Abstract
Targeting the immune system with nanomaterials is an intensely active area of research. Specifically, the capability to induce immunosuppression is a promising complement for drug delivery and regenerative medicine therapies. Many novel strategies for immunosuppression rely on nanoparticles as delivery vehicles for small-molecule immunosuppressive compounds. As a consequence, efforts in understanding the mechanisms in which nanoparticles directly interact with the immune system have been overshadowed. The immunological activity of nanoparticles is dependent on the physiochemical properties of the nanoparticles and its subsequent cellular internalization. As the underlying factors for these reactions are elucidated, more nanoparticles may be engineered and evaluated for inducing immunosuppression and complementing immunosuppressive drugs. This review will briefly summarize the state-of-the-art and developments in understanding how nanoparticles induce immunosuppressive responses, compare the inherent properties of nanomaterials which induce these immunological reactions, and comment on the potential for using nanomaterials to modulate and control the immune system.
Keywords: Nanoparticles, immunosuppression, bionanoscience, nanotoxicology, immunotoxicity, anti-inflammatory
Introduction
Immunosuppression is the effect of a drug or other material to reduce the activation or efficacy of the immune system. Conventionally, immunosuppression is considered an immunotoxic effect implicated in an array of human pathologies. Immunosuppressive therapies often rely on the use of small molecule medications and biologics, and although the acute efficacy of these therapies has improved over the recent decade, long-term use of an immunosuppressant results in co-morbidities due to systemic toxicity or immunodeficiency.1–3 Immunosuppressive strategies that pin-point specific tissue or pathways, while reducing systemic side effects, are a potential panacea for treating autoimmune disorders and complementing pharmaceuticals. To achieve these goals, many chemical and biomedical researchers have engineered nanoparticles to carry and locally deliver immunosuppressive agents.4–10 This can be considered “indirect immunosuppression,” where the nanoparticle solely serves as the delivery vehicle.7 As an alternative, a small but prevailing body of literature is reassessing direct immunosuppression by nanoparticles to be exploited as a compliment for drug therapies or organ/tissue transplantation that otherwise would be rendered ineffective or rejected by the native immune response.11–15 Reports in the fields of nanotoxicology and nanomedicine are exploring the fundamental mechanisms for both intended and unintended immunosuppression by nanomaterials.16–19 Understanding the direct immunosuppressive effect of nanoparticles and resultant development of new immunosuppressive therapies can impact both pharmaceutical and regenerative medicine technologies.9,20,21
When developing methods to suppress immune response, it is important to consider both innate and adaptive immune responses. Innate immunity involves the first response activation of macrophages and neutrophils that secrete specific cytokines, which produce local inflammation at the site of reaction. This inflammation is usually followed by the adaptive immune response that activates antigen specific cells, T cells and B cells to target the pathogen or other instigator of the immune response. Both innate and adaptive responses are intertwined, and responsivity to any immunosuppressive agent is highly dependent on which pathway in the immune system is affected.22 For the purposes of this discussion, both suppression of innate and adaptive immune response will be considered, and particular attention will be paid to suppression of the inflammatory reaction by nanoparticles (i.e., anti-inflammatory activity).
The explosion of research into engineered nanomaterials and nanomedicine has resulted in the discovery of unique immunological responses elicited by nanomaterials. A pre-eminent goal of nanomedicine had always been to engineer nanomaterials that target specific tissue and deliver drugs, while simultaneously avoiding undesirable immunostimulatory or immunosuppressive reactions.23–25 However, the inherent immunological response to nanomaterials may be also exploited for novel therapies. Immunological response can be modulated and suppressed through multiple pathways by the engineering the physiochemical properties of the nanomaterials. Between 2007 and 2010, Dobrovolskaia et al. produced comprehensive summaries of the basic structure–activity relationships of engineered nanoparticles and the immune system.26–28 These reviews concluded that the immunological response to engineered nanomaterials can be attributed to the milieu of physiochemical properties, such as particle size, shape, charge, and surface chemistry.23,29,30 For example, simple differences in nanoparticle size can alter cellular internalization, such that larger nanoparticles (>100 nm) are internalized and transported by macrophages and dendritic cells of the innate immune system, while smaller nanoparticles can easily travel to and accumulate in lymph nodes and effect B cells and T cells.31,32 Modifying the surface of nanomaterials also alters the nanoparticle/biofluid interface which influences the way nanoparticles interact with and are labeled by plasma proteins. This effect is critical in determining the nanoparticles’ interaction with cells, in vivo organ distribution and clearance pathways.33–36
While the fundamental structure–activity responses are still being investigated there is concurrent research into exploiting the immunological response to nanomaterials for novel direct immunostimulatory to immunosuppressive therapies.30,37 Many researchers are investigating novel immunostimulants, such as nanoparticle-based or nanoparticle-supported vaccines.28,38,39 Alternatively, the immunosuppressive capabilities of nanoparticles are garnering much attention. These research efforts involve understanding the direct interactions and effect of nanomaterials to suppress immunological signaling pathways. Direct immunosuppression can be applicable in overcoming autoimmune disease, reducing allergic reactions, and anti-rejection treatment. In the past year, Dobrovolskaia et al. also highlighted the burgeoning exploration of immunosuppressive and anti-inflammatory properties of nanomaterials.37,40 Herein, we review recent reports on nanomaterials and methods to induce an immunosuppressive reaction through direct nanoparticle interactions with immune cells. We take in to account different nanoparticle chemistries and note the differences of immunosuppressive interactions with both innate immune cells (macrophages, dendritic cells, neutrophils, mast cells, and natural killer cell) and adaptive immune cells (T cells and B cells). We also discuss the effect of nanoparticles on the common immune reaction of inflammation.
Immunosuppressive nanomaterials
The size, shape, and chemistry of nanoparticles facilitate the binding of blood, cellular, and protein components which facilitate interactions with the immune cells and result in the immunological response. In general, the physiochemical properties of nanoparticles are important factors that significantly influence the interaction of nanoparticles and cells. Gold nanoparticles are particularly exemplary systems to illustrate these effects.41,42 Spherical gold nanoparticles between 5 and 30 nm in diameter are capable of interacting with cells by passive means; however, larger nanoparticles and rod-like nanoparticles are more commonly internalized via complex uptake processes.43–45 The surface-coating of the gold nanoparticles also effect cellular uptake. Where small-molecule organic ligands like citrate or lipids may promote stability and passive cellular uptake, macromolecular coatings like poly(ethylene glycol) may result in protein adsorption and reduction in cellular uptake.44,46 Accordingly, the wide variation in size, shape, and surface coating of nanoparticles limit broad generalizations about the interactions of nanoparticles with the immune system. In the following discussion, we classify the nanoparticles into four groups based on chemical constituency: 1. metal nanoparticles, 2. metal-oxide nanoparticles, 3. carbon nanomaterials, and 4. polymer nanoparticles and macromolecules, and we described how they modulate and immunosuppressive response related to their physicochemical properties. Figure 1 illustrates the representative pathways nanoparticles may directly interact with the immune system and instigate an immunosuppressive response.
Figure 1.
Direct interactions of nanoparticles with the immune system. Dependent on physiochemical properties, the nanoparticles interact with the constituents of the immune response, including macrophages, antigen presenting cells, B cells or T cells. Direct effects of carbon nanomaterials include the upregulation of transforming growth factor-β (TGFβ), interleukin-10 (IL-10), decreased B cell activity, as well as apoptosis. Metal-oxide nanoparticles can directly affect adaptive immune cells, and materials like cerium oxide nanoparticles scavenge reactive oxygen species (ROS) and operate as anti-inflammatory agents. Polymer nanoparticles and macromolecules (dendrimers) exhibit an array of immunosuppressive effects. The pathways shown are representative examples by which different nanoscale products might suppress the immune system. Immunosuppressive effects are modified from Smith et al.30 and Jiao et al.37 (A color version of this figure is available in the online journal.)
Metal nanoparticles
Metal and metal-oxide nanoparticles make up a significant share of engineered nanomaterials that are produced.47 Accordingly, most efforts in to understanding the immunological response to nanomaterials have been related to occupational and environmental exposure to metal and metal-oxide nanoparticles.
Noble metal nanoparticles, such as gold and silver, interact with both the innate and adaptive immune systems,48–51 but there are few reports that uncover the mechanism behind noble metal nanoparticles’ ability to elicit an immunosuppressive response. Injection of organo-gold compounds has been utilized for nearly a century to treat inflammation, and only recent reports provide the first analysis of the biochemical pathway in which gold nanoparticles may reduce inflammation.52,53 Citrate-coated gold nanoparticles were reported to not cause detectable cell or organ toxicity in mice, but the report concluded that citrate-coated gold nanoparticles showed anti-inflammatory activity and inhibited cellular responses induced by interleukin 1 beta (IL-1β). IL-1β is an inflammatory cytokine that acts as an arbiter between the innate and adaptive immune response; moreover, common inflammatory disorders, for example, rheumatoid arthritis, are mediated by IL-1β production. Monodispersed citrate-coated gold nanoparticles of different sizes between 5 and 35 nm were evaluated for their ability to modulate the pro-inflammatory function mediated by IL-1β production. The smallest nanoparticles, 5 nm, exhibited complete disruption of the IL-1β pathway. Larger nanoparticles, >10 nm, exhibited lesser effect, and 35 nm particles showed no effect on the IL-1β pathway.45,54 The biochemical mechanism of the down-regulation of IL-1β induced inflammation pathways is assigned to extracellular interaction of citrate-coated gold nanoparticles with IL-1β. These results support prior research concluding that gold nanoparticles smaller than 10 nm exhibit unique interactions with cells.55 Gold nanoparticles with diameter of approximately 200 nm and coated with poly(acrylic acid) showed the opposite response, as they promoted inflammation in the same THP-1 cell line.56 These reports suggest the surface coating of the gold nanoparticles determine the cellular and immune system interaction that results from exposure to gold nanoparticles. The surface charge of stabilizing ligands also determine the immunological fate of the gold nanoparticles. Ultra-small zwitterion-stabilized gold nanoparticles (< 3 nm) were reported to directly instigate an immunosuppressive response without any measurable cytotoxicity.57 Alternatively, other polymer and organic coatings did not produce comparable anti-inflammatory responses.58–60 This suggests the size and coating either perform in unison or direct different pathways that induce the immunosuppressive response. Decoupling of these mechanisms would provide a clear direction to engineer better gold-based nanomedicines with the ability to regulate the IL-1β pathways for the treatment of chronic rheumatic disease.
Unlike gold nanoparticles, immunosuppression caused by silver nanoparticles has not been as widely reported. Silver nanoparticles have been reported to stimulate the production of cytokines, including TNF-α, IL-1, IL-6, IL-8, and IL-11.61–63 Cytokines play an important role in wound inflammation, and Tian et al. reported the modulation of cytokines at a wound site by topical application of silver nanoparticles. Expression levels of IL-6 mRNA were significantly lower during the healing process while TGF-β1 levels were higher.64 The reduction of inflammation with topical application65 and systemic application66 has also been reported. It is important to note, topical application of silver nanoparticles exhibited reduction of inflammatory cytokines, but also showed extensive apoptosis of inflammatory cells.65 Continued exploration of the interaction of silver nanoparticles with the immune system will benefit our understanding in designing for the application of silver nanoparticles in fields such as medical devices, antimicrobial systems, and drug delivery.
Metal-oxide nanoparticles
Metal-oxide nanoparticles have been reported to induce both immunosuppressive and anti-inflammatory responses. Iron oxide nanoparticles have been shown to reduce the humoral immune response. The humoral response involves the recognition of antigens, allergens, pathogens, or foreign bodies in the blood with B cells. In comparative reports by Liao et al. and Shen et al., ovalbumin-sensitized mice were administered ovalbumin, a T cell-dependent antigen, following a dose of iron oxide nanoparticles. The production of antibodies specific to the antigen was significantly reduced along with spleen production of antigen-specific cytokines. Specifically, the iron oxide nanoparticle treatment blocked the activity of Thelper cells and macrophages and reduced the expression of interferon-γ, interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α).67,68 Both IL-6 and TNF-α are cytokines involved in systemic inflammation. The iron oxide nanoparticles also shifted the Thelper cell balance and suppressed the allergic reaction.68 In an alternate mouse model, treatment with iron oxide particles prior to immune stimulation with an endotoxin reduced the production of IL-1β in microglia cells. The iron oxide nanoparticles inhibited pathways involved in cytokine processing which mediated the attenuation of IL-1β.69 In a corollary report, human dendritic cells exhibited decreased antigen processing and decreased T cell stimulation when treated with poly(vinyl alcohol)-coated iron oxide nanoparticles. Additional injections of iron oxide nanoparticles resulted in a reduction of inflammation.70 Iron-oxide nanoparticles have been reported to induce immunosuppression via multiple mechanism, as previously described; however, the review literature does not thoroughly differentiate the mechanism of action in which the iron-oxide nanoparticles interrupt or modulate the immune response. Jan et al. do report that the iron-oxide nanoparticles accumulated in lysosomes, which increased lysosomal permeability and decreased the activity of cathepsin B.69 Cathepsin B is a secretory lysosomal enzyme involved in the activity of IL-1β, and its dysfunction may lead to the exhibited disruption of IL-1β production. A concerted effort to similarly explore and elucidate the fundamental interaction of iron-oxide nanoparticles with the biochemistry of immune cells or immune pathways would be beneficial to the current state-of-the art.
Metal-oxide nanoparticles also support an alternative anti-inflammatory response. Free-radical formation in the body has been attributed to inflammation, tissue damage, and development of diseases. Cerium can stably switch oxidation states between Ce4+ and Ce3+, which enables cerium oxide nanoparticles to readily absorb free radicals. The oxidation and reduction of cerium oxide produces oxygen vacancies in the crystal lattice as it switches oxidation states. In nanoparticles, these vacancies are concentrated at the surface; therefore, cerium oxide nanoparticles have a unique capability to quickly react with free radicals. These antioxidant capabilities have been repeatedly demonstrated,71 although the mechanism of their antioxidant capabilities and free radical scavenging in biological systems has not been isolated. Cerium oxide particles have been reported to mimic the superoxide dismutase active scavenging of superoxide (O2−).72 Many other free radicals are in biological systems that can be implicated in the antioxidant and anti-inflammatory properties of cerium oxide nanoparticles. Of particular interest, cerium oxide nanoparticles scavenge highly reactive hydroxyl radicals.73 In any case, the ability to scavenge reactive oxygen species can significantly reduce inflammation antagonized by reactive oxygen species.74 Anti-oxidant properties of cerium oxide nanoparticles allow it to also inhibit inflammation generated from nitric oxide production by nitric oxide synthase.75 Cerium oxide nanoparticles also caused the upregulation of superoxide dismutase-2, a mediator of oxidative stress.76 Similar to many nanoparticles, antioxidant properties of cerium oxide nanoparticles depend on size and surface properties. As such, smaller cerium oxide nanoparticles possessed higher antioxidant capacity than their larger counterparts.77
Unlike cerium oxide nanoparticles, the single defining characteristics of the immunological response to zinc oxide nanoparticles is cytotoxicity related to oxidative stress from the increased production of reactive oxygen species.78–80 Kim et al. explored the effect of surface charge and size on the immunological response to zinc oxide nanoparticles, and they concluded positively charged zinc oxide nanoparticles exerted higher cytotoxicity than the negatively charged zinc oxide nanoparticles. Further evaluation showed natural killer cell activity was suppressed; the CD4+/CD8+ T cell ratio (a measure of the ratio of Thelper to cytotoxic T cells) was slightly reduced, nitric oxide production from splenocyte culture was lower; and IL-1β, TNF-α, IL-10, and interferon-γ in zinc oxide were significantly suppressed. Each are cytokines related to pro-inflammatory response. Collectively, these responses suggest the charged surface of zinc nanoparticles elicit both dramatic and systemic immunosuppression, which correlated to the observed cytotoxicity in the same cells. The effect of nanoparticle size on immune cell cytotoxicity was evaluated, and similar to prior efforts,81–83 it was reported that smaller particles exhibited a slightly higher EC50 than smaller zinc oxide nanoparticles The size of the nanoparticle also correlated to cytokine concentration in the serum; the serum levels of pro-inflammatory cytokines were reduced with increased particle size.84
Another common metal-oxide nanoparticle, titanium oxide, are constituents of many cosmetic and commercial colorants, included in products such as paints and plastics. Unlike zinc oxide nanoparticles, titanium oxide nanoparticles have been reported as non-cytotoxic in vitro,85,86 but they can induce an immunosuppressive response. In a tumor model, titanium oxide nanoparticles inhibited T-cells, B-cells, macrophages, and natural killer cells.87
Carbon nanomaterials
Carbon nanomaterials exhibit strong free-radical scavenger properties and can be utilized as anti-inflammatory agents.88–90 Similar to cerium oxide nanoparticles, fullerenes (C60) decrease the level of reactive oxygen species by efficiently scavenging free-radicals. Unlike cerium oxide nanoparticles, fullerenes distribute the free-radicals through their aromatic structure.91 Fullerenes reduce reactive oxygen species of both hydroxyl and superoxide radicals, and many of the radical scavenging properties are moderated by the functionalization of fullerenes with water-soluble ligands.71,92 Fullerenes have been shown to suppress oxidative stress in vitro and in vivo models, but immunological response is highly dependent on dosing and delivery.93 Both intraperitoneal injection and inhalation delivery of low doses of fullerenes exhibited reduction in oxidative stress. Hydroxylated fullerenes were used to protect against oxidative stress in RAW 264.6 cells in vitro and ischemia-perfused lungs in rats.94 Reduction in signaling pathways associated with reactive oxygen species levels also enable fullerenes to inhibit human mast cell and peripheral blood basophil release of meditators. Mast cells and peripheral blood basophils are cells involved in the initiation of inflammatory response and Type I hypersensitivity allergies. Type I hypersensitivity allergies are the result of repeated exposure to an allergen. After initial exposure, B cells produce the allergen-specific Immunoglobulin E (IgE) which then binds the receptors and sensitizes mast cells and peripheral blood basophils. Subsequent exposure to the allergen causes crosslinking of the IgE and activates mediators such as histamine and prostaglandins. After treatment with fullerenes, the mast cells and peripheral blood basophils decreased this IgE signaling and decreased ROS production, which prevented histamine release.89 Many allergy medications are aimed at a similar response for neutralizing the IgE response. Studies examining the toxicity of fullerenes on biological systems are still ongoing, and an in vivo evaluation of the immunological response to fullerenes may still pave the way for other treatments of autoimmune disorders.
Other allotropes of carbon nanomaterials exhibit a less clear mechanism for immunosuppressive action. Mitchell et al. have reported the inhalation of multi-walled carbon nanotubes at low concentrations in mouse models lead to stimulation of pathways responsible for T-cell dysfunction and suppressed immune response from spleen cells; however, they report no cytotoxicity or immunosuppression in the lungs. Exposure to the multi-walled carbon nanotubes also resulted in decreased natural killer cell function, increased production of prostaglandin and increased IL-10.95,96 The signaling and transference of immunological response from the exposed lungs to the spleen is very interesting. For comparison, single-walled carbon nanotubes were reported to suppress immune system mediators in human lung epithelial cells.97 Other studies showed that inhalation of single-wall carbon nanotubes in mouse models induced pulmonary inflammation and suppressed the responsiveness of T cells after exposure; this immunosuppression was associated with the direct effects of single-wall carbon nanotubes on dendritic cells.98
The reported differences in immunological response between fullerenes, multi-wall and single-wall carbon nanotubes may be attributed to their geometries as well as their different electrochemical properties. Future efforts to decouple these effects may lead to a better understanding of surface charge and inherent conductivity on immunological response.
Polymer nanoparticles, macromolecules, and liposomes
A popular alternative to inorganic and carbon nanomaterials are polymer nanoparticles and macromolecules. A majority of polymer nanoparticles and macromolecules have been utilized as carriers of drugs for indirect immunosuppression. Polymer and other macromolecular nanomaterials are also very popular for induction of immunostimulatory response. There have been a few nanotoxicology studies to explore the immunological response to simple polymer nanomaterials. One particular class of polymer nanoparticles to be examined for immunological response is polystyrene latexes. Polystyrene latexes are very common industrial polymers that provide a simple model to study the bio-activity of nanoparticles dependence on size and surface chemistry. Unfortunately, the tunability of polystyrene latexes are their only simple characteristic. Polystyrene latexes have been reported to induce both immunostimulatory and immunosuppressive responses. During an allergen challenge, polystyrene nanoparticles inhibited lung inflammation and this reaction was attributed to the inhibited expansion of dendritic cells in the lungs.99 Another study with polystyrene nanoparticles emphasized the importance of surface charge on immunological response. Frick et al. altered the surface charge of polystyrene nanoparticles by decorating them with charged sulfonate and phosphonate groups. This resulted in immunostimulation by dendritic cell maturation and enhanced CD4+ T cell activity.100 Finally, antigen-decorated polystyrene nanoparticles also induced T-cell tolerance and suppressed autoimmune encephalomyelitis by inactivating pathogenic T cells.101 The significant impact of surface groups on the immunological response to simple polystyrene nanoparticles illustrates the necessity for continued research into the immunology of polymer nanoparticles that are so often praised and developed for drug deliver and other nanomedicine applications.
Polymer macromolecules, specifically dendrimers, have been explored as potential anti-inflammatory agents.102 Anti-inflammatory properties of polyamidoamine dendrimers were unexpectedly discovered while investigating their application for a drug delivery system. Polyamidoamine dendrimers possessing amine or hydroxyl surface groups substantially reduced pro-inflammatory responses. Dendrimers presenting carboxylate surface groups did not show any enhanced anti-inflammatory properties.103 Hydroxyl-terminus polyamidoamine dendrimers also blocked the release of pro-inflammatory regulators nitric oxide and IL-6 in microglia cells.104 Hayder et al. also modified the surface charge of dendrimers with anionic azabisphosphonate group. Arthritic mouse models were treated with these anionic dendrimers and they inhibited the secretion of pro-inflammatory cytokines and stunted the osteoclastogenesis process.105
Dendrimers also enable a unique hybrid of direct and indirect immunosuppression. As previously mentioned, indirect immunosuppression involves the delivery of an immunosuppressant, for example, gluco-steroid or cyclosporine A, with a nanoparticle as the delivery vehicle. Dendrimers have the unique capability to be both the delivery vehicle and payload, since they can be composed of repeating small-molecule or incorporate immunosuppressive molecules within their macrostructure. For example, carboxylic acid terminated polyamidoamine dendrimers did not significantly reduce inflammatory response; however, when these dendrimers were conjugated with glucosamine, dendritic cell, and macrophage activity was inhibited.106,107 The polyamidoamine-glucosamine dendrimers inhibit toll-like receptor (TLR) mediated inflammatory responses, and they have been shown to decrease IL-6 and IL-8 production.108 TLR mediate the innate immune system response, and they are expressed in macrophages and dendritic cells. Analogous to the polyamidoamine-glucosamine dendrimers are imidazoquinoline-based dendrimers, which are dendrimers with the base constituent being imidazoquinoline instead of glucosamine. Imidazoquinoline is an agonist for TLR7 and TLR8; thus, the imidazoquinoline-based dendrimers suppress both TLR7 and TLR8 activity.109 Although the mechanisms by which particular pathways are suppressed are not fully understood, it is clear that the surface charge and increased chemical density of the dendrimers play a very impactful role. This highlights how simple surface charge and surface chemistry manipulation at the nanoscale level can be used to engineer nanoparticles that finely tune the innate immunological response.
Liposomes are another class engineered nanoparticles; liposomes are lipid bi-layer spheres. Liposomes can be used to enhance localized delivery of encapsulated immunosuppressive agents. Hong et al. evaluated the efficacy of delivering an IL-10 gene within cationic liposomes to increase allograft survival following a heart transplant. Delivery using liposomes resulted in local overexpression of the IL-10 and a reduction in lymphocyte responsiveness.110 Similarly, canines were administered liposomal tacrolimus, an immunosuppressive drug, following a liver transplant and survived significantly longer than canines who were given tacrolimus intravenously.111 Glucocorticoids encapsulated within liposomes provided for dose reduction when applied to rheumatoid arthritis in rats, resulting in reduced toxicity and increased suppression of anti-inflammatory cytokines when compared to administration of free drug.112 As this illustrates, liposomes have been utilized as a carrier for multiple immunosuppressive therapies; however, a more probing evaluation of the direct interaction of “cargo-free” liposomes with the immune system after systemic or environmental exposure will be of particular interest in the future.
Similar to liposomes, peptide and peptide-based amphiphiles are macromolecules which self-assemble into either nanoparticles or nanofibers in situ, and current research efforts are focused on utilizing these nanomaterials for delivery of immunotherapeutics. As described by Tirrell et al., a majority of peptide amphiphile delivery systems are developed to induce immunostimulatory effects in the innate and adaptive immune system.113–115 Although these reports describe the capability of peptide amphiphile micelles to cause immunostimulation, based on their direct interaction with the immune system, there may be future potential to use similar nanomaterials generate immunosuppressive responses.
As demonstrated by the myriad macromolecular nanoparticles discussed heretofore, there is significant potential to decorate and functionalize macromolecular nanoparticles to modulate the immune response. Similar to metal and metal-oxide nanoparticles, the surface chemistry of macromolecular nanoparticles direct cellular targeting, uptake, and bioactivity. Accordingly, researchers are exploring nanoparticle chemistries and coatings to modulate immune responses.
Complications with nanoparticle immunosuppresion
In the best circumstances, immunosuppressive therapies would be targeted to hyperactive components of the immune system in the case of an autoimmune disorder, or the immunosuppressive therapy could be acutely active to coincide with pharmaceutical treatment and then immediately dissipate. In both scenarios, the immunosuppressive therapy and would not result in any immunodeficiency. Nonetheless, current immunosuppressive therapies have the potential to cause immunodeficiency which results in increased susceptibility to opportunistic pathogens, degradation of bone marrow (myelosuppression), and increased cytotoxicity and genotoxicity to the active nanomaterial.
Since small changes in nanoparticle size, shape, charge, and constituency result in magnified immunological response, it will be necessary to continue expansive research into the toxicology and immunology of nanoparticles. In 2009, Bregoli et al. reported a toxicity analysis of different metal and metal-oxide nanoparticles that highlights the dramatic variability of immunological response to nanoparticles, and frames the current viability of nanoparticles as immunomodulators. Variants of iron oxide, antimony oxide, gold, titanium oxide, and cobalt and silver nanoparticles were introduced to hematopoietic progenitor cells. Hematopoietic progenitor cells are bone marrow derived cells that give rise to myeloid and lymphoid lineages of immune cells. Only antimony oxide and cobalt nanoparticles exhibited a toxic effect. Antimony oxide nanoparticles were specifically toxic to erythroid progenitors. Cobalt nanoparticles were toxic to both erythroid and granulocytic-monocytic progenitors. On the other hand, in a complimentary assay in which antimony oxide nanoparticles were tested against immortalized cell lines of hematopoietic origins, there was no displayed toxicity.116 Different types of immune cells and their lineage may exhibit different sensitivities to the same nanoparticle type. Analysis of the immunological response of zinc oxide nanoparticles has resulted in similarly complex results; reports have described immunosuppression, immunostimulation, and general cytotoxicity of zinc oxide nanoparticles.84,117 Zinc oxide nanoparticles induced high levels of toxicity and increased ROS production in monocytes, while lymphocytes remained relatively resistant to the toxic effects.80 These contradictory results are unfortunately the hallmark of nanotoxicology research and it raises questions on the utility and methods of analyzing immunological response to nanoparticles with in vitro cell cultures.
As the factors effecting immunological response to nanoparticles are further investigated at the fundamental level, it should be noted that in application, these factors may exhibit a compounding effect. For example, if the nanoparticle delivery vehicle for an immunosuppressant or chemotherapeutic creates its own immunosuppressive response, the drug’s activity may be exponentially compounded. It has been reported that doxorubicin bound to polymer nanoparticles both stimulated greater myelo-suppresive affects than doxorubicin alone, and the nanoparticles released stimulating factors they were attributed to an increased toxicity of doxorubicin.118 This can cause potential problems with nanoparticles due to their ability to deliver precise and localized drug dosing.119,120 It should be considered that the nanoparticle formulation of a drug may not only increase the efficacy of the drug by precise delivery and increased circulation time, but there may be a synergistic immunological response caused by the nanoparticle.
Outlook
Heretofore, we have discussed direct immunosuppression induced by nanoparticles. There is a great potential to expand our understanding of immunosuppression by nanoparticles, and further studies need to be conducted to completely evaluate fundamental mechanisms in more relevant models. These efforts will assist in understanding why some nanoparticle complexes are immuno-stimulatory in vivo while immunosuppressive in vitro or other combinations thereof.
Ultimately, associations of pathway regulation with nanomaterials size, structure, and method of introduction to tissue needs to be comprehensively evaluated in order to introduce these nanomaterial applications clinically.9,121,122 While immunological reaction to nanoparticles are being intensely investigated, their direct immunosuppressive and anti-inflammatory properties may hold the necessary keys to the next generation drug delivery vehicles and complimentary treatments for regenerative medicine and transplantation. The cooperative research among materials scientists, immunologists, and toxicologists is a very exciting field expected to progress alongside the development of new nanomedicines.
Acknowledgments
All authors participated in the research and writing of the manuscript. The authors are funded by North Carolina State University. Funders had no role in the preparation of the review or the decision to submit for publication.
Declaration of conflicting interests
The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
References
- 1.Hartono C, Muthukumar T, Suthanthiran M. Immunosuppressive drug therapy. Cold Spring Harb Perspect Med 2013; 3: a015487–a015487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Gummer JF, Ikonen T, Morris RE. New immunosuppressive drugs: a review. J Am Soc Nephrol 1999; 10: 15–15. [DOI] [PubMed] [Google Scholar]
- 3.van Gelder T, van Schaik RH, Hesselink DA. Pharmacogenetics and immunosuppressive drugs in solid organ transplantation. Nat Rev Nephrol 2014; 10: 725–731. [DOI] [PubMed] [Google Scholar]
- 4.Gharagozloo M, Majewski S, Foldvari M. Therapeutic applications of nanomedicine in autoimmune diseases: from immunosuppression to tolerance induction. Nanomed Nanotechnol Biol Med 2015; 11: 1003–1018. [DOI] [PubMed] [Google Scholar]
- 5.Guada M, Sebastian V, Irusta S, Feijoo E, Dios-Vieitez MD, Blanco-Prieto MJ. Lipid nanoparticles for cyclosporine A administration: development, characterization, and in vitro evaluation of their immunosuppression activity. Int J Nanomed 2015; 10: 6541–6553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Nadig SN, Dixit SK, Levey N, Esckilsen S, Miller K, Dennis W, Atkinson C, Broome AM. Immunosuppressive nano-therapeutic micelles downregulate endothelial cell inflammation and immunogenicity. Rsc Adv 2015; 5: 43552–43562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Ilinskaya AN, Dobrovolskaia MA. Immunosuppressive and anti-inflammatory properties of engineered nanomaterials. Br J Pharmacol 2014; 171: 3988–4000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Xu W, Ling PX, Zhang TM. Toward immunosuppressive effects on liver transplantation in rat model: tacrolimus loaded poly(ethylene glycol)-poly(D,L-lactide) nanoparticle with longer survival time. Int J Pharm 2014; 460: 173–180. [DOI] [PubMed] [Google Scholar]
- 9.Broome AM, Dixit S, Levey N, Atkinson C, Nadig S. Towards targeted drug delivery in transplantation: use of immunosuppressant nanoparticle therapy. Am J Transplant 2014; 14: 63–64. [Google Scholar]
- 10.Chen WC, May JP, Li SD. Immune responses of therapeutic lipid nanoparticles. Nanotechnol Rev 2013; 2: 201–213. [Google Scholar]
- 11.Maldonado RA, LaMothe RA, Ferrari JD, Zhang AH, Rossi RJ, Kolte PN, Griset AP, O’Neil C, Altreuter DH, Browning E, Johnston L, Farokhzad OC, Langer R, Scott DW, von Andrian UH, Kishimoto TK. Polymeric synthetic nanoparticles for the induction of antigen-specific immunological tolerance. Proc Natl Acad Sci USA 2015; 112: E156–E165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Zhang PP, Chiu YC, Tostanoski LH, Jewell CM. Polyelectrolyte multilayers assembled entirely from immune signals on gold nanoparticle templates promote antigen-specific T cell response. ACS Nano 2015; 9: 6465–6477. [DOI] [PubMed] [Google Scholar]
- 13.Wang J, Zhu RR, Sun DM, Sun XY, Geng ZS, Liu H, Wang SL. Intracellular uptake of curcumin-loaded solid lipid nanoparticles exhibit anti-inflammatory activities superior to those of curcumin through the NF-kappa B signaling pathway. J Biomed Nanotechnol 2015; 11: 403–415. [DOI] [PubMed] [Google Scholar]
- 14.Wang J, Wang HY, Zhu RR, Liu Q, Fei J, Wang SL. Anti-inflammatory activity of curcumin-loaded solid lipid nanoparticles in IL-1 beta transgenic mice subjected to the, lipopolysaccharide-induced sepsis. Biomaterials 2015; 53: 475–483. [DOI] [PubMed] [Google Scholar]
- 15.Silva CO, Rijo P, Molpeceres J, Figueiredo IV, Ascensao L, Fernandes AS, Roberto A, Reis CP. Polymeric nanoparticles modified with fatty acids encapsulating betamethasone for anti-inflammatory treatment. Int J Pharm 2015; 493: 271–284. [DOI] [PubMed] [Google Scholar]
- 16.Elder A. How do nanotubes suppress T cells? Nat Nanotechnol 2009; 4: 409–410. [DOI] [PubMed] [Google Scholar]
- 17.Shvedova AA, Kagan VE. The role of nanotoxicology in realizing the ‘helping without harm’ paradigm of nanomedicine: lessons from studies of pulmonary effects of single-walled carbon nanotubes. J Intern Med 2010; 267: 106–118. [DOI] [PubMed] [Google Scholar]
- 18.Gernand JM, Casman EA. A meta-analysis of carbon nanotube pulmonary toxicity studies—how physical dimensions and impurities affect the toxicity of carbon nanotubes. Risk Anal 2014; 34: 583–597. [DOI] [PubMed] [Google Scholar]
- 19.Handy RD, Shaw BJ. Toxic effects of nanoparticles and nanomaterials: implications for public health, risk assessment and the public perception of nanotechnology. Health Risk Soc 2007; 9: 125–144. [Google Scholar]
- 20.Zakrzewski JL, van den Brink MRM, Hubbell JA. Overcoming immunological barriers in regenerative medicine. Nat Biotechnol 2014; 32: 786–794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Clemente-Casares X, Santamaria P. Nanomedicine in autoimmunity. Immunol Lett 2014; 158: 167–174. [DOI] [PubMed] [Google Scholar]
- 22.Janeway CAJ, Travers P, Walport M. Principals of inate and adaptive immunity. In: Immunobiology: the immune system in health and disease, New York, NY: Garland Science, 2001. [Google Scholar]
- 23.Aggarwal P, Hall JB, McLeland CB, Dobrovolskaia MA, McNeil SE. Nanoparticle interaction with plasma proteins as it relates to particle biodistribution, biocompatibility and therapeutic efficacy. Adv Drug Deliv Rev 2009; 61: 428–437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Luk BT, Zhang L. Cell membrane-camouflaged nanoparticles for drug delivery. J Control Release 2015; 220(Pt B): 600–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Caracciolo G, Palchetti S, Colapicchioni V, Digiacomo L, Pozzi D, Capriotti AL, La Barbera G, Lagana A. Stealth effect of biomolecular corona on nanoparticle uptake by immune cells. Langmuir 2015; 31: 10764–10773. [DOI] [PubMed] [Google Scholar]
- 26.Dobrovolskaia MA, Aggarwal P, Hall JB, McNeil SE. Preclinical studies to understand nanoparticle interaction with the immune system and its potential effects on nanoparticle biodistribution. Mol Pharm 2008; 5: 487–495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Dobrovolskaia MA, Mcneil SE. Immunological properties of engineered nanomaterials. Nat Nanotechnol 2007; 2: 469–478. [DOI] [PubMed] [Google Scholar]
- 28.Zolnik BS, Gonzalez-Fernandez A, Sadrieh N, Dobrovolskaia MA. Minireview: nanoparticles and the immune system. Endocrinology 2010; 151: 458–465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Moyano DF, Goldsmith M, Solfiell DJ, Landesman-Milo DJ, Miranda OR, Peer D, Rotello VM. Nanoparticle hydrophobicity dictates immune response. J Am Chem Soc 2012; 134: 3965–3967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Smith DM, Simon JK, Baker JR. Applications of nanotechnology for immunology. Nat Rev Immunol 2013; 13: 592–605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Shang L, Nienhaus K, Nienhaus GU. Engineered nanoparticles interacting with cells: size matters. J Nanobiotechnol 2014; 12: b67–b67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Manolova V, Flace A, Bauer M, Schwarz K, Saudan P, Bachmann MF. Nanoparticles target distinct dendritic cell populations according to their size. Eur J Immunol 2008; 38: 1404–1413. [DOI] [PubMed] [Google Scholar]
- 33.Tenzer S, Docter D, Kuharev J, Musyanovych A, Fetz V, Hecht R, Schlenk F, Fischer D, Kiouptsi K, Reinhardt C, Landfester K, Schild H, Maskos M, Knauer SK, Stauber RH. Rapid formation of plasma protein corona critically affects nanoparticle pathophysiology. Nat Nanotechnol 2013; 8: 772–781. [DOI] [PubMed] [Google Scholar]
- 34.Lundqvist M, Stigler J, Elia G, Lynch I, Cedervall T, Dawson KA. Nanoparticle size and surface properties determine the protein corona with possible implications for biological impacts. Proc Natl Acad Sci U S A 2008; 105: 14265–14270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Mirshafiee V, Kim R, Park S, Mahmoudi M, Kraft ML. Impact of protein pre-coating on the protein corona composition and nanoparticle cellular uptake. Biomaterials 2016; 75: 295–304. [DOI] [PubMed] [Google Scholar]
- 36.Gunawan C, Lim M, Marquis CP, Amal R. Nanoparticle-protein corona complexes govern the biological fates and functions of nanoparticles. J Mater Chem B 2014; 2: 2060–2083. [DOI] [PubMed] [Google Scholar]
- 37.Jiao Q, Li LW, Mu QX, Zhang Q. Immunomodulation of nanoparticles in nanomedicine applications. Biomed Res Inter 2014; 2014: 1–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Foerster J, Bachman M. Beyond passive immunization: toward a nanoparticle-based IL-17 vaccine as first in class of future immune treatments. Nanomedicine 2015; 10: 1361–1369. [DOI] [PubMed] [Google Scholar]
- 39.Talekar M, Tran TH, Amiji M. Translational nano-medicines: targeted therapeutic delivery for cancer and inflammatory diseases. AAPS J 2015; 17: 813–827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Ilinskaya A, Dobrovolskaia MA. Immunosuppressive and anti-inflammatory properties of engineered nanomaterials. Br J Pharmacol 2014; 171: 13–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Dykman L, Khlebtsov N. Gold nanoparticles in biomedical applications: recent advances and perspectives. Chem Soc Rev 2012; 41: 2256–2282. [DOI] [PubMed] [Google Scholar]
- 42.Zhang XY. Gold nanoparticles: recent advances in the biomedical applications. Cell Biochem Biophys 2015; 72: 771–775. [DOI] [PubMed] [Google Scholar]
- 43.Pissuwan D, Valenzuela SM, Killingsworth MC, Xu XD, Cortie MB. Targeted destruction of murine macrophage cells with bioconjugated gold nanorods. J Nanopart Res 2007; 9: 1109–1124. [Google Scholar]
- 44.Chithrani BD, Ghazani AA, Chan WCW. Determining the size and shape dependence of gold nanoparticle uptake into mammalian cells. Nano Lett 2006; 6: 662–668. [DOI] [PubMed] [Google Scholar]
- 45.Chen H, Dorrigan A, Saad S, Hare DJ, Cortie MB, Valenzuela SM. In vivo study of spherical gold nanoparticles: inflammatory effects and distribution in mice. PLoS One 2013; 8: e58208–e58208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Lin IC, Liang MT, Liu TY, Monteiro MJ, Toth I. Cellular transport pathways of polymer coated gold nanoparticles. Nanomed Nanotechnol Biol Med 2012; 8: 8–11. [DOI] [PubMed] [Google Scholar]
- 47.Petrarca C, Clemente E, Amato V, Pedata P, Sabbioni E, Bernardini G, Iavicoli I, Cortese S, Niu Q, Otsuki T, Paganelli R, Di Gioacchino M. Engineered metal based nanoparticles and innate immunity. Clin Mol Allergy 2015; 13: 13–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Sur A, Pradhan B, Banerjee A, Aich P. Immune activation efficacy of indolicidin is enhanced upon conjugation with carbon nanotubes and gold nanoparticles. PLoS One 2015; 10: e0123905–e0123905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Fytianos K, Rordriguez-Lorenzo L, Blank F, von Garnier C, Petri-Fink A, Rothen-Rutishauser B. Surface modified gold nanoparticles to specifically target lung immune cells in vitro. J Aerosol Med Pulm Drug Deliv 2015; 28: A35–A35. [Google Scholar]
- 50.Kendall E, Liptrott N, Owen A. Investigating the interaction of gold nanoparticles with the human immune system. Scott Med J 2014; 59: E34–E34. [Google Scholar]
- 51.Garcia CP, Sumbayev V, Gilliland D, Yasinska IM, Gibbs BF, Mehn D, Calzolai L, Rossi F. Microscopic analysis of the interaction of gold nanoparticles with cells of the innate immune system. Sci Rep 2013; 3: 1–7. [Google Scholar]
- 52.Thakor AS, Jokerst J, Zavaleta C, Massoud TF, Gambhir SS. Gold Nanoparticles: a revival in precious metal administration to patients. Nano Lett 2011; 11: 4029–4036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.James LRA, Xu ZQ, Sluyter R, Hawksworth EL, Kelso C, Lai B, Paterson DJ, de Jonge MD, Dixon NE, Beck JL, Ralph SF, Dillon CT. An investigation into the interactions of gold nanoparticles and anti-arthritic drugs with macrophages, and their reactivity towards thioredoxin reductase. J Inorg Biochem 2015; 142: 28–38. [DOI] [PubMed] [Google Scholar]
- 54.Sumbayev VV, Yasinska IM, Garcia CP, Gilliland D, Lall GS, Gibbs BF, Bonsall DR, Varani L, Rossi F, Calzolai L. Gold nanoparticles downregulate interleukin-1-induced pro-inflammatory responses. Small 2013; 9: 472–477. [DOI] [PubMed] [Google Scholar]
- 55.Huang KY, Ma HL, Liu J, Huo SD, Kumar A, Wei T, Zhang X, Jin SB, Gan YL, Wang PC, He ST, Zhang XN, Liang XJ. Size-dependent localization and penetration of ultrasmall gold nanoparticles in cancer cells, multicellular spheroids, and tumors in vivo. ACS Nano 2012; 6: 4483–4493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Goodman CM, McCusker CD, Yilmaz T, Rotello VM. Toxicity of gold nanoparticles functionalized with cationic and anionic side chains. Bioconjugate Chem 2004; 15: 897–900. [DOI] [PubMed] [Google Scholar]
- 57.Fernandez TD, Pearson JR, Leal MP, Torres MJ, Blanca M, Mayorga C, Le Guevel X. Intracellular accumulation and immunological properties of fluorescent gold nanoclusters in human dendritic cells. Biomaterials 2015; 43: 1–12. [DOI] [PubMed] [Google Scholar]
- 58.Liptrott NJ, Kendall E, Nieves DJ, Farrell J, Rannard S, Fernig DG, Owen A. Partial mitigation of gold nanoparticle interactions with human lymphocytes by surface functionalization with a ‘mixed matrix’. Nanomedicine 2014; 9: 2467–2479. [DOI] [PubMed] [Google Scholar]
- 59.Deng ZJ, Liang MT, Monteiro M, Toth I, Minchin RF. Nanoparticle-induced unfolding of fibrinogen promotes Mac-1 receptor activation and inflammation. Nat Nanotechnol 2011; 6: 39–44. [DOI] [PubMed] [Google Scholar]
- 60.Fytianos K, Rodriguez-Lorenzo L, Clift MJ, Blank F, Vanhecke D, von Garnier C, Petri-Fink A, Rothen-Rutishauser B. Uptake efficiency of surface modified gold nanoparticles does not correlate with functional changes and cytokine secretion in human dendritic cells in vitro. Nanomedicine 2015; 11: 633–644. [DOI] [PubMed] [Google Scholar]
- 61.Greulich C, Diendorf, Gessmann J, Simon T, Habijan T, Eggeler G, Schildhauer TA, Epple M, Koller M. Cell type-specific responses of peripheral blood mononuclear cells to silver nanoparticles. Acta Biomaterialia 2011; 7: 3505–3514. [DOI] [PubMed] [Google Scholar]
- 62.Shin SH, Ye MK, Kim HS, Kang HS. The effects of nano-silver on the proliferation and cytokine expression by peripheral blood mononuclear cells. Int Immunopharmacol 2007; 7: 1813–1818. [DOI] [PubMed] [Google Scholar]
- 63.Yen HJ, Hsu SH, Tsai CL. Cytotoxicity and immunological response of gold and silver nanoparticles of different sizes. Small 2009; 5: 1553–1561. [DOI] [PubMed] [Google Scholar]
- 64.Tian J, Wong KKY, Ho CM, Lok CN, Yu WY, Che CM, Chiu JF, Tam PKH. Topical delivery of silver nanoparticles promotes wound healing. ChemMedChem 2007; 2: 129–136. [DOI] [PubMed] [Google Scholar]
- 65.Bhol KC, Schechter PJ. Topical nanocrystalline silver cream suppresses inflammatory cytokines and induces apoptosis of inflammatory cells in a murine model of allergic contact dermatitis. Br J Dermatol 2005; 152: 1235–1242. [DOI] [PubMed] [Google Scholar]
- 66.Bhol KC, Schechter PJ. Effects of nanocrystalline silver (NPI 32101) in a rat model of ulcerative colitis. Dig Dis Sci 2007; 52: 2732–2742. [DOI] [PubMed] [Google Scholar]
- 67.Liao MH, Jan TR. A single exposure to iron oxide nanoparticles attenuates antigen-specific antibody production and T-cell reactivity in ovalbumin-sensitized BALB/c mice. Int J Nanomed 2011; 6: 1229–1235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Shen CC, Liang HJ, Wang CC, Liao MH, Jan TR. Iron oxide nanoparticles suppressed T helper 1 cell-mediated immunity in a murine model of delayed-type hypersensitivity. Int J Nanomed 2012; 7: 2729–2729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Wu HY, Chung MC, Wang CC, Huang CH, Liang HJ, Jan TR. Iron oxide nanoparticles suppress the production of IL-1beta via the secretory lysosomal pathway in murine microglial cells. Part Fibre Toxicol 2013; 10: 46–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Blank F, Gerber P, Rothen-Rutishauser B, Sakulkhu U, Salaklang J, De Peyer K, Gehr P, Nicod LP, Hofmann H, Geiser T, Petri-Fink A, Von Garnier C. Biomedical nanoparticles modulate specific CD4(+) T cell stimulation by inhibition of antigen processing in dendritic cells. Nanotoxicology 2011; 5: 606–621. [DOI] [PubMed] [Google Scholar]
- 71.Karakoti A, Singh S, Dowding JM, Seal S, Self WT. Redox-active radical scavenging nanomaterials. Chem Soc Rev 2010; 39: 4422–4432. [DOI] [PubMed] [Google Scholar]
- 72.Korsvik C, Patil S, Seal S, Self WT. Superoxide dismutase mimetic properties exhibited by vacancy engineered ceria nanoparticles. Chem Commun 2007; 10: 1056–1058. [DOI] [PubMed] [Google Scholar]
- 73.Xue Y, Luan QF, Yang D, Yao X, Zhou KB. Direct evidence for hydroxyl radical scavenging activity of cerium oxide nanoparticles. J Phys Chem C 2011; 115: 4433–4438. [Google Scholar]
- 74.Heckman KL, DeCoteau W, Estevez A, Reed KJ, Costanzo W, Sanford D, Leiter JC, Clauss J, Knapp K, Gomez C. Custom cerium oxide nanoparticles protect against a free radical mediated autoimmune degenerative disease in the brain. ACS Nano 2013; 7: 10582–10596. [DOI] [PubMed] [Google Scholar]
- 75.Hirst SM, Karakoti AS, Tyler RD, Sriranganathan N, Seal S, Reilly CM. Anti-inflammatory properties of cerium oxide nanoparticles. Small 2009; 5: 2848–2856. [DOI] [PubMed] [Google Scholar]
- 76.Colon J, Hsieh N, Ferguson A, Kupelian P, Seal S, Jenkins DW, Baker CH. Cerium oxide nanoparticles protect gastrointestinal epithelium from radiation-induced damage by reduction of reactive oxygen species and upregulation of superoxide dismutase 2. Nanomed Nanotechnol Biol Med 2010; 6: 698–705. [DOI] [PubMed] [Google Scholar]
- 77.Lee SS, Song WS, Cho MJ, Puppala HL, Nguyen P, Zhu HG, Segatori L, Colvin VL. Antioxidant properties of cerium oxide nanocrystals as a function of nanocrystal diameter and surface coating. ACS Nano 2013; 7: 9693–9703. [DOI] [PubMed] [Google Scholar]
- 78.Song WH, Zhang JY, Guo J, Zhang JH, Ding F, Li LY, Sun ZT. Role of the dissolved zinc ion and reactive oxygen species in cytotoxicity of ZnO nanoparticles. Toxicol Lett 2010; 199: 389–397. [DOI] [PubMed] [Google Scholar]
- 79.Lipovsky A, Nitzan Y, Gedanken A, Lubart R. Antifungal activity of ZnO nanoparticles—the role of ROS mediated cell injury. Nanotechnology 2011; 22: 1–5. [DOI] [PubMed] [Google Scholar]
- 80.Hanley C, Thurber A, Hanna C, Punnoose A, Zhang JH, Wingett DG. The influences of cell type and ZnO nanoparticle size on immune cell cytotoxicity and cytokine induction. Nanoscale Res Lett 2009; 4: 1409–1420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Hanley C, Thurber A, Hanna C, Punnoose A, Zhang J, Wingett DG. The influences of cell type and ZnO nanoparticle size on immune cell cytotoxicity and cytokine induction. Nanoscale Res Lett 2009; 4: 1409–1420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Saptarshi SR, Feltis BN, Wright PFA, Lopata AL. Investigating the immunomodulatory nature of zinc oxide nanoparticles at sub-cytotoxic levels in vitro and after intranasal instillation in vivo. J Nanobiotechnol 2015; 13: 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Feltis BN, OKeefe SJ, Harford AJ, Piva TJ, Turney TW, Wright PFA. Independent cytotoxic and inflammatory responses to zinc oxide nanoparticles in human monocytes and macrophages. Nanotoxicology 2012; 6: 757–765. [DOI] [PubMed] [Google Scholar]
- 84.Kim CS, Nguyen HD, Ignacio RM, Kim JH, Cho HC, Maeng EH, Kim YR, Kim MK, Park BK, Kim SK. Immunotoxicity of zinc oxide nanoparticles with different size and electrostatic charge. Int J Nanomed 2014; 9: 195–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Andersson-Willman B, Gehrmann U, Cansu Z, Buerki-Thurnherr T, Krug HF, Gabrielsson S, Scheynius A. Effects of subtoxic concentrations of TiO2 and ZnO nanoparticles on human lymphocytes, dendritic cells and exosome production. Toxicol Appl Pharmacol 2012; 264: 94–103. [DOI] [PubMed] [Google Scholar]
- 86.Tuomela S, Autio R, Buerki-Thurnherr T, Arslan O, Kunzmann A, Andersson-Willman B, Wick P, Mathur S, Scheynius A, Krug HF, Fadeel B, Lahesmaa R. Gene expression profiling of immune-competent human cells exposed to engineered zinc oxide or titanium dioxide nanoparticles. PLoS One 2013; 8: e68415–e68415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Moon EY, Yi GH, Kang JS, Lim JS, Kim HM, Pyo S. An increase in mouse tumor growth by an in vivo immunomodulating effect of titanium dioxide nanoparticles. J Immunotoxicol 2011; 8: 56–67. [DOI] [PubMed] [Google Scholar]
- 88.Mitchell LA, Lauer FT, Burchiel SW, McDonald JD. Mechanisms for how inhaled multiwalled carbon nanotubes suppress systemic immune function in mice. Nat Nanotechnol 2009; 4: 451–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Ryan JJ, Bateman HR, Stover A, Gomez G, Norton SK, Zhao W, Schwartz LB, Lenk R, Kepley CL. Fullerene nanomaterials inhibit the allergic response. J Immunol 2007; 179: 665–672. [DOI] [PubMed] [Google Scholar]
- 90.Yudoh K, Karasawa R, Masuko K, Kato T. Water-soluble fullerene (C60) inhibits the development of arthritis in the rat model of arthritis. Int J Nanomed 2009; 4: 217–225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Gharbi N, Pressac M, Hadchouel M, Szwarc H, Wilson SR, Moussa F. [60] fullerene is a powerful antioxidant in vivo with no acute or subacute toxicity. Nano Lett 2005; 5: 2578–2585. [DOI] [PubMed] [Google Scholar]
- 92.Dugan LL, Turetsky DM, Du C, Lobner D, Wheeler M, Almli CR, Shen CKF, Luh TY, Choi DW, Lin TS. Carboxyfullerenes as neuroprotective agents. Proc Natl Acad Sci U S A 1997; 94: 9434–9439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Zogovic NS, Nikolic NS, Vranjes-Djuric SD, Harhaji LM, Vucicevic LM, Janjetovic KD, Misirkic MS, Todorovic-Markovic BM, Markovic ZM, Milonjic SK, Trajkovic VS. Opposite effects of nanocrystalline fullerene (C-60) on tumour cell growth in vitro and in vivo and a possible role of immunosupression in the cancer-promoting activity of C-60. Biomaterials 2009; 30: 6940–6946. [DOI] [PubMed] [Google Scholar]
- 94.Chen Y-W, Hwang KC, Yen C-C, Lai Y-L. Fullerene derivatives protect against oxidative stress in RAW 264.7 cells and ischemia-reperfused lungs. Am J Physiol Regul Integr Comp Physiol 2004; 287: R21–R26. [DOI] [PubMed] [Google Scholar]
- 95.Mitchell L, Lauer F, Burchiel S, McDonald J. Mechanisms for how inhaled multiwalled carbon nanotubes suppress systemic immune function in mice. Nat Nanotechnol 2009; 4: 451–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Mitchell LA, Gao J, Vander Wal R, Gigliotti A, Burchiel SW, McDonald JD. Pulmonary and systemic immune response to inhaled multiwalled carbon nanotubes. Toxicol Sci 2007; 100: 203–214. [DOI] [PubMed] [Google Scholar]
- 97.Herzog E, Byrne HJ, Casey A, Davoren M, Lenz A-G, Maier KL, Duschl A, Oostingh GJ. SWCNT suppress inflammatory mediator responses in human lung epithelium in vitro. Toxicol Appl Pharmacol 2009; 234: 378–390. [DOI] [PubMed] [Google Scholar]
- 98.Tkach AV, Shurin GV, Shurin MR, Kisin ER, Murray AR, Young SH, Star A, Fadeel B, Kagan VE, Shvedova AA. Direct effects of carbon nanotubes on dendritic cells induce immune suppression upon pulmonary exposure. ACS Nano 2011; 5: 5755–5762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Hardy CL, LeMasurier JS, Belz GT, Scalzo-Inguanti K, Yao J, Xiang SD, Kanellakis P, Bobik A, Strickland DH, Rolland JM, O’Hehir RE, Plebanski M. Inert 50-nm polystyrene nanoparticles that modify pulmonary dendritic cell function and inhibit allergic airway inflammation. J Immunol 2012; 188: 1431–1441. [DOI] [PubMed] [Google Scholar]
- 100.Frick SU, Bacher N, Baier G, Mailander V, Landfester K, Steinbrink K. Functionalized polystyrene nanoparticles trigger human dendritic cell maturation resulting in enhanced CD4+T cell activation. Macromol Biosci 2012; 2: 1637–1647. [DOI] [PubMed] [Google Scholar]
- 101.Getts DR, Martin AJ, McCarthy DP, Terry RL, Hunter ZN, Yap WT, Getts MT, Pleiss M, Luo XR, King NJC, Shea LD, Miller SD. Microparticles bearing encephalitogenic peptides induce T-cell tolerance and ameliorate experimental autoimmune encephalomyelitis. Nat Biotechnol 2013; 31: 565–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Bosch X. Dendrimers to treat rheumatoid arthritis. ACS Nano 2011; 5: 6779–6785. [DOI] [PubMed] [Google Scholar]
- 103.Chauhan AS, Diwan PV, Jain NK, Tomalia DA. Unexpected in vivo anti-inflammatory activity observed for simple, surface functionalized poly (amidoamine) dendrimers. Biomacromolecules 2009; 10: 1195–1202. [DOI] [PubMed] [Google Scholar]
- 104.Boridy S, Soliman GM, Maysinger D. Modulation of inflammatory signaling and cytokine release from microglia by celastrol incorporated into dendrimer nanocarriers. Nanomedicine 2012; 7: 1149–1165. [DOI] [PubMed] [Google Scholar]
- 105.Hayder M, Poupot M, Baron M, Nigon D, Turrin CO, Caminade AM, Majoral JP, Eisenberg RA, Fournie JJ, Cantagrel A, Poupot R, Davignon JL. A phosphorus-based dendrimer targets inflammation and osteoclastogenesis in experimental arthritis. Sci Transl Med 2011; 3: 1–13. [DOI] [PubMed] [Google Scholar]
- 106.Barata TS, Teo I, Brocchini S, Zloh M, Shaunak S. Partially glycosylated dendrimers block MD-2 and prevent TLR4-MD-2-LPS complex mediated cytokine responses. PLos Comput Biol 2011; 7: e1002095–e1002095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Shaunak S, Thomas S, Gianasi E, Godwin A, Jones E, Teo I, Mireskandari K, Luthert P, Duncan R, Patterson S. Polyvalent dendrimer glucosamine conjugates prevent scar tissue formation. Nat Biotechnol 2004; 22: 977–984. [DOI] [PubMed] [Google Scholar]
- 108.Teo I, Toms SM, Marteyn B, Barata TS, Simpson P, Johnston KA, Schnupf P, Puhar A, Bell T, Tang C, Zloh M, Matthews S, Rendle PM, Sansonetti PJ, Shaunak S. Preventing acute gut wall damage in infectious diarrhoeas with glycosylated dendrimers. EMBO Mol Med 2012; 4: 866–881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Shukla NM, Salunke DB, Balakrishna R, Mutz CA, Malladi SS, David SA. Potent adjuvanticity of a pure TLR7-agonistic imidazoquinoline dendrimer. PLoS One 2012; 7: e43612–e43612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Hong YS, Laks H, Cui GG, Chong T, Sen LY. Localized immunosuppression in the cardiac allograft induced by a new liposome-mediated IL-10 gene therapy. J Heart Lung Transplant 2002; 21: 1188–1200. [DOI] [PubMed] [Google Scholar]
- 111.Ko S, Nakajima Y, Kanehiro H, Horikawa M, Yoshimura A, Taki J, Aomatsu Y, Kin T, Yagura K, Nakano H. The enhanced immunosuppressive efficacy of newly developed liposomal Fk506 in canine liver-transplantation. Transplantation 1995; 59: 1384–1388. [DOI] [PubMed] [Google Scholar]
- 112.Metselaar JM, Wauben MHM, Wagenaar-Hilbers JPA, Boerman OC, Storm G. Complete remission of experimental arthritis by joint targeting of glucocorticoids with long-circulating liposomes. Arthritis Rheum 2003; 48: 2059–2066. [DOI] [PubMed] [Google Scholar]
- 113.Black M, Trent A, Kostenko Y, Lee JS, Olive C, Tirrell M. Self-assembled peptide amphiphile micelles containing a cytotoxic T-cell epitope promote a protective immune response in vivo. Adv Mater 2012; 24: 3845–3849. [DOI] [PubMed] [Google Scholar]
- 114.Mitchell D, Yong M, Raju J, Willemsen N, Black M, Trent A, Tirrell M, Olive C. Toll-like receptor-mediated adjuvanticity and immunomodulation in dendritic cells: implications for peptide vaccines. Hum Vaccin 2011; 7: 85–93. [DOI] [PubMed] [Google Scholar]
- 115.Rudra JS, Tian YF, Jung JP, Collier JH. A self-assembling peptide acting as an immune adjuvant. Proc Natl Acad Sci U S A 2010; 107: 622–627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Bregoli L, Chiarini F, Gambarelli A, Sighinolfi G, Gatti AM, Santi P, Martelli AM, Cocco L. Toxicity of antimony trioxide nanoparticles on human hematopoietic progenitor cells and comparison to cell lines. Toxicology 2009; 262: 121–129. [DOI] [PubMed] [Google Scholar]
- 117.Matsumura M, Takasu N, Nagata M, Nakamura K, Kawai M, Yoshino S. Effect of ultrafine zinc oxide (ZnO) nanoparticles on induction of oral tolerance in mice. J Immunotoxicol 2010; 7: 232–237. [DOI] [PubMed] [Google Scholar]
- 118.Gibaud S, Andreux J, Weingarten C, Renard M, Couvreur P. Increased bone marrow toxicity of doxorubicin bound to nanoparticles. Eur J Cancer 1994; 30: 820–826. [DOI] [PubMed] [Google Scholar]
- 119.Desai N. Challenges in development of nanoparticle-based therapeutics. AAPS J 2012; 14: 282–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Stern ST, McNeil SE. Nanotechnology safety concerns revisited. Toxicol Sci 2008; 101: 4–21. [DOI] [PubMed] [Google Scholar]
- 121.Zajicova A, Javorkova E, Trosan P, Krulova M, Holan V. Drug-loaded nanofiber scaffolds for a simultaneous delivery of stem cells and immunosuppressive drugs in cell-based therapy and regenerative medicine. J Tissue Eng Regen Med 2014; 8: 277–278. [Google Scholar]
- 122.Saito N, Haniu H, Usui Y, Aoki K, Hara K, Takanashi S, Shimizu M, Narita N, Okamoto M, Kobayashi S, Nomura H, Kato H, Nishimura N, Taruta S, Endo M. Safe clinical use of carbon nanotubes as innovative biomaterials. Chem Rev 2014; 114: 6040–6079. [DOI] [PMC free article] [PubMed] [Google Scholar]

