Masood Alaei†
ab,
Khadijeh Koushki†
c,
Kimia Taebi†
ab,
Mahdieh Yousefi Taba
d,
Samaneh Keshavarz Hedayati
e and
Sanaz Keshavarz Shahbaz‡
*fb
aStudent Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
bUSERN Office, Qazvin University of Medical Science, Qazvin, Iran. E-mail: Sanaz.ks_023@yahoo.com; sanaz.keshavarzshahbaz@jax.org
cDepartment of Neurosurgery, University of Texas Houston Health Science Center (UTHealth), Houston, TX, USA
dDepartment of Anatomy and Cell Biology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
eDepartment of Pharmacology, Faculty of Medicine, Qazvin University of Medical Science, Qazvin, Iran
fCellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, 34197-59811, Iran
First published on 18th February 2025
Microglia, the primary immune cells of the central nervous system (CNS), are crucial in maintaining brain homeostasis and responding to pathological changes. While they play protective roles, their activation can lead to neuroinflammation and the progression of neurodegenerative diseases. Metal nanoparticles (NPs), due to their unique ability to cross the blood–brain barrier (BBB), have emerged as promising agents for drug delivery to the CNS. In this way, we aim to review the dual role of metal-containing NPs, gold (AuNPs), silver (AgNPs), iron oxide (IONPs), zinc oxide (ZnONPs), cobalt (CoNPs), titanium dioxide (TiO2NPs), and silica (SiO2NPs) in modulating microglial activity. Some NPs promote anti-inflammatory effects, while others exacerbate neuroinflammation. We examine how these NPs influence microglial activation, focusing on their potential therapeutic benefits and risks. A deeper understanding of NP-microglia interactions is crucial for developing safe and efficient treatments for neuroinflammatory and neurodegenerative disorders.
Recent advancements in nanomedicine have highlighted the potential of polymer and metal nanoparticles (NPs) as innovative therapeutic agents capable of modulating microglial activity and influencing neuroinflammatory responses. Recently, we reviewed polymeric NP carriers applied to deliver microglial inhibition in neurological disorders with remarkable results, showcasing the promising role of these NPs in microglial modulation during drug delivery.2
In addition, due to the unique physicochemical properties—such as high surface area, tunable size, and functional versatility—Metal NPs can effectively cross the blood–brain barrier (BBB),3 presenting new opportunities for targeted drug delivery in treating CNS disorders. Various metal-containing nanoparticles, including gold (AuNPs),4 silver (AgNPs), iron oxide (IONPs), zinc oxide (ZnONPs), cobalt (CoNPs), titanium dioxide (TiO2NPs), and silica (SiO2NPs), have been shown to interact with microglia, either promoting anti-inflammatory effects or exacerbating neuroinflammation.5
In this way, this review aims to provide a comprehensive analysis of metal nanoparticles' impact on microglial dynamics and their implications for CNS function. By examining the complex interplay between these nanomaterials and microglial cells, we seek to elucidate their potential therapeutic benefits and risks in addressing neuroinflammatory conditions. Understanding these interactions is crucial for developing safe and effective treatments for neurodegenerative diseases and advancing the field of nanomedicine.
Neuroprotective functions of microglia include maintaining homeostasis by regulating the brain's internal environment, metabolic regulation, and facilitating immune responses.14,15 In their resting state, known as the M0 phenotype, microglia clear apoptotic debris, remove dysfunctional synapses, and support pruning in developing brains. This process involves synaptic remodeling, phagocytosis of cells with intracellular inclusions, neuronal feedback,16 facilitating myelination,17 neurogenesis, and trophic maintenance of neurons.18,19 These functions are crucial for preserving a healthy brain environment.
Microglia also have two activated states: the “M2” anti-inflammatory phenotype and the “M1” proinflammatory phenotype. M2 microglia are responsible for healing-related actions, such as maintaining homeostasis and promoting anti-inflammatory processes. They contribute to the generation of anti-inflammatory cytokines and neurotrophic agents.10,20,21 In contrast, M1 microglia are the first line of defense, responsible for homeostasis and pro-killing functions. They can produce proinflammatory cytokines interleukin-1b (IL-1b), IL-17, IL-12, IL-6, IL-18, IFN-c, IL-23, inducible nitric oxide synthase (iNOS), tumour necrosis factor-alpha (TNF-α), cyclooxygenase-2 (COX-2), reactive oxygen species (ROS), prostaglandin E2 (PGE2), and (MHC-II).22,23
Although the pro-inflammatory functions of the M1 phenotype are protective in certain situations, excessive release of cytotoxic substances has been attributed to the development of neuroinflammatory disorders.24 Many studies have shown that microglial activation plays a pivotal role in the pathogenesis of neurodegenerative disorders, including Parkinson's disease, Alzheimer's disease (AD), psychiatric disease, ischemic disease, traumatic brain injury, and stroke.10,25–27 Microglia become activated in response to pathogen-associated molecular patterns (PAMPs), danger-associated molecular patterns (DAMPs), and certain nanostructures, triggering a pro-inflammatory response.
To perform their surveillance functions, microglia are equipped with different signaling immunoreceptors, such as Toll-like receptors (TLR4 and TLR2), complement phagocytic receptors (CR4 and CR3), and scavenger receptors (Cluster of Differentiation-36 (CD36) and CD204) to interact with extracellular species.25,28,29 When neurons are exposed to harmful stimuli, they begin to generate “help me” signals, including fractalkine, interleukin-34 (IL-34), and CX3C chemokine.30,31 In response, microglia become activated and release proinflammatory cytokines, including TNF-α and IL-1β, along with neurotoxic molecules, such as ROS.32 Moreover, activated microglia can release glutamate, which causes an increase in the neuronal and neurite number, thereby contributing to neurodegenerative disorder exacerbation.25,33
Upon stimulation of microglial surface receptors, several signaling pathways become activated. They induce the production of inflammatory cytokines, the NLRP inflammasome activation, and beta-secretase enzyme (BACE) expression,29 ultimately driving neuroinflammation and neuronal death. Microglia can also accidently damage neurons while attempting to limit infections by producing cathepsin B, superoxide, nitric oxide, and derivative oxidants.34 Furthermore, microglia-mediated reduction in insulin-like growth factor 1 (IGF-1) and nutritional brain-derived neurotropic factor (BDNF) contributes to neuronal death35,36 (Fig. 1). These mechanisms highlight the potential of microglia M1 in the beginning and progression of neurodegenerative diseases.
Given the crucial role of microglia, particularly M1 microglia, in neuroinflammatory processes, potent inhibitors that target microglial immune activity may offer promising strategies for addressing these issues.
Resting microglia, known as M0 microglia, perform multiple health-promoting functions. They have the potential to support neuronal progenitors in the neurogenesis process. Additionally, they can appropriately eliminate cell debris and dead cell bodies through phagocytosis. Moreover, M0 microglia can phagocytose unnecessary synaptic connections (called synaptic pruning) to leave more space to form new connections between neurons. An oligodendrocyte is a cell that is responsible for forming myelin around axons. Interestingly, M0 microglia participate in this process by assisting oligodendrocytes. Microglia M0 also contribute to the neuronal feedback mechanism. When neurons are activated, microglia use negative neuronal feedback to prevent neuronal overactivation, thus leading to a balanced neuronal environment. Additionally, resting microglia can play a role in myelin clearance by attempting to phagocytose myelin debris to prevent impaired neurogenesis.
Resveratrol, a polyphenol abundant in peanuts, raisins, red grapes, and berries, possesses anti-inflammatory, antioxidant, and antiapoptotic properties.43,44 Studies have illustrated that resveratrol can limit microglial activity and mitigate rotenone-induced neurotoxicity, CD11 (a microglial activity marker), TNF- α, and IL-1β.45 Curcumin, another well-known inhibitor, is a potent anti-inflammatory and antioxidant agent found in turmeric. Interestingly, curcumin plays a suppressive role in microglial activity and decreases microglia-induced inflammatory cytokines.46 It can also block the MAPK signaling pathway, which further inhibits the activation of NF-κβ.47 Curcumin suppresses the production of COX-2 and other inflammatory cytokines by modulating TLR4 signaling.48 Quercetin, a natural flavonoid present in vegetables and fruits, namely green tea, onions, apples, red grapes, and berries, is also a potent inhibitor of microglial activity due to its antioxidant and anti-inflammatory properties.41 The treatment with quercetin reduces TLR-4 expression in the hippocampus and cortex, a key receptor involved in microglial activation.49 In addition, it can reduce the expression of ionized calcium-binding adapter molecule 1 (Iba-1), IL-1β, TNFα, and COX2.50 Minocycline, a well-known antibiotic, has also been extensively studied for its ability to inhibit microglial function, reducing the number of microglial inflammatory mediators.51,52 Minocycline can also reduce MHC-II expression in microglia.42
While these drugs offer significant potential in mitigating microglial immune responses, drug delivery to the CNS is still a considerable challenge because of the restrictive nature of the blood–brain barrier (BBB). Its specific structure, surrounding cells, and molecular transport mechanisms limit the efficient delivery of many therapeutic agents. Therefore, the following section further discusses these challenges and potential solutions.
Various transport systems control the traffic of substances across the BBB, including fenestra, transendothelial channels, pinocytotic vesicles, active efflux transport proteins, and breast cancer resistance proteins.58 These systems are crucial in controlling the flow of specific drugs and essential nutrients into the CNS. Passive distribution via a paracellular or transcellular pathway for low molecular weight or lipophilic substances (the majority of CNS-targeting drugs); vesicular trafficking, such as adsorptive-mediated transcytosis (for positively-charged substances); receptor-mediated transcytosis (an energy-dependent pathway for proteins hormones and proteins); and carrier-mediated transport (for amino acids and glucose) are some examples.59–61 The BBB's relative impermeability is primarily due to tight junctions between endothelial cells.62,63 The mentioned mechanisms and complexes are potential obstacles for most drugs to enter the CNS, making drug delivery challenging with high failure rates and increased costs.64 Even if drugs manage to cross the BBB, achieving therapeutic concentrations within the CNS can be difficult. Ensuring that drugs are effective without causing adverse side effects and neurotoxicity remains a critical challenge.65 Also, CNS disorders often require targeted treatments that are tailored to individual patients. Achieving this specificity in drug delivery systems is complex and ongoing.66 Moreover, many conventional therapeutic agents suffer from low bioavailability due to rapid metabolism or elimination, leading to degradation. Therefore, encapsulating drugs can protect them from degradation and improve their pharmacokinetic profiles.67 The ability to control drug release is also vital for maintaining therapeutic concentrations over time and avoiding excessive drug dosages, which many conventional drugs lack this ability.68 These problems can lead to ineffective treatment for CNS disorders such as cerebral malignancies due to low brain penetration. Over the last decade, researchers have developed various technologies to overcome these challenges. One promising approach is using targeted vectors (peptides, proteins, antibodies, or specific formulations) to aid the transport of drugs across the BBB.69 Additionally, nano-delivery systems have gained attention as a novel strategy. These systems offer the possibility to improve therapeutic precision and maintain medicine efficacy while minimizing toxicity.23,70
Some NPs can traverse vessel walls and enter brain tissue. This ability is mainly attributed to their physiochemical characteristics, including size, shape, surface chemistry, surface charge, and surface traits.76 Therefore, engineering the NPs to modify their morphological features enhances their ability to cross the BBB. One strategy involves designing the NPs in combination with elements specific to pathological sites and BBB-penetrating molecules, such as the spontaneous exploitation of NPs with trans-Golgi network (TGN) peptides and the cancer cell-specific aptamer AS1411. Such modification can remarkably improve BBB penetration and target delivery.77 The shape and surface characteristics of NPs also influence how microglia internalize them;78 for instance, spiky “urchin-shaped” gold NPs(AuNps) show higher levels of microglial uptake in contrast to rod or spherical-shaped AuNPs.72,79 Hence, the way microglia take NPs up is highly dependent on the design of the NP surface and properties.80 Microglia internalize NPs primarily through active processes like invagination and endocytosis.69 However, certain NPs may also passively diffuse across the cell membrane. Furthermore, the uptake of NPs varies between microglial phenotypes. Compared with resting microglia, lipopolysaccharide (LPS)-activated microglia display greater dendrimer uptake, which is associated with increased endocytosis.81
Once NPs reach the brain tissue, they can release their therapeutic cargo at the target location in a time-dependent way and navigate the drugs (genes, small molecular agents, and biomolecules) to the target organelle without being trapped in endo/lysosomes. Various strategies have been developed to facilitate lysosomal escape. One common approach is the proton sponge effect, where pH-sensitive nanoparticles swell and disrupt the lysosomal membrane upon exposure to the acidic environment, leading to the release of their contents into the cytosol. Other methods include osmotic lysis, which results from the disassembly of nanoparticles in response to low pH, and mechanical disruption techniques that utilize nanomechanical actions or photochemical processes to destabilize lysosomal membranes.82 This controlled release mechanism decreases the required drug dosage and side effects of using nanomaterials.56,83,84 Engineering NPs is also critical for drug delivery. One example involves using stimulus-sensitive bonds to ensure the accurate release of cargo in the expected areas in response to spatial variations in redox capacity.85 Additionally, NPs designed with epidermal growth factor (EGF) and two types of bioresponsive bonds that enhance vascular permeability86 can deliver drugs directly to subcellular organelles, improving drug efficacy in the brain.87,88 Such designs have been employed to deliver DNA-binding agents and ROS-generating drugs into mitochondria,76,89 facilitating the treatment of stroke, glioma, epilepsy, and AD. Besides, the unique electrical and optical traits of some nanoparticles enable them to treat CNS disorders.71
Put simply, NPs favor the treatment of neurodegenerative disorders by delivering essential drugs to the brain, which is partially impossible for macromolecular drugs to reach alone. Nonetheless, metal-containing NPs hold promise for enhancing drug delivery performance compared to NPs alone. However, they also present side effects such as toxicity in the brain by switching M0 microglia to the M1 phenotype. Alternatively, they may alleviate the neuroinflammation by promoting the M2 phenotype. Therefore, the following section further discusses how metal-containing NPs, as multifaceted substances, influence brain health and neuroinflammatory conditions in detail.
Once inside the brain, metal-containing NPs are immediately internalized by microglia and astrocyte-like (ALT) cells. M1 microglia produce CCL2 and proinflammatory cytokines, for instance, NO, IL-12, IL-1β, and IL-6, which result in acute neuroinflammatory reactions. On the contrary, the M2 phenotype releases anti-inflammatory cytokines such as TGF-β, IL-10, and IL-4, aiding the resolution of neuroinflammation and repairing the damaged brain.92 Metal-containing NPs can influence this polarization, with some promoting M1 activation and exacerbating inflammation, while others encourage the M2 phenotype, facilitating neuroprotection and recovery.93
Metal NP-induced M1 phenotype can exert neurotoxic effects through various mechanisms, including the generation of ROS, which leads to oxidative stress, inducing neuronal apoptosis and necrosis and contributing to neurodegenerative diseases.94 Furthermore, metal NPs can trigger the activation of microglia, leading to the release of pro-inflammatory cytokines, exacerbating neuroinflammation, and compromising neuronal health.95 The neurotoxic potential of metal NPs is significantly influenced by their physicochemical properties. Smaller NPs generally exhibit higher reactivity and greater cellular uptake, which can enhance their therapeutic efficacy but also increase the risk of toxicity. Surface modifications, such as PEGylation, can alter NP-cell interactions, improve circulation time, and reduce immunogenicity, potentially mitigating some toxic effects.96 The neurotoxicity of metal NPs is often dose-dependent, with low concentrations potentially eliciting beneficial effects by modulating microglial activity and promoting anti-inflammatory responses. Higher concentrations can overwhelm cellular defense mechanisms, leading to toxicity. This highlights the crucial need to optimize dosages in therapeutic applications to maximize efficacy while minimizing adverse effects.97 Chronic exposure to metal NPs may lead to cumulative neurotoxic effects that are not immediately apparent. NP prolonged exposure potentially results in significant alterations in microglial function and neuronal integrity, leading to long-term cognitive deficits or the exacerbation of existing neurological conditions.98
For instance, silver nanoparticles (AgNPs) are able to promote M1 polarization and induce neurotoxicity. Duffy et al. reported that AgNPs triggered the production of proinflammatory cytokines like TNF-α in BV2 microglial cells, leading to neuroinflammatory responses.99 Additionally, AgNPs have been shown to enhance the level of the proinflammatory chemokine CXCL13 in microglia, astrocytes, and Neuro2a (N2a) cells, further elevating the levels of IL-1β.100 Besides silver, other metal-based NPs, like titanium dioxide nanoparticles (TiO2NPs), have been linked to neuroinflammatory responses. TiO2NPs can activate inflammasomes and nuclear factor-κB (NF-κB), activating microglia and subsequent inflammation.75
Despite these proinflammatory effects, some metal-containing NPs also exhibit anti-inflammatory properties. AuNPs, for instance, have been evidenced to prevent the proinflammatory reactions in microglia by promoting M2 phenotype, thereby contributing to CNS repair.101 Similarly, exposure to AgNPs has been found in another article to be associated with a reduction in inflammation. This could be possibly due to the detoxification of silver ions through sulfidation and the activation of hydrogen sulfide-synthesizing enzymes.102 Moreover, iron oxide nanoparticles (IONPs) have been demonstrated to inhibit the production of IL-1β in LPS-stimulated microglia, further underscoring the potential neuroprotective role of metal-based NPs.103 (Fig. 2).
Although remarkable endeavors have been made to understand the neurotoxic and neuroprotective effects of metal-containing NPs, much remains to be learned about their role in neuroinflammation. Additionally, since studies have shown that microglia activation can serve as an early warning and defensive response against exogenous NPs that invade and accumulate in the CNS, it is crucial to investigate the interaction between various metal-containing NPs conjugated with specific drugs and microglia. Hence, additional exploration is needed to fully exploit the ability of nanotechnology to treat CNS disorders.
In the following section, we aim to comprehensively discuss some well-known metal-containing NPs and their conjugation with certain drugs, highlighting how NP uptake by microglia influences treatment outcomes (Table 1).
NP type | The surface coating | NP properties | Cell type/animal models | Mechanisms & outcomes | Ref |
---|---|---|---|---|---|
a Abbreviations: Au, gold; Ag, silver; IO, iron oxide; Co, cobalt; ZnO, zinc oxide; TiO2, titanium dioxide; SiO2, silica; Si, silicon. | |||||
Au | Gold-quercetin NPs | 27 nm, 100, 200, 400 μg mL−1 | LPS-stimulated microglia | Significant decrease in both the transcriptional and translational levels of inducible NO synthase, cyclooxygenase-2; COX-2 and iNOS | 110 |
Inhibiting the release of NO and proinflammatory PGE2 from LPS-stimulated microglia without causing any cytotoxic effect | |||||
Au | AuNP-FIB-BS hard corona | 50–3 nm, dose: 26 μg mL−1 | Murine BV2 microglia | Significant decrease in oxidative stress and ROS generation | 111 |
Significant increase in cellular uptake | |||||
Au | Ephedra sinica Stapf extract extract-capped AuNPs | 57.6 ± 3.07 nm | Microglia | Decrease in the production of the proinflammatory cytokines, including IL-1β, IL-6, and TNF-α in LPS-stimulated microglia through the downregulation of JAK/STAT, NF-κB, IKK-α/β, JNK, and p38 MAPK signaling pathways | 112 |
Upregulation of NQO1 and HO-1 | |||||
Activation of AMPK and Nrf2 | |||||
Au | Polyethyleneglycol-coupled GNPs | 8.09 ± 3.60 nm (85 × 106 nL−1) | Microglia | Self-limited, transient, and predominantly localized cellular response of microglia at the injection site within 3 to 90 days following intracerebral injection | 113 |
No chronic microglial response by NPs | |||||
Au | Paeonia moutan-functionalized GNPs | 100 nm, 5, 10 and 20 μg mL−1 | BV2 microglia and C57BL/6 mice/mouse model of parkinsonian | Inhibiting the inflammatory cytokines (IL-1β, IL-6, and TNFα) and NO synthesis | 115 |
Trapping the reactive oxygen in LPS-stimulated BV2 murine microglia suffering from PD | |||||
Significant reduction in the expression of inflammatory COX2 and iNOS | |||||
Increase in the expression of tyrosine hydroxylase | |||||
Improving motor coordination and alleviating the neuroinflammation in the Parkinson model | |||||
Au | Paeonia moutan-functionalized GNPs | 190–450 nm | BV2 microglia | Decrease in formation of intracellular α-syn oligomers, pro-inflammatory cytokines (IL-6 and TNF-α) secretion, and α-syn internalization, in vitro | 116 |
Lowered α-syn-induced production of ROS and NO in microglia | |||||
Nuclear translocation of NF-κB | |||||
Suppression the expression of Iba-1 by α-syn-challenged microglia | |||||
Au | Anthocyanin-loaded poly (ethylene glycol)-AuNPs | 135 ± 5 nm | Aβ1-42-induced mouse model and BV2 microglia | Reduction in Aβ1-42-induced neuroapoptotic markers and neuroinflammation by restricting the p-GSK3β/NF-κB/p-JNK pathways in both in vivo and in vitro AD models | 118 |
Significant mitigation in Aβ-induced apoptosis in both BV2 microglia and the mouse hippocampus by reducing Cyt. c release, Bax protein expression, and increasing Bcl2 protein levels | |||||
Reduction in the production of Iba-1 and GFAP in microglia | |||||
Mitigating the expression of iNOS and p-NF-κB proteins | |||||
Ag | AgNPs | 20 nm, 50 μg mL−1 | Mouse BV-2 microglia | Decrease in microglial growth by AgNPs and CdTe-QDs by stopping the cells in the G1 phase (CdTe-QDs) or S phase (AgNPs and CeO2NPs) of the cell cycle | 129 |
Ce | Cerium oxide NPs | 25 nm, 100 μg mL−1 | Significant reduction in Aβ uptake by BV-2 microglia with AgNPs and CeO2NPs, but not CdTe-QDs | ||
Cadmium telluride quantum dots | 3.8 nm, 3 or 10 μg mL−1 | No impacts on the secretion of IL-6, IL-1b, and IFNg by Aβ, nor NPs or their combinations | |||
Significant increase in TNFa secretion by CeO2NPs | |||||
Ag | — | 20 nm, dose: 50 μg mL−1 | Mouse BV-2 microglia | Efficiently blocking the Aβ uptake by microglia | 130 |
Ag | — | 20 nm | Mouse BV-2 microglia | Impairing Aβ clearance by BV-2 microglia by competing with Aβ for scavenger receptors | 131 |
Ag | — | 10 nm, 6, 3, and 1 μg mL−1 | BV2 microglia | Releasing soluble factors like NO and H2O2 from glial cells | 121 |
Significantly inhibiting the induced ROS and cytokines (TNF-α, MCP-1, and IL-6) from LPS-activated BV-2 | |||||
Decreasing cell viability of BV-2 by releasing H2O2 from ALT cells through indirect AgNP exposure | |||||
Ag | — | 23 nm diameter | Glial cells | Destruction of the cerebellum granular layer, causing cerebellar ataxia-like symptoms in rats | 124 |
Ag | — | 23.44 ± 4.92 nm, 5 μg mL−1 | BV2 microglia cell lines of mouse | AgNPs-induced M1 polarization of microglia in a time- and dose-dependent way by inhibiting the fusion of autophagosomes with lysosomes | 126 |
Increasing the expression of pro-inflammatory genes such as IL-1β, TNF-α, Iba-1, NF-κB, and MCP-1 in BV2 cells | |||||
Reducing the mRNA expression of anti-inflammatory cytokines | |||||
Ag | — | 3–5 nm, dose: 5–12.5 μg mL−1 | Murine BV-2 microglia | Inducing pro-inflammatory cytokine secretion such as IL-1β secretion and gene expression of CXCL13, GSS, and macrophage MARCO | 100 |
Altering protein and gene expressions of Aβ deposition by inducing the expression of amyloid precursor protein (APP) gene | |||||
Ag | — | 49.7 ± 10.5 nm | Microglia | Reducing LPS-stimulated NO, TNF-α, and ROS production | 133 |
Significant anti-inflammatory effects | |||||
Reducing microglial toxicity to dopaminergic neurons | |||||
Ag | — | Human microglia cells (HMC3) | M1 to M2 phenotype switch | 134 | |
Enhancing the expressions of anti-inflammatory markers including transforming TGF-β and IL-10 | |||||
A significant reduction in mRNA expressions of TNF-α and IL-6 | |||||
Biogenic AgNPs were protected against oxidative stress and neuroinflammation by targeting Nrf2/HO-1 and TLR4/MyD88 signaling pathways | |||||
Ag | — | 18 ± 1.8 nm | Microglia | Neurobehavioral alterations in offspring | 172 |
Body fat increase | |||||
Long-term gut dysbiosis | |||||
Reducing the microglial counts | |||||
IO | — | ∼65 nm | Cultured rat microglia | Time- and concentration-dependent uptake | 136 |
Longer incubation periods of exposure or higher concentrations or severely attenuated cell viability | |||||
IO | — | 58.7 nm, 1–510 μ Fe/mL | Primary murine microglia | Attenuation of the IL-1β production, but not TNF-α, mediated by their accumulation in lysosomes and affecting the secretory lysosomal pathway of cytokine recessing | 137 |
Suppression of IL-1β converting enzyme in IONP-treated murine microglia by decreasing the activity of cathepsin B | |||||
Magnetic iron oxide (γ-Fe2O3) | — | 11 ± 3.5 nm | rTg4510 tau-mutant mice | A significant decrease in the number of activated microglia in comparison with the same concentration of the free peptides by stabilizing the peptide to the γ-Fe2O3 NPs | 138 |
Fe2O3 | — | γ -Fe2O3: (31 ± 17) nm | BV2 microglia | Proliferation of microglia | 139 |
α-Fe2O3NP: (22 ± 5) nm | Increased phagocytosis | ||||
Dose: 0.02, 0.2, 2 mol Fe/L of Fe2O3-NP suspensions or FeCl3 solution | Higher release of ROS and NO by microglia | ||||
Co | — | 50 nm | C57BL/6J mice brain | Toxic effects and inflammatory responses in BV2 microglia and mice by activating the NOX2 (NADPH oxidase 2) | 142 |
Dose: 1.25, 2.5 and 5 μg mL−1 | BV2 microglia | Catalyzing ROS production, IL-1β, NLRP3, accompanied by tau phosphorylation | |||
Co | — | 96 and 123 nm | Microglia | Microglial activation | 143 |
ZnO | — | 38.52 ± 2.82 nm, dose: 6.6 μg mL−1 | Mouse microglia N9 cell line | Disrupting the MMP activity and subsequently inducing the apoptotic pathway in the microglia by NADPH oxidase-independent ROS and ATP depletion | 146 |
Disrupting mitochondrial membrane potential | |||||
Microglia apoptosis, involving altered intracellular calcium (Ca2+) level, mitochondrial ROS production, caspase-9 and -3 activation, ERK and p38 phosphorylation, and cytochrome-c release | |||||
ZnO | — | 50 nm, dose: 10 μg mL−1 | Murine BV-2 microglia | Increase in the ROS levels and oxidative stress in BV-2 cells in a time-dependent manner through autophagy and PINK1/parkin-mediated mitophagy | 150 |
Increasing count of swollen mitochondria and autophagosomes | |||||
ZnO | — | 20 nm (5, 10, 20, 40, and 80 μg mL−1) | Murine BV-2 microglia | Influencing the lysosomal destabilization | 151 |
Inducing extensive cellular and organelle (mitochondria, lysosome), ROS accumulation, and consequently nonapoptotic cell death, leading to the release of lysosomal enzymes | |||||
Promoting inflammation by cell debris and accumulating ROS at the CNS level | |||||
ZnO | — | 42.31 ± 17.94 nm, dose: 30 μg mL−1 | BV2 microglia cell line | Driving microglia and inflammatory responses in the CNS by activating the Ca2+-dependent ERK, p38, NF-κB pathways | 152 |
ZnO | — | 26.4 ± 2.3 nm, 5 μg mL−1 | BV2 microglia | Microglial activation and proliferation by ERK and Akt signaling pathways | 153 |
ZnO | — | 50 nm | Microglia | Induction of tau protein expression, microglia activation, and oxidative stress in the brain, resulting in neurotoxicity | 154 |
ZnO | Luteolin/ZnO NPs | 17 nm | Microglia | Regulating microglia polarization by targeting C/EBPA and alleviating inflammatory injury by modulating the redox-sensitive signal transduction pathways | 155 |
TiO2 | TiO2 NPs (Degussa P25) | 330 nm, dose: 2.5–120 ppm | BV2 microglia | Upregulation of NF-κB and ERK/MAPK | 148 |
Stimulating BV2 microglia to have an prolonged and immediate release of ROS | |||||
Damaging neurons at low concentrations in cultures of the brain striatum, probably by microglial-generated ROS | |||||
Influencing genomic pathways linked to cell cycling, upregulation of apoptotic pathways, inflammation, mitochondrial bioenergetics, and downregulation of energy metabolism | |||||
TiO2 | — | 20–30 nm, 0.1 to 200 μg mL−1 | BV2 microglia | TiO2NP accumulation in BV-2 cells | 157 |
Induction of oxidative stress and mitochondrial dysfunctions | |||||
Damaging the permeability of cell membranes, inhibiting cell adhesion with a loss of mitochondrial transmembrane potential. Induction of ROS overproduction and reducing cell viability | |||||
TiO2 | — | 1–100 nm | Microglia | Producing excessive ROS via the oxidative burst | 158 |
Interfering with mitochondrial energy production in vitro | |||||
Damaging membrane integrity | |||||
TiO2 | TiO2 NPs (Degussa P25) | 30 nm | BV2 microglia | Formation of free radicals in cellular and morphological expressions | 159 |
TiO2 | — | 21 nm, 25–200 μg mL−1 | Male C57BL/6 mice and murine BV2 microglia cell line | Stimulating inflammatory mediators in the brain and neurons in vitro | 160 |
Significantly elevating pro-inflammatory cytokine (TNF-α and IL-1β) mRNAs and IL-1β protein levels in the brains of LPS-exposed mice | |||||
Enhancing TNF-α production and NF-κB binding activity by LPS-stimulated BV2 microglia | |||||
Causing neuroinflammatory responses by enhancing microglial activation in the preinflamed brain and leading microglia N9 to apoptosis | |||||
TiO2 | TiO2NPs | 20–60 nm/0.25 mg mL−1 and 0.5 mg mL−1 | Primary microglia | Inducing a significant expression of iNOS and subsequent NO secretion | 162 |
HAP-NPs | Upregulating the expression levels of MIP-1 and MCP-1 from NP-stimulated microglia by inducing NF-κB activation | ||||
Increasing the production of TNF-α, IL-6, and IL-1β by TiO2-NPs and HAP-NPs | |||||
TiO2 | — | 35 nm, dose: 4–125 μg mL−1 | Microglia N9 | Inducing TiO2-induced apoptosis | 173 |
TiO2 | — | 6 nm, dose: 100–5 μg mL−1 | BV-2 microglia | Inducing IL-1β production and ROS production | 174 |
Clathrin-dependent endocytosis, phagocytosis, and a slow translocation to the lysosome in BV2 cells | |||||
More TiO2NP uptake in LPS-activated BV-2 than normal BV-2, resulting in more released ROS, IL-6, IL-1β, and MCP-1 levels | |||||
SiO2 | Fluorescein isothiocyanate -tagged SiO2 NPs | 115 nm | Male C57BL/6N mice & microglia | Increasing Iba-1 antibody in the hippocampus | 166 |
SiO2 | Silica-coated magnetic NPs containing rhodamine B isothiocyanate dye | 50 nm | Murine BV2 microglia | Increasing the expression of Iba1 | 169 |
Increasing the serine protein, especially excitotoxic D-serine secretion in the growth medium of activated microglia from primary rat microglia | |||||
Activation of primary microglia | |||||
Accumulation of ubiquitinated proteins and increasing the inclusion bodies in primary cortical and dopaminergic neurons, cocultured with activated primary microglia | |||||
Reduction of intracellular ATP levels and proteasome activity in cocultured neuronal cells, especially in primary cortical neurons, by D-serine secretion | |||||
Si | — | 48.53 ± 3.12 nm | Murine BV2 microglia | Significantly increasing caspase-1, ASC, and NLRP3 after stimulation by LPS and SiNPs | 170 |
Raising the production of inflammatory factors, including IL-6, IL-1β, and TNF-α | |||||
Decreasing the cell viability by increasing the concentration of NP | |||||
Changing the ultrastructure | |||||
Invading the cytoplasm | |||||
Activating the NLRP3 inflammasome | |||||
Releasing a large number of inflammatory factors | |||||
Disrupting cellular antioxidant function | |||||
Inducing ferroptosis | |||||
Increasing intracellular ferrous ion levels | |||||
SiO2 | — | — | Microglia | Elevating oxidative stress levels | |
Activation of microglial functions | |||||
Si | — | 150–200 nm | Primary rat microglia | Significantly increasing intracellular RNS and ROS productions | 149 |
Decreasing TNF-α gene expression | |||||
Increasing the expression of COX-2 gene | |||||
Inducing a small but detectable IL-1β release |
Metal-containing nanoparticles have serious impacts on microglia in the CNS. They can directly influence resting microglia (M0) to switch them to the M1 or M2 phenotype. They can also induce this effect on the M1 and M2 phenotypes, resulting in a phenotypic switch between M2 and M1. In case of a switch to the M1 phenotype, proinflammatory and inflammatory cytokines are likely to be released, resulting in an inflammatory state in the CNS and, consequently, neuronal death. On the other hand, if a phenotype switch occurs to the M2 phenotype, it is probable that anti-inflammatory cytokines will be released, leading to a protective state in the CNS and healthy neurons.
Several researchers have confirmed the anti-inflammatory function of AuNPs in the CNS by influencing microglia. For example, Ozdal et al. revealed that gold-quercetin NPs exhibited superior anti-inflammatory and therapeutic effects compared to free quercetin. These NPs notably reduced the translational and transcriptional levels of inflammation-associated enzymes, PGE2, and nitric oxide (NO) in LPS-stimulated microglia without causing cytotoxic effects. This highlights the potential of AuNPs as carriers to address solubility issues of therapeutic compounds like quercetin.110 Another example involves dihydrolipoic acid (DHLA)-functionalized AuNPs, which act as neuroprotective antioxidants. These GNPs polarize microglia to M2-like phenotype, effectively reducing oxidative stress and NF-κB signaling. Additionally, they support microglial survival by preventing apoptosis.101 Moreover, Kuschnerus and colleagues reported that AuNPs coated with a hard corona composed of fibrinogen (FIB) and bovine serum (BS), in vitro, significantly enhanced cellular uptake and lowered oxidative stress and ROS production in microglia more effectively than AuNPs-FIB, protein corona (PC), and BS-T120W3-AuNPs each alone. This selective formation of AuNP-corona complexes may offer a promising strategy for controlling oxidative stress and improving cellular uptake.111 Additionally, in an in vitro study, E. sinica Stapf extract (ES)-functionalized AuNPs lowered proinflammatory cytokine levels by downregulating the NF-κB and Janus kinase/signal transducers and activators of transcription (JAK/STAT) in LPS-stimulated microglia, thereby suggesting that ES-functionalized AuNPs may mitigate neuroinflammation and neurodegenerative disorders.112 Also, Diaz and colleagues113 evaluated microglial responses to intracerebrally injected PEGylated AuNPs (polyethylene glycol-coupled AuNPs). The results indicated a transient and predominantly localized cellular response of microglia and astrocytes at the injection site with minimal harmful effects on the brain for 3 to 90 days. It was suggested that neural tissues could tolerate PEGylated GNPs well. Interestingly, Hutter et al. discovered that AuNP exposure caused a limited and transient upregulation of TLR-2 and inflammatory markers like IL-1α, NO, and GM-CSF in microglia. Notably, microglial activation occurred in a limited number at a slow pace, highlighting their ability for long-term drug delivery to the CNS.114
AuNPs have also shown privileges in treating neurodegenerative diseases like Parkinson's disease (PD). In this regard, a study utilizing Paeonia moutan-functionalized AuNPs (PM-AuNPs) in a PD mouse model showed that these NPs significantly inhibited the production of NO and inflammatory cytokines and scavenged ROS in LPS-stimulated BV2 microglia all without causing cytotoxic effects. PM-AuNPs reduced levels of COX2 and iNOS, key markers of inflammation, while improving motor coordination in the PD model.115 Consistently, Zhao et al. observed that PM-AuNPs reduced α-synuclein internalization and oligomer formation, as well as decreasing TNF-α and IL-6 levels in vitro. This supports PM-AuNPs’ role in managing PD-related neuroinflammation.116 Additionally, AuNPs have been used to treat Alzheimer's disease (AD). As a study has shown, anthocyanins-loaded GNPs successfully crossed the BBB. They inhibited a key inflammatory pathway in microglia-induced neuroinflammation, p-GSK3β, without harming neurons. GSK-3β mainly regulates the balance between proinflammatory and anti-inflammatory agents in microglia. Intriguingly, it activates the JNK and NF-κB pathways, resulting in enhanced chemokine and cytokine production117 by microglia in AD models. Therefore, anthocyanins-loaded GNPs could also indirectly inhibit NF-κB and JNK pathways. Anthocyanins conjugated with PEG-GNPs (AnPEG-GNPs) could also reduce the expression of Aβ1–42-escalated neuroapoptotic markers in BV2 microglia in mouse AD models, making it a promising therapeutic approach.118
Overall, AuNPs offer considerable advantages, including their anti-inflammatory properties and ability to cross the BBB, making them promising carriers for drug delivery in neurological disorders like AD and PD, with minimal microglia-mediated side effects for neurons and the brain. Nonetheless, more investigation is necessary to address the potential for even mild and transient neuroinflammatory responses associated with AuNPs, ensuring their safety and efficacy in CNS treatments.
Several studies have shown that AgNPs can induce neurotoxic effects through microglial activation. For instance, one study evidenced that prenatal AgNP exposure led to cognitive dysfunctions and abnormal behaviors in adults, which were linked to microglial activation.123 Another survey by Hsiao et al. found the toxic effects of AgNPs on neurons were indirectly mediated by the release of NO and H2O2 from glial cells. Although, cytokines, namely IL-6 and TNF-α, were not involved in this process.121 Additionally, in an animal study, intranasal administration of 23 nm AgNPs resulted in microglial activation, destructing the cerebellum granular layer. This process caused cerebellar ataxia-like symptoms in rats, as well as motor dysfunction and impaired locomotor activity.124 Autophagy significantly affects microglial inflammation and phenotype transformation.125 Shang et al.126 explained that AgNPs promoted M1 polarization and inflammation in microglia in a time- and dose-dependent manner by avoiding the fusion of autophagosomes with lysosomes, thereby altering the lysosomal function and impairing autophagy. This finding provides insights into the molecular mechanisms behind AgNP-induced neurotoxicity.127 Moreover, Huang et al. reported that AgNPs promoted neuroinflammation, oxidative stress, and Aβ deposition in microglia, which was mediated by the secretion of IL-1β, the production of CXCL13 (C-X-C motif chemokine 13), macrophage receptor with collagenous structure (MARCO), and glutathione synthetase (GSS).100 One of the key concerns about AgNPs is their potential to exacerbate neurodegenerative diseases like AD. Aβ deposits cause toxicity to neurons as they cause proinflammatory responses and oxidative stress in the CNS.128 Since AgNPs and Aβ both are taken up by microglia via the scavenger receptor 1 (Scara1), AgNPs may compete with Aβ for uptake, potentially impairing Aβ clearance and worsening AD pathology.129–131 Sikorska et al. also observed that the AgNPs accompanied by cerium oxide nanoparticles (CeO2NPs) reduced microglial phagocytic activity and amyloid-β (Aβ) uptake by BV-2 microglia, which may assist in the AD pathogenesis. AgNPs also attenuated the microglial viability once combined with cadmium telluride quantum dots (CdTe-QDs), favoring the pathogenesis of AD.129
On the contrary, some studies suggest that AgNPs can exhibit anti-inflammatory and neuroprotective properties. For example, AgNPs, in combination with CdTe-QDs or CeO2NPs, even at relatively nontoxic concentrations, could decrease microglial growth by arresting the cell cycle at the G1 phase or S phase, respectively. This suggests a new approach to alleviate neuroinflammation and further disorders.129 Likewise, Lyu et al. confirmed that using AgNPs reduced the number of microglia, supporting their anti-inflammatory role.132 Moreover, citrate-capped AgNPs have demonstrated both anti-inflammatory and antioxidant effects in microglia. These AgNPs were specifically absorbed by microglia and further reduced LPS-stimulated NO, ROS, and TNFα production, leading to less neurotoxicity of microglia for dopaminergic neurons. Also, LDH release, following AgNP treatment, showed a significant reduction, underscoring the role of AgNPs in heightening neuronal cell viability (Fig. 3).133 Furthermore, the inhibitory role of biogenic AgNPs in LPS-induced neuro-inflammation by HMC3 microglial cells was studied. Cotreatment with AgNPs significantly decreased the production of inflammatory markers while increasing anti-inflammatory markers, facilitating a shift from M1 to M2 phenotype in microglia. Therefore, biogenic AgNPs are able to defend CNS against oxidative stress and neuroinflammation.134
![]() | ||
Fig. 3 Anti-inflammatory effects of AgNPs on microglial cells (a and b) N9 microglial cells were treated with LPS (500 ng mL−1) with or without AgNPs for a 1 h pulse period. Then, a 24 h chase period was considered. Microglial inflammation was assessed through evaluation of (a) ROS production and (b) TNFα release following AgNP treatment. (c) N9 microglia were treated for 1 h (pulse) with AgNP (50 μg mL−1) and/or LPS (500 ng mL−1). Then, a 24 hours chase period was considered with only LPS present. The medium was then transferred to N27 neurons and incubated for 48 hours. Afterward, the cell viability was assessed through LDH release examination. Adapted from Gonzalez-Carter et al.133 |
Consequently, the effects of AgNPs on microglia in the context of neurodegenerative diseases remain controversial. While some studies highlight the proinflammatory and neurotoxic potential of AgNPs, others demonstrate their anti-inflammatory and neuroprotective effects. Therefore, AgNPs act as a double-edged sword in neuroinflammation. Further investigation is required to better perceive their mechanisms and ensure their safe and appropriate use to treat CNS disorders.
Iron oxide-based metal NPs are primarily internalized by microglia, mainly through clathrin-mediated endocytosis and macropinocytosis, after which they accumulate in the lysosomal compartment.136 This accumulation in lysosomes plays a crucial role in modulating microglial activity. Interestingly, Wu et al. showed that accumulated IONP attenuated the expression of IL-1β in LPS-stimulated microglia by affecting the secretory lysosomal pathway. In addition, the IL-1β converting enzyme (ICE) was inhibited in microglia following the treatment with IONP by preventing the activity of cathepsin B, an enzyme responsible for IL-1β activation. These findings suggest that IONPs efficiently suppress IL-1β production, highlighting their potential for use in drug-delivery systems that target inflammation.137 Another critical area of investigation is the ability of microglia to alleviate tau pathology in neurodegenerative conditions such as AD. In this regard, Glat et al.138 showed the efficacy of iron oxide (γ-Fe2O3) NPs in delivering fibrin γ377–395 peptides to nervous tissue. Stabilizing the peptide to γ-Fe2O3 NPs significantly decreased the activated microglia in comparison with the same concentration of the free peptides. Therefore, the authors suggested γ-Fe2O3 NPs as suitable carriers for the controlled release of medicine in the CNS. Wang et al.139 also observed that Fe2O3 NPs induced the proliferation of microglia, enhanced phagocytosis, and increased ROS and NO production in microglia. However, the study noted no significant production of inflammatory cytokines, namely, IL-6, IL-1β, and TNF-α, implying that IONPs may boost some microglial functions without causing overt inflammation.
Despite their promising applications, many studies have evidenced the potential detrimental impacts of IONPs on microglia. As reported by Petters et al.,140 IONP exposure triggered ROS production by microglia, causing cellular and tissue damage. Additionally, the IONP accumulation in microglia could lead to changes in microglial morphology and function, potentially disrupting their usual tasks in the brain. Similarly, Luther et al. noted that prolonged exposure to IONPs compromised microglial cell viability, raising concerns about long-term use of these NPs in the brain.136
IONPs offer a noteworthy ability to deliver drugs, particularly in their ability to regulate microglial activity and mitigate neuroinflammation. However, the dual nature of their effects—both as modulators of inflammation and potential toxicity sources—necessitates further investigation. While some studies have shown their efficacy in reducing proinflammatory cytokine production and promoting drug delivery to the CNS, others have indicated possible adverse outcomes, such as the generation of ROS and compromised microglial viability. Consequently, more comprehensive research is required to make sure IONPs are safe and efficient as therapeutic agents for CNS-related conditions.
Recent studies have paved the way for understanding the potential neurotoxic and pro-inflammatory effects of CoNPs. In this way, Li et al. illustrated that CoNPs induced toxicity and inflammatory responses in microglial BV2 cells by activating NADPH oxidase 2 (NOX2). CoNPs in both BV2 cells and mouse brains (the hippocampus and cortex) further catalyzed ROS production and upregulation of IL-1β and NLRP3, which are inflammation-related proteins. Additionally, CoNP exposure was linked to increased tau phosphorylation, which is a hallmark of neurodegenerative diseases.142 Similarly, a survey by Zheng et al. reported that CoNPs were capable of inducing microglial activation, leading to the expression of oxidative stress-related substances NRF2, heme oxygenase-1 (HO-1), and malondialdehyde (MDA) in the hippocampus and cortex of the rat brain.143 These results indicate that CoNPs can induce significant inflammation and oxidative stress in the brain.
While these studies provide initial insights into the impacts of CoNPs, the limited research on this topic underscores the need for further investigations. The potential proinflammatory effects of CoNPs during drug delivery warrant a deeper understanding of their interactions with microglia and their long-term implications for neurodegenerative processes. Continued research is essential to elucidate how CoNPs may contribute to neuroinflammation and cognitive decline, ultimately identifying their safe application in biomedical contexts.
Several studies have shown that ZnONPs trigger apoptosis in the murine microglial cell line N9 by generating ROS and depleting cellular energy,146,147 leading to neuronal damage148 or self-destructive processes.149 Wei et al. indicated that ZnONPs significantly raised ROS levels and oxidative stress in a time-dependent way in BV-2 cells, which occurred through autophagy and PINK1/parkin-mediated mitophagy.150 Moreover, Sharma et al. found that ZnONPs disrupted matrix metalloproteinases (MMPs) and subsequently activated the apoptotic pathway in microglia via NADPH oxidase-independent ROS generation and ATP depletion. This microglial apoptosis exacerbates the existing neuroinflammation.146 Also, it has been reported that ZnONPs induce a nonapoptotic mode of cell death in microglia, which is probably driven by ROS accumulation, leading to lysosomal destabilization and extensive damage to mitochondria and lysosomes. This nonapoptotic cell death can severely damage the brain by accumulating ROS and releasing lysosomal enzymes and cell debris, resulting in severe neuroinflammation.151 ZnONPs have also triggered several inflammatory responses. For instance, a study claimed that ZnONPs activate NF-κB, Ca2+-dependent extracellular signal-regulated kinase (ERK), and p38 pathways in BV2 microglia following tongue instillation.152 Similarly, Liu et al. revealed that even nontoxic concentrations of ZnONPs led to BV2 proliferation and activation through the Akt (protein kinase B) and ERK signaling pathways.153 Another paper studied the acute outcomes of pulmonary exposure to ZnONPs in a rat model. The results indicated that acute exposure to ZnONPs induces microglial activation, tau protein expression, and oxidative stress in the brain, contributing to neurotoxicity.154
Despite the concerns associated with ZnONPs, some studies have reported potential benefits. In this respect, a survey by Moustafa et al. on diabetic patients revealed that luteolin/ZnONPs could regulate microglial polarization by targeting brain CCAAT/enhancer-binding protein (C/EBPA mRNA). These NPs also alleviated inflammation by modulating redox-sensitive signal transduction pathways. Therefore, it was concluded that luteolin/ZnONPs may offer a novel approach to protecting BBB and preventing neurological complications.155
The adverse effects of ZnONPs on microglial neuroinflammation appear to outweigh their potential benefits in promoting brain health. Urgent and comprehensive studies should be conducted to thoroughly investigate the possible positive and negative effects of ZnONPs on microglia-related neuroinflammation. This is crucial to determine a safer dosage with minimal side effects for patients suffering from neurodegenerative disorders.
Rihane et al. evidenced that TiO2NPs predominantly accumulate in BV-2 cells, promoting mitochondrial dysfunction following oxidative stress. These NPs also cause various side effects, such as damaging the permeability of cell membranes, ROS overproduction, and inhibiting cell adhesion with a loss of mitochondrial transmembrane potential, thereby leading to microglia apoptosis.157 Additionally, Sheng et al. highlighted that TiO2NPs contribute to the apoptosis of primary hippocampal neurons and microglia.158 On top of that, recent reports have indicated that low concentrations of TiO2 stimulate BV2 microglia to undergo immediate and prolonged release of ROS, damaging neurons in brain striatum cultures.148,159 Shin et al.160 reported that ultrafine TiO2NPs stimulate the release of inflammatory mediators, encompassing IL-1β, TNF-α, and mRNA in the brains of LPS-exposed mice. These NPs also enhanced NF-κB binding activity in LPS-stimulated BV2 microglia. Therefore, the study suggests that nanosized TiO2NPs promote exaggerated neuroinflammatory responses by activating microglia. In line with this, another study found that LPS-activated BV-2 cells took more TiO2NPs up compared to non-activated cells, leading to increased IL-6, ROS, MCP-1, and IL-1β.161 Along with it, Xue et al.162 showed that TiO2NPs induced significant iNOS expression and subsequent NO secretion, accompanied by upregulation of chemokines through NF-κB activation in NP-stimulated microglia in vitro. This study also indicated raised levels of pro-inflammatory cytokines.
To conclude, TiO2NPs activate microglia and neuroinflammation using various pathways. Given the small number of papers written on TiO2NPs and the scarcity of positive findings, additional research is essential to comprehensively understand the mechanisms in charge of the harmful effects of TiO2NPs on microglia-related neuroinflammation. These findings will facilitate the evolution of strategies for the secure and efficient employment of TiO2NPs in the treatment of neurodegenerative diseases.
SiO2NPs have shown potential in activating microglia. In line with this, in a study, after exposure to fluorescein isothiocyanate-tagged SiO2NPs (FITC-SiO2-NPs),167,168 the number of Iba-1- stained microglia significantly rose in the hippocampus in comparison with the controls.166 Consistently, a study showed that silica-coated magnetic NPs containing rhodamine B isothiocyanate dye (MNPs@SiO2(RITC)) morphologically activated BV2 murine microglia and increased Iba1 expression, an activation marker protein.169 The study also demonstrated that microglia activation elevated serine protein levels in the growth medium. Notably, the secretion of excitotoxic D-serine from primary rat microglia was significantly upregulated, which in turn decreased intracellular ATP and activity of the proteasome in cocultured neuronal cells, particularly in primary cortical neurons. This led to the accumulation of ubiquitinated proteins and the formation of inclusion bodies in cortical and primary dopaminergic neurons cocultured with activated microglia. Thus, the activation of microglia by MNPs@SiO2(RITC) initiates excitotoxicity in neurons through the secretion of D-serine, underscoring the neurotoxic processes triggered by microglial activation.156 SiO2NPs are likely to produce inflammatory agents and cause neuroinflammation. In this way, Xue et al. demonstrated that SiO2NPs enhance the proinflammatory cytokines (TNF-α, IL-1β, and IL-6).162 Additionally, the findings of another study illustrated that deficient SiNP levels could change microglial function. In turn, alteration in proinflammatory genes, cytokine release, and heightened RNS and ROS production adversely affect not only microglial function but also surrounding neurons.170 Correspondingly, MNPs@SiO2(RITC) were shown by another study to enhance ROS production in a dose-dependent manner.171 The findings of another research indicated that the viability of MNPs@SiO2(RITC) was gradually reduced with increasing SiO2NP concentration and exposure duration. The findings indicated that SiNPs could penetrate the cytoplasm, alter the ultrastructure, activate the NLRP3 inflammasome, release a multitude of inflammatory molecules, and start inflammatory reactions. SiNPs were also discovered to induce ferroptosis, increase intracellular ferrous ion levels, and disrupt cellular antioxidant function.170
In summary, the studies reviewed suggest that SiO2NPs possess the potential to induce microglial activation and neuroinflammation, negatively impacting neuronal health. Due to the scarcity of studies on the positive influences of these NPs on microglia-related neuroinflammation, the potential of SiO2NPs for treating neuroinflammation deserves further research to fully elucidate the risks of exploiting this nanoparticle.
Regarding the explained studies, Fig. 4 illustrates the advantages and disadvantages of each metal NP in the inhibition of microglia-mediated neuroinflammation.
![]() | ||
Fig. 4 The advantages and disadvantages of drug-loaded metal NPs in microglia-induced neuroinflammation. |
By addressing these challenges and exploring these prospective directions, nanoparticle-based therapies could revolutionize the treatment of neuroinflammation and related CNS disorders.
CNS | Central nervous system |
AuNP | Gold nanoparticle |
AgNP | Silver nanoparticle |
IONP | Iron oxide nanoparticle |
SiO2NP | Silica nanoparticle |
ZnONP | Zinc oxide nanoparticle |
CoNP | Cobalt nanoparticle |
TiO2NP | Titanium oxide nanoparticle |
TNF-α | Tumour necrosis factor alpha |
DAMPs | Danger-associated molecular patterns |
PGE2 | Prostaglandin E2 |
PAMPs | Pathogen-associated molecular patterns |
CR and CR | Complement receptors |
CD | Cluster of differentiation |
IL | Interleukin |
ROS | Reactive oxygen species |
TLR and TLR | Toll-like receptors |
BACE | Beta-secretase enzyme |
iNOS | Inducible nitric oxide synthase |
COX2 | Cyclooxygenase-2 |
BBB | Blood–brain-barrier |
ECM | Extracellular matrix |
NPs | Nanoparticles |
TGN | Trans-Golgi network |
Iba-1 | Ionized calcium-binding adapter molecule |
ALTs | Astrocyte-like |
N2a | Neuro2a |
NF-κB | Nuclear factor-κB |
SPIONPs | Superparamagnetic iron oxide nanoparticles |
MPI | Magnetic particle imaging |
DHLA | Dihydrolipoic acid |
FIB | Fibrinogen |
BS | Bovine serum |
PC | Protein corona |
JAK/STAT | Janus kinase/signal transducers and activators of transcription |
PEGylated AuNPs | polyethyleneglycol-coupled AuNPs |
PM-AuNPs | Paeonia moutan to functionalize GNPs |
CeO2NPs | Cerium oxide nanoparticles |
Aβ | Amyloid-β |
GSS | Glutathione synthetase |
AD | Alzheimer's disease |
MMP | Matrix metalloproteinases |
AnPEG-GNPs | Anthocyanins conjugated with PEG-GNPs |
MAPKs | Mitogen-activated protein kinases |
MARCO | Macrophage receptor with collagenous structure |
FITC | Fluorescein isothiocyanate |
ERK | Extracellular signal-regulated kinase |
Footnotes |
† Should be considered joint author. |
‡ Current affiliation: The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA Phone (Fax): +982833790620-59 (cell). |
This journal is © The Royal Society of Chemistry 2025 |