598-017-09312-zSuche in Google Scholar PubMed PubMed Central
[140] Bai Q, Xiao Y, Hong H, Cao X, Zhang L, Han R, et al. Scavenger receptor-targeted plaque delivery of microRNA-coated nanoparticles for alleviating atherosclerosis. Proc Natl Acad Sci. 2022;119:e2201443119.10.1073/pnas.2201443119Suche in Google Scholar PubMed PubMed Central
[141] Zaleski MH, Chase LS, Hood ED, Wang Z, Nong J, Espy CL, et al. Conjugation chemistry markedly impacts toxicity and biodistribution of targeted nanoparticles, mediated by complement activation. Adv Mater. 2025;37:2409945.10.1002/adma.202409945Suche in Google Scholar PubMed PubMed Central
[142] Shannahan J. The biocorona: A challenge for the biomedical application of nanoparticles. Nanotechnol Rev. 2017;6:345–53.10.1515/ntrev-2016-0098Suche in Google Scholar PubMed PubMed Central
[143] Wang J, Xu Y, Zhou Y, Zhang J, Jia J, Jiao P, et al. Modulating the toxicity of engineered nanoparticles by controlling protein corona formation: Recent advances and future prospects. Sci Total Environ. 2024;914:169590.10.1016/j.scitotenv.2023.169590Suche in Google Scholar PubMed
[144] Yeo ELL, Azman NA, Kah JCY. Stealthiness and Hematocompatibility of Gold Nanoparticles with Pre-Formed Protein Corona. Langmuir. 2021;37:4913–23.10.1021/acs.langmuir.1c00151Suche in Google Scholar PubMed
[145] Ge C, Du J, Zhao L, Wang L, Liu Y, Li D, et al. Binding of blood proteins to carbon nanotubes reduces cytotoxicity. Proc Natl Acad Sci. 2011;108:16968–73.10.1073/pnas.1105270108Suche in Google Scholar PubMed PubMed Central
[146] Chandran P, Riviere JE, Monteiro-Riviere NA. Surface chemistry of gold nanoparticles determines the biocorona composition impacting cellular uptake, toxicity and gene expression profiles in human endothelial cells. Nanotoxicology. 2017;11:507–19.10.1080/17435390.2017.1314036Suche in Google Scholar PubMed
[147] Escamilla-Rivera V, Uribe-Ramírez M, González-Pozos S, Lozano O, Lucas S, De Vizcaya-Ruiz A. Protein corona acts as a protective shield against Fe3O4-PEG inflammation and ROS-induced toxicity in human macrophages. Toxicol Lett. 2016;240:172–84.10.1016/j.toxlet.2015.10.018Suche in Google Scholar PubMed
[148] Ding Y, Tian R, Yang Z, Chen J, Lu N. Effects of serum albumin on the degradation and cytotoxicity of single-walled carbon nanotubes. Biophys Chem. 2017;222:1–6.10.1016/j.bpc.2016.12.002Suche in Google Scholar PubMed
[149] Leibe R, Hsiao IL, Fritsch-Decker S, Kielmeier U, Wagbo AM, Voss B, et al. The protein corona suppresses the cytotoxic and pro-inflammatory response in lung epithelial cells and macrophages upon exposure to nanosilica. Arch Toxicol. 2019;93:871–85.10.1007/s00204-019-02422-9Suche in Google Scholar PubMed
[150] Liu N, Liang Y, Wei T, Zou L, Bai C, Huang X, et al. Protein corona mitigated the cytotoxicity of CdTe QDs to macrophages by targeting mitochondria. NanoImpact. 2022;25:100367.10.1016/j.impact.2021.100367Suche in Google Scholar PubMed
[151] Yoshida T, Yoshioka Y, Morishita Y, Aoyama M, Tochigi S, Hirai T, et al. Protein corona changes mediated by surface modification of amorphous silica nanoparticles suppress acute toxicity and activation of intrinsic coagulation cascade in mice. Nanotechnology. 2015;26:245101.10.1088/0957-4484/26/24/245101Suche in Google Scholar PubMed
[152] Vlasova II, Mikhalchik EV, Barinov NA, Kostevich VA, Smolina NV, Klinov DV, et al. Adsorbed plasma proteins modulate the effects of single-walled carbon nanotubes on neutrophils in blood. Nanomed: Nanotechnol Biol Med. 2016;12:1615–25.10.1016/j.nano.2016.02.012Suche in Google Scholar PubMed
[153] Liu S, Wang Z, Jiang X, Gan J, Tian X, Xing Z, et al. Denatured corona proteins mediate the intracellular bioactivities of nanoparticles via the unfolded protein response. Biomaterials. 2021;265:120452.10.1016/j.biomaterials.2020.120452Suche in Google Scholar PubMed
[154] Pavlin M, Lojk J, Strojan K, Hafner-Bratkovič I, Jerala R, Leonardi A, et al. The relevance of physico-chemical properties and protein corona for evaluation of nanoparticles immunotoxicity—in vitro correlation analysis on THP-1 macrophages. Int J Mol Sci. 2022;23:6197.10.3390/ijms23116197Suche in Google Scholar PubMed PubMed Central
[155] Deng ZJ, Liang M, Monteiro M, Toth I, Minchin RF. Nanoparticle-induced unfolding of fibrinogen promotes Mac-1 receptor activation and inflammation. Nat Nanotechnol. 2011;6:39–44.10.1038/nnano.2010.250Suche in Google Scholar PubMed
[156] Escamilla-Rivera V, Solorio-Rodríguez A, Uribe-Ramírez M, Lozano O, Lucas S, Chagolla-López A, et al. Plasma protein adsorption on Fe3O4-PEG nanoparticles activates the complement system and induces an inflammatory response. Int J Nanomed. 2019;14:2055–67.10.2147/IJN.S192214Suche in Google Scholar PubMed PubMed Central
[157] Chen F, Wang G, Griffin JI, Brenneman B, Banda NK, Holers VM, et al. Complement proteins bind to nanoparticle protein corona and undergo dynamic exchange in vivo. Nat Nanotechnol. 2017;12:387–93.10.1038/nnano.2016.269Suche in Google Scholar PubMed PubMed Central
[158] Ho YT, Azman NA, Loh FWY, Ong GKT, Engudar G, Kriz SA, et al. Protein corona formed from different blood plasma proteins affects the colloidal stability of nanoparticles differently. Bioconjugate Chem. 2018;29:3923–34.10.1021/acs.bioconjchem.8b00743Suche in Google Scholar PubMed
[159] Mahmoudi M, Sheibani S, Milani AS, Rezaee F, Gauberti M, Dinarvand R, et al. Crucial role of the protein corona for the specific targeting of nanoparticles. Nanomedicine. 2015;10:215–26.10.2217/nnm.14.69Suche in Google Scholar PubMed
[160] Chang J, Paillard A, Passirani C, Morille M, Benoit J-P, Betbeder D, et al. Transferrin adsorption onto PLGA nanoparticles governs their interaction with biological systems from blood circulation to brain cancer cells. Pharm Res. 2012;29:1495–505.10.1007/s11095-011-0624-1Suche in Google Scholar PubMed
[161] Cox A, Andreozzi P, Dal Magro R, Fiordaliso F, Corbelli A, Talamini L, et al. Evolution of nanoparticle protein corona across the blood–brain barrier. ACS Nano. 2018;12:7292–300.10.1021/acsnano.8b03500Suche in Google Scholar PubMed
[162] Mao X, Liu Y, Wei Y, Li X, Liu Y, Su G, et al. Threats of per- and poly-fluoroalkyl pollutants to susceptible populations. Sci Total Environ. 2024;921:171188.10.1016/j.scitotenv.2024.171188Suche in Google Scholar PubMed
[163] Li Y, Zhang Y, Yan B. Nanotoxicity overview: Nano-threat to susceptible populations. Int J Mol Sci. 2014;15:3671–97.10.3390/ijms15033671Suche in Google Scholar PubMed PubMed Central
[164] Kang GS, Gillespie PA, Gunnison A, Moreira AL, Tchou-Wong K-M, Chen L-C. Long-term inhalation exposure to nickel nanoparticles exacerbated atherosclerosis in a susceptible mouse model. Environ Health Perspect. 2011;119:176–81.10.1289/ehp.1002508Suche in Google Scholar PubMed PubMed Central
[165] Li Z, Hulderman T, Salmen R, Chapman R, Leonard SS, Young S-H, et al. Cardiovascular effects of pulmonary exposure to single-wall carbon nanotubes. Environ Health Perspect. 2007;115:377–82.10.1289/ehp.9688Suche in Google Scholar PubMed PubMed Central
[166] Dekkers S, Miller MR, Schins RPF, Römer I, Russ M, Vandebriel RJ, et al. The effect of zirconium doping of cerium dioxide nanoparticles on pulmonary and cardiovascular toxicity and biodistribution in mice after inhalation. Nanotoxicology. 2017;11:794–808.10.1080/17435390.2017.1357214Suche in Google Scholar PubMed
[167] Ramirez-Lee MA, Espinosa-Tanguma R, Mejía-Elizondo R, Medina-Hernández A, Martinez-Castañon GA, Gonzalez C. Effect of silver nanoparticles upon the myocardial and coronary vascular function in isolated and perfused diabetic rat hearts. Nanomed: Nanotechnol Biol Med. 2017;13:2587–96.10.1016/j.nano.2017.07.007Suche in Google Scholar PubMed
[168] Ramirez-Lee MA, Aguirre-Bañuelos P, Martinez-Cuevas PP, Espinosa-Tanguma R, Chi-Ahumada E, Martinez-Castañon GA, et al. Evaluation of cardiovascular responses to silver nanoparticles (AgNPs) in spontaneously hypertensive rats. Nanomed: Nanotechnol Biol Med. 2018;14:385–95.10.1016/j.nano.2017.11.013Suche in Google Scholar PubMed
[169] Ge C, Meng L, Xu L, Bai R, Du J, Zhang L, et al. Acute pulmonary and moderate cardiovascular responses of spontaneously hypertensive rats after exposure to single-wall carbon nanotubes. Nanotoxicology. 2012;6:526–42.10.3109/17435390.2011.587905Suche in Google Scholar PubMed
[170] Inoue K-I, Takano H, Yanagisawa R, Hirano S, Sakurai M, Shimada A, et al. Effects of airway exposure to nanoparticles on lung inflammation induced by bacterial endotoxin in mice. Environ Health Perspect. 2006;114:1325–30.10.1289/ehp.8903Suche in Google Scholar PubMed PubMed Central
[171] Inoue K-I, Takano H, Koike E, Yanagisawa R, Sakurai M, Tasaka S, et al. Effects of pulmonary exposure to carbon nanotubes on lung and systemic inflammation with coagulatory disturbance induced by lipopolysaccharide in mice. Exp Biol Med. 2008;233:1583–90.10.3181/0805-RM-179Suche in Google Scholar PubMed
[172] Inoue K, Takano H, Ohnuki M, Yanagisawa R, Sakurai M, Shimada A, et al. Size effects of nanomaterials on lung inflammation and coagulatory disturbance. Int J Immunopathol Pharmacol. 2008;21:197–206.10.1177/039463200802100122Suche in Google Scholar PubMed
[173] Bian Y, Kim K, Ngo T, Kim I, Bae O-N, Lim K-M, et al. Silver nanoparticles promote procoagulant activity of red blood cells: a potential risk of thrombosis in susceptible population. Part Fibre Toxicol. 2019;16:9.10.1186/s12989-019-0292-6Suche in Google Scholar PubMed PubMed Central
[174] Chen Z, Meng H, Xing G, Yuan H, Zhao F, Liu R, et al. Age-related differences in pulmonary and cardiovascular responses to SiO2 nanoparticle inhalation: Nanotoxicity has susceptible population. Environ Sci Technol. 2008;42:8985–92.10.1021/es800975uSuche in Google Scholar PubMed
[175] Deng R, Lin D, Zhu L, Majumdar S, White JC, Gardea-Torresdey JL, et al. Nanoparticle interactions with co-existing contaminants: Joint toxicity, bioaccumulation and risk. Nanotoxicology. 2017;11:591–612.10.1080/17435390.2017.1343404Suche in Google Scholar PubMed
[176] Forest V. Combined effects of nanoparticles and other environmental contaminants on human health - an issue often overlooked. NanoImpact. 2021;23:100344.10.1016/j.impact.2021.100344Suche in Google Scholar PubMed
[177] Hu H, Shi Y, Zhang Y, Wu J, Asweto CO, Feng L, et al. Comprehensive gene and microRNA expression profiling on cardiovascular system in zebrafish co-exposured of SiNPs and MeHg. Sci Total Environ. 2017;607-608:795–805.10.1016/j.scitotenv.2017.07.036Suche in Google Scholar PubMed
[178] Duan J, Hu H, Li Q, Jiang L, Zou Y, Wang Y, et al. Combined toxicity of silica nanoparticles and methylmercury on cardiovascular system in zebrafish (Danio rerio) embryos. Environ Toxicol Pharmacol. 2016;44:120–7.10.1016/j.etap.2016.05.004Suche in Google Scholar PubMed
[179] Liu P, Wang S, Chang Z, Li L, Xing H, Dong W-F. Combined toxicity of silica nanoparticles and cadmium chloride on the cardiovascular system of zebrafish (Danio rerio) larvae. Comp Biochem Physiol Part C: Toxicol Pharmacol. 2021;239:108895.10.1016/j.cbpc.2020.108895Suche in Google Scholar PubMed
[180] Yang X, Feng L, Zhang Y, Hu H, Shi Y, Liang S, et al. Co-exposure of silica nanoparticles and methylmercury induced cardiac toxicity in vitro and in vivo. Sci Total Environ. 2018;631-632:811–21.10.1016/j.scitotenv.2018.03.107Suche in Google Scholar PubMed
[181] Feng L, Yang X, Shi Y, Liang S, Zhao T, Duan J, et al. Co-exposure subacute toxicity of silica nanoparticles and lead acetate on cardiovascular system. Int J Nanomed. 2018;13:7819–34.10.2147/IJN.S185259Suche in Google Scholar PubMed PubMed Central
[182] Zhang B, Chen L, Choi JJ, Hennig B, Toborek M. Cerebrovascular toxicity of PCB153 is enhanced by binding to silica nanoparticles. J Neuroimmune Pharmacol. 2012;7:991–1001.10.1007/s11481-012-9403-ySuche in Google Scholar PubMed PubMed Central
[183] Trevisan R, Voy C, Chen S, Di Giulio RT. Nanoplastics decrease the toxicity of a complex PAH mixture but impair mitochondrial energy production in developing zebrafish. Environ Sci Technol. 2019;53:8405–15.10.1021/acs.est.9b02003Suche in Google Scholar PubMed PubMed Central
© 2025 the author(s), published by De Gruyter
This work is licensed under the Creative Commons Attribution 4.0 International License.
Artikel in diesem Heft
- Research Articles
- MHD radiative mixed convective flow of a sodium alginate-based hybrid nanofluid over a convectively heated extending sheet with Joule heating
- Experimental study of mortar incorporating nano-magnetite on engineering performance and radiation shielding
- Multicriteria-based optimization and multi-variable non-linear regression analysis of concrete containing blends of nano date palm ash and eggshell powder as cementitious materials
- A promising Ag2S/poly-2-amino-1-mercaptobenzene open-top spherical core–shell nanocomposite for optoelectronic devices: A one-pot technique
- Biogenic synthesized selenium nanoparticles combined chitosan nanoparticles controlled lung cancer growth via ROS generation and mitochondrial damage pathway
- Fabrication of PDMS nano-mold by deposition casting method
- Stimulus-responsive gradient hydrogel micro-actuators fabricated by two-photon polymerization-based 4D printing
- Physical aspects of radiative Carreau nanofluid flow with motile microorganisms movement under yield stress via oblique penetrable wedge
- Effect of polar functional groups on the hydrophobicity of carbon nanotubes-bacterial cellulose nanocomposite
- Review in green synthesis mechanisms, application, and future prospects for Garcinia mangostana L. (mangosteen)-derived nanoparticles
- Entropy generation and heat transfer in nonlinear Buoyancy–driven Darcy–Forchheimer hybrid nanofluids with activation energy
- Green synthesis of silver nanoparticles using Ginkgo biloba seed extract: Evaluation of antioxidant, anticancer, antifungal, and antibacterial activities
- A numerical analysis of heat and mass transfer in water-based hybrid nanofluid flow containing copper and alumina nanoparticles over an extending sheet
- Investigating the behaviour of electro-magneto-hydrodynamic Carreau nanofluid flow with slip effects over a stretching cylinder
- Electrospun thermoplastic polyurethane/nano-Ag-coated clear aligners for the inhibition of Streptococcus mutans and oral biofilm
- Investigation of the optoelectronic properties of a novel polypyrrole-multi-well carbon nanotubes/titanium oxide/aluminum oxide/p-silicon heterojunction
- Novel photothermal magnetic Janus membranes suitable for solar water desalination
- Green synthesis of silver nanoparticles using Ageratum conyzoides for activated carbon compositing to prepare antimicrobial cotton fabric
- Activation energy and Coriolis force impact on three-dimensional dusty nanofluid flow containing gyrotactic microorganisms: Machine learning and numerical approach
- Machine learning analysis of thermo-bioconvection in a micropolar hybrid nanofluid-filled square cavity with oxytactic microorganisms
- Research and improvement of mechanical properties of cement nanocomposites for well cementing
- Thermal and stability analysis of silver–water nanofluid flow over unsteady stretching sheet under the influence of heat generation/absorption at the boundary
- Cobalt iron oxide-infused silicone nanocomposites: Magnetoactive materials for remote actuation and sensing
- Magnesium-reinforced PMMA composite scaffolds: Synthesis, characterization, and 3D printing via stereolithography
- Bayesian inference-based physics-informed neural network for performance study of hybrid nanofluids
- Numerical simulation of non-Newtonian hybrid nanofluid flow subject to a heterogeneous/homogeneous chemical reaction over a Riga surface
- Enhancing the superhydrophobicity, UV-resistance, and antifungal properties of natural wood surfaces via in situ formation of ZnO, TiO2, and SiO2 particles
- Synthesis and electrochemical characterization of iron oxide/poly(2-methylaniline) nanohybrids for supercapacitor application
- Impacts of double stratification on thermally radiative third-grade nanofluid flow on elongating cylinder with homogeneous/heterogeneous reactions by implementing machine learning approach
- Synthesis of Cu4O3 nanoparticles using pumpkin seed extract: Optimization, antimicrobial, and cytotoxicity studies
- Cationic charge influence on the magnetic response of the Fe3O4–[Me2+ 1−y Me3+ y (OH2)] y+(Co3 2−) y/2·mH2O hydrotalcite system
- Pressure sensing intelligent martial arts short soldier combat protection system based on conjugated polymer nanocomposite materials
- Magnetohydrodynamics heat transfer rate under inclined buoyancy force for nano and dusty fluids: Response surface optimization for the thermal transport
- Fly ash and nano-graphene enhanced stabilization of engine oil-contaminated soils
- Enhancing natural fiber-reinforced biopolymer composites with graphene nanoplatelets: Mechanical, morphological, and thermal properties
- Performance evaluation of dual-scale strengthened co-bonded single-lap joints using carbon nanotubes and Z-pins with ANN
- Computational works of blood flow with dust particles and partially ionized containing tiny particles on a moving wedge: Applications of nanotechnology
- Hybridization of biocomposites with oil palm cellulose nanofibrils/graphene nanoplatelets reinforcement in green epoxy: A study of physical, thermal, mechanical, and morphological properties
- Design and preparation of micro-nano dual-scale particle-reinforced Cu–Al–V alloy: Research on the aluminothermic reduction process
- Spectral quasi-linearization and response optimization on magnetohydrodynamic flow via stenosed artery with hybrid and ternary solid nanoparticles: Support vector machine learning
- Review Articles
- A comprehensive review on hybrid plasmonic waveguides: Structures, applications, challenges, and future perspectives
- Nanoparticles in low-temperature preservation of biological systems of animal origin
- Fluorescent sulfur quantum dots for environmental monitoring
- Nanoscience systematic review methodology standardization
- Nanotechnology revolutionizing osteosarcoma treatment: Advances in targeted kinase inhibitors
- AFM: An important enabling technology for 2D materials and devices
- Carbon and 2D nanomaterial smart hydrogels for therapeutic applications
- Principles, applications and future prospects in photodegradation systems
- Do gold nanoparticles consistently benefit crop plants under both non-stressed and abiotic stress conditions?
- An updated overview of nanoparticle-induced cardiovascular toxicity
- Arginine as a promising amino acid for functionalized nanosystems: Innovations, challenges, and future directions
- Advancements in the use of cancer nanovaccines: Comprehensive insights with focus on lung and colon cancer
- Membrane-based biomimetic delivery systems for glioblastoma multiforme therapy
- The drug delivery systems based on nanoparticles for spinal cord injury repair
- Green synthesis, biomedical effects, and future trends of Ag/ZnO bimetallic nanoparticles: An update
- Application of magnesium and its compounds in biomaterials for nerve injury repair
- Micro/nanomotors in biomedicine: Construction and applications
- Hydrothermal synthesis of biomass-derived CQDs: Advances and applications
- Research progress in 3D bioprinting of skin: Challenges and opportunities
- Review on bio-selenium nanoparticles: Synthesis, protocols, and applications in biomedical processes
- Gold nanocrystals and nanorods functionalized with protein and polymeric ligands for environmental, energy storage, and diagnostic applications: A review
- An in-depth analysis of rotational and non-rotational piezoelectric energy harvesting beams: A comprehensive review
- Advancements in perovskite/CIGS tandem solar cells: Material synergies, device configurations, and economic viability for sustainable energy
- Deep learning in-depth analysis of crystal graph convolutional neural networks: A new era in materials discovery and its applications
- Review of recent nano TiO2 film coating methods, assessment techniques, and key problems for scaleup
- Antioxidant quantum dots for spinal cord injuries: A review on advancing neuroprotection and regeneration in neurological disorders
- Special Issue on Advanced Nanomaterials for Carbon Capture, Environment and Utilization for Energy Sustainability - Part III
- Efficiency optimization of quantum dot photovoltaic cell by solar thermophotovoltaic system
- Exploring the diverse nanomaterials employed in dental prosthesis and implant techniques: An overview
- Electrochemical investigation of bismuth-doped anode materials for low‑temperature solid oxide fuel cells with boosted voltage using a DC-DC voltage converter
- Synthesis of HfSe2 and CuHfSe2 crystalline materials using the chemical vapor transport method and their applications in supercapacitor energy storage devices
- Special Issue on Green Nanotechnology and Nano-materials for Environment Sustainability
- Influence of nano-silica and nano-ferrite particles on mechanical and durability of sustainable concrete: A review
- Surfaces and interfaces analysis on different carboxymethylation reaction time of anionic cellulose nanoparticles derived from oil palm biomass
- Processing and effective utilization of lignocellulosic biomass: Nanocellulose, nanolignin, and nanoxylan for wastewater treatment
- Retraction
- Retraction of “Aging assessment of silicone rubber materials under corona discharge accompanied by humidity and UV radiation”
Artikel in diesem Heft
- Research Articles
- MHD radiative mixed convective flow of a sodium alginate-based hybrid nanofluid over a convectively heated extending sheet with Joule heating
- Experimental study of mortar incorporating nano-magnetite on engineering performance and radiation shielding
- Multicriteria-based optimization and multi-variable non-linear regression analysis of concrete containing blends of nano date palm ash and eggshell powder as cementitious materials
- A promising Ag2S/poly-2-amino-1-mercaptobenzene open-top spherical core–shell nanocomposite for optoelectronic devices: A one-pot technique
- Biogenic synthesized selenium nanoparticles combined chitosan nanoparticles controlled lung cancer growth via ROS generation and mitochondrial damage pathway
- Fabrication of PDMS nano-mold by deposition casting method
- Stimulus-responsive gradient hydrogel micro-actuators fabricated by two-photon polymerization-based 4D printing
- Physical aspects of radiative Carreau nanofluid flow with motile microorganisms movement under yield stress via oblique penetrable wedge
- Effect of polar functional groups on the hydrophobicity of carbon nanotubes-bacterial cellulose nanocomposite
- Review in green synthesis mechanisms, application, and future prospects for Garcinia mangostana L. (mangosteen)-derived nanoparticles
- Entropy generation and heat transfer in nonlinear Buoyancy–driven Darcy–Forchheimer hybrid nanofluids with activation energy
- Green synthesis of silver nanoparticles using Ginkgo biloba seed extract: Evaluation of antioxidant, anticancer, antifungal, and antibacterial activities
- A numerical analysis of heat and mass transfer in water-based hybrid nanofluid flow containing copper and alumina nanoparticles over an extending sheet
- Investigating the behaviour of electro-magneto-hydrodynamic Carreau nanofluid flow with slip effects over a stretching cylinder
- Electrospun thermoplastic polyurethane/nano-Ag-coated clear aligners for the inhibition of Streptococcus mutans and oral biofilm
- Investigation of the optoelectronic properties of a novel polypyrrole-multi-well carbon nanotubes/titanium oxide/aluminum oxide/p-silicon heterojunction
- Novel photothermal magnetic Janus membranes suitable for solar water desalination
- Green synthesis of silver nanoparticles using Ageratum conyzoides for activated carbon compositing to prepare antimicrobial cotton fabric
- Activation energy and Coriolis force impact on three-dimensional dusty nanofluid flow containing gyrotactic microorganisms: Machine learning and numerical approach
- Machine learning analysis of thermo-bioconvection in a micropolar hybrid nanofluid-filled square cavity with oxytactic microorganisms
- Research and improvement of mechanical properties of cement nanocomposites for well cementing
- Thermal and stability analysis of silver–water nanofluid flow over unsteady stretching sheet under the influence of heat generation/absorption at the boundary
- Cobalt iron oxide-infused silicone nanocomposites: Magnetoactive materials for remote actuation and sensing
- Magnesium-reinforced PMMA composite scaffolds: Synthesis, characterization, and 3D printing via stereolithography
- Bayesian inference-based physics-informed neural network for performance study of hybrid nanofluids
- Numerical simulation of non-Newtonian hybrid nanofluid flow subject to a heterogeneous/homogeneous chemical reaction over a Riga surface
- Enhancing the superhydrophobicity, UV-resistance, and antifungal properties of natural wood surfaces via in situ formation of ZnO, TiO2, and SiO2 particles
- Synthesis and electrochemical characterization of iron oxide/poly(2-methylaniline) nanohybrids for supercapacitor application
- Impacts of double stratification on thermally radiative third-grade nanofluid flow on elongating cylinder with homogeneous/heterogeneous reactions by implementing machine learning approach
- Synthesis of Cu4O3 nanoparticles using pumpkin seed extract: Optimization, antimicrobial, and cytotoxicity studies
- Cationic charge influence on the magnetic response of the Fe3O4–[Me2+ 1−y Me3+ y (OH2)] y+(Co3 2−) y/2·mH2O hydrotalcite system
- Pressure sensing intelligent martial arts short soldier combat protection system based on conjugated polymer nanocomposite materials
- Magnetohydrodynamics heat transfer rate under inclined buoyancy force for nano and dusty fluids: Response surface optimization for the thermal transport
- Fly ash and nano-graphene enhanced stabilization of engine oil-contaminated soils
- Enhancing natural fiber-reinforced biopolymer composites with graphene nanoplatelets: Mechanical, morphological, and thermal properties
- Performance evaluation of dual-scale strengthened co-bonded single-lap joints using carbon nanotubes and Z-pins with ANN
- Computational works of blood flow with dust particles and partially ionized containing tiny particles on a moving wedge: Applications of nanotechnology
- Hybridization of biocomposites with oil palm cellulose nanofibrils/graphene nanoplatelets reinforcement in green epoxy: A study of physical, thermal, mechanical, and morphological properties
- Design and preparation of micro-nano dual-scale particle-reinforced Cu–Al–V alloy: Research on the aluminothermic reduction process
- Spectral quasi-linearization and response optimization on magnetohydrodynamic flow via stenosed artery with hybrid and ternary solid nanoparticles: Support vector machine learning
- Review Articles
- A comprehensive review on hybrid plasmonic waveguides: Structures, applications, challenges, and future perspectives
- Nanoparticles in low-temperature preservation of biological systems of animal origin
- Fluorescent sulfur quantum dots for environmental monitoring
- Nanoscience systematic review methodology standardization
- Nanotechnology revolutionizing osteosarcoma treatment: Advances in targeted kinase inhibitors
- AFM: An important enabling technology for 2D materials and devices
- Carbon and 2D nanomaterial smart hydrogels for therapeutic applications
- Principles, applications and future prospects in photodegradation systems
- Do gold nanoparticles consistently benefit crop plants under both non-stressed and abiotic stress conditions?
- An updated overview of nanoparticle-induced cardiovascular toxicity
- Arginine as a promising amino acid for functionalized nanosystems: Innovations, challenges, and future directions
- Advancements in the use of cancer nanovaccines: Comprehensive insights with focus on lung and colon cancer
- Membrane-based biomimetic delivery systems for glioblastoma multiforme therapy
- The drug delivery systems based on nanoparticles for spinal cord injury repair
- Green synthesis, biomedical effects, and future trends of Ag/ZnO bimetallic nanoparticles: An update
- Application of magnesium and its compounds in biomaterials for nerve injury repair
- Micro/nanomotors in biomedicine: Construction and applications
- Hydrothermal synthesis of biomass-derived CQDs: Advances and applications
- Research progress in 3D bioprinting of skin: Challenges and opportunities
- Review on bio-selenium nanoparticles: Synthesis, protocols, and applications in biomedical processes
- Gold nanocrystals and nanorods functionalized with protein and polymeric ligands for environmental, energy storage, and diagnostic applications: A review
- An in-depth analysis of rotational and non-rotational piezoelectric energy harvesting beams: A comprehensive review
- Advancements in perovskite/CIGS tandem solar cells: Material synergies, device configurations, and economic viability for sustainable energy
- Deep learning in-depth analysis of crystal graph convolutional neural networks: A new era in materials discovery and its applications
- Review of recent nano TiO2 film coating methods, assessment techniques, and key problems for scaleup
- Antioxidant quantum dots for spinal cord injuries: A review on advancing neuroprotection and regeneration in neurological disorders
- Special Issue on Advanced Nanomaterials for Carbon Capture, Environment and Utilization for Energy Sustainability - Part III
- Efficiency optimization of quantum dot photovoltaic cell by solar thermophotovoltaic system
- Exploring the diverse nanomaterials employed in dental prosthesis and implant techniques: An overview
- Electrochemical investigation of bismuth-doped anode materials for low‑temperature solid oxide fuel cells with boosted voltage using a DC-DC voltage converter
- Synthesis of HfSe2 and CuHfSe2 crystalline materials using the chemical vapor transport method and their applications in supercapacitor energy storage devices
- Special Issue on Green Nanotechnology and Nano-materials for Environment Sustainability
- Influence of nano-silica and nano-ferrite particles on mechanical and durability of sustainable concrete: A review
- Surfaces and interfaces analysis on different carboxymethylation reaction time of anionic cellulose nanoparticles derived from oil palm biomass
- Processing and effective utilization of lignocellulosic biomass: Nanocellulose, nanolignin, and nanoxylan for wastewater treatment
- Retraction
- Retraction of “Aging assessment of silicone rubber materials under corona discharge accompanied by humidity and UV radiation”