6,629
Views
33
CrossRef citations to date
0
Altmetric
Reviews

Human macrophage responses to metal-oxide nanoparticles: a review

, , , &
Pages 694-703 | Received 27 Mar 2018, Accepted 20 Apr 2018, Published online: 04 May 2018

Abstract

Nanomaterials have been widely used in our daily lives in medicine, cosmetics, paints, textiles and food products. Many studies aim to determine their biological effects in different types of cells. The interaction of these materials with the immune system leads to reactions by modifying the susceptibility or resistance of the host body which could induce adverse health effects. Macrophages, as specific cells of the innate immune response, play a crucial role in the human defence system to foreign agents. They can be used as a reliable test object for the investigation of immune responses under nanomaterials exposure displayed by expression of a variety of receptors and active secretion of key signalling substances for these processes. This report covers studies of human macrophage behaviours upon exposure of nanomaterials. We focused on their interaction with metal-oxide nanoparticles as these are largely used in medical and cosmetics applications. The discussion and summary of these studies can guide the development of new nanomaterials, which are, at the same time, safe and useful for new purposes, especially for health applications.

Introduction

Nanomaterials have been broadly used in different fields owing to their large surface to volume ratio. Their key properties are often compared to their bulk counterparts [Citation1]. Metal-oxide nanomaterials, in particular, have found numerous applications mainly in the medical, cosmetic and food areas. Titanium dioxide nanoparticles act as photocatalysts and their optical and chemical properties make them attractive for applications in cosmetics and as coating materials for numerous purposes [Citation2–4]. Iron oxide, zinc oxide and cobalt oxide nanomaterials have been extensively studied due to their magnetic properties [Citation5–7]. The superparamagnetic behaviour of iron oxide nanoparticles allows their use in magnetic resonance imaging, drug delivery, biosensors and many other biomedical applications [Citation8,Citation9]. Zirconium oxides have demonstrated antibacterial activity [Citation10]. Besides, cerium oxide (CeO2), a potential catalytic antioxidant is another example of metal-oxide nanoparticles extensively studied from the past few years [Citation11,Citation12].

The remarkable variety of nanoparticle applications brings up the concern on their safety for human health. Biological effects can be triggered by nanoparticle exposure thanks to their unique physical and chemical properties. Cells in contact with foreign particles may induce an immunological response. The immune system consists of a group of cells and molecules responsible for defending the body against these foreign substances. presents the different cells of the immune system in various organs of the human body. The immune system is mainly divided into two subsystems: the innate immune system and the adaptive immune system. The innate immune system comprises the first front of immunological defence of the body after the anatomical and physical barriers, working in the recognition of the invader. On the other hand, the adaptive immune system is a more specific line of defence attacking the invader in different ways and generating immunological memories. Cells composing the innate immune system are phagocytic leukocytes, dendritic cells and natural killer cells.

Figure 1. Distribution of immune cells in various organs of the human body. Reproduced with permission from [Citation13]. Copyright 2013 Royal Society of Chemistry.

Figure 1. Distribution of immune cells in various organs of the human body. Reproduced with permission from [Citation13]. Copyright 2013 Royal Society of Chemistry.

Macrophages are leukocytes, which are originated by primitive cells from yolk sac, can be self-renew or derivate from bone marrow-derived monocytes [Citation14–17]. They accumulate in different organs of the body, as shown in , acting as scavengers and antigen-presenting cells [Citation13]. During inflammation or tissue injury, macrophages are commonly exhibiting pro- or anti-inflammatory characteristics depending on the stimuli and their origins. Phenotype 1 (M1) is an inflammatory macrophage while phenotype 2 (M2) has a regulatory function acting as an anti-inflammatory macrophage and is involved in tissue repair. Mantovani et al. [Citation18] reported a model presenting polarized macrophage and their specific properties under different stimuli (). Monocytic cell lines submitted to differentiation treatments are usually used to investigate macrophage functions in vitro. Exposure of the cell to different stimuli implies the development of specific phenotypes. Human monocytic THP-1 cells and human peripheral blood monocytes differentiated by phorbol 12-myristate 13-acetate (PMA) and colony-stimulating factors are broadly used in studies as macrophage models. In humans, the granulocyte macrophage colony-stimulating factor (GM-CSF) leads to M1 and the macrophage colony-stimulating factor (M-CSF) to M2 being able to polarize towards M1 after specific stimulation [Citation19].

Figure 2. Responses of various macrophage populations under different stimuli. DTH: delayed-type hypersensitivity; IC: immune complexes; IFN-g: interferon-g; iNOS: inducible nitric oxide synthase; LPS: lipopolysaccharide; MR: mannose receptor; PTX3: the long pentraxin PTX3; RNI: reactive nitrogen intermediates; ROI: reactive oxygen intermediates; SLAM: signalling lymphocytic activation molecule; SRs: scavenger receptors; TLR: Toll-like receptor. Reproduced with permission from [Citation18]. Copyright 2004 Elsevier.

Figure 2. Responses of various macrophage populations under different stimuli. DTH: delayed-type hypersensitivity; IC: immune complexes; IFN-g: interferon-g; iNOS: inducible nitric oxide synthase; LPS: lipopolysaccharide; MR: mannose receptor; PTX3: the long pentraxin PTX3; RNI: reactive nitrogen intermediates; ROI: reactive oxygen intermediates; SLAM: signalling lymphocytic activation molecule; SRs: scavenger receptors; TLR: Toll-like receptor. Reproduced with permission from [Citation18]. Copyright 2004 Elsevier.

Macrophages are characterized by the expression of specific proteins on their membranes, such as the glycoprotein CD14, which is a well-established differentiation marker for monocytes/macrophages [Citation20]. CD80 and CD200R are specific markers for M1 and M2, respectively [Citation18,Citation21]. Human macrophages express different markers compared to murine macrophages. The chemokine CCL2 is considered as a marker for polarization in murine macrophages but not in human macrophages [Citation19]. Functionally, these proteins act as receptors recognizing foreign particles and substances activating immune responses. The activation of receptors induces macrophage phagocytosis as well as the production of cytokines and chemokines [Citation22]. The investigation of the receptor expression and the presence of cytokines are useful parameters to evaluate the macrophages functions of defence. Accordingly, the interaction of nanomaterials with the human body can be evaluated by analysing how these nanomaterials interact with such cells.

In this review, we summarize the different reactions of human macrophages to metal-oxide nanoparticles and discuss the effect of modulated nanomaterial properties on this interaction.

Nanomaterial recognition by macrophages

The first step of the immune response is related to the recognition of the foreign compounds by macrophages. When nanomaterials enter a biological environment, they interact with molecules presented in this system, coating their surface and forming the protein corona. A wide range of studies suggests that the protein corona of nanoparticles formed in the biological environment plays an important role in the macrophage behaviour [Citation23–25]. Our group recently investigated the characteristics of the protein corona of TiO2 nanoparticles and the interaction of these particles with primary cells of human macrophages. We observed preferentially secondary modified protein in the corona of nanoparticles and IL-6 gene secretion by the macrophages even in low nanoparticle concentrations [Citation26]. Vogt et al. [Citation27] have shown that the protein corona modulates cellular uptake of silica-coated superparamagnetic iron oxide nanoparticles (SPIONs) in primary human macrophages. The uptake of these nanoparticles was enhanced by the presence of a protein corona. On the other hand, no difference was noticed in the uptake of dextran-coated SPIONs with or without corona.

Macrophages recognize foreign molecules expressing immune receptors. Among these are toll-like receptors (TLRs), nucleotide oligomerization domain-like receptors (NLRs), Fc receptors (FcRs), scavenger receptors (e.g. MARCO, SR-A1), integrins and carbohydrate receptors. The evaluation of these expressions is useful for the modulation of macrophage responses when exposed to nanoparticles.

TLR signalling is related to the initiation of the innate immune system reactions. The activation of TLR starts a complex intracellular signalling pathway. TLR1 to TLR10 have been well characterized in humans [Citation22]. Each TLR recognizes specific substances inducing different responses. Some of these receptors were predicted to recognize viruses. TLR7 expression is related to single stranded RNA (ssRNA) recognition and TLR3 responds to double-stranded RNA (dsRNA), produced by viruses during their replication. TLR9 is a receptor for bacterial- and viral-derived CpG DNA being its agonists. This aspect is of interest in vaccine development [Citation28,Citation29].

Lucarelli et al. [Citation30] have observed that the level of TLR7 is increasing in a population of human macrophages (PMA-differentiated U-937 cells) when in contact with TiO2 and ZrO2 at non-toxic concentrations. Moreover, it has been shown that ZrO2 nanoparticles induced TLR3 expression and TiO2 nanoparticles impaired by TLR9 expression. This finding demonstrates that these nanoparticles, especially ZrO2 nanoparticles, can enhance the reactivity of macrophages against viral infections and that TiO2 nanoparticles can decrease their ability to respond to vaccination. The increase in TLR10 expression also seen in their study possibly corroborates the hyperactivity of the body to viral infections in contact with both of metal-oxide nanoparticles. Even though the receptor TLR10 still has unknown functions, a study has demonstrated its role as a sensing receptor for virus in human macrophages [Citation31].

While TLRs are receptors located in the cell membrane, NLRs (nod-like receptors) are cytoplasmic receptors that oligomerize to form multiprotein complexes called inflammasome, activating inflammatory signallings [Citation32]. Yazdi et al. [Citation33] have investigated the inflammatory ability of TiO2 nanoparticles by the activation of the inflammasome NLRP3 (NLR pyrin domain containing 3) in human macrophages (PMA-differentiated THP-1 cells). Compared to different nanoparticles, TiO2 nanoparticles trigger NLRP3 activation in contrast to ZnO counterparts. Later on, Baron et al. [Citation34] have shown that the mechanisms mediating NLRP3 inflammasome activation by TiO2 nanoparticles involve exogenous adenosine. The authors showed that extracellular adenosine at high concentrations enhance ATP release and promote NLRP3 inflammasome activation.

Besides the ability to recognize foreign compounds through receptors, another feature of macrophages is the ability to internalize compounds and communicate with other cells through endocytosis. The internalization of these compounds not only permits their clearance from the body but also their presentation to specialized cells of the immune system allowing the so-called “immunological memory”. Endocytosis includes non-specific endocytosis such as phagocytosis and pinocytosis, and receptor-mediated endocytosis () [Citation35].

Figure 3. Mechanisms of endocytosis of nanoparticles (NP) and cellular components involved. Reproduced with permission from [Citation35]. Copyright 2013 American Chemical Society.

Figure 3. Mechanisms of endocytosis of nanoparticles (NP) and cellular components involved. Reproduced with permission from [Citation35]. Copyright 2013 American Chemical Society.

An efficient recognition of some particles by macrophages associated with the high endocytic activity of these cells can be useful for cell imaging in the diagnosis of specific diseases characterized by macrophage accumulation. Among these nanoparticles are the superparamagnetic iron oxide nanoparticles (SPIONs), used in the detection of atherosclerotic plaques and lymph-node metastases [Citation36,Citation37]. By using uptake inhibitors it is possible to determine which mechanism and cellular receptor are involved in the cellular uptake of nanomaterials. SPIONs and ultrasmall SPIONs (USPIONs) with a carboxydextran coating with 60 and 20 nm in diameter, respectively, were uptaken by primary human monocyte-derived macrophages (M-CSF-differentiated) via endocytosis mediated by the protein clathrin, as demonstrated by Lunov et al. [Citation38].

This mechanism is also involved in the uptake of ZnO nanoparticles by THP-1-derived macrophages (PMA-differentiated) as shown by James et al. [Citation39]. Lunov et al. [Citation38] have also detected the involvement of the scavenger receptor A (SR-A) in the uptake of SPION and USPION nanoparticles by primary human monocyte-derived macrophages (M-CSF-differentiated). SR-A also mediated the uptake of dextran-coated SPIONs (120–180 nm) by THP-1-derived macrophages (PMA-differentiated) [Citation40]. A peptidic SR-A type I ligand conjugated in USPIONs (15–30 nm) increased the uptake of these particles by macrophages (PMA-differentiated THP-1 and U937 cells) in the study of Segers et al. [Citation41] reinforcing the involvement of this receptor in the internalization of some SPIONs used as magnetic resonance contrasts.

SR-A are phagocytic recognition receptors, as well as opsonic FcR, and bind a broad range of ligands such as modified low-density lipoprotein (LDL), polynucleic acids and bacterial components [Citation42]. The pathogenesis of some diseases with high macrophage accumulation can be related to the high expression of this receptor [Citation43,Citation44]. ZnO nanoparticles upregulated the expression of SR-A and CD36 (SR-B) in THP-1-derived macrophages (PMA-differentiated cells) increasing the uptake of modified LDL by these cells. On the other hand, TiO2 nanoparticles presented the opposite effect on CD36 expression and have no effect on SR-A expression. The effect promoted by ZnO nanoparticles suggests that these particles induce the progression of atherosclerosis in humans [Citation45].

The interaction of macrophages with SPIONs can vary depending on the functionalization of these nanoparticles. The presence of amino groups on the surface of SPIONs allows their uptake by primary human monocyte-derived macrophages (PMA-differentiated) through the integrin receptor Mac-1 [Citation46]. This receptor consists of integrins (αM and β2) involved in several immune responses besides phagocytosis. Zhou et al. [Citation47] reported that Mac-1 receptors have also been involved in the extracellular dsRNA recognition. Folic acid functionalization of iron oxide nanoparticles with an organic and inorganic layers leads to the uptake by primary human macrophages (M-CSF differentiated) through endocytosis and folate receptor-α independently [Citation48].

The charge of the nanoparticle will also define the nature of the protein adsorbed to their surface in a biological medium leading their interactions with cells. Deng et al. [Citation49] suggested that negatively charged nanoparticles interact with Mac-1 receptor on THP-1 cells due to binding and unfolding induction of fibrogen by these nanoparticles. Conformational changes of proteins expose buried sequences influencing the interaction of nanoparticles with cells [Citation50].

The recognition of nanomaterials by macrophage receptors activates intracellular pathways leading to the production of cytokines and chemokines, and are promoting the sequence of immune reactions.

Macrophage activation by metal-oxide nanomaterials

The macrophages activation has several mechanisms. Elucidation of signalling pathways of macrophages upon a specific material is essential for understanding the inflammatory process. Therewith, regulatory mechanisms can be studied to tune macrophages activation.

Nanoparticles can elicit immune-modulatory effects on the body. Induction of inflammatory and anti-inflammatory cytokines reveals the immune-stimulation and immune-suppression ability of nanoparticles, respectively. Nanoparticles with high potential for imaging applications, for example, SPIONs, must avoid immune recognition. Müller et al. [Citation51] have demonstrated that dextran-coated USPIONs (30 nm) did not trigger inflammatory responses on primary human macrophages. The authors have investigated the secretion of IL-12, IL-6, TNF-α and IFN-γ by the cells. Vogt et al. [Citation27] also have not observed release of TNF-α on primary human macrophages under exposure of dextran-coated SPIONs (multiple iron core clusters of ∼50 nm) and silica-coated SPIONs (single core shell nanoparticles of ∼50 nm). On the other hand, a desired immune-activation can be useful in the development of therapeutic nanoparticles. Chattopadhyay et al. [Citation52] demonstrated that primary human macrophages were activated by CoO nanoparticles loaded with tumour-associated antigens, under non-toxic concentrations, releasing TNF-α, IFN-γ and IL-12. The human oral carcinoma cell line KB was used to investigate antitumor effects under nanoparticles activation. Since macrophages are present in or around tumours, this augment of the cytokine production induces the adaptive immunity resulting in an efficient immune activity around the region to be treated [Citation52,Citation53]. Upon studying cell elasticity of human macrophages, Gonnissen et al. [Citation54] demonstrated a decrease of the elasticity in the presence of starch coated iron oxide nanoparticles, which also resulted in a decrease of transmigratory activity of such cells. The latter effects might be useful, for example, to develop anti-inflammatory active nanoparticles, and demonstrate that the effects of nanoparticles can be more delicate than just affecting the survival of cells.

The ability of some metal-oxide nanoparticles to induce cytokine production was presented by several authors. In situ studies performed by Andujar et al. [Citation55] showed that the chemical ingredient of metal-oxide nanoparticles detected in macrophages presented in the lung tissue of welders, workers commonly exposed to metal fumes, were mainly Fe, Cr and Mn. By analysing THP-1 macrophages (PMA-differentiated cells) in vitro the authors demonstrated that Fe2O3 and alloyed metal oxide MnFe2O4 nanoparticles induce mainly the secretion of pro-inflammatory cytokines TNF-α, IL-1β, CCL-2, CCL-3, CCL-4 and CXCL-9. Fe3O4 was the lowest reactive nanoparticle.

Yazdi et al. [Citation33] observed that anatase TiO2 nanoparticles (20 nm and rutile 80 nm) trigger the secretion of IL-1β in the same type of macrophages, whereas ZnO nanoparticles (15 nm) present no effects on these cells. The inflammatory pathway of IL-1β starts with the activation of the adaptor protein Myd88 to the receptor leading to the translocation of the transcription factor NF-κB into the nucleus. IL-1β pathway also comprises the proteinase caspase-1 activation. Thereafter, Baron et al. [Citation34] have also shown the involvement of adenosine on the IL-1β secretion. TiO2 nanoparticles induced the release of ATP leading to IL-1β secretion. The authors demonstrated that the inhibition of the P2X7 receptor (purinergic receptor for ATP) of THP-1 macrophages reduced IL-1β secretion. Therefore, investigation on the activity of these compounds is important to determine the secretion of mature forms of cytokines [Citation56,Citation57]. It is well known that IL-1β contributes to inflammatory diseases [Citation56,Citation58]. Using the same type of macrophages, the results by Yazdi et al. [Citation33] for TiO2 nanoparticles and ZnO nanoparticles were corroborated by Morishige et al. [Citation59] and Xia et al. [Citation60], respectively. On the other hand, TiO2 with a belt shape (7000 nm length ×200 nm width ×10 nm thickness), rather than particles (81/19% anatase/rutile, 24 nm and 100% anatase, 28 nm) was the only nanomaterial that presented the ability to induce IL-1β expression in the study of Xia et al. [Citation60]. Considering that the studies of Yazdi et al. [Citation33] and Morishige et al. [Citation59] range TiO2 nanoparticle concentrations of 20–500 µg/mL for cell stimulation, the inconsistency of their results with those obtained by Xia et al. [Citation60] (nanoparticle concentrations up to 100 µg/mL) is possibly due to the characteristics of the cell population investigated. While the latter authors used 5 nM PMA associated with vitamin D3 on THP-1 cells differentiated by 10 ng/mL LPS, the former authors used a 100 times higher PMA concentration. Concerning PMA treatment on THP-1, the conditions for the culture have to be carefully monitored to obtain a homogeneous culture expressing reliable responses [Citation61]. Thus, the precise definition of a cell population in studies involving bioactivity is essential.

Macrophages also differ in their ability to respond to nanoparticles depending on their activation state. TiO2 (70 nm) and ZrO2 (5–30 nm) nanoparticles have increased the level of anti-inflammatory IL-1Ra, the receptor antagonist of IL-1, on PMA-differentiated U-937 macrophages and decreased its level on the same cells exposed to LPS (M1 type macrophage) in the study of Lucarelli et al. [Citation30]. ZrO2 nanoparticles have enhanced IL-1β production on LPS-activated macrophages. On the other hand, some metal-oxide materials demonstrate effects on macrophage polarization. SPIONs formulated with different coatings evince distinct reactions on these cells. Laskar et al. [Citation62] have demonstrated the carboxydextran-coated SPIONs (45–60 nm) accumulation in M2-differentiated THP-1 cells affecting their phenotype and leading to a shift towards an M1-like phenotype. Later, Rojas et al. [Citation63] showed that the same cells have their activation profiles changed by DMSA-, APS- and AD-SPIONs without switching to M1 phenotype. The authors also observed differences in the gene expression profiles of these cells and murine primary IL-4-activated bone marrow-derived macrophages differentiated with M-CSF after interaction with SPIONs, suggesting that the choice of the model to be studied and its initial activation state are of great importance in the evaluation of the nanoparticles effects.

Nanomaterials can induce the production of some toxic mediators, such as reactive oxygen species (ROS), since they are able to reach mitochondria directly, and thereby induce inflammatory pathways on activated macrophages. Morishige et al. [Citation59] and Yazdi et al. [Citation33] demonstrated that ROS are responsible for inflammasome activation leading to IL-1β maturation in PMA-differentiated THP-1 macrophages, under TiO2 nanoparticle stimulation.

Oxidative stress occurs when the concentration of oxidizing compounds increases causing inflammatory and toxic effects to the cells () [Citation64].

Figure 4. Responses of the cells under oxidative stress induced by nanoparticles (NP). Reproduced with permission from [Citation64]. Copyright 2008 Elsevier.

Figure 4. Responses of the cells under oxidative stress induced by nanoparticles (NP). Reproduced with permission from [Citation64]. Copyright 2008 Elsevier.

Rahman et al. [Citation65] showed that human alveolar macrophages produce more ROS than rat alveolar macrophages in contact with ultrafine TiO2 nanoparticles making them a more reliable indicator for the toxicity of some nanomaterials.

The ability of metal ions to enhance the ROS response on macrophages has been observed by Mokgobu et al. [Citation66] and Petit et al. [Citation67]. These studies revealed that uncoated iron oxide nanoparticles are degraded into toxic metal ions inside the cells [Citation68–70].

The toxicity is enhanced in the case of macrophages that have higher ability to reduce iron than other cells being the ferrous state catalyser of hydroxyl radical generation via the Fenton reaction [Citation71,Citation72]. Lunov et al. [Citation73] showed that carboxydextran-coated SPIONs, small (∼60 nm) and ultrasmall (∼20 nm), also induce toxicity on monocyte-derived macrophages (M-CSF-differentiated) by ROS production after exposure times over 24 h. These nanoparticles trigger cell apoptosis due to the degeneration of the coating and exposure of the reactive core. Exposure times under 24 h did not induce any cytotoxicity on these cells as was also shown by other studies for human fibroblasts and human carcinoma cells [Citation74–76].

Strehl et al. [Citation77] recently demonstrated that amino-polyvinyl alcohol-coated SPIONs even increase the same type of cell survival under 20 h of exposure.

ROS scavengers are useful to reduce the damage of oxidative stress. The body has an antioxidant defence system composed of enzymes (e.g. superoxide dismutase) and substances like glutathione and some vitamins. However, facing the abundance of ROS, several substances such as Trolox, PEG, BHA and NAPDH oxidase have been used in studies presenting efficient ROS scavenging abilities [Citation59,Citation73,Citation78–80]. Interestingly, some metal oxide nanoparticles present this property, as is the case of cerium oxide nanoparticles. Cerium oxide exhibit catalytic properties since they present a reversibility of the oxidation states Ce+3 and Ce+4 that leads to oxygen vacancies [Citation81]. Lord et al. [Citation82] investigated the antioxidant properties of cerium oxide nanoparticles in U-937 derived macrophages (PMA-differentiated) exposing the cells for up to 72 h to the nanoparticle. The authors demonstrated the uptake of the nanoparticles by the cells leading to a continuous intracellular ROS scavenger action.

For an overview, some interactions of metal-oxide nanoparticles with human macrophages are listed in .

Table 1. Interaction of different metal-oxide nanomaterials with human macrophages.

Physicochemical properties of metal-oxide nanomaterials on macrophage behaviour

As mentioned previously, the characterization of the working population of cells is highly important for the correct analysis of macrophage responses upon contact with nanoparticles. Besides, the properties of the nanoparticles can greatly affect their interaction with macrophages and sequential intracellular signalling.

Studies involving nanomaterials had aroused the interest of the scientific community mainly due to their nanoscale sizes (i.e. high surface area to volume ratio), which lets them display quite different properties from their bulk counterparts. The ability of ultrasmall metal-oxide nanoparticles (<30 nm) to induce more pronounced effect on human macrophages than small (∼60 nm) nanoparticles was demonstrated in some studies [Citation40,Citation73]. An impairment of primary human monocyte-derived macrophage (M-CSF-differentiated) function and an enhanced long-term toxicity (>24 h) after exposure of carboxydextran-coated ultrasmall SPIONs (20 nm) was observed by Lunov et al. [Citation73] measuring iron uptake and ROS generation compared to carboxydextran-coated small SPIONs (60 nm). Cell viability measurements demonstrated that macrophages only exhibit translocation of membrane components after 3-days exposure of small SPIONs, characteristic of the early signs of cell death by apoptosis, while the same time exposure with ultrasmall SPIONs resulted in a loss of integrity of the cell membrane, an evidence of the late stage of apoptosis. These results were confirmed by the detection of a higher activity of caspase-3 on macrophages exposed to ultrasmall than small SPIONs. This enzyme is activated by mitochondrial components released during apoptosis [Citation72]. The authors also developed the mathematical modelling of the uptake of these nanoparticles by the macrophages enabling the prediction of parameters such as wrapping time and uptake rate of the nanoparticles. The uptake mechanism was the same for both nanoparticles being clathrin-mediated endocytosis, however, ultrasmall SPIONs were uptaken faster than small SPIONs [Citation38].

Of note, studies with alternatively labelling human monocytes for imaging with SPION and USPIONs also showed higher internalization of SPIONs than USPIONs by these cells [Citation83].

Some metal-oxide nanomaterials present unique characteristics that have to be taken into account together with the size on the evaluation of their effects. Among these are TiO2 nanoparticles. TiO2 may exhibit three different crystal structures: anatase, rutile and brookite, with anatase and rutile vastly studied. Morishige et al. [Citation59] have reported that smaller anatase and larger rutile induced IL-β secretion by PMA-differentiated THP-1 macrophages. The authors also demonstrated higher stimulation ability of rutile than anatase TiO2 nanoparticles on the macrophages.

Proteins bound to the nanomaterial surface mediate immune responses, as discussed previously. The physicochemical properties of materials influence the amount and the profile of proteins adsorbed to their surfaces leading to different cell outcomes [Citation24,Citation83]. Surface coating, a preset to alter undesirable properties of nanoparticles, can highly affect the nanoparticle-protein corona since the surface is the first contact site of the nanoparticles with the cellular environment. Consequently, the interaction of nanoparticles with cells occurs via this protein corona [Citation26].

Uncoated TiO2 nanoparticles adsorbed more proteins than alumina and silicone-coated particles in contact with primary human monocyte-derived macrophages (GM-CSF-differentiated) as demonstrated by Sund et al. [Citation84]. Later on, the authors studied the cytoplasmic protein changes of the macrophages in contact with uncoated- and silica coated-TiO2 nanoparticles. These proteomic changes of macrophages can be considered as an indicator of certain biological activities, for example, phagocytosis. This study has shown that TiO2 nanoparticles induce pronounced changes in the proteome of the same human macrophages when they are coated [Citation85]. The difference on the protein binding due to different surface charges of silica-coated (30–120 nm) and dextran-coated (20 and 50 nm) SPIONs was suggested by Kunzmann [Citation86] to be the reason for the higher uptake detected for silica-coated SPIONs by primary human monocyte-derived macrophages (M-CSF differentiated). Using inductively coupled plasma-mass spectrometry (ICP-MS) with an inhibitor of actin polymerization, the authors monitored the uptake of these particles and verified that silica-coated SPIONs were taken up to a higher degree using an actin-cytoskeleton dependent mechanism. On the other hand, Deng et al. [Citation87] showed that some metal oxide nanoparticles, such as TiO2 and ZnO, have the ability to bind different proteins in contact with human plasma even though they present similar surface charges. Therefore, the degree of complexity for protein–nanoparticle interaction is high and many factors have to be considered together with the nanoparticle characteristics, like the agglomeration state in biological systems, and further in vivo studies have to be developed since the protein binding status is highly variable within protean biological conditions.

Compared to the size, the surface properties of SPIONs have higher impact on the phagocytosis in human monocytes and rat macrophages cell lines [Citation83].

The shape of nanomaterials is another factor to consider since the material surface area interacting with the environmental changes. The study of Xia et al. [Citation60] showed that the cytokine release by PMA-differentiated THP-1 macrophages was influenced by their shape once exposed to TiO2 nanomaterials (nanoparticles and nanobelts). TiO2 nanospheres attract qualitatively more protein than TiO2 nanorods (ellipsoid shaped nanomaterials) and TiO2 nanotubes, which will trigger different immune responses [Citation87].

Therefore, the characterization of the nanomaterials is of great importance and a prerequisite for their safe and actual medical application toward human beings.

Conclusion

Evaluation of the interaction of macrophages with different metal-oxide nanomaterials, as discussed in this report, is important for the handling the risk in the development of new nanomaterials. Interactions between cells and nanoparticles are regulated by a plethora of factors including the design, shape and surface chemistry of the nanoparticle on the one hand and the characteristics of the cells and the experimental conditions on the other hand. That is why general conclusions can only be made including all of the abovementioned factors.

Disclosure statement

The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. No writing assistance was utilized in the production of this manuscript.

References

  • Riehemann K, Schneider SW, Luger TA, et al. Nanomedicine-challenge and perspectives. Angew Chem Int Ed Engl. 2009;48:872–897.
  • Leyland NS, Podporska-Carroll J, Browne J, et al. Highly efficient F, Cu doped TiO2 anti-bacterial visible light active photocatalytic coatings to combat hospital-acquired infections. Sci Rep. 2016;6:24770.
  • Pakdel E, Daoud WA, Sun L, et al. Reprint of: photostability of wool fabrics coated with pure and modified TiO2 colloids. J Colloid Interface Sci. 2015;447:191–201.
  • Weir A, Westerhoff P, Fabricius L, et al. Titanium dioxide nanoparticles in food and personal care products. Environ Sci Technol. 2013;46:2242–2250.
  • Shi D, Sadat ME, Dunn AW, et al. Photo-fluorescent and magnetic properties of iron oxide nanoparticles for biomedical applications. Nanoscale. 2015;7:8209–8232.
  • Xiong HM. ZnO nanoparticles applied to bioimaging and drug delivery. Adv Mater. 2013;25:5329–5335.
  • Yang H, Zhou H, Zhang C, et al. Water-soluble magnetic CoO nanocrystals functionalized with surfactants as T2-weighed MRI contrast agents in vitro. Dalton Trans. 2011;40:3616–3621.
  • Laurent S, Saei AA, Behzadi S, et al. Superparamagnetic iron oxide nanoparticles for delivery of therapeutic agents: opportunities and challenges. Expert Opin Drug Deliv. 2014;11:1449–1470.
  • Likhitkar S, Bajpai AK. Magnetically controlled release of cisplatin from superparamagnetic starch nanoparticles. Carbohydr Polym. 2012;87:300–308.
  • Jangra SL, Stalin K, Dilbaghi N, et al. Antimicrobial activity of zirconia (ZrO2) nanoparticles and zirconium complexes. J Nanosci Nanotechnol. 2012;12:7105–7112.
  • Walkey C, Das S, Seal S, et al. Catalytic properties and biomedical applications of cerium oxide nanoparticles. Environ Sci Nano. 2015;2:33–53.
  • Li K, Xie Y, You M, et al. Plasma sprayed cerium oxide coating inhibits H2O2-induced oxidative stress and supports cell viability. J Mater Sci Mater Med. 2016;27:100.
  • Elsabahy M, Wooley KL. Cytokines as biomarkers of nanoparticle immunotoxicity. Chem Soc Rev. 2013;42:5552–5576.
  • Ginhoux F, Greter M, Leboeuf M, et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science. 2010;330:841–845.
  • Jenkins SJ, Ruckerl D, Cook PC, et al. Local macrophage proliferation, rather than recruitment from the blood, is a signature of Th2 inflammation. Science. 2011;332:1284–1288.
  • Schulz C, Gomez PE, Chorro L, et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science. 2012;336:86–90.
  • Hashimoto D, Chow A, Noizat C, et al. Tissue-resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity. 2013;38:792.
  • Mantovani A, Sica A, Sozzani S, et al. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004;25:677–686.
  • Jaguin M, Houlbert N, Fardel O, et al. Polarization profiles of human M-CSF-generated macrophages and comparison of M1-markers in classically activated macrophages from GM-CSF and M-CSF origin. Cell Immunol. 2013;281:51–61.
  • Ziegler-Heitbrock HWL, Ulevitch RJ. CD14: cell surface receptor and differentiation marker. Immunol Today. 1993;14:121–125.
  • Ambarus CA, Krausz S, van Eijk M, et al. Systematic validation of specific phenotypic markers for in vitro polarized human macrophages. J Immunol Methods. 2012;375:196–206.
  • Janeway CA, Medzhitov R. Innate immune recognition. Annu Rev Immunol. 2002;20:197–216.
  • Fedeli C, Segat D, Tavano R, et al. Variations of the corona HDL: albumin ratio determines distinct effects of amorphous SiO2 nanoparticles on monocytes and macrophages in serum. Nanomedicine. 2014;9:2481–2497.
  • Landfester K, Simmet T. Differential uptake of functionalized polystyrene nanoparticles by human macrophages and a monocytic cell line. ACS Nano. 2011;5:1657–1669.
  • Saptarshi SR, Duschl A, Lopata AL. Interaction of nanoparticles with proteins: relation to bio-reactivity of the nanoparticle. J Nanobiotechnol. 2013;11:26.
  • Borgognoni CF, Mormann M, Qu Y, et al. Reaction of human macrophages on protein corona covered TiO2 nanoparticles. Nanomedicine. 2015;11:275–282.
  • Vogt C, Pernemalm M, Kohonen P, et al. Proteomics analysis reveals distinct corona composition on magnetic nanoparticles with different surface coatings: implications for interactions with primary human macrophages. PLoS One. 2015;10:1–20.
  • Akira S. Innate immunity and adjuvants. Philos Trans Roy Soc Lond, B, Biol Sci. 2011;366:2748–2755.
  • Duthie MS, Windish HP, Fox CB, et al. Use of defined TLR ligands as adjuvants within human vaccines. Immunol Rev. 2011;239:178–196.
  • Lucarelli M, Gatti AM, Savarino G, et al. Innate defence functions of macrophages can be biased by nano-sized ceramic and metallic particles. Eur Cytokine Netw. 2004;15:339–346.
  • Lee SMY, Kok K-H, Jaume M, et al. Toll-like receptor 10 is involved in induction of innate immune responses to influenza virus infection. Proc Natl Acad Sci USA. 2014;111:3793–3798.
  • Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-β. Mol Cell. 2002;10:417–426.
  • Yazdi AS, Guarda G, Riteau N, et al. Nanoparticles activate the NLR pyrin domain containing 3 (Nlrp3) inflammasome and cause pulmonary inflammation through release of IL-1α and IL-1β. Proc Natl Acad Sci USA. 2010;107:19449–19454.
  • Baron L, Gombault A, Fanny M, et al. The NLRP3 inflammasome is activated by nanoparticles through ATP, ADP and adenosine. Cell Death Dis. 2015;6:1–15.
  • Zhu M, Nie G, Meng H, et al. Physicochemical properties determine nanomaterial cellular uptake, transport and fate. Acc Chem Res. 2013;46:622–631.
  • Morishige T, Yoshioka Y, Tanabe A, et al. Titanium dioxide induces different levels of IL-1beta production dependent on its particle characteristics through caspase-1 activation mediated by reactive oxygen species and cathepsin B. Biochem Biophys Res Commun. 2010;392:160–165.
  • Vu-Quang H, Yoo MK, Jeong HJ, et al. Targeted delivery of mannan-coated superparamagnetic iron oxide nanoparticles to antigen-presenting cells for magnetic resonance-based diagnosis of metastatic lymph nodes in vivo. Acta Biomater. 2011;7:3935–3945.
  • Lunov O, Zablotskii V, Syrovets T, et al. Modelling receptor-mediated endocytosis of polymer-functionalized iron oxide nanoparticles by human macrophages. Biomaterials. 2011;32:547–555.
  • James SA, Feltis BN, de Jonge MD, et al. Quantification of ZnO nanoparticle uptake, distribution and dissolution within individual human macrophages. ACS Nano. 2013;7:10621–10635.
  • Raynal I, Prigent P, Peyramaure S, et al. Macrophage endocytosis of superparamagnetic iron oxide nanoparticles: mechanisms and comparison of ferumoxides and ferumoxtran-10. Invest Radiol. 2004;39:56–63.
  • Segers FME, den Adel B, Bot I, et al. Scavenger receptor-AI-targeted iron oxide nanoparticles for in vivo MRI detection of atherosclerotic lesions. Arterioscler Thromb Vasc Biol. 2013;33:1812–1819.
  • Bowdish DME, Gordon S. Conserved domains of the class A scavenger receptors: evolution and function. Immunol Rev. 2009;227:19–31.
  • Matsumoto A, Naito M, Itakura H, et al. Human macrophage scavenger receptors: primary structure, expression, and localization in atherosclerotic lesions. Proc Natl Acad Sci USA.1990;7:9133–9137.
  • Suzuki H, Kurihara Y, Takeya M, et al. A role for macrophage scavenger receptors in atherosclerosis and susceptibility to infection. Nature. 1997;386:292–296.
  • Suzuki Y, Tada-Oikawa S, Ichihara G, et al. Zinc oxide nanoparticles induce migration and adhesion of monocytes to endothelial cells and accelerate foam cell formation. Toxicol Appl Pharmacol. 2014;278:16–25.
  • Von zur Muhlen C, von Elverfeldt D, Bassler N, et al. Superparamagnetic iron oxide binding and uptake as imaged by magnetic resonance is mediated by the integrin receptor Mac-1 (CD11b/CD18): implications on imaging of atherosclerotic plaques. Atherosclerosis. 2007;193:102–111.
  • Zhou H, Liao J, Aloor J, et al. CD11b/CD18 (Mac-1) is a novel surface receptor for extracellular double-stranded RNA to mediate cellular inflammatory responses. J Immunol. 2013;190:115–125.
  • Krais A, Wortmann L, Hermanns L, et al. Targeted uptake of folic acid-functionalized iron oxide nanoparticles by ovarian cancer cells in the presence but not in the absence of serum. Nanomedicine. 2014;10:1421–1431.
  • Deng ZJ, Liang M, Monteiro M, et al. Nanoparticle-induced unfolding of fibrinogen promotes Mac-1 receptor activation and inflammation. Nature Nanotech. 2011;6:39–44.
  • Lynch I, Salvati A, Dawson KA. Protein-nanoparticle interactions: what does the cell see? Nat Nanotechnol. 2009;4:546–547.
  • Müller K, Skepper JN, Posfai M, et al. Effect of ultrasmall superparamagnetic iron oxide nanoparticles (Ferumoxtran-10) on human monocyte-macrophages in vitro. Biomaterials. 2007;28:1629–1642.
  • Chattopadhyay S, Dash SK, Ghosh T, et al. Anticancer and immunostimulatory role of encapsulated tumour antigen containing cobalt oxide nanoparticles. J Biol Inorg Chem. 2013;18:957–973.
  • Kohchi C, Inagawa H, Hino M, et al. Utilization of macrophages in anticancer therapy: the macrophage network theory. Anticancer Res. 2004;24:3311–3320.
  • Gonnissen D, Qu Y, Langer K, et al. Comparison of cellular effects of starch coated SPIONs and PLGA matrix nanoparticles in human monocytes. Int J Nanomed. 2016;11:5221–5236.
  • Andujar P, Simon-Deckers A, Galateau-Sallé F, et al. Role of metal oxide nanoparticles in histopathological changes observed in the lung of welders. Part Fibre Toxicol. 2014;11:23
  • Dinarello CA,. Interleukin-1 in the pathogenesis and treatment of inflammatory diseases. A Review. Blood. 2011;117:3720–3732.
  • Kim SB, Natori Y, Ozawa T, et al. A method for determining the activities of cytokines based on the nuclear transport of nuclear factor-kappaB. Anal Biochem. 2006;359:147–149.
  • Rider P, Carmi Y, Guttman O, et al. IL-1α and IL-1β recruit different myeloid cells and promote different stages of sterile inflammation. J Immunol. 2011;187:4835–4843.
  • Morishige K, Kacher DF, Libby P, et al. High-resolution magnetic resonance imaging enhanced with superparamagnetic nanoparticles measures macrophage burden in atherosclerosis. Circulation. 2010;122:1707–1715.
  • Xia T Jr, Hamilton RF, Bonner JC, et al. Interlaboratory evaluation of in vitro cytotoxicity and inflammatory responses to engineered nanomaterials: the NIEHS Nano GO Consortium. Environ Health Perspect. 2013;121:683–690.
  • Aldo PB, Craveiro V, Guller S, et al. Effect of culture conditions on the phenotype of THP-1 monocyte cell line. Am J Reprod Immunol. 2013;70:80–86.
  • Laskar A, Eilertsen J, Li W, et al. SPION primes THP1 derived M2 macrophages towards M1-like macrophages. Biochem Biophys Res Commun. 2013;441:737–742.
  • Rojas JM, Sanz-Ortega L, Mulens-Arias V, et al. Superparamagnetic iron oxide nanoparticle uptake alters M2 macrophage phenotype, iron metabolism, migration and invasion. Nanomed Nanotechnol Biol Med. 2016;12:1127–1138.
  • Sanvicens N, Marco MP. Multifunctional nanoparticles – properties and prospects for their use in human medicine. Trends Biotechnol. 2008;26:425–433.
  • Rahman Q, Norwood J, Hatch G. Evidence that exposure of particulate air pollutants to human and rat alveolar macrophages leads to differential oxidative response. Biochem Biophys Res Commun. 2007;240:669–672.
  • Mokgobu MI, Anderson R, Steel HC, et al. Manganese promotes increased formation of hydrogen peroxide by activated human macrophages and neutrophils in vitro. Inhalation Toxicol. 2012;24:634–644.
  • Petit A, Mwale F, Tkaczyk C, et al. Induction of protein oxidation by cobalt and chromium ions in human U937 macrophages. Biomaterials. 2005;26:4416–4422.
  • Apopa PL, Qian Y, Shao R. Iron oxide nanoparticles induce human microvascular endothelial cell permeability through reactive oxygen species production and microtubule remodelling. Part Fibre Toxicol. 2009;6:1.
  • Dwivedi S, Siddiqui MA, Farshori NN, et al. Synthesis, characterization and toxicological evaluation of iron oxide nanoparticles in human lung alveolar epithelial cells. Colloids Surf B Biointerfaces. 2014;122:209–215.
  • Pisanic TR II, Blackwell JD, Shubayev VI, et al. Nanotoxicity of iron oxide nanoparticle internalization in growing neurons. Biomaterials. 2007;28:2572–2581.
  • Partridge J, Wallace DF, Raja KB, et al. Monocyte-macrophage ferric reductase activity is inhibited by iron and stimulated by cellular differentiation. Biochem J. 1998;336:541–543.
  • Stohs SJ, Bagchi D. Oxidative mechanisms in the toxicity of metal ions. Free Radic Biol Med. 1995;18:321–336.
  • Lunov O, Syrovets T, Büchele B, et al. The effect of carboxydextran-coated superparamagnetic iron oxide nanoparticles on c-Jun N-terminal kinase-mediated apoptosis in human macrophages. Biomaterials. 2010;31:5063–5071.
  • Auffan M, Decome L, Rose J, et al. In vitro interactions between DMSA-coated maghemite nanoparticles and human fibroblasts: a physicochemical and cyto-genotoxical study. Environ Sci Technol. 2006;40:4367–4373.
  • Hong SC, Lee JH, Lee J, et al. Subtle cytotoxicity and genotoxicity differences in superparamagnetic iron oxide nanoparticles coated with various functional groups. Int J Nanomedicine. 2011;6:3219–3231.
  • Villanueva A, Cañete M, Roca AG, et al. The influence of surface functionalization on the enhanced internalization of magnetic nanoparticles in cancer cells. Nanotechnology. 2009;20:115103.
  • Strehl C, Gaber T, Maurizi L, et al. Effects of PVA coated nanoparticles on human immune cells. Int J Nanomedicine. 2015;10:3429–3445.
  • Liu-Snyder P, Logan MP, Shi R, et al. Neuroprotection from secondary injury by polyethylene glycol requires its internalization. J Exp Biol. 2007;210:1455–1462.
  • Mulens-Arias V, Rojas JM, Pérez-Yagüe S, et al. Polyethylenimine-coated SPIONs trigger macrophage activation through TLR-4 signalling and ROS production and modulate podosome dynamics. Biomaterials. 2015;52:494–506.
  • Urner M, Schlicker A, Z'graggen BR, et al. Inflammatory response of lung macrophages and epithelial cells after exposure to redox active nanoparticles: effect of solubility and antioxidant treatment. Environ Sci Technol. 2014;48:13960–13968.
  • Shoko E, Smith MF, McKenzie RH. Charge distribution near bulk oxygen vacancies in cerium oxides. J Phys: Condens Matter. 2010;22:223201.
  • Lord MS, Jung M, Teoh WY, et al. Cellular uptake and reactive oxygen species modulation of cerium oxide nanoparticles in human monocyte cell line U937. Biomaterials. 2012;33:7915–7924.
  • Metz S, Bonaterra G, Rudelius M, et al. Capacity of human monocytes to phagocytose approved iron oxide MR contrast agents in vitro. Eur Radiol. 2004;14:1851–1858.
  • Sund J, Alenius H, Vippola M, et al. Proteomic characterization of engineered nanomaterial–protein interactions in relation to surface reactivity. ACS Nano. 2011;5:4300–4309.
  • Sund J, Palomäki J, Ahonen N, et al. Phagocytosis of nano-sized titanium dioxide triggers changes in protein acetylation. J Proteomics. 2014;108:469–483.
  • Kunzmann A, Andersson B, Vogt C, et al. Efficient internalization of silica-coated iron oxide nanoparticles of different sizes by primary human macrophages and dendritic cells. Toxicol Appl Pharmacol. 2011;253:81–93.
  • Deng ZJ, Mortimer G, Schiller T, et al. Differential plasma protein binding to metal oxide nanoparticles. Nanotechnology. 2009;20:455101.