Abstract
Effective and safe pharmacotherapies for central nervous system (CNS) disorders remain a major obstacle to human health worldwide. Nanotechnology offers promise in addressing this challenge by enabling the transport of large molecules across the blood–brain barrier (BBB) and the delivery of multiple drugs. Numerous studies have demonstrated the efficacy of nanodrugs in animal models of various CNS disorders, including Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, amyotrophic lateral sclerosis, stroke, lysosomal storage disease, and gliomas. Nanoparticles (NPs), tailor-made to enhance drug enrichment locally and promote extended drug release, can prevent neuronal death, decrease neurotoxicity, and inhibit tumor growth. In addition to drug delivery, gene therapy using nanotechnology has emerged as a potentially curative option for neurodegenerative diseases. In this review, we summarize how NPs have successfully crossed the BBB and specifically targeted different cells in various CNS disease models over the past decade. Although nanotechnology holds great promise, several drawbacks and challenges must be addressed before its effective implementation in the treatment of CNS diseases.
1 Introduction
Central nervous system (CNS) diseases comprise a diverse range of traumatic neurological disorders including Alzheimer’s disease (AD) [1], Parkinson’s disease (PD) [2,3], multiple sclerosis (MS) [4], amyotrophic lateral sclerosis (ALS) [5], epilepsy [6], and brain tumors. These conditions are characterized by progressive degeneration of the central or peripheral nervous system, widespread neuronal loss, and inflammation [7,8,9,10]. Among the malignant primary brain tumors, glioblastomas, astrocytomas, and oligodendrogliomas are the most common types of gliomas, representing approximately 20% of all malignant gliomas [11] and characterized by nuclear atypia, increased proliferation activity, and microvessel necrosis, depending on the tumor grade (I–IV) [12]. Despite the FDA’s approval of several new treatments in 2017 and 2022, such as Aducanumab for AD, Epidiolex for Dravet and Lennox-Gastaut syndromes, Inebilizumab for neuromyelitis optica spectrum disorder, Brexanolone for postpartum depression, and Esketamine for treatment-resistant depression (ClinicalTrials.gov), CNS diseases remain challenging to treat, and the currently available treatments mainly relieve symptoms. Additionally, the pharmacological kinetics and targeting mechanisms of approved therapeutics are not well understood [13,14,15].
Nanoparticles (NPs), a type of material that includes particulate substances, are typically defined to be between 1 and 100 nm in size [16]. Due to their ability to be readily taken up by cells, NPs are extensively used for targeting different types of cells in disease therapy [17]. These NPs are either used alone (e.g., exosomes and liposomes) or as part of a high-loaded drug delivery system (e.g., polymeric NPs and solid–lipid NPs) and have been employed as nanomedicines for various human diseases [18]. Recently, nanotechnology has emerged as a promising option for improving the diagnosis and treatment of CNS diseases by facilitating drug transport across the blood–brain barrier (BBB) and targeting specific cell types [19,20,21]. Polymer- and lipid-based NPs have shown great potential for clinical use [22,23,24].
Several studies have explored the progress of nanotechnology in treating CNS diseases from various perspectives, including the types of nanomaterials [25] and CNS diseases [26,27]. Notably, one of the advantages of NPs is their ability to be customized for targeting specific cell types based on their unique requirements. However, there have been few reviews summarizing this topic in the past decade. In this review, we aim to assess the latest advancements and categorize the applications of nanotechnology in the treatment of neurodegenerative and other CNS diseases, including gliomas. To fully leverage the potential of nanomedicine for treating CNS-related diseases, we provide a summary of various types of NPs that target specific cell types, such as neurons, microglia, astrocytes, epithelial cells, and oligodendrocytes, as well as the use of nanotechnology in gene therapy for CNS diseases.
2 Pathways of NPs across the BBB
BBB is a crucial interface of capillary endothelial cells that carefully regulates the transfer of substances between the periphery and the brain, safeguarding the brain from circulating pathogens [28]. BBB is composed of various components, including tight junctions between endothelial cells, pericytes, astrocyte endfeet, microglia, interneurons, basement membranes, and extracellular matrix (ECM) proteins, which collaborate to maintain a semipermeable border (Figure 1). Tight junctions, barrier permeability, and endothelial expression of receptors and transporters help to maintain endothelial cell function [28–30]. Molecules can cross the BBB mainly through receptor-mediated transcytosis and adsorptive-mediated transcytosis facilitated by these receptors and transporters. Astrocytes are glial cells that connect brain capillaries and neurons. The basement membrane encircles the endothelial cells and pericytes and contains various ECM proteins such as heparan sulfate proteoglycans, laminin, and collagen type IV [31,32].

BBB structure and NP transport pathways. TfR1, transferrin receptor 1; IR, insulin receptor; LRP, LDLR-related protein; LfR, lactoferrin receptor; LDLR, low-density lipoprotein receptor; GLUT1, glucose transporter-1; ACST2, amino acid transporter; PLGA/PLA, poly(lactic-co-glycolic acid)/polylactic acid; PBCA, poly(butylcyanoacrylate); CDs, carbon dots.
The highly selective semi-permeable border poses a challenge for the transport of certain drugs to the brain, hindering their effectiveness in treating CNS diseases. In recent years, several new strategies have emerged to overcome this barrier [33]. Invasive approaches, such as intracerebroventricular infusion, have been used, but non-invasive therapies such as oral, peripheral, or intranasal administration are preferred [34]. While most drugs diffuse throughout the brain after crossing the BBB, they often do not accumulate at a specific site [35]. Apomorphine (AMP), a drug employed to treat PD and other movement disorders [36], is believed to encounter blockage or degradation before reaching the intended target site [37]. In contrast, NPs can deliver drugs to a specific site, protecting their functional groups and preventing premature drug release [7,8,33,38]. Organic NPs, such as liposomes and polymeric NPs, are biocompatible and biodegradable, and can target cell-specific ligands, crossing the BBB mainly through receptor-mediated transcytosis or carrier-mediated transport. In contrast, inorganic NPs use transcellular or paracellular transport to cross the BBB (Figure 1) [39,40]. NPs can also be modified with specific ligands to leverage transporters in the BBB, such as glucose transporter 1 [41] and amino acid transporter [42]. The favorable properties of organic NPs make them an effective and non-invasive drug delivery tool for the treatment of CNS diseases [43]. Furthermore, NPs can be used wirelessly to stimulate nerve cells in the deep brain [44,45]. Overall, the emergence of NPs as a drug delivery tool offers a promising solution to the challenge of transporting drugs across the BBB to treat CNS diseases effectively.
Effective drug delivery to the brain remains challenging due to several factors. Although researchers have identified potential targets for modifying the surface of NPs, the protein corona can mask functional ligands and prevent NPs from crossing the BBB and being cleared [46,47]. Ligand-modified NPs may also become trapped within brain capillary endothelial cells due to the high binding affinity of ligands with receptors, reducing the amount of NPs that cross the BBB [48,49], and increasing their clearance by microglia before reaching the target site [50]. Additionally, the efflux mechanisms of drug metabolites and waste products remain unknown [51–53].
3 Nano-strategies targeting different cell types
Although some drugs can cross the BBB owing to their favorable properties or the use of nano-delivery systems, their effectiveness in treating CNS diseases has been unsatisfactory. In light of the varying roles played by different cell types and regions in the progression of these diseases, recent therapeutic strategies have focused on the development of nanomedicines that target specific cell populations in the CNS. In this article, we provide a brief overview of the cells that are of interest in the design of nanodrugs based on their physiological functions (Figure 2).

Specific ligands for cell-type targeting in NP designation. Ab: antibody; Apo-E: apolipoprotein E; B2R: bradykinin receptor B2; CD11b: integrin alpha-M; CR: complement receptor; CD204: cluster of differentiation 204; CD36: cluster of differentiation 36; ECM: extracellular matrix; g7: a glycosylated 7-amino acid-long peptide; RGD: arginyl-glycyl-aspartic acid; RVG: rabies viral glycoprotein; LRP1: LDL receptor related protein 1; SSTR2: somatostatin receptor 2; Tet1: Tet methylcytosine dioxygenase 1; Tf: transferrin; TLR: toll-like receptor; TrkA: tyrosine kinase A; Ts1: tityusserrulatus (a neurotoxin binding voltage-gated sodium channel).
3.1 Neurons
Functional neurons make up only approximately 10% of the cells in the brain, but they are critical therapeutic targets as they are often impaired in many types of brain pathology. Manipulating the growth and activity of neurons is a promising strategy for treating patients with neurodegenerative diseases or nerve damage resulting from trauma. However, targeting disease-related neurons in a clinical setting is challenging due to their non-phagocytic nature and diversity of neuron types [54].
One approach involves leveraging the intrinsic ability of vehicles to interact with neurons. For example, apolipoprotein E (ApoE) is the primary lipoprotein produced by astrocytes in the brain, and it delivers essential lipids to neurons by binding to the cell membrane. Wang et al. conjugated ApoE to a lysosomal enzyme, α-l-iduronidase, to treat lysosomal storage disease (LSD), and they observed that this complex accumulated in neurons and astrocytes [55]. After long-term treatment, 2–3% of normal brain α-l-iduronidase activities were recovered from the mouse models of mucopolysaccharidosis type I [55]. Under inflammatory conditions, monocytes and macrophages secrete exosomes that can cross the BBB [56]. In addition to negative targeting approaches, NPs can be designed to recognize neurons based on potential ligands such as dopamine [57], Tet1 peptide (binds to gangliosides and sphingophospholipids, specifically expressed in neuronal cells) [58–60], tyrosine kinase A [61], lactoferrin, rabies virus glycoprotein (RVG)-29 peptide [62], Ts1 (a neurotoxin binding voltage-gated sodium channel) [63,64], antisauvagine-30 [65], and arginyl-glycyl-aspartic acid peptide [66,67]. Pathological neurons also provide targets, such as the hydroxyl-terminated generation of four polyamidoamine (PAMAM) dendrimers, which accumulate specifically in injured neurons [68].
3.2 Microglia
Microglia are long-surviving, self-renewing innate immune cells in the CNS that play crucial roles in maintaining a healthy microenvironment that enables adequate neuronal activity by clearing debris and invading pathogens. However, overactivated microglia can exacerbate the progression of most CNS diseases by inducing neurotoxicity and inflammation [69–71]. Recently, NPs have been used to specifically downregulate the immune state via gene editing [72–74]. In addition, compounds that target microglial inflammasomes, such as rosiglitazone [75], MCC950 [76], Sargramostim, and Fingolimod [77], can be delivered using nanocarriers to prevent dopaminergic (DA) neuron loss in the SNC in PD. Thus, NPs offer promising potential for re-normalizing microglia in CNS disorders [78].
Microglia are crucial players in the pathogenesis of several CNS diseases including MS [79]. Current treatments for MS are not curative [80–82]; therefore, nanomedicines targeting microglia have emerged as promising therapeutic options [83]. For example, novel biomimetic Cu2−x Se-PVP-Qe NPs have been coated with neural cell membranes to specifically target microglia and modulate their polarization into an anti-inflammatory M2-like phenotype to relieve neuroinflammation [84]. The 2.8 ± 0.2 nm sized Cu2−x Se-PVP-Qe NPs specifically recognized microglial molecule-1 and α4β1 integrin, enhanced the expression of the M2-like biomarker CD206, and restored the dopamine level in the cerebrospinal fluid (CSF) and tyrosine hydroxylase (TH) back to the normal level [84]. Curcumin-loaded mPEG-b-PLA NPs effectively reduced oxidative stress in endothelial cells and inhibited M1-microglial activation in a mouse model of ischemia/reperfusion injury [85]. Similarly, dihydrolipoic acid-containing gold NPs changed microglia from polarized M1 microglia to M2 microglia in an ex vivo brain slice stroke model [86]. Despite their phagocytic nature, specific targeting ligands (TLR2, TLR4, CR3, CR4, CD36, and CD204) have also been modified to enhance microglial recognition [87].
Although microglial targeting holds immense potential for treating CNS-related diseases, several challenges still need to be addressed.
Owing to its phagocytic and digestive nature, how can lysosomal degradation be avoided to ensure drug utilization?
How can pathological or activated microglia be targeted?
Like a double-edged sword, NPs themselves (like metal-containing NPs) are also one of the potential factors that cause an immune response in the CNS [88].
How can a delivery system targeting other cell types be designed to prevent microglial clearance?
3.3 Astrocytes
Astrocytes are specialized glial cells that provide support to neurons and outnumber neurons over fivefold [89]. They are also critical components of the BBB, contributing to tight junctions between specialized endothelial cells by secreting soluble factors and enveloping the junctions with their endfeet [90,91]. Notably, astrogliosis is a reliable and sensitive marker of the diseased CNS [92], making targeting astrocytes an increasingly attractive approach for treating diseases.
Several strategies have been employed to specifically target astrocytes using nanomedicine. For example, apolipoprotein E was modified on the surface of lipid NPs for delivering mRNA via intracranial injection [93]. Chitosan NPs containing small interfering RNA (siRNA) were also modified with transferrin receptor (TfR) and bradykinin B2 receptor (B2R) antibodies to specifically downregulate gene expression in astrocytes [94]. In another study, Vismara et al. grafted primary amines to the surface of rolipram-containing nanogel (NG)-based delivery systems, which specifically modulated A1 astrocytes to downregulate their inflammatory toxic effects in a mouse model of spinal cord injury [95]. By co-culturing with primary neurons, astrocytes, and microglia from the spinal cords of mouse embryos, they found that NG accumulated in astrocytes much more than in microglia following exposure to the cells for 24 h [95]. However, it should be noted that astrocytes do not always protect neurons. In a study by Hawkins et al., maternal exposure to cobalt and chromium NPs resulted in neuronal DNA damage via astrocytes [96]. In summary, nanomedicine holds great promise in normalizing pathological astrocytes in CNS diseases.
In addition to their supportive functions for neurons, astrocytes also play a crucial role in protecting against metal-related toxicity by absorbing metal NPs such as iron, copper, and silver NPs [97,98]. However, exposure to titanium dioxide NPs has been found to cause mitochondrial injury in primary astrocytes [99]. Conversely, certain types of NPs, such as selenium NPs, have been shown to exert a protective effect on primary cortical neurons and astrocytes during oxygen-glucose deprivation and reoxygenation [100]. Furthermore, in various CNS disorders, astrocytes have been targeted to reduce neuronal inflammation and oxidative stress [101].
3.4 The basal membrane: endothelial cells and pericytes
The vascular basement membrane, predominantly composed of brain capillary endothelial cells, plays a crucial role in maintaining BBB integrity [102]. Pericytes embedded in the basement membrane and astrocyte endfeet provide support to the endothelial cells. Under neuropathological conditions, changes in the molecular composition of the endothelial cell–cell junctions can result in the loss of BBB integrity [103].
The endothelium represents the first obstacle to be overcome by NPs attempting to cross the BBB via active pathways. To improve drug delivery efficiency and minimize off-target effects in other organs, NPs can be designed to target brain endothelial cells by exploiting the differential endocytic rates of brain and peripheral endothelium [104]. However, a significant proportion of NPs taken up by endothelial cells are degraded by lysosomes, leading to both a failure in the escape of these cells and cytotoxicity in the endothelium [105]. Rassu et al. demonstrated that chitosan-coated or -uncoated solid lipid NPs are a viable option for the penetration of siRNA across epithelial cells following nasal administration [106]. Khan et al. modified NPs with an MMP-1-sensitive fusion peptide, HER2-targeting K, and low-density lipoprotein receptor-related protein-1 (LRP1)-targeting angiopep-2, and demonstrated their ability to escape the BBB [107]. Liposomes have been shown to remain intact inside lysosomes, enabling their escape from endothelial cells [108]. Saturation of endolysosomes by large amounts of Tf-modified liposomes can facilitate the passage of remaining NPs through the BBB and their subsequent release into the brain parenchyma [109]. Gold NPs injected into the mouse CSF have been observed to accumulate in the basement membrane on the outer side of cortical arteries, highlighting the basement membrane as a potential pathway for NPs to exit the brain [110]. However, other studies have demonstrated that Au NPs can impair tight junctions and increase BBB permeability in mice [111].
The toxicity of NPs to endothelial cells is another crucial concern that must be addressed. Studies have shown that concentrated non-toxic engineered carbon and iron oxide NP can cause oxidative/nitrosative stress, energy metabolism imbalance, and mitochondrial dysfunction in endothelial cells [112,113]. Additionally, silica NPs were found to induce oxidative stress through the JNK/P53 and NF-κB pathways in endothelial cells [114]. Although the uptake of NPs by endothelial cells is inevitable, they activate the cells [115]. The effects on endothelial cells are determined by factors such as size, shape, surface ligands, and charge [116].
Pericytes, which are highly expressed in small blood vessels, play a crucial role in maintaining the integrity of the BBB, increasing trans-endothelial electrical resistance, inhibiting immune cell trafficking, and regulating the transcytosis of endothelial cells [117]. However, pericytes are also vulnerable to damage [118]. Another study reported a reduction in the number of astrocytes and degeneration of pericytes in a diabetic mouse model [119,120]. The depletion of pericytes (e.g., PDGFRbC+/−) can lead to impaired BBB functionality [117]. Although an approach to activate or inhibit pericyte function has enormous potential to help treat cellular pathologies in CNS diseases [121], it is difficult to target pericytes using NPs due to the lack of specific markers. In contrast, targeting the basal membrane is comparatively easier, but crossing the BBB to increase drug delivery efficiency using NPs without damaging the BBB remains a challenge.
3.5 Oligodendrocytes and oligodendrocyte precursor cells
Oligodendrocytes are highly vulnerable cells in the CNS that undergo a precise program of proliferation, migration, differentiation, and myelination [122]. Once damaged, they are difficult to repair. This is particularly evident in age-related demyelinating diseases such as MS, where remyelination is challenging [123]. Rittchen et al. developed poly(lactic-co-glycolic acid)-based NPs, modified with NG-2 chondroitin sulfate proteoglycan antibody, to deliver leukemia inhibitory factor to oligodendrocyte precursor cells, successfully increasing myelin repair in a model of focal CNS demyelination [124]. In this study, oligodendrocyte-targeting NPs were able to increase both the number of myelinated axons and the thickness of myelin per axon [124]. Similarly, chitosan NPs were shown to deliver LIN-GO-1-directed siRNA to the brain, promoting central remyelination in a rat model of demyelination [125]. Retinoic acid-containing NFL-lipid NPs (RA–NFL–LNC) were reported to promote the differentiation of primary neural stem cells toward oligodendrocytes [126]. This was demonstrated by the results showing that the Oligo2+ cell numbers in the brain sections from the mice treated with RA–NFL–LNC after lysolecithin-induced focal lesions were more than three times that of the mice treated with vehicle, indicating oligodendrocyte repopulation by RA–NFL–LNC [126]. Additionally, a type of extracellular vesicle containing MiR-219a-5p showed better efficacy in OPC differentiation and experimental autoimmune encephalomyelitis (EAE) cure than liposomes and polymeric NPs [127]. Li et al. designed DNA NPs that could activate transit transcription, promoting the differentiation of human pluripotent stem cells toward oligodendrocytes [128]. However, due to the vulnerability of oligodendrocytes, researchers must exercise caution in ensuring NPs’ safety. Jenkins et al. demonstrated that “stealth” PEG-modified NPs could evade major brain cells, including oligodendrocytes [129]. Sruthi et al. also demonstrated that functionalized super-paramagnetic iron oxide NPs had no toxicological or morphological effect on oligodendrocytes, with no oxidative stress or inflammatory response to the brain [130]. In summary, the design of NPs for targeting oligodendrocytes and oligodendrocyte precursor cells is still a long way because of their vulnerability and potential adverse effects.
3.6 Brain microenvironment
In addition to the BBB, the brain microenvironment poses another challenge for drug delivery to the CNS [131]. It comprises a porous and electrostatically charged brain ECM, soluble molecules, and other components that can influence drug diffusion into the brain. Furthermore, the surface properties of NPs can affect cellular tropism in the brain. Stealth NPs were found not to be taken up by any of the cell types, remaining in the extracellular space in the brain, unless they were functionalized with bio-adhesive end-groups [132].
The brain microenvironment is also affected during brain injury, including the compromised BBB, reactive astrocytes, activated microglia, and necrotic/degenerating neurons [133]. A reactive oxygen species (ROS)-responsive polymeric micelle system has been reported to remodel microglia and correct the inflammatory status in the early stages of AD [134]. In another study, Shi et al. developed a novel biomimetic decoy-integrated NP for managing the over-activated brain microenvironment in ischemic stroke mouse models. The multifunctional NPs featured a shell composed of CXCR4-expressed mesenchymal stem cells membrane, promoting their accumulation in cerebral ischemic lesions and disrupting the infiltration of neutrophils and macrophages. Additionally, the NPs absorbed and neutralized CXCL12, preventing the polarization of microglia. The cargos carried by the NPs, such as A151, effectively induced the polarization of microglia towards the M2 phenotype. This innovative approach demonstrates promising potential for precise immune modulation in the context of ischemic stroke, offering a targeted strategy for therapeutic intervention [135]. The tumor microenvironment in the brain is similar to that in the periphery, consisting of neighboring cells, molecules, and vascular and lymphatic networks. Wang et al. used Pep-1 and CREKA peptides to modify paclitaxel-containing NPs to enhance their ability to cross the BBB, penetrate into the parenchymal cell, and accumulate in the fibrin–fibronectin complex-rich microenvironment in glioblastoma [136]. Although researchers have begun to evaluate the brain microenvironment, there are still few biomarkers or proven nanomedicines available to target this complex system.
The fate of NPs after crossing the BBB and being taken up by neuronal cells is a crucial area of study that has been largely neglected. Recent evidence suggests that NPs may be transported between different cells within the brain, independent of their surface properties, following uptake by neurons or glial cells [137]. Such cell-to-cell transport could have significant implications for the distribution and accumulation of NPs within the brain, as well as their potential toxicity and therapeutic efficacy. Further research is needed to fully understand the mechanisms of cell-to-cell transport and their impact on NP-based drug delivery to the CNS.
4 Nanotechnology targeting CNS diseases
Neurodegenerative diseases are characterized by progressive dysfunction and loss of neurons in the CNS [3,138], including PD, AD, MS, and ALS. Chronic immune activation is another common characteristic of neurodegenerative diseases, in which reactive microglial cells play a major role [139–144]. Thus, cell type-specific targeting is crucial for treating CNS diseases, and NPs can be designed to achieve this goal [38,145,146] (as shown in Figure 3). Various types of NPs have been studied to improve drug delivery to the neurons. Table 1 summarizes the different types of NPs applied to neurodegenerative diseases based on their target cell types.

Cell-type NPs for treating neurodegenerative diseases.
Neurons and microglia targeting NPs for treating CNS diseases
Targeting cell | Targeting markers | NP type | Surface modification | Delivered compounds | BBB transport pathway | Administration | Encephalic region | Disease | Bio-safety | Animal | Year | Ref |
---|---|---|---|---|---|---|---|---|---|---|---|---|
Neuron or microglia | — | Ag NPs | — | — | — | Nose inhalation | Cerebrum and cerebellum | Neurotoxicity and immunotoxicity | C57BL/6 mice | 2010 | [249] | |
Neuron | — | Ferulic acid (FA) and glycol chitosan | — | Chitosan and FA | Through the ruptured blood capillaries and interrupted brain-spinal cord barriers | Intravenously | Gray matter and white matter | Spinal cord contusion injury | Neuroprotective | Long-Evans rats | 2014 | [250] |
Neuron | Nicotinic acetylcholine receptor | mPEG-PLGA | RVG peptide | DFO | Possibly via nAchR-mediated | Intravenously | Substantia nigra and striatum | PD | Biocompatible of RNP-DFO in brain and other organs | C57BL/6 mice | 2018 | [62) |
— | — | Fe3O4-NPs, 30 nm | — | — | — | Intranasally instilled | Hippocampus | Potential oxidative damage in the striata | SD rats | 2013 | [251) | |
Ischemic neuron | Glutamate receptor | A dextran polymer core modified with ROS-responsive boronic ester and a red blood cell membrane shell | Stroke homing peptide | NR2B9C | Stroke homing peptide-mediated transcytosis | Intravenous injection | Ischemic brain region | Middle cerebral artery occlusion | BBB | Rats | 2018 | [225] |
Neuron | Transferrin receptor | Magnetic NPs | Transferrin, FITC | — | Receptor-mediated transcytosis | Perfusion into the internal carotid artery (ICA) | Corpus striatum, hippocampus, thalamus, substantia nigra, mesencephalon, and cerebellum | — | — | Wistar rats | 2013 | [252] |
Neuron | — | Au NPs, 5 nm | Positively charged | — | Olfactory pathway | Aerosol, locust antenna exposure | Whole brain | — | No significant alterations in their spontaneous and odor- evoked spiking properties | Locust | 2017 | [253] |
Neuron | — | RVG-TP peptide NPs | RVG and transportation peptide | CASP3 siRNA | Via vicinity of areas with a compromised BBB | Tail intravenous injection | Injured hemisphere | Traumatic brain injuries | — | C57BL/6 mice | 2016 | [254] |
Retinal neuron | — | P3HT NPs | Thiophene | — | — | Subretinal injection | Entire subretinal space | Retinitis pigmentosa, and age-related macular degeneration | — | Rat | 2020 | [255] |
Dopamine neuron | — | Electromagnetized Au NPs | — | — | — | Implant | Striatum | PD | Mice | 2017 | [256] | |
Blood-borne inflammatory monocytes | — | Carboxylated PLG NPs, 500 nm | — | — | — | Intravenous infusion | Injury site | Spinal cord injury | No toxicity found | C57BL/6 mice | 2017 | [257] |
Microglia | — | PLGA-PEG NPs | CD47 extracellular domain, BBB-penetrating peptide CRTIGPSVC (CRT) | Nec-1s | Transferrin receptor-mediated transcytosis | Intravenous injection | Whole brain | AD | — | C57B6/J mice | 2020 | [258] |
Neuron and microglia | Amyloid | Superparamagnetic iron oxide NPs | Amyloid Ab | — | — | Intravenous injection | Amyloid plaques | AD | — | Mice | 2017 | [259] |
Microglia | Ionized calcium binding adaptor molecule 1 (Iba1) | FePt-NPs | Iba1 Ab | — | — | Intravenous injection | Amyloid plaques | AD | — | Mice | 2017 | [260] |
Microglia | — | Silica NPs | Gadolinium or tetramethyl rhodamine iso-thiocyanate | — | — | Intravenous injection | Tumor | Glioma | — | Nude mice | 2011 | [260] |
Microglia | Scavenger receptor class A | Iron oxide NPs | Sulfated dextran | — | — | Intravenous injection | Inflammation region | Cerebral inflammation | Biocompatible to microglia | BALB/c mice | 2018 | [261] |
Microglia | CX3CR1 | PLGA NPs | CX3CR1 siRNA | — | Intrathecal injection | Injection sites | Spinal nerve ligation | — | Rat | 2020 | [262] | |
Microglia | Iba1 | Superparamagnetic iron–platinum (FePt) NPs | Iba1 Ab | — | — | Intravenous injection | Infarcted region in the ischemic hemisphere | Cerebral ischemia | — | Rat | 2020 | [74] |
Microglia | — | Curdlan NPs | — | NF-κB siRNA or p65 siRNA | Via vicinity of areas with acompromised BBB | Intravenous injection | Stroke lesion | Stroke | — | Balb/c mice | 2020 | [263] |
4.1 PD: DA neurons
PD is a common multisystem neurodegenerative disorder characterized by progressive degeneration of DA neurons in the substantia nigra and striatum [2,147]. The severity of motor and non-motor symptoms is directly correlated with the extent of DA neuron loss [148]. In addition to DA loss, PD is characterized by the accumulation of α-synuclein and Lewy bodies in the neurons [148,149]. Although levodopa preparations, dopamine agonists, and monoamine oxidase-B inhibitors are the primary medical therapies [150,151], more effective treatments are needed to improve PD therapy.
Nanotechnology has emerged as a promising strategy for delivering dopamine agonists or α-synuclein inhibitors to neurons or the microenvironment in the CNS of animal models of PD. For instance, one approach involves implanting an intracranial nano-enabled scaffold device that carries DA and releases it consistently in the parenchyma of the frontal lobe [152]. Another approach uses polyethyleneimine polymeric NPs (PEG-NPs) to carry RA to DA neurons by inducing the expression of Nurr1 and Pitx3 in 1-methyl-4-phenyl-1,2,3,6 tetrahydropyridine (MPTP)-induced PD mice following intrastriatal stereotaxic injection [153]. PEG-NPs have also been reported to enhance the penetration of curcumin across the BBB and prolong its retention time in the cerebral cortex and hippocampus [154]. The curcumin-loaded NPs were demonstrated to prolong t 1/2 of curcumin in the cerebral cortex to 19.9 ± 2.63 min and in the hippocampus to 16.7 ± 2.56 min [154]. Additionally, PEG-NPs have been used to protect AMP from oxidation before accumulation in the brain via intranasal delivery [155]. Interestingly, acidic PLGA-NPs have been found to rescue compromised lysosomes and protect DA neurons via intraperitoneal injection in healthy mice [156]. Similarly, chitosan NPs encapsulating pramipexole and lactoferrin NPs loaded with rotigotine have successfully preserved DA levels and increased functioning and activity in PD animal models following intranasal administration [157,158]. Polysorbate (PS) 80 cerasome NPs encapsulating curcumin have shown similar abilities to recover DA neurons in a PD mouse model following intravenous administration [159]. Furthermore, RVG 29-modified PLGA NPs (RVG-NPs) have successfully delivered an iron chelator, deferrioxamine (DFO), to the stratium and substantia nigra in PD mice and rescued DA neuronal damage [62]. RVG-NPs were demonstrated to target NeuN+ neurons in the stratum and TH+ neurons in the substantia nigra, decrease their cellular iron levels by delivering DFO, and rescue TH levels in MPTP mice [62].
However, some NPs have been developed to target microglia, with the aim of downregulating inflammation and treating PD [160]. Overall, nanotechnology provides promising tools for more effective and precise treatment of PD.
4.2 AD: neurons and microglia
AD is the most prevalent neurodegenerative disease among the elderly and is characterized by progressive cognitive and memory decline, functional impairment, and neuropsychiatric symptoms [161–163]. Key pathological features of AD include the accumulation of extracellular amyloid plaques and intraneuronal hyperphosphorylated tau proteins [164,165]. In recent years, nanotechnology has been used to eliminate these pathological hallmarks in AD animal models.
Most treatments, including nanotechnology, focus on eliminating amyloid beta (Aβ) [1,15]. Intracerebral injection of hydrogen-containing NPs has been shown to restore learning properties, synaptic function, and neuronal death [166]. These NPs prolonged the release of hydrogen, reducing Aβ aggregation and oxidative stress by inducing the antioxidative pathway and improving mitochondrial dysfunction in AD mice [166]. To develop milder treatments, PLGA NPs encapsulating vitamin D-binding protein (DBP) were intravenously administered to 5XFAD transgenic AD mice, resulting in decreased Aβ plaque formation and AD pathology [167]. Chitosan-l-valine NPs were also used to deliver the poorly-permeable drug saxagliptin throughout the brain by intraperitoneal delivery [168]. Recently, a novel nanocomposite was developed to combine Aβ aggregates on the surface to form nanoclusters following intravenous tail injection in AD mice. Aβ nanoclusters are then phagocytosed and digested by microglia, thereby mitigating Aβ-induced neurotoxicity [154]. In another study, anti-Aβ1-42-functionalized NPs were intravenously injected into AD mice to clear brain Aβ, leading to significant correction of memory defects. However, the poor penetration of anti-Aβ NPs into the brain suggests that the treatment mainly clears peripheral Aβ [169]. In brief, nanodrugs target the Aβ pathway mainly by reducing the continuous synthesis and clearing the abnormal aggregation of Aβ in animal models of AD.
Recent studies have also aimed to target the tau pathway, which is closely associated with the symptoms of AD. One promising approach involves designing a methylene blue-loaded NP (CeNC/IONC/MSN-T807), which was injected stereotactically and demonstrated the ability to either bind to hyperphosphorylated tau or inhibit other tau-associated pathways [170]. The nanocomposite was shown to target Tau+ neurons, downregulate p396-Tau and p205-Tau, decrease the Tau filaments in TEM images, and rescue other cell populations, such as Iba-1+ microglia and GFAP+ astrocytes in the brains of AD mice [170]. Another study in 2019 reported on a novel mesoporous nano-selenium (MSe) multifunctional delivery system (MSe-Res/Fc-β-CD/Bor) which had borneol (Bor) conjugated to its surface to enable it to cross the BBB, and β-cyclodextrinnanovalves (Fc-β-CD) were added as a “lock” to control the opening of specific sites by redox to release resveratrol [171]. Intravenous administration of MSe-Res/Fc-β-CD/Bor was found to protect nerve cells and improve memory by reducing oxidative stress, inhibiting Aβ aggregation, and suppressing tau hyperphosphorylation [171].
Noninvasive strategies, such as oral administration, are preferred over invasive approaches when treating patients. Memantine hydrochloride (MEM) encapsulated in PLGA-PEG-NPs has been reported to increase its efficacy and drug release profile while decreasing its required dose through oral administration in transgenic AD mice [172]. The MEM-PLGA-PEG NPs can cross the BBB, reach the hippocampus, and significantly improve functionality compared to free MEM and control treatments. Histochemical staining has shown that animals treated with NPs exhibit less Aβ plaque formation and decreased inflammation [172]. In addition, oral administration of either pioglitazone (PGZ) or PGZ-loaded PLGA-PEG NPs can decrease memory impairment and Aβ deposition in APP/PS1 transgenic mice. Treatment with PGZ-NPs results in less Aβ deposition than treatment with free PGZ [173]. Furthermore, nanotechnology has been applied to the early diagnosis of AD [174,175].
A sensitive peptide nanonetwork biosensor was designed to detect peripheral platelet-secreted Aβ, and showed promising results for early disease detection [175]. In conclusion, there is increasing focus on the use of nanotechnology to treat AD. However, it remains challenging to precisely target the critical region and distinguish between soluble and insoluble Aβ.
4.3 MS: oligodendrocytes, astrocytes, phagocytic cells, and T-cells
MS is a chronic neurodegenerative disorder characterized by demyelinated lesions and widespread inflammatory processes within the CNS [176]. In patients with MS, widespread lymphocyte infiltration, microglial, and macrophage activation induce demyelination, synaptic, axonal, and neuronal loss [177–179]. Nanotechnology has provided a novel therapeutic approach for MS by manipulating the polarization states of macrophages [179].
Nanoformulations have been used to enhance the efficacy of existing drugs and to reduce their adverse effects. In EAE animal models, which are widely used to study MS [13,180], liposomes loaded with various immunodominant peptides of myelin basic protein have shown efficacy in treating EAE rats, possibly due to the mannosylation of liposomes and the encapsulation of peptides [181]. Another study demonstrated that encapsulation of dimethyl fumarate (DMF) in solid lipid-based NPs (SLNs) improved its effect on disease modification in cuprizone-induced demyelinated mice following oral administration [182]. Furthermore, histological staining of the stomach showed that mice treated with cuprizone and free DMF exhibited mucosal damage, whereas DMF-SL-NP-treated mice did not show any malignancies [182]. In addition, curcumin-encapsulated chitosan-alginate-sodium tripolyphosphate NPs effectively reduced immune cell infiltration, astrocytic and microglial activation, and demyelination in mice with LPC-induced demyelination following intraperitoneal injection [183]. NPs carrying glucocorticoids (GCs), which are commonly used to treat acute relapses in MS, have also been shown to be effective in treating EAE in mice. In one study, dexamethasone was encapsulated in inorganic-organic hybrid NPs and injected intraperitoneally into EAE mice [184]. BMP-NPs are mainly endocytosed by phagocytic cells rather than by T cells, thus decreasing the side effects of GCs [184].
Several recent studies have reported tolerogenic effects in murine relapse-remitting EAE models through the delivery of antigens using polymeric PLG-NPs, which is an interesting finding [185–187]. For example, PLG-NPs have been utilized to deliver proteolipid peptides (PLP139–151) and ovalbumin to antigen-presenting cells, resulting in immune tolerance observed in various signaling pathways, including transcription factor activity, cytokine secretion, and T-cell activation [185–187]. These studies suggest that nano-carrier systems in MS disease models may act on a wide variety of cells, including oligodendrocytes, astrocytes, phagocytes, and T-cells. Although reducing the inflammatory response is the main target in treating MS, improving tolerogenic effects may be a promising strategy for future research.
4.4 ALS: microglia
ALS is a devastating neurodegenerative disease that targets upper and lower motor neurons, leading to severe muscle atrophy and weakness [188,189]. In addition, inflammatory processes within the CNS are believed to contribute to disease pathology, as evidenced by the presence of reactive microglia in neurodegenerative areas of the ALS brain [188,190]. However, the number of nanodrugs used to treat ALS is limited. One of the few nanodrugs used in ALS treatment is cerium-oxide NPs (CeNPs), an antioxidant that was orally administered to SOD1G93A mice, which has been shown to extend the survival time from 22 to 33 days [191]. The beneficial effects observed in the CeNP-treated group can be attributed to a decrease in reactive oxygen and nitrogen species [191].
4.5 Stroke: neuron and microglia
Stroke is a devastating global cerebrovascular disease characterized by interruption of blood supply, resulting in neurological dysfunction [192–194]. Under ischemic stroke conditions, decreased BBB integrity results in increased paracellular permeability, which directly contributes to cerebral vasogenic edema, hemorrhagic transformation, and increased mortality [195]. Although most patients survive after the first onset, only a few are cured during long-term rehabilitation. The rapid development of nanotechnology in other areas of modern medicine has ignited widespread interest in its potential in the field of stroke treatment [196–198]. The identification of stroke biomarkers and the microenvironment also provides possibilities for designing stimuli-responsive nanodrugs [199,200].
Recent advances in nanotechnology for stroke treatment include global [201–204] or targeted [205–209] contrast enhancement of imaging, nanosensors [210–213], theranostics [214–216], improved drug delivery [217–224], and tunable drug release [225,226]. Different types of nanomaterials, such as perfluorocarbon NPs, iron oxide NPs, and gold NPs, have been developed for the diagnosis and treatment of strokes [196–198]. Among these, polymeric and lipidic NPs are the most commonly used vehicles for compounds and have been applied in the co-delivery of chemotherapeutic agents, clot-busting drugs, and neuroprotective drugs [227]. For example, the effective delivery of complement component C3 (C3)-targeted siRNA via cationic lipid-assisted PEG-PLA NPs successfully traversed the BBB. This delivery system significantly suppressed C3 expression in microglia, thereby preventing neuronal apoptosis and reducing the volume of the ischemic zone [228]. Their immunofluorescence staining results unequivocally indicated the specific uptake of C3 siRNA-loaded NPs by Iba-1+ microglia in the ischemic penumbra. Furthermore, these NPs downregulated C3 levels to nearly half of those in the control group [228]. The combination of stem cells and nano-capsules is also promising for treating stroke. Alginate hydrogel microcapsules were used to carry EGF-loaded NPs, which were then trapped in palmitic acid-CLEVSRKNC peptide-coated mesenchymal stem cells to target the stroke region [229]. This smart multiscale system showed better strength, stability, specificity, and retention time than hydrogel microcapsules. Local implantation of the multiscale system facilitates tissue regeneration and effectively restores blood perfusion within 4 weeks without evident side effects [229]. Although a wide repertoire of nanocarriers has been demonstrated to be effective in animal models, it is still challenging to choose the right carrier, identify the appropriate route of administration, and meet clinical needs.
4.6 LSDs: neuron
LSDs are rare inherited metabolic disorders that result in defects in lysosome function [230–233]. These disorders occur due to deficiencies in enzymes required for lipid, glycoprotein, or mucopolysaccharide metabolism, which results in the accumulation of large undigested or partially digested molecules [231–233]. Therapeutic options for LSDs currently available include enzyme replacement therapy or chaperone therapy [234]. NPs have contributed significantly to the delivery of enzymes or chaperones across the BBB and protect them before reaching the target site.
For the treatment of mucopolysaccharidosis type II (MPSII), a type of LSD, brain-targeted polymeric NPs loaded with iduronate 2-sulfatase (IDS) have been shown to successfully remove the deposited glycosaminoglycans [235]. IDS enzyme-delivering NPs effectively removed GVG in fibroblasts from MPSII patients and MPSII mice [235]. Similarly, RVG peptide (RVG29-Cyspeptide)-modified chitosan NPs were used to deliver β-galactosidase (β-Gal) to nude mice, and the accumulation of active β-Gal in the brain indicates the potential of RVG-chitosan NPs in treating β-Gal-related diseases such as Hurler’s syndrome and Krabbe disease [236–238]. Different types of NPs have been tested to absorb or conjugate arylsulfatase A (ASA) on the surface and were intravenously injected into the ASA −/− MLD mouse model. Unfortunately, no significant change was observed compared to the free ASA-administered group [239]. This failure was attributed to the interference of glycoprotein-ASA during receptor-mediated transcytosis of NPs crossing the BBB [240].
4.7 Primary brain tumors: glial cells
Primary brain tumors are a relatively uncommon form of cancer but are the most common and fatal solid tumors among children [241]. In adults, gliomas and CNS lymphomas are the most frequent primary brain tumors [242]. Gliomas can originate from various cell types, including neural stem cells and progenitor cells, but their underlying causes are still largely unknown.
Drug nanoformulations offer several advantages for treating gliomas, including improved efficiency across the BBB, deeper tumor penetration, and decreased adverse effects. For instance, cisplatin-loaded PAA-PEG NPs improved the long-term survivors from none to 80% of F98-tumor-bearing rats when administered locally [243]. Non-invasive therapy using camptothecin-encapsulated PLGA-NPs administered via tail vein injection significantly reduced tumor growth in GL261-bearing mice [244]. To achieve better brain penetration, An-PEG-DOX-Au NPs, loaded with DOX and modified with angiopep-2, were developed, resulting in increased median survival time to 2.89-fold longer than that of saline in a Kunming glioma mouse model after intraperitoneal injection [245]. The ability of these NPs to cross the BBB via the LRP1 receptor and specifically target glioma cells is the most likely cause of their improved anti-glioma effect [245]. Similarly, transferrin-modified PEG NPs encapsulating temozomolide (TMZ) and a bromo-domain inhibitor (JQ1) increased the median survival time of U87MG and GL261 tumor-bearing mice when administered intravenously [146]. Inhibition of tumor growth was also demonstrated in U87 glioma-bearing mice treated with TMZ and vincristine loaded in solid lipid NPs (SLNs) and nanostructured lipid carriers (NLCs), and tumor growth inhibition was greater in mice treated with VT-NLCs than in those treated with VT-SLNs and T-NLCs [149]. The receptor-mediated transcytosis pathway is the mechanism by which most of the functionalized NPs mentioned above cross the BBB.
The cell-mediated transport of NPs provides another option for penetrating brain tumors. In one study, scientists loaded catalase into polyion complex micelles (nanozymes) [246] and found that bone marrow-derived macrophages delivering this nanozyme protected the nigrostriatum against MPTP intoxication. Further evidence has shown that nanozymes can be transported by bone marrow-derived macrophages to contiguous neural cells, including brain microvascular endothelial cells, neurons, and astrocytes [246]. Neutrophils, the most abundant type of immune cell known to penetrate inflamed brain tumors [247,248], have also been shown to be excellent carriers of nanodrugs [248]. For example, neutrophils carrying liposomes containing paclitaxel can penetrate the brain and suppress glioma recurrence in mice whose tumors have been surgically resected [248]. Cancer therapeutic molecule-delivering NPs have also been shown to penetrate brain tumors and efficiently inhibit tumor growth compared with free drugs.
In summary, poor penetration into the tumor site and rapid resistance to a single drug remain limiting factors in brain tumor therapy, and the development of nanocarriers is needed to address these issues. Owing to the critical role of the CNS, specific targeting is more urgent in treating brain tumors than in other types of tumors.
5 Gene tools by nanotechnology targeting neurodegenerative diseases
Gene therapy holds great promise for the treatment of neurodegenerative and CNS diseases by modulating the expression of specific genes and changing the phenotype [264]. Although several genetic medicines have been introduced, their high costs and safety concerns continue to be significant considerations for the wider community [265]. The latest advancements in gene delivery have led to the emergence of virus-based vectors, particularly the adeno-associated virus, which has gained regulatory approval. However, clinical trials have revealed certain limitations of virus-based gene delivery systems, including cost, packaging capacity, toxicity, and immunogenicity.
NPs provide an alternative to viral vectors for delivering genetic material to the nervous system. Notably, lipid-based NPs have shown remarkable potential for gene delivery over the past 5 years [266,267]. However, gene therapy using nanotechnology remains challenging due to the need for a large number of cells to counteract or reverse the malfunction, escape immune responses, and survive for longer periods [268].
Silencing genes through siRNAs, CRISPR, or enhancing them through antisense oligonucleotides (ASOs) are common strategies for gene modulation. However, the invasive administration of these molecules through injection or direct infusion into the brain carries certain risks, making this strategy impractical and unacceptable. Therefore, combining gene therapy with nanotechnology is a promising approach for the treatment of CNS diseases [268]. Recent experiments have used NPs to deliver genes to the brain. Table 2 lists the specifications of these NPs, the genetic products they carry, their cell targets, and the disease and animal models used in the in vivo experiments.
Evaluation of NPs used for gene therapy in recent in vivo experiments
NPs | Surface modification | Gene tool and target gene | Disease model | Cell target | Animal model | Published | Year |
---|---|---|---|---|---|---|---|
Polymer (PLA) | PEG | siRNA; C3 | Cerebral ischemia | Microglia | C57BL/6 mice | 2018 | [228] |
Polymer (PDMAEMA) | PEG | siRNA; BACE1 | AD | Neurons | APP/PS1 double transgenic mice | 2018 | [275] |
Exosome | RVG | siRNA; α-synuclein | PD | Neurons | C57BL/6 (transgenic) mice | 2014 | [272] |
Solid–lipid (T7-LPC) | Transferrin protein (T7) | siRNA; EGFR | Glioma | — | nu/nu mice | 2016 | [273] |
Cationic nano-emulsion | siRNA; anti-TNFα | Neuro-inflammation | Microglia | Sprague–Dawley rats | 2016 | [270] | |
Lipid NP | Calcium phosphate-coated | ASO; SOD1 | ALS | Motor neurons | Danio rerio | 2017 | [275] |
Lipid vector (SNALP) | Chlorotoxin | AMO; anti-miR-21 | Glioblastoma | — | C57BL/6 mice | 2015 | [276] |
Liposome | Transferrin protein | Plasmid DNA; β-galactosidase | Neurons | C57BL/6 mice | 2018 | [279] | |
Polyethylene glycol substituted lysine 30-mers | Plasmid DNA; hGDNF | PD | — | Sprague–Dawley rats | 2019 | [280] | |
GNP | NGF | Plasmid DNA; SNCA | PD | DA neurons | C57BL/6 mice | 2018 | [274] |
Polymer (DGL) | RVG | Plasmid shRNA; BACE1-AS | AD | Neurons | C57BL/6 APP/PS1 double transgenic mice | 2016 | [281] |
Poly(β-l-malic acid)-trileucine polymer | Six peptide vectors | Vectors | AD and tumor mouse models | — | C57BL/6 J mouse model with AD, or brain tumors | [282] |
Abbreviations: BACE1, B-site amyloid precursor protein cleaving enzyme 1; BACE1-AS, B-site amyloid precursor protein cleaving enzyme 1 antisense; C3, complement component 3; DGL, dendrigraft poly-L-lysine; EGFR, epidermal growth factor receptor; GFP, green fluorescent protein; GNP, gold NPs; hGDNF, human glial-cell line-derived neurotrophic factor; IVT-mRNA, in vitro transcribed mRNA; NGF, nerve growth factor; PEG, polyethylene glycol; RVG, rabies-virus glycoprotein; shRNA, short hairpin RNA; SNALP, stable nucleic acid lipid NPs; SNCA, α-synuclein; SOD1, superoxide dismutase 1; TNFα, tumor necrosis factor-α; T7-LPC, T7 peptide-liposome-protamine-chondroitin sulfate.
5.1 siRNA
SiRNAs are commonly delivered using nanotechnology techniques, such as intranasally administered manganese chitosan-matrix NPs encapsulating anti-eGFP siRNAs or RFP dsDNAs that successfully transfected neurons along the rostral-caudal axis of the brain [269]. Exosomes are also well-verified nano-tools for siRNA delivery [269].
Recently, functional siRNA carriers have been reported, such as anti-tumor necrosis factor (TNF) siRNA delivered by a cationic nano-emulsion, which downregulates TNF expression via intranasal administration [270]. Nanocarriers for intravenous or intraperitoneal injections have also been optimized to cross the BBB and accumulate at specific sites or cells in different disease models. For instance, β-site amyloid precursor protein cleaving enzyme 1 (BACE1) siRNAs containing NPs were modified with the CGN peptide for BBB penetration and the Tet1 peptide for neuron-specific binding, enabling them to escape from the lysosomes of neurons and induce gene silencing in an AD mouse model [271]. This resulted in hippocampal neurogenesis, fewer amyloid plaques, and lower phosphorylated tau levels [271]. RVG-conjugated exosomes encapsulating α-synuclein siRNA were shown to decrease α-synuclein levels in the striatum and mid-brain following peripheral injections [272]. Using a transgenic mouse model of PD, α-synuclein siRNA-RVG-NPs were found to decrease α-synuclein aggregation throughout the brain [272]. Transferrin-modified core–shell (T7-LPC) NPs were designed to deliver epidermal growth factor receptor (EGFR) siRNA to the glioma site, resulting in prolonged survival in mice compared to control treatment [273]. In this study, live imaging of mice bearing glioma showed that T7-LPC NPs delivered siRNA deeper into the brain than the NPs without transferrin modification, as well as prolonged retention time in the brain tumor. In addition, the T7-LPC NPs were demonstrated to downregulate EGFR level to almost half of the control group in glioma [273]. Polymeric PLA-PEGylated NPs loaded with siRNA were shown to be transported across the BBB and taken up by microglia in the ischemic penumbra of cerebral ischemia and reperfusion injury mice, and delivery of C3 siRNA to microglia reduced microglial neurotoxicity by downregulating C3 expression [274]. In conclusion, stable and traditional nanotools, such as polymeric NPs, chitosan-matrix NPs, and lipid NPs, can effectively deliver siRNA across the BBB after modification without causing significant side effects. Furthermore, these siRNA delivery tools have been applied to various CNS disease models.
5.2 Oligonucleotides
Gene therapy for CNS diseases has seen a surge in the use of ASOs in recent years, leading to FDA approval of an SMA therapy in 2016. However, the use of nanotechnology to administer ASOs has been limited. Studies have reported the successful delivery of ASOs against SOD1 using solid–lipid phosphate calcium lipid NPs, which diffuse throughout the brain and spinal cord of zebrafish [275]. Additionally, stable nucleic acid lipid particles (SNALP) have been used to deliver MiR-21 anti-mRNA oligonucleotides (AMOs) to glioma cells using chlorotoxin-induced glioma-specific targeting, resulting in effective therapy [276]. In fact, NPs delivering AMOs inside tumor cells have been shown to downregulate MiR-21 mRNA expression and repress tumor activity [276]. Recently, a G-Quadruplex-forming aptamer was reported to exhibit resistance to nuclease in the serum and demonstrated its efficacy in enhancing motor neuronal function in a Drosophila melanogaster model expressing the mutated form of human huntingtin [277,278].
5.3 Plasmid DNAs
Multiple studies have demonstrated the potential of functional NPs for delivering plasmid DNAs to the CNS [274,279–281]. For instance, transferrin and penetratin-modified liposomes (PenTf-liposomes) have been designed for better penetration into brain cells [279]. Intravenous injection of PenTf-liposomes encapsulating pGFP DNAs demonstrated their accumulation in the brain and transfection in neurons [279]. In another study, a glial cell line-derived neurotrophic factor (GDNF) plasmid delivered by poly-ethylene glycol-substituted lysine was administered intranasally to rats, resulting in global and long-term expression of GDNF in the brain [280]. Additionally, NPs offer the advantage of combination therapies. RVG-coated dendrigraft poly-lysine NPs were designed to encapsulate both therapeutic genes and peptides to target two key hallmarks of AD pathology [281]. BACE1 short hairpin RNA (shRNA) plasmid and a peptide inhibiting tau-related fibril formation were loaded together into RVG NPs. These multifunctional RVG NPs successfully reduced neurofibrillary tangles and rescued memory loss in AD mice [93]. However, plasmid delivery still faces challenges such as nanosize and low transfection efficacy.
In conclusion, nanotechnology-based gene editing holds tremendous potential for the treatment of CNS diseases. However, their biosafety must be thoroughly evaluated before conducting clinical trials. For instance, although rare, neurological side effects of the SARS-CoV-2 mRNA vaccine have been reported [283]. Additionally, an adeno-associated virus vector, which is an effective gene delivery tool, caused the death of two patients who received a high dose of AT132 during investigational therapy [284]. Therefore, it is crucial to carefully assess the safety and potential risks of these gene-editing tools to minimize any potential adverse effects in patients.
6 Clinical translations of NPs in treating CNS diseases
Although significant progress has been made in the treatment of CNS disorders in animal models, relatively few clinical trials have been conducted. However, some promising results have been reported [285,286]. For example, a novel gadolinium-based NP (AGuIX) combined with radiotherapy was shown to be safe and feasible in a phase I clinical trial [287]. Exosomes have also shown promise as nanocarriers in clinical trials due to their ability to be taken up by immature myeloid cells [288]. They have been used to deliver therapeutics across the BBB to treat neuralgia and refractory depression (NCT04202783 and NCT04202770). Additionally, autologous plasma-derived exosomes have been used to treat chronic multisystem autoimmune disorders (NCT02565264) [288], an early clinical trial, a spherical nucleic acid was reported to cross the BBB and kill tumor cells in humans [289].
7 Conclusions and future directions
7.1 Outcomes and potential targets
To date, various NPs have been designed to exploit transporter proteins, channels, and receptors to cross the BBB. Diagnostic imaging often employs gold, quantum dots, and magnetic iron NPs. Liposomes, exosomes, dendrimers, and polymeric NPs are commonly used as drug carriers due to their biocompatibility. Nano-gels [290] and nano-emulsions [291] have emerged as ideal candidates for CNS drug storage and delivery. Self-assembled peptides and therapeutic compounds have also demonstrated potential for less invasive applications [292,293]. Although most NPs are administered intravascularly, intranasal administration through the olfactory system is another option for NP transport to the brain [294].
In the treatment of CNS diseases, therapeutic targets have received significant attention. For instance, NPs have been developed in animal models of PD that target DA neurons to modify disease pathology by enhancing the development of these neurons. Besides targeting dopamine or oxidative stress, cellular pathways such as α-synuclein, glucocerebrosidase, and leucine-rich repeat kinase in neurons have also been explored as potential targets [151]. In addition, autoimmunity, neuroinflammation [295], and chaperone-mediated autophagy [296] observed in PD offer other possibilities for synergistic therapy using nanotechnology.
Recent advances in nanocarriers have improved their efficacy in the treatment of AD by delivering compounds that target Aβ deposits and hyperphosphorylated tau proteins. As our understanding of AD pathogenesis deepens, the ubiquitin proteasomal system [297], mitochondria [298], and microglia [299] have been found to play indispensable roles in AD progression. Notably, curcumin has been demonstrated to be effective in treating both PD [149] and AD [300] by exerting a neuro-protective action through multiple pathways, including AD-related enzymes, Aβ, tau-related proteins, mitochondria, and metal chelation [300]. Despite the promising results of the described nanocarriers, there is currently no perfect type of NPs (mainly polymeric but composed of different materials) or widely used route of administration for AD therapy. In the case of MS treatment, most NPs are designed to target inflammation and are taken up by a wide range of cells, including oligodendrocytes, astrocytes, phagocytic cells, and T cells. However, there has been limited research on NPs targeting ALS.
NPs have demonstrated their effectiveness in delivering drugs into the CNS, modifying disease pathology, and delivering genes to various cell types. The multifunctionality of drugs in nano-formulation, including improved efficiency across the BBB, co-delivery of drugs, and specific targeting, highlights their potential in treating CNS diseases. Nevertheless, while NPs can be modified and tailored to target different cell types, it remains challenging to accurately target them in different encephalic regions (Figure 4).

CNS-targeting NP design criteria.
7.2 Safety concerns and challenges
Although the use of NPs has demonstrated effectiveness in carrying drugs across the BBB in CNS diseases, their development is still in its early stages, and there are concerns regarding their safety. These concerns are primarily related to biocompatibility, biodegradability, and potential immune responses to administered NPs. Most studies have been conducted using animal disease models, and although some studies have included histopathological analysis and evaluation of inflammatory cytokines, further long-term evaluation is necessary. Despite the demonstrated efficacy of nanodrugs for treating CNS diseases, the exact fate of NPs after crossing the BBB remains unclear, leading to unforeseen safety concerns.
Free drugs can cause toxicity in organs and cells owing to their rapid spread throughout the body. NPs can overcome this problem by increasing targeting specificity, which has been demonstrated in many studies to decrease the toxicity and side effects of free drugs [301]. However, the adverse effects of NPs must also be evaluated. To study the safety of NPs more extensively, Knudsen et al. measured several parameters, such as histopathology, gene toxicity, and gene expression, after administering liposomes and micelles to mice. They observed that micelle treatment increased cytokine (IL-6 and CCL2) gene expression in the liver, as well as oxidative stress response gene (HMOX1) and DNA repair enzyme gene (OGG1). Histopathological assays showed no toxicological effects of NPs on various tissues, such as the liver and lungs [301]. However, other studies have reported cytotoxicity of certain types of NPs, such as PAMAM dendrimers [302], silica NPs [283,284], metal-containing NPs [88,305], and gold NPs [306]. Various reviews have highlighted that cytotoxicity may be influenced by the NP dose, NP coating, size, shape, and distribution [302–304,307]. Despite the potential of NPs to reduce drug toxicity, nanomaterials themselves may pose new safety concerns, requiring comprehensive and long-term monitoring.
Other concerns regarding NPs in CNS therapy include their pharmacokinetics, particularly their ability to penetrate the BBB. Although NPs have been shown to effectively deliver drugs into the CNS, their efficiency in crossing the BBB still needs to be optimized. However, the process of transferring waste products from the brain to the periphery remains unclear. To accurately assess the efficacy of NP penetration, it is important to study their behavior in the same disease model for which they are intended for therapy, rather than in healthy animals. In certain diseases such as stroke, where the BBB is compromised, there may be more opportunities for NPs to cross the barrier, but repairing the BBB is also a crucial consideration.
Another issue that needs to be addressed is the protein corona, which is formed by biomolecules that bind to the surface of NPs during circulation in biological fluids [308,309]. Disturbed by the steric hindrance from the protein corona, the functionalized NPs were unable to cross the BBB [47], target specific cell types, and distribute among the organs unpredictably [310–312]. Recent studies have shown that the protein corona perturbs proteostasis and remodels cell metabolism [313]. To solve these problems, it is better to change the surface chemistry rather than the charge [314]. PEGlaytion could help partially repulse the binding of biomolecules from biological fluids, thus protecting the exposure of the targeting ligands [315]. The NPs could also be designed to attract more dysopsonins, such as albumin [316–318] or clusterin [319,320], on the surface to maintain longer circulation and better drug delivery efficiency. An advisable option is to manipulate the protein corona for brain-targeting. Zhang et al. modified an Aβ1–42 derived peptide onto the surface to absorb plasma apolipoproteins, and thus exposed the receptor-binding domain of apolipoproteins via the receptor-mediated transcytosis to cross BBB [312]. Tween 80 has also been shown to absorb serum apolipoproteins [321].
After crossing the BBB, CSF is another factor that leads to the formation of a protein corona, which obviously affects the targeting, cellular uptake, and drug release of NPs [322]. Wang et al. demonstrated both in vivo and in vitro, the formation of CSF protein corona on the surface of polystyrene NPs led to the loss of specific targeting neuronal cells [322]. Interestingly, Pinals et al. quantized the composition of protein corona on the surface of DNA-functionalized single-walled carbon nanotubes in plasma and CSF and found that the main types of proteins were involved in lipid binding, complement activation, and coagulation [323]. However, the knowledge of protein corona composition, formation mechanisms, and dynamics is still poor, which remains an obstacle in the clinical application of NPs.
In conclusion, this review highlights the potential of NPs to target different types of cells for the treatment of CNS diseases, indicating significant promise in the development of precise medicine. Nanotechnology enables flexible treatment strategies by combining drugs, enzymes, and gene tools, providing stability in circulation, improving capacity to cross the BBB, and allowing for regional and cell type targeting to enhance the disease conditions while reducing adverse effects on other tissues. Although several polymeric NPs and exosomes have undergone clinical trials at different stages, most nanodrugs for treating CNS diseases are still at the laboratory level. Therefore, further studies are required to assess the dynamics and biosafety of these nanodrugs before proceeding to clinical trials.
-
Funding information: This work was supported by the GuangDong Basic and Applied Basic Research Foundation –Youth Fund (S.G., 2022A1515110230), and a grant from The Netherlands Organisation for Health Research and Development (ZonMw) (C.-X.Y., 459001004).
-
Author contributions: S.G. and C.X.Y. conceptualized the review topic and outline. S.G. drafted the manuscript and C.X.Y. provided critical revisions and edits. Both authors contributed equally to the final version of the manuscript.
-
Conflict of interest: The authors state no conflict of interest.
References
[1] Sharma P, Srivastava P, Seth A, Tripathi PN, Banerjee AG, Shrivastava SK. Comprehensive review of mechanisms of pathogenesis involved in Alzheimer’s disease and potential therapeutic strategies. Prog Neurobiol. 2019;174:53–89.10.1016/j.pneurobio.2018.12.006Search in Google Scholar PubMed
[2] Poewe W, Seppi K, Tanner CM, Halliday GM, Brundin P, Volkmann J, et al. Parkinson disease. Nat Rev Dis Primers. 2017;3:17013.10.1038/nrdp.2017.13Search in Google Scholar PubMed
[3] Martin JB. Molecular basis of the neurodegenerative disorders. New Engl J Med. 1999;340:1970–80.10.1056/NEJM199906243402507Search in Google Scholar PubMed
[4] Dobson R, Giovannoni G. Multiple sclerosis–a review. Eur J Neurol. 2019;26(1):27–40.Search in Google Scholar
[5] Feldman EL, Goutman SA, Petri S, Mazzini L, Savelieff MG, Shaw PJ, et al. Amyotrophic lateral sclerosis. Lancet. 2022;400(10360):1363–80.10.1016/S0140-6736(22)01272-7Search in Google Scholar PubMed PubMed Central
[6] Moshe SL, Perucca E, Ryvlin P, Tomson T. Epilepsy: new advances. Lancet. 2015;385:884–98.10.1016/S0140-6736(14)60456-6Search in Google Scholar PubMed
[7] Zhang B, Yan W, Zhu Y, Yang W, Le W, Chen B, et al. Nanomaterials in neural-stem-cell-mediated regenerative medicine: imaging and treatment of neurological diseases. Adv Mater. 2018;30:e1705694.10.1002/adma.201705694Search in Google Scholar PubMed
[8] Teleanu DM, Chircov C, Grumezescu AM, Teleanu RI. Neuronanomedicine: an up-to-date overview. Pharmaceutics. 2019;11(3):101.10.3390/pharmaceutics11030101Search in Google Scholar PubMed PubMed Central
[9] Sweeney MD, Sagare AP, Zlokovic BV. Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat Rev Neurol. 2018;14:133–50.10.1038/nrneurol.2017.188Search in Google Scholar PubMed PubMed Central
[10] Daneman R, Prat A. The blood-brain barrier. Cold Spring Harb Perspect Biol. 2015;7:a020412.10.1101/cshperspect.a020412Search in Google Scholar PubMed PubMed Central
[11] Banks WA. From blood-brain barrier to blood-brain interface: new opportunities for CNS drug delivery. Nat Rev Drug Discov. 2016;15:275–92.10.1038/nrd.2015.21Search in Google Scholar PubMed
[12] Lapointe S, Perry A, Butowski NA. Primary brain tumours in adults. Lancet. 2018;392:432–46.Search in Google Scholar
[13] Dobson R, Giovannoni G. Multiple sclerosis - a review. Eur J Neurol. 2019;26:27–40.10.1111/ene.13819Search in Google Scholar PubMed
[14] Vissers C, Ming GL, Song H. Nanoparticle technology and stem cell therapy team up against neurodegenerative disorders. Adv Drug Deliv Rev. 2019;19(3):279–98.10.1016/j.addr.2019.02.007Search in Google Scholar PubMed PubMed Central
[15] Van Bulck M, Sierra-Magro A, Alarcon-Gil J, Perez-Castillo A, Morales-Garcia JA. Novel approaches for the treatment of Alzheimer’s and Parkinson’s disease. Int J Mol Sci. 2019;20(3):719.10.3390/ijms20030719Search in Google Scholar PubMed PubMed Central
[16] Selmani A, Kovačević D, Bohinc K. Nanoparticles: From synthesis to applications and beyond. Adv Colloid Interface Sci. 2022;303:102640.10.1016/j.cis.2022.102640Search in Google Scholar PubMed
[17] Mitchell MJ, Billingsley MM, Haley RM, Wechsler ME, Peppas NA, Langer R. Engineering precision nanoparticles for drug delivery. Nat Rev Drug Discov. 2021;20(2):101–24.10.1038/s41573-020-0090-8Search in Google Scholar PubMed PubMed Central
[18] Seaberg J, Montazerian H, Hossen MN, Bhattacharya R, Khademhosseini A, Mukherjee P. Hybrid nanosystems for biomedical applications. ACS Nano. 2021;15(2):2099–142.10.1021/acsnano.0c09382Search in Google Scholar PubMed PubMed Central
[19] Riccardi C, Napolitano F, Montesarchio D, Sampaolo S, Melone MAB. Nanoparticle-guided brain drug delivery: Expanding the therapeutic approach to neurodegenerative diseases. Pharmaceutics. 2021;13(11):1897.10.3390/pharmaceutics13111897Search in Google Scholar PubMed PubMed Central
[20] Feng L, Wang H, Xue X. Recent progress of nanomedicine in the treatment of central nervous system diseases. Adv Ther. 2020;3(5):1900159.10.1002/adtp.201900159Search in Google Scholar
[21] Saeedi M, Eslamifar M, Khezri K, Dizaj SM. Applications of nanotechnology in drug delivery to the central nervous system. Biomed Pharmacother. 2019;111:666–75.10.1016/j.biopha.2018.12.133Search in Google Scholar PubMed
[22] Zhang WS, Mehta A, Tong ZQ, Esser L, Voelcker NH. Development of polymeric nanoparticles for blood-brain barrier transfer-strategies and challenges. Adv Sci. 2021;8(10):2003937.10.1002/advs.202003937Search in Google Scholar PubMed PubMed Central
[23] Satapathy MK, Yen TL, Jan JS, Tang RD, Wang JY, Taliyan R, et al. Solid lipid nanoparticles (SLNs): an advanced drug delivery system targeting brain through BBB. Pharmaceutics. 2021;13(8):1183.10.3390/pharmaceutics13081183Search in Google Scholar PubMed PubMed Central
[24] Amiri M, Jafari S, Kurd M, Mohamadpour H, Khayati M, Ghobadinezhad F, et al. Engineered solid lipid nanoparticles and nanostructured lipid carriers as new generations of blood-brain barrier transmitters. ACS Chem Neurosci. 2021;12(24):4475–90.10.1021/acschemneuro.1c00540Search in Google Scholar PubMed
[25] Spandana KMA, Bhaskaran M, Karri VR, Natarajan J. A comprehensive review of nano drug delivery system in the treatment of CNS disorders. J Drug Deliv Sci Technol. 2020;57:101628.10.1016/j.jddst.2020.101628Search in Google Scholar
[26] Gianni C, Campisi M, Mattu C, Kamm RD, Cjiono V, Raynold AAM, et al. Roadmap on Nanomedicine for the Central Nervous System-Section 2: Microphysiological systems for preclinical testing of drug-loaded nanoparticle transport across the human blood-brain barrier. J Phys Mater. 2022;6:5–10.10.1088/2515-7639/acab88Search in Google Scholar
[27] Nguyen TT, Nguyen TTD, Vo TK, Tran NMA, Nguyen MK, Vo TV, et al. Nanotechnology-based drug delivery for central nervous system disorders. Biomed Pharmacother. 2021;143:112117.10.1016/j.biopha.2021.112117Search in Google Scholar PubMed
[28] Sweeney MD, Zhao Z, Montagne A, Nelson AR, Zlokovic BV. Blood-brain barrier: From physiology to disease and back. Physiol Rev. 2019;99:21–78.10.1152/physrev.00050.2017Search in Google Scholar PubMed PubMed Central
[29] Wohlfart S, Gelperina S, Kreuter J. Transport of drugs across the blood-brain barrier by nanoparticles. J Control Release. 2012;161(2):264–73.10.1016/j.jconrel.2011.08.017Search in Google Scholar PubMed
[30] Moura RP, Martins C, Pinto S, Sousa F, Sarmento B. Blood-brain barrier receptors and transporters: an insight on their function and how to exploit them through nanotechnology. Expert Opin Drug Del. 2019;16(3):271–85.10.1080/17425247.2019.1583205Search in Google Scholar PubMed
[31] Tanjore H, Kalluri R. The role of type IV collagen and basement membranes in cancer progression and metastasis. Am J Pathol. 2006;168(3):715–7.10.2353/ajpath.2006.051321Search in Google Scholar PubMed PubMed Central
[32] Aumailley M, Smyth N. The role of laminins in basement membrane function. J Anat. 1998;193(1):1–21.10.1046/j.1469-7580.1998.19310001.xSearch in Google Scholar PubMed PubMed Central
[33] Teleanu DM, Negut I, Grumezescu V, Grumezescu AM, Teleanu RI. Nanomaterials for drug delivery to the central nervous system. Nanomaterials (Basel). 2019;9(3):371.10.3390/nano9030371Search in Google Scholar PubMed PubMed Central
[34] Bellettato CM, Scarpa M. Possible strategies to cross the blood-brain barrier. Ital J Pediatr. 2018;44(Suppl 2):131.10.1186/s13052-018-0563-0Search in Google Scholar PubMed PubMed Central
[35] Furtado D, Bjornmalm M, Ayton S, Bush AI, Kempe K, Caruso F. Overcoming the blood-brain barrier: the role of nanomaterials in treating neurological diseases. Adv Mater. 2018;30(46):e1801362.10.1002/adma.201801362Search in Google Scholar PubMed
[36] Carbone F, Djamshidian A, Seppi K, Poewe W. Apomorphine for Parkinson’s disease: efficacy and safety of current and new formulations. CNS Drugs. 2019;33:905–18.10.1007/s40263-019-00661-zSearch in Google Scholar PubMed PubMed Central
[37] Singh AP, Biswas A, Shukla A, Maiti P. Targeted therapy in chronic diseases using nanomaterial-based drug delivery vehicles. Signal Transduct Target Ther. 2019;4(1):33.10.1038/s41392-019-0068-3Search in Google Scholar PubMed PubMed Central
[38] Kreuter J. Drug delivery to the central nervous system by polymeric nanoparticles: what do we know? Adv Drug Deliv Rev. 2014;71:2–14.10.1016/j.addr.2013.08.008Search in Google Scholar PubMed
[39] Cena V, Jativa P. Nanoparticle crossing of blood-brain barrier: a road to new therapeutic approaches to central nervous system diseases. Nanomedicine. 2018;13(13):1513–6.10.2217/nnm-2018-0139Search in Google Scholar PubMed
[40] Patel MM, Patel BM. Crossing the blood-brain barrier: recent advances in drug delivery to the brain. CNS Drugs. 2017;31(2):109–33.10.1007/s40263-016-0405-9Search in Google Scholar PubMed
[41] Devraj K, Klinger ME, Myers RL, Mokashi A, Hawkins RA, Simpson IA. GLUT-1 glucose transporters in the blood-brain barrier: Differential phosphorylation. J Neurosci Res. 2011;89:1913–25.10.1002/jnr.22738Search in Google Scholar PubMed PubMed Central
[42] Gliddon CM, Shao Z, LeMaistre JL, Anderson CM. Cellular distribution of the neutral amino acid transporter subtype ASCT2 in mouse brain. J Neurochem. 2009;108:372–83.10.1111/j.1471-4159.2008.05767.xSearch in Google Scholar PubMed
[43] Zhou YQ, Peng ZL, Seven ES, Leblanc RM. Crossing the blood-brain barrier with nanoparticles. J Control Release. 2018;270:290–303.10.1016/j.jconrel.2017.12.015Search in Google Scholar PubMed
[44] Chen R, Romero G, Christiansen MG, Mohr A, Anikeeva P. Wireless magnetothermal deep brain stimulation. Science. 2015;347(6229):1477–80.10.1126/science.1261821Search in Google Scholar PubMed
[45] Carvalho-de-Souza JL, Treger JS, Dang B, Kent SBH, Pepperberg DR, Bezanilla F. Photosensitivity of neurons enabled by cell-targeted gold nanoparticles. Neuron. 2015;86(1):207–17.10.1016/j.neuron.2015.02.033Search in Google Scholar PubMed PubMed Central
[46] Naidu PSR, Gavriel N, Gray CGG, Bartlett CA, Toomey LM, Kretzmann JA, et al. Elucidating the inability of functionalized nanoparticles to cross the blood-brain barrier and target specific cells in vivo. ACS Appl Mater Inter. 2019;11(25):22085–95.10.1021/acsami.9b01356Search in Google Scholar PubMed
[47] Xiao W, Wang Y, Zhang H, Liu Y, Xie R, He X, et al. The protein corona hampers the transcytosis of transferrin-modified nanoparticles through blood–brain barrier and attenuates their targeting ability to brain tumor. Biomaterials. 2021;274:120888.10.1016/j.biomaterials.2021.120888Search in Google Scholar PubMed
[48] Yu YJ, Zhang Y, Kenrick M, Hoyte K, Luk W, Lu YM, et al. Boosting brain uptake of a therapeutic antibody by reducing its affinity for a transcytosis target. Sci Transl Med. 2011;3(84):84ra44.10.1126/scitranslmed.3002230Search in Google Scholar PubMed
[49] Widera A, Norouziyan F, Shen WC. Mechanisms of TfR-mediated transcytosis and sorting in epithelial cells and applications toward drug delivery. Adv Drug Deliv Rev. 2003;55(11):1439–66.10.1016/j.addr.2003.07.004Search in Google Scholar PubMed
[50] Gu X, Song QX, Zhang Q, Huang M, Zheng MN, Chen J, et al. Clearance of two organic nanoparticles from the brain via the paravascular pathway. J Control Release. 2020;322:31–41.10.1016/j.jconrel.2020.03.009Search in Google Scholar PubMed
[51] Taylor EM. The impact of efflux transporters in the brain on the development of drugs for CNS disorders. Clin Pharmacokinet. 2002;41:81–92.10.2165/00003088-200241020-00001Search in Google Scholar PubMed
[52] Sharom FJ. The P-glycoprotein multidrug transporter. Essays Biochem. 2011;50:161–78.10.1042/bse0500161Search in Google Scholar PubMed
[53] Potschka H. Role of CNS efflux drug transporters in antiepileptic drug delivery: Overcoming CNS efflux drug transport. Adv Drug Deliv Rev. 2012;64:943–52.10.1016/j.addr.2011.12.007Search in Google Scholar PubMed
[54] Zhang F, Lin YA, Kannan S, Kannan RM. Targeting specific cells in the brain with nanomedicines for CNS therapies. J Control Release. 2016;240:212–26.10.1016/j.jconrel.2015.12.013Search in Google Scholar PubMed PubMed Central
[55] Wang D, El-Amouri SS, Dai M, Kuan CY, Hui DY, Brady RO, et al. Engineering a lysosomal enzyme with a derivative of receptor-binding domain of apoE enables delivery across the blood-brain barrier. Proc Natl Acad Sci U S A. 2013;110(8):2999–3004.10.1073/pnas.1222742110Search in Google Scholar PubMed PubMed Central
[56] Haney MJ, Klyachko NL, Zhaoa YL, Gupta R, Plotnikova EG, He ZJ, et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J Control Release. 2015;207:18–30.10.1016/j.jconrel.2015.03.033Search in Google Scholar PubMed PubMed Central
[57] Zhang HL, Jiang YH, Zhao SG, Jiang LQ, Meng Y, Liu P, et al. Selective neuronal targeting, protection and signaling network analysis via dopamine-mediated mesoporous silica nanoparticles. Medchemcomm. 2015;6:1117–29.10.1039/C5MD00038FSearch in Google Scholar
[58] Chu DSH, Schellinger JG, Bocek MJ, Johnson RN, Pun SH. Optimization of Tet1 ligand density in HPMA-co-oligolysine copolymers for targeted neuronal gene delivery. Biomaterials. 2013;34(37):9632–7.10.1016/j.biomaterials.2013.08.045Search in Google Scholar PubMed PubMed Central
[59] Wei H, Schellinger JG, Chu DSH, Pun SH. Neuron-targeted copolymers with sheddable shielding blocks synthesized using a reducible, RAFT-ATRP double-head agent. J Am Chem Soc. 2012;134(40):16554–7.10.1021/ja3085803Search in Google Scholar PubMed PubMed Central
[60] Kwon EJ, Lasiene J, Jacobson BE, Park IK, Horner PJ, Pun SH. Targeted nonviral delivery vehicles to neural progenitor cells in the mouse subventricular zone. Biomaterials. 2010;31(8):2417–24.10.1016/j.biomaterials.2009.11.086Search in Google Scholar PubMed PubMed Central
[61] Kurakhmaeva KB, Djindjikhashvili IA, Petrov VE, Balabanyan VU, Voronina TA, Trofimov SS, et al. Brain targeting of nerve growth factor using poly(butyl cyanoacrylate) nanoparticles. J Drug Target. 2009;17(8):564–74.10.1080/10611860903112842Search in Google Scholar PubMed
[62] You LH, Wang J, Liu TQ, Zhang YL, Han XX, Wang T, et al. Targeted brain delivery of rabies virus glycoprotein 29-modified deferoxamine-loaded nanoparticles reverses functional deficits in Parkinsonian mice. ACS Nano. 2018;12(5):4123–39.10.1021/acsnano.7b08172Search in Google Scholar PubMed
[63] Nag OK, Muroski ME, Hastman DA, Almeida B, Medintz IL, Huston AL, et al. Nanoparticle-mediated visualization and control of cellular membrane potential: strategies, progress, and remaining issues. ACS Nano. 2020;14(3):2659–77.10.1021/acsnano.9b10163Search in Google Scholar PubMed
[64] Young AT, Cornwell N, Daniele MA. Neuro-Nano Interfaces: Utilizing Nano-Coatings and Nanoparticles to Enable Next-Generation Electrophysiological Recording, Neural Stimulation, and Biochemical Modulation. Adv Funct Mater. 2018;28(12):1700239.10.1002/adfm.201700239Search in Google Scholar PubMed PubMed Central
[65] Vinzant N, Scholl JL, Wu CM, Kindle T, Koodali R, Forster GL. Iron oxide nanoparticle delivery of peptides to the brain: reversal of anxiety during drug withdrawal. Front Neurosci-Switz. 2017;11:608.10.3389/fnins.2017.00608Search in Google Scholar PubMed PubMed Central
[66] Shen ZY, Liu T, Li Y, Lau J, Yang Z, Fan WP, et al. Fenton-reaction-acceleratable magnetic nanoparticles for ferroptosis therapy of orthotopic brain tumors. ACS Nano. 2018;12(11):11355–65.10.1021/acsnano.8b06201Search in Google Scholar PubMed
[67] Chen CT, Duan ZQ, Yuan Y, Li RX, Pang L, Liang JM, et al. Peptide-22 and cyclic RGD functionalized liposomes for glioma targeting drug delivery overcoming BBB and BBTB. ACS Appl Mater Inter. 2017;9(7):5864–73.10.1021/acsami.6b15831Search in Google Scholar PubMed
[68] Mishra MK, Beaty CA, Lesniak WG, Kambhampati SR, Zhang F, Wilson MA, et al. Dendrimer brain uptake and targeted therapy for brain injury in a large animal model of hypothermic circulatory arrest. ACS Nano. 2014;8(3):2134–47.10.1021/nn404872eSearch in Google Scholar PubMed PubMed Central
[69] Zhu FD, Hu YJ, Yu L, Zhou XG, Wu JM, Tang Y, et al. Nanoparticles: a hope for the treatment of inflammation in CNS. Front Pharmacol. 2021;12:683935.10.3389/fphar.2021.683935Search in Google Scholar PubMed PubMed Central
[70] Garden GA, Moller T. Microglia biology in health and disease. J Neuroimmune Pharm. 2006;1(2):127–37.10.1007/s11481-006-9015-5Search in Google Scholar PubMed
[71] Lehnardt S. Innate immunity and neuroinflammation in the CNS: the role of microglia in toll-like receptor-mediated neuronal injury. Glia. 2010;58(3):253–63.10.1002/glia.20928Search in Google Scholar PubMed
[72] Guo SS, Cazarez-Marquez F, Jiao H, Foppen E, Korpel NL, Grootemaat AE, et al. Specific silencing of microglial gene expression in the rat brain by nanoparticle-based small interfering RNA delivery. ACS Appl Mater Inter. 2022;14(4):5066–79.10.1021/acsami.1c22434Search in Google Scholar PubMed PubMed Central
[73] Nadjar A. Nanoparticles: The new promise for region-specific targeting of microglia. Matter-Us. 2022;5:2529–30.10.1016/j.matt.2022.06.048Search in Google Scholar
[74] Noh C, Shin HJ, Lee S, Kim SI, Kim YH, Lee WH, et al. CX3CR1-targeted PLGA nanoparticles reduce microglia activation and pain behavior in rats with spinal nerve ligation. Int J Mol Sci. 2020;21(10):3469.10.3390/ijms21103469Search in Google Scholar PubMed PubMed Central
[75] Carta AR, Pisanu A. Modulating microglia activity with PPAR-gamma agonists: a promising therapy for Parkinson’s Disease? Neurotox Res. 2013;23(2):112–23.10.1007/s12640-012-9342-7Search in Google Scholar PubMed
[76] Gordon R, Albornoz EA, Christie DC, Langley MR, Kumar V, Mantovani S, et al. Inflammasome inhibition prevents alpha-synuclein pathology and dopaminergic neurodegeneration in mice. Sci Transl Med. 2018;10(465):eaah4066.10.1126/scitranslmed.aah4066Search in Google Scholar PubMed PubMed Central
[77] Martinez B, Peplow PV. Neuroprotection by immunomodulatory agents in animal models of Parkinson’s disease. Neural Regen Res. 2018;13(9):1493–506.10.4103/1673-5374.237108Search in Google Scholar PubMed PubMed Central
[78] Korshoj LE, Shi W, Duan B, Kielian T. The prospect of nanoparticle systems for modulating immune cell polarization during central nervous system infection. Front Immunol. 2021;12:670931.10.3389/fimmu.2021.670931Search in Google Scholar PubMed PubMed Central
[79] McGinley MP, Goldschmidt CH, Rae-Grant AD. Diagnosis and treatment of multiple sclerosis: a review. Jama. 2021;325(8):765–79.10.1001/jama.2020.26858Search in Google Scholar PubMed
[80] Brownlee WJ, Hardy TA, Fazekas F, Miller DH. Diagnosis of multiple sclerosis: progress and challenges. Lancet. 2017;389(10076):1336–46.10.1016/S0140-6736(16)30959-XSearch in Google Scholar PubMed
[81] Oh J, Vidal-Jordana A, Montalban X. Multiple sclerosis: clinical aspects. Curr Opin Neurol. 2018;31(6):752–59.10.1097/WCO.0000000000000622Search in Google Scholar PubMed
[82] Hauser SL, Cree BAC. Treatment of Multiple Sclerosis: areview. Am J Med. 2020;133(12):1380–90.10.1016/j.amjmed.2020.05.049Search in Google Scholar PubMed PubMed Central
[83] Rahiman N, Mohammadi M, Alavizadeh SH, Arabi L, Badiee A, Jaafari MR. Recent advancements in nanoparticle-mediated approaches for restoration of multiple sclerosis. J Control Release. 2022;343:620–44.10.1016/j.jconrel.2022.02.009Search in Google Scholar PubMed
[84] Liu HH, Han YB, Wang TT, Zhang H, Xu Q, Yuan JX, et al. Targeting microglia for therapy of Parkinson’s Disease by using biomimetic ultrasmall nanoparticles. J Am Chem Soc. 2020;142(52):21730–42.10.1021/jacs.0c09390Search in Google Scholar PubMed
[85] Wang Y, Luo J, Shi YL. Nano-curcumin simultaneously protects the blood-brain barrier and reduces m1 microglial activation during cerebral ischemia-reperfusion injury. ACS Appl Mater Inter. 2019;11(4):3763–70.10.1021/acsami.8b20594Search in Google Scholar PubMed
[86] Xiao L, Wei F, Zhou YH, Anderson GJ, Frazer DM, Lim YC, et al. Dihydrolipoic acid-gold nanoclusters regulate microglial polarization and have the potential to alter neurogenesis. Nano Lett. 2020;20(1):478–95.10.1021/acs.nanolett.9b04216Search in Google Scholar PubMed
[87] Zhao NX, Francis NL, Calvelli HR, Moghe PV. Microglia-targeting nanotherapeutics for neurodegenerative diseases. Appl Bioeng. 2020;4(3):030902.10.1063/5.0013178Search in Google Scholar PubMed PubMed Central
[88] Chang X, Li J, Niu S, Xue Y, Tang M. Neurotoxicity of metal-containing nanoparticles and implications in glial cells. J Appl Toxicol. 2021;41(1):65–81.10.1002/jat.4037Search in Google Scholar PubMed
[89] Pekny M, Nilsson M. Astrocyte activation and reactive gliosis. Glia. 2005;50(4):427–34.10.1002/glia.20207Search in Google Scholar PubMed
[90] Persidsky Y, Ramirez SH, Haorah J, Kanmogne GD. Blood-brain barrier: structural components and function under physiologic and pathologic conditions. J Neuroimmune Pharmacol. 2006;1(3):223–36.10.1007/s11481-006-9025-3Search in Google Scholar PubMed
[91] Moura RP, Almeida A, Sarmento B. The role of non-endothelial cells on the penetration of nanoparticles through the blood brain barrier. Prog Neurobiol. 2017;159:39–49.10.1016/j.pneurobio.2017.09.001Search in Google Scholar PubMed
[92] Sofroniew MV. Astrogliosis. Cold Spring Harb Perspect Biol. 2015;7(2):a020420.10.1101/cshperspect.a020420Search in Google Scholar PubMed PubMed Central
[93] Tanaka H, Nakatani T, Furihata T, Tange K, Nakai Y, Yoshioka H, et al. In vivo introduction of mrna encapsulated in lipid nanoparticles to brain neuronal cells and astrocytes via intracerebroventricular administration. Mol Pharm. 2018;15(5):2060–7.10.1021/acs.molpharmaceut.7b01084Search in Google Scholar PubMed
[94] Gu J, Al-Bayati K, Ho EA. Development of antibody-modified chitosan nanoparticles for the targeted delivery of siRNA across the blood-brain barrier as a strategy for inhibiting HIV replication in astrocytes. Drug Deliv Transl Res. 2017;7(4):497–506.10.1007/s13346-017-0368-5Search in Google Scholar PubMed
[95] Vismara I, Papa S, Veneruso V, Mauri E, Mariani A, De Paola M, et al. Selective modulation of a1 astrocytes by drug-loaded nano-structured gel in spinal cord injury. ACS Nano. 2020;14(1):360–71.10.1021/acsnano.9b05579Search in Google Scholar PubMed
[96] Hawkins SJ, Crompton LA, Sood A, Saunders M, Boyle NT, Buckley A, et al. Nanoparticle-induced neuronal toxicity across placental barriers is mediated by autophagy and dependent on astrocytes. Nat Nanotechnol. 2018;13(5):427–33.10.1038/s41565-018-0085-3Search in Google Scholar PubMed
[97] Hohnholt MC, Dringen R. Uptake and metabolism of iron and iron oxide nanoparticles in brain astrocytes. Biochem Soc Trans. 2013;41(6):1588–92.10.1042/BST20130114Search in Google Scholar PubMed
[98] Fernandez-Bertolez N, Costa C, Brando F, Duarte JA, Teixeira JP, Pasaro E, et al. Evaluation of cytotoxicity and genotoxicity induced by oleic acid-coated iron oxide nanoparticles in human astrocytes. Environ Mol Mutagen. 2019;60(9):816–29.10.1002/em.22323Search in Google Scholar PubMed
[99] Wilson CL, Natarajan V, Hayward SL, Khalimonchuk O, Kidambi S. Mitochondrial dysfunction and loss of glutamate uptake in primary astrocytes exposed to titanium dioxide nanoparticles. Nanoscale. 2015;7(44):18477–88.10.1039/C5NR03646ASearch in Google Scholar
[100] Turovsky EA, Mal’tseva VN, Sarimov RM, Simakin AV, Gudkov SV, Plotnikov EY. Features of the cytoprotective effect of selenium nanoparticles on primary cortical neurons and astrocytes during oxygen-glucose deprivation and reoxygenation. Sci Rep-UK. 2022;12(1):1710.10.1038/s41598-022-05674-1Search in Google Scholar PubMed PubMed Central
[101] Surnar B, Shah AS, Park M, Kalathil AA, Kamran MZ, Ramirez, Jaime R, et al. Brain-accumulating nanoparticles for assisting astrocytes to reduce human immunodeficiency virus and drug abuse-induced neuroinflammation and oxidative stress. ACS Nano. 2021;15(10):15741–53.10.1021/acsnano.0c09553Search in Google Scholar PubMed PubMed Central
[102] Thomsen MS, Routhe LJ, Moos T. The vascular basement membrane in the healthy and pathological brain. J Cereb Blood Flow Metab. 2017;37(10):3300–17.10.1177/0271678X17722436Search in Google Scholar PubMed PubMed Central
[103] Dejana E. Endothelial cell-cell junctions: Happy together. Nat Rev Mol Cell Bio. 2004;5(4):261–70.10.1038/nrm1357Search in Google Scholar PubMed
[104] Gonzalez-Carter D, Liu X, Tockary TA, Dirisala A, Toh K, Anraku Y, et al. Targeting nanoparticles to the brain by exploiting the blood-brain barrier impermeability to selectively label the brain endothelium. Proc Natl Acad Sci U S A. 2020;117(32):19141–50.10.1073/pnas.2002016117Search in Google Scholar PubMed PubMed Central
[105] Eldridge BN, Xing F, Fahrenholtz CD, Singh RN. Evaluation of multiwalled carbon nanotube cytotoxicity in cultures of human brain microvascular endothelial cells grown on plastic or basement membrane. Toxicol In Vitro. 2017;41:223–31.10.1016/j.tiv.2017.03.002Search in Google Scholar PubMed PubMed Central
[106] Rassu G, Soddu E, Posadino AM, Pintus G, Sarmento B, Giunchedi P, et al. Nose-to-brain delivery of BACE1 siRNA loaded in solid lipid nanoparticles for Alzheimer’s therapy. Colloids Surf B Biointerfaces. 2017;152:296–301.10.1016/j.colsurfb.2017.01.031Search in Google Scholar PubMed
[107] Khan NEEL, Ni J, Ju XF, Miao TT, Chen HY, Han L. Escape from abluminal LRP1-mediated clearance for boosted nanoparticle brain delivery and brain metastasis treatment. Acta Pharm Sin B. 2021;11(5):1341–54.10.1016/j.apsb.2020.10.015Search in Google Scholar PubMed PubMed Central
[108] Gao JQ, Lv Q, Li LM, Tang XJ, Li FZ, Hu YL, et al. Glioma targeting and blood-brain barrier penetration by dual-targeting doxorubincin liposomes. Biomaterials. 2013;34(22):5628–39.10.1016/j.biomaterials.2013.03.097Search in Google Scholar PubMed
[109] Markoutsa E, Pampalakis G, Niarakis A, Romero IA, Weksler B, Couraud PO, et al. Uptake and permeability studies of BBB-targeting immunoliposomes using the hCMEC/D3 cell line. Eur J Pharm Biopharm. 2011;77(2):265–74.10.1016/j.ejpb.2010.11.015Search in Google Scholar PubMed
[110] Morris AWJ, Sharp MM, Albargothy NJ, Fernandes R, Hawkes CA, Verma A, et al. Vascular basement membranes as pathways for the passage of fluid into and out of the brain. Acta Neuropathol. 2016;131(5):725–36.10.1007/s00401-016-1555-zSearch in Google Scholar PubMed PubMed Central
[111] Li CH, Shyu MK, Jhan C, Cheng YW, Tsai CH, Liu CW, et al. Gold nanoparticles increase endothelial paracellular permeability by altering components of endothelial tight junctions, and increase blood-brain barrier permeability in mice. Toxicol Sci. 2015;148(1):192–203.10.1093/toxsci/kfv176Search in Google Scholar PubMed
[112] Fresta CG, Chakraborty A, Wijesinghe MB, Amorini AM, Lazzarino G, Lazzarino G, et al. Non-toxic engineered carbon nanodiamond concentrations induce oxidative/nitrosative stress, imbalance of energy metabolism, and mitochondrial dysfunction in microglial and alveolar basal epithelial cells. Cell Death Dis. 2018;9(2):245.10.1038/s41419-018-0280-zSearch in Google Scholar PubMed PubMed Central
[113] Zhu MT, Wang B, Wang Y, Yuan L, Wang HJ, Wang M, et al. Endothelial dysfunction and inflammation induced by iron oxide nanoparticle exposure: Risk factors for early atherosclerosis. Toxicol Lett. 2011;203(2):162–71.10.1016/j.toxlet.2011.03.021Search in Google Scholar PubMed
[114] Liu X, Sun JA. Endothelial cells dysfunction induced by silica nanoparticles through oxidative stress via JNK/P53 and NF-kappa B pathways. Biomaterials. 2010;31(32):8198–209.10.1016/j.biomaterials.2010.07.069Search in Google Scholar PubMed
[115] Engin AB, Neagu M, Golokhvast K, Tsatsakis A. Nanoparticles and endothelium: an update on the toxicological interactions. Farmacia. 2015;63:792–804.Search in Google Scholar
[116] Ye D, Raghnaill MN, Bramini M, Mahon E, Åberg C, Salvati A, et al. Nanoparticle accumulation and transcytosis in brain endothelial cell layers. Nanoscale. 2013;5(22):11153–65.10.1039/c3nr02905kSearch in Google Scholar PubMed
[117] Armulik A, Genove G, Mae M, Nisancioglu MH, Wallgard E, Niaudet C, et al. Pericytes regulate the blood-brain barrier. Nature. 2010;468(7323):557–61.10.1038/nature09522Search in Google Scholar PubMed
[118] Montagne A, Barnes SR, Sweeney MD, Halliday MR, Sagare AP, Zhao Z, et al. Blood-Brain Barrier breakdown in the aging human hippocampus. Neuron. 2015;85(2):296–302.10.1016/j.neuron.2014.12.032Search in Google Scholar PubMed PubMed Central
[119] Price TO, Eranki V, Banks WA, Ercal N, Shah GN. Topiramate treatment protects blood-brain barrier pericytes from hyperglycemia-induced oxidative damage in diabetic mice. Endocrinology. 2012;153(1):362–72.10.1210/en.2011-1638Search in Google Scholar PubMed PubMed Central
[120] Salameh TS, Shah GN, Price TO, Hayden MR, Banks WA. Blood-Brain Barrier disruption and neurovascular unit dysfunction in diabetic mice: protection with the mitochondrial carbonic anhydrase inhibitor topiramate. J Pharmacol Exp Ther. 2016;359(3):452–9.10.1124/jpet.116.237057Search in Google Scholar PubMed PubMed Central
[121] Brown LS, Foster CG, Courtney JM, King NE, Howells DW, Sutherland BA. Pericytes and Neurovascular Function in the Healthy and Diseased Brain. Front Cell Neurosci. 2019;13:282.10.3389/fncel.2019.00282Search in Google Scholar PubMed PubMed Central
[122] Skaper SD. Oligodendrocyte precursor cells as a therapeutic target for demyelinating diseases. Prog Brain Res. 2019;245:119–44.10.1016/bs.pbr.2019.03.013Search in Google Scholar PubMed
[123] Gomez-Pinedo U, Matias-Guiu JA, Benito-Martin MS, Moreno-Jimenez L, Sanclemente-Alaman I, Selma-Calvo B, et al. Intranasal administration of undifferentiated oligodendrocyte lineage cells as a potential approach to deliver oligodendrocyte precursor cells into brain. Int J Mol Sci. 2021;22(19):10738.10.3390/ijms221910738Search in Google Scholar PubMed PubMed Central
[124] Rittchen S, Boy A, Burns A, Park J, Fahmy TM, Metcalfe S, et al. Myelin repair in vivo is increased by targeting oligodendrocyte precursor cells with nanoparticles encapsulating leukaemia inhibitory factor (LIF). Biomaterials. 2015;56:78–85.10.1016/j.biomaterials.2015.03.044Search in Google Scholar PubMed PubMed Central
[125] Youssef AEH, Dief AE, El Azhary NM, Abdelmonsif DA, El-fetiany OS. LINGO-1 siRNA nanoparticles promote central remyelination in ethidium bromide-induced demyelination in rats. J Physiol Biochem. 2019;75(1):89–99.10.1007/s13105-018-00660-6Search in Google Scholar PubMed
[126] Carradori D, Labrak Y, Miron VE, Saulnier P, Eyer J, Preat V, et al. Retinoic acid-loaded NFL-lipid nanocapsules promote oligodendrogenesis in focal white matter lesion. Biomaterials. 2020;230:119653.10.1016/j.biomaterials.2019.119653Search in Google Scholar PubMed
[127] Osorio-Querejeta I, Carregal-Romero S, Ayerdi-Izquierdo A, Mager I, Nash LA, Wood M, et al. MiR-219a-5p enriched extracellular vesicles induce opc differentiation and EAE improvement more efficiently than liposomes and polymeric nanoparticles. Pharmaceutics. 2020;12(2):186.10.3390/pharmaceutics12020186Search in Google Scholar PubMed PubMed Central
[128] Li XW, Tzeng SY, Zamboni CG, Koliatsos VE, Ming GL, Green JJ, et al. Enhancing oligodendrocyte differentiation by transient transcription activation via DNA nanoparticle-mediated transfection. Acta Biomater. 2017;54:249–58.10.1016/j.actbio.2017.03.032Search in Google Scholar PubMed PubMed Central
[129] Jenkins SI, Weinberg D, al-Shakli AF, Fernandes AR, Yiu HHP, Telling ND, et al. ‘Stealth’ nanoparticles evade neural immune cells but also evade major brain cell populations: Implications for PEG-based neurotherapeutics. J Control Release. 2016;224:136–45.10.1016/j.jconrel.2016.01.013Search in Google Scholar PubMed
[130] Sruthi S, Maurizi L, Nury T, Sallem F, Boudon J, Riedinger JM, et al. Cellular interactions of functionalized superparamagnetic iron oxide nanoparticles on oligodendrocytes without detrimental side effects: Cell death induction, oxidative stress and inflammation. Colloid Surf B. 2018;170:454–62.10.1016/j.colsurfb.2018.06.041Search in Google Scholar PubMed
[131] Nance E. Brain-penetrating nanoparticles for analysis of the brain microenvironment. Methods Mol Biol. 2017;1570:91–104.10.1007/978-1-4939-6840-4_6Search in Google Scholar PubMed
[132] Song E, Gaudin A, King AR, Seo YE, Suh HW, Deng Y, et al. Surface chemistry governs cellular tropism of nanoparticles in the brain. Nat Commun. 2017;8:15322.10.1038/ncomms15322Search in Google Scholar PubMed PubMed Central
[133] Bharadwaj VN, Nguyen DT, Kodibagkar VD, Stabenfeldt SE. Nanoparticle-based therapeutics for brain injury. Adv Healthc Mater. 2018;7(1):10.10.1002/adhm.201700668Search in Google Scholar PubMed PubMed Central
[134] Lu YF, Guo ZY, Zhang YJ, Li C, Zhang Y, Guo Q, et al. Microenvironment remodeling micelles for Alzheimer’s Disease therapy by early modulation of activated microglia. Adv Sci. 2019;6(4):1801586.10.1002/advs.201801586Search in Google Scholar PubMed PubMed Central
[135] Shi J, Yang Y, Yin N, Liu C, Zhao Y, Cheng H, et al. Engineering CXCL12 biomimetic decoy-integrated versatile immunosuppressive nanoparticle for ischemic stroke therapy with management of overactivated brain immune microenvironment. Small Methods. 2022;6(1):e2101158.10.1002/smtd.202101158Search in Google Scholar PubMed
[136] Wang X, Zhang Q, Lv L, Fu J, Jiang Y, Xin H, et al. Glioma and microenvironment dual targeted nanocarrier for improved antiglioblastoma efficacy. Drug Deliv. 2017;24(1):1401–9.10.1080/10717544.2017.1378940Search in Google Scholar PubMed PubMed Central
[137] Tosi G, Vilella A, Chhabra R, Schmeisser MJ, Boeckers TM, Ruozi B, et al. Insight on the fate of CNS-targeted nanoparticles. Part II: Intercellular neuronal cell-to-cell transport. J Control Release. 2014;177:96–107.10.1016/j.jconrel.2014.01.004Search in Google Scholar PubMed
[138] Gitler AD, Dhillon P, Shorter J. Neurodegenerative disease: models, mechanisms, and a new hope. Dis Model Mech. 2017;10(5):499–502.10.1242/dmm.030205Search in Google Scholar PubMed PubMed Central
[139] Sochocka M, Diniz BS, Leszek J. Inflammatory response in the CNS: friend or foe? Mol Neurobiol. 2017;54(10):8071–89.10.1007/s12035-016-0297-1Search in Google Scholar PubMed PubMed Central
[140] Wyss-Coray T. Inflammation in Alzheimer disease: driving force, bystander or beneficial response? Nat Med. 2006;12(9):1005–15.Search in Google Scholar
[141] Machado-Santos J, Saji E, Troscher AR, Paunovic M, Liblau R, Gabriely G, et al. The compartmentalized inflammatory response in the multiple sclerosis brain is composed of tissue-resident CD8+T lymphocytes and B cells. Brain. 2018;141(7):2066–82.10.1093/brain/awy151Search in Google Scholar PubMed PubMed Central
[142] Salter MW, Stevens B. Microglia emerge as central players in brain disease. Nat Med. 2017;23(9):1018–27.10.1038/nm.4397Search in Google Scholar PubMed
[143] Graeber MB, Streit WJ. Microglia: Immune network in the CNS. Brain Pathol. 1990;1(1):2–5.10.1111/j.1750-3639.1990.tb00630.xSearch in Google Scholar PubMed
[144] Streit WJ, Graeber MB, Kreutzberg GW. Functional plasticity of microglia – a review. Glia. 1988;1(5):301–7.10.1002/glia.440010502Search in Google Scholar PubMed
[145] Umlauf BJ, Shusta EV. Exploiting BBB disruption for the delivery of nanocarriers to the diseased CNS. Curr Opin Biotechnol. 2019;60:146–52.10.1016/j.copbio.2019.01.013Search in Google Scholar PubMed PubMed Central
[146] Khan FA, Almohazey D, Alomari M, Almofty SA. Impact of nanoparticles on neuron biology: current research trends. Int J Nanomed. 2018;13:2767–76.10.2147/IJN.S165675Search in Google Scholar PubMed PubMed Central
[147] Calne DB, Langston JW. Aetiology of Parkinson’s disease. Lancet. 1983;386(9996):896–912.10.1016/S0140-6736(14)61393-3Search in Google Scholar PubMed
[148] Candel PE, Córcoles JMP, Delicado UV, Hernández MA, Molina AM. Relationship between motor and nonmotor symptoms and quality of life in patients with parkinson’s disease. Nurs Repo. 2021;12(1):1–12.10.3390/nursrep12010001Search in Google Scholar PubMed PubMed Central
[149] Ozansoy M, Başak AN. The central theme of Parkinson’s disease: α-synuclein. Mol Neurobiol. 2013;47(2):460–5.10.1007/s12035-012-8369-3Search in Google Scholar PubMed
[150] Armstrong MJ, Okun MS. Diagnosis and treatment of Parkinson Disease: areview. JAMA. 2020;323(6):548–60.10.1001/jama.2019.22360Search in Google Scholar PubMed
[151] Sardi SP, Cedarbaum JM, Brundin P. Targeted therapies for Parkinson’s Disease: from genetics to the clinic. Mov Disord. 2018;33(5):684–96.10.1002/mds.27414Search in Google Scholar PubMed PubMed Central
[152] Pillay S, Pillay V, Choonara YE, Naidoo D, Khan RA, du Toit LC, et al. Design, biometric simulation and optimization of a nano-enabled scaffold device for enhanced delivery of dopamine to the brain. Int J Pharm. 2009;382(1–2):277–90.10.1016/j.ijpharm.2009.08.021Search in Google Scholar PubMed
[153] Esteves M, Cristovao AC, Saraiva T, Rocha SM, Baltazar G, Ferreira L, et al. Retinoic acid-loaded polymeric nanoparticles induce neuroprotection in a mouse model for Parkinson’s disease. Front Aging Neurosci. 2015;7:20.10.3389/fnagi.2015.00020Search in Google Scholar PubMed PubMed Central
[154] Tsai YM, Chien CF, Lin LC, Tsai TH. Curcumin and its nano-formulation: the kinetics of tissue distribution and blood-brain barrier penetration. Int J Pharm. 2011;416(1):331–8.10.1016/j.ijpharm.2011.06.030Search in Google Scholar PubMed
[155] Tan JPK, Voo ZX, Lim S, Venkataraman S, Ng KM, Gao SJ, et al. Effective encapsulation of apomorphine into biodegradable polymeric nanoparticles through a reversible chemical bond for delivery across the blood-brain barrier. Nanomed Nanotechnol. 2019;17:236–45.10.1016/j.nano.2019.01.014Search in Google Scholar PubMed
[156] Bourdenx M, Daniel J, Genin E, Soria FN, Blanchard-Desce M, Bezard E, et al. Nanoparticles restore lysosomal acidification defects: Implications for Parkinson and other lysosomal-related diseases. Autophagy. 2016;12(3):472–83.10.1080/15548627.2015.1136769Search in Google Scholar PubMed PubMed Central
[157] Raj R, Wairkar S, Sridhar V, Gaud R. Pramipexole dihydrochloride loaded chitosan nanoparticles for nose to brain delivery: Development, characterization and in vivo anti-Parkinson activity. Int Biol Macromol. 2018;109:27–35.10.1016/j.ijbiomac.2017.12.056Search in Google Scholar PubMed
[158] Yan X, Xu L, Bi C, Duan D, Chu L, Yu X, et al. Lactoferrin-modified rotigotine nanoparticles for enhanced nose-to-brain delivery: LESA-MS/MS-based drug biodistribution, pharmacodynamics, and neuroprotective effects. Int J Nanomedicine. 2018;13:273–81.10.2147/IJN.S151475Search in Google Scholar PubMed PubMed Central
[159] Zhang NS, Ya F, Liang XL, Wu MX, Shen YY, Chen M, et al. Localized delivery of curcumin into brain with polysorbate 80-modified cerasomes by ultrasound-targeted microbubble destruction for improved Parkinson’s disease therapy. Theranostics. 2018;8(8):2264–77.10.7150/thno.23734Search in Google Scholar PubMed PubMed Central
[160] Duwa R, Jeong JH, Yook S. Development of immunotherapy and nanoparticles-based strategies for the treatment of Parkinson’s disease. J Pharm Invest. 2021;51:465–81.10.1007/s40005-021-00521-3Search in Google Scholar
[161] Lane CA, Hardy J, Schott JM. Alzheimer’s disease. Eur J Neurol. 2018;25:59–70.10.1111/ene.13439Search in Google Scholar PubMed
[162] Soria Lopez JA, González HM, Léger GC. Alzheimer’s disease. Handb Clin Neurol. 2019;167:231–55.10.1016/B978-0-12-804766-8.00013-3Search in Google Scholar PubMed
[163] Bellenguez C, Grenier-Boley B, Lambert JC. Genetics of Alzheimer’s disease: Where we are, and where we are going. Curr Opin Neurobiol. 2020;61:40–8.10.1016/j.conb.2019.11.024Search in Google Scholar PubMed
[164] LaFerla FM, Green KN, Oddo S. Intracellular amyloid-beta in Alzheimer’s disease. Nat Rev Neurosci. 2007;8(7):499–509.10.1038/nrn2168Search in Google Scholar PubMed
[165] Dubois B, Feldman HH, Jacova C, Cummings JL, DeKosky ST, Barberger-Gateau P, et al. Revising the definition of Alzheimer’s disease: a new lexicon. Lancet Neurol. 2010;9(11):1118–27.10.1016/S1474-4422(10)70223-4Search in Google Scholar PubMed
[166] Zhang L, Zhao PH, Yue CP, Jin ZK, Liu Q, Du XB, et al. Sustained release of bioactive hydrogen by Pd hydride nanoparticles overcomes Alzheimer’s disease. Biomaterials. 2019;197:393–404.10.1016/j.biomaterials.2019.01.037Search in Google Scholar PubMed
[167] Jeon SG, Cha MY, Kim JI, Hwang TW, Kim KA, Kim TH, et al. Vitamin D-binding protein-loaded PLGA nanoparticles suppress Alzheimer’s disease-related pathology in 5XFAD mice. Nanomed Nanotechnol. 2019;17:297–307.10.1016/j.nano.2019.02.004Search in Google Scholar PubMed
[168] Fernandes J, Ghate MV, Mallik SB, Lewis SA. Amino acid conjugated chitosan nanoparticles for the brain targeting of a model dipeptidyl peptidase-4 inhibitor. Int J Pharm. 2018;547(1–2):563–71.10.1016/j.ijpharm.2018.06.031Search in Google Scholar PubMed
[169] Carradori D, Balducci C, Re F, Brambilla D, Le Droumaguet B, Flores O, et al. Antibody-functionalized polymer nanoparticle leading to memory recovery in Alzheimer’s disease-like transgenic mouse model. Nanomed Nanotechnol. 2018;14(2):609–18.10.1016/j.nano.2017.12.006Search in Google Scholar PubMed
[170] Chen L, Du Y, Zhang K, Liang ZY, Li JQ, Yu H, et al. Tau-targeted multifunctional nanocomposite for combinational therapy of Alzheimer’s disease. ACS Nano. 2018;12(2):1321–38.10.1021/acsnano.7b07625Search in Google Scholar PubMed
[171] Sun J, Wei CF, Liu YA, Xie WJ, Xu MM, Zhou H, et al. Progressive release of mesoporous nano-selenium delivery system for the multi-channel synergistic treatment of Alzheimer’s disease. Biomaterials. 2019;197:417–31.10.1016/j.biomaterials.2018.12.027Search in Google Scholar PubMed
[172] Sanchez-Lopez E, Ettcheto M, Egea MA, Espina M, Cano A, Calpena AC, et al. Memantine loaded PLGA PEGylated nanoparticles for Alzheimer’s disease: in vitro and in vivo characterization. J Nanobiotechnol. 2018;16(1):32.10.1186/s12951-018-0356-zSearch in Google Scholar PubMed PubMed Central
[173] Silva-Abreu M, Calpena AC, Andres-Benito P, Aso E, Romero IA, Roig-Carles D, et al. PPAR gamma agonist-loaded PLGA-PEG nanocarriers as a potential treatment for Alzheimer’s disease: in vitro and in vivo studies. Int J Nanomed. 2018;13:5577–90.10.2147/IJN.S171490Search in Google Scholar PubMed PubMed Central
[174] Ahmed A, Ghallab EH, Shehata M, Zekri AN, Ahmed OS. Impact of nano-conjugate on Drosophila for early diagnosis of Alzheimer’s disease. Nanotechnology. 2020;31(36):365102.10.1088/1361-6528/ab7535Search in Google Scholar PubMed
[175] Zhang K, Yang Q, Fan Z, Zhao J, Li H. Platelet-driven formation of interface peptide nano-network biosensor enabling a non-invasive means for early detection of Alzheimer’s disease. Biosens Bioelectron. 2019;145:111701.10.1016/j.bios.2019.111701Search in Google Scholar PubMed
[176] Lassmann H, van Horssen J, Mahad D. Progressive multiple sclerosis: pathology and pathogenesis. Nat Rev Neurol. 2012;8(11):647–56.10.1038/nrneurol.2012.168Search in Google Scholar PubMed
[177] Lassmann H, Bruck W, Lucchinetti C. The immunopathology of multiple sclerosis: An overview. Brain Pathol. 2007;17(2):210–8.10.1111/j.1750-3639.2007.00064.xSearch in Google Scholar PubMed PubMed Central
[178] Trapp BD, Nave KA. Multiple sclerosis: An immune or neurodegenerative disorder? Annu Rev Neurosci. 2008;31:247–69.10.1146/annurev.neuro.30.051606.094313Search in Google Scholar PubMed
[179] Nally FK, De Santi C, McCoy CE. Nanomodulation of macrophages in multiple sclerosis. Cells. 2019;8(6):543.10.3390/cells8060543Search in Google Scholar PubMed PubMed Central
[180] Dolati S, Babaloo Z, Jadidi-Niaragh F, Ayromlou H, Sadreddini S, Yousefi M. Multiple sclerosis: Therapeutic applications of advancing drug delivery systems. Biomed Pharmacother. 2017;86:343–53.10.1016/j.biopha.2016.12.010Search in Google Scholar PubMed
[181] Belogurov AA Jr, Stepanov AV, Smirnov IV, Melamed D, Bacon A, Mamedov AE, et al. Liposome-encapsulated peptides protect against experimental allergic encephalitis. FASEB J. 2013;27(1):222–31.10.1096/fj.12-213975Search in Google Scholar PubMed PubMed Central
[182] Kumar P, Sharma G, Gupta V, Kaur R, Thakur K, Malik R, et al. Preclinical explorative assessment of dimethyl fumarate-based biocompatible nanolipoidal carriers for the management of Multiple Sclerosis. ACS Chem Neurosci. 2018;9(5):1152–8.10.1021/acschemneuro.7b00519Search in Google Scholar PubMed
[183] Naeimi R, Safarpour F, Hashemian M, Tashakorian H, Ahmadian SR, Ashrafpour M, et al. Curcumin-loaded nanoparticles ameliorate glial activation and improve myelin repair in lyolecithin-induced focal demyelination model of rat corpus callosum. Neurosci Lett. 2018;674:1–10.10.1016/j.neulet.2018.03.018Search in Google Scholar PubMed
[184] Montes-Cobos E, Ring S, Fischer HJ, Heck J, Strauss J, Schwaninger M, et al. Targeted delivery of glucocorticoids to macrophages in a mouse model of multiple sclerosis using inorganic-organic hybrid nanoparticles. J Control Release. 2017;245:157–69.10.1016/j.jconrel.2016.12.003Search in Google Scholar PubMed
[185] McCarthy DP, Yap JWT, Harp CT, Song WK, Chen J, Pearson RM, et al. An antigen-encapsulating nanoparticle platform for T(H)1/17 immune tolerance therapy. Nanomed Nanotechnol. 2017;13(1):191–200.10.1016/j.nano.2016.09.007Search in Google Scholar PubMed PubMed Central
[186] Pearson RM, Casey LM, Hughes KR, Wang LZ, North MG, Getts DR, et al. Controlled delivery of single or multiple antigens in tolerogenic nanoparticles using peptide-polymer bioconjugates. Mol Ther. 2017;25(7):1655–64.10.1016/j.ymthe.2017.04.015Search in Google Scholar PubMed PubMed Central
[187] Kuo R, Saito E, Miller SD, Shea LD. Peptide-conjugated nanoparticles reduce positive co-stimulatory expression and T cell activity to induce tolerance. Mol Ther. 2017;25(7):1676–85.10.1016/j.ymthe.2017.03.032Search in Google Scholar PubMed PubMed Central
[188] Rothstein JD. Current hypotheses for the underlying biology of Amyotrophic Lateral Sclerosis. Ann Neurol. 2009;65(Suppl 1):S3–9.10.1002/ana.21543Search in Google Scholar PubMed
[189] Gordon PH. Amyotrophic Lateral Sclerosis: an update for 2013 clinical features, pathophysiology, management and therapeutic trials. Aging Dis. 2013;4(5):295–310.10.14336/AD.2013.0400295Search in Google Scholar PubMed PubMed Central
[190] McGeer PL, McGeer EG. Inflammatory processes in amyotrophic lateral sclerosis. Muscle Nerve. 2002;26(4):459–70.10.1002/mus.10191Search in Google Scholar PubMed
[191] DeCoteau W, Heckman KL, Estevez AY, Reed KJ, Costanzo W, Sandford D, et al. Cerium oxide nanoparticles with antioxidant properties ameliorate strength and prolong life in mouse model of amyotrophic lateral sclerosis. Nanomed Nano Technol. 2016;12(8):2311–20.10.1016/j.nano.2016.06.009Search in Google Scholar PubMed
[192] Langhorne P, Bernhardt J, Kwakkel G. Stroke rehabilitation. Lancet. 2011;377(9778):1693–702.10.1016/S0140-6736(11)60325-5Search in Google Scholar PubMed
[193] Bonita R, Mendis S, Truelsen T, Bogousslavsky J, Toole J, Yatsu F. The global stroke initiative. Lancet Neurol. 2004;3(7):391–3.10.1016/S1474-4422(04)00800-2Search in Google Scholar PubMed
[194] Donnan GA, Fisher M, Macleod M, Davis SM. Stroke. Lancet. 2008;371(9624):1612–23.10.1016/S0140-6736(08)60694-7Search in Google Scholar PubMed
[195] Sandoval KE, Witt KA. Blood-brain barrier tight junction permeability and ischemic stroke. Neurobiol Dis. 2008;32(2):200–19.10.1016/j.nbd.2008.08.005Search in Google Scholar PubMed
[196] Kyle S, Saha S. Nanotechnology for the detection and therapy of stroke. Adv Healthc Mater. 2014;3(11):1703–20.10.1002/adhm.201400009Search in Google Scholar PubMed
[197] Chen L, Gao X. The application of nanoparticles for neuroprotection in acute ischemic stroke. Ther Deliv. 2017;8(10):915–28.10.4155/tde-2017-0023Search in Google Scholar PubMed
[198] Landowski LM, Niego B, Sutherland BA, Hagemeyer CE, Howells DW. Applications of nanotechnology in the diagnosis and therapy of stroke. Semin Thromb Hemost. 2020;46(5):592–605.10.1055/s-0039-3399568Search in Google Scholar PubMed
[199] Simpkins AN, Janowski M, Oz HS, Roberts J, Bix G, Dore S, et al. Biomarker application for precision medicine in stroke. Transl Stroke Res. 2020;11(4):615–27.10.1007/s12975-019-00762-3Search in Google Scholar PubMed PubMed Central
[200] Jickling GC, Sharp FR. Biomarker panels in ischemic stroke. Stroke. 2015;46(3):915–20.10.1161/STROKEAHA.114.005604Search in Google Scholar PubMed PubMed Central
[201] Das NM, Hatsell S, Nannuru K, Huang L, Wen X, Wang L, et al. In vivo quantitative microcomputed tomographic analysis of vasculature and organs in a normal and diseased mouse model. PLoS One. 2016;11(2):e0150085.10.1371/journal.pone.0150085Search in Google Scholar PubMed PubMed Central
[202] Qin J, Zhou S, Li Z, Chen Y, Qin Q, Ai T. Combination of magnetic resonance imaging and targeted contrast agent for the diagnosis of myocardial infarction. Exp Ther Med. 2018;16(4):3303–8.10.3892/etm.2018.6600Search in Google Scholar PubMed PubMed Central
[203] Toth GB, Varallyay CG, Horvath A, Bashir MR, Choyke PL, Daldrup-Link HE, et al. Current and potential imaging applications of ferumoxytol for magnetic resonance imaging. Kidney Int. 2017;92(1):47–66.10.1016/j.kint.2016.12.037Search in Google Scholar PubMed PubMed Central
[204] Joo J, Liu X, Kotamraju VR, Ruoslahti E, Nam Y, Sailor MJ. Gated luminescence imaging of silicon nanoparticles. ACS Nano. 2015;9(6):6233–41.10.1021/acsnano.5b01594Search in Google Scholar PubMed PubMed Central
[205] Kim T, Lemaster JE, Chen F, Li J, Jokerst JV. Photoacoustic imaging of human mesenchymal stem cells labeled with prussian blue-poly(l-lysine) nanocomplexes. ACS Nano. 2017;11(9):9022–32.10.1021/acsnano.7b03519Search in Google Scholar PubMed PubMed Central
[206] Farr TD, Lai CH, Grunstein D, Orts-Gil G, Wang CC, Boehm-Sturm P, et al. Imaging early endothelial inflammation following stroke by core shell silica superparamagnetic glyconanoparticles that target selectin. Nano Lett. 2014;14(4):2130–4.10.1021/nl500388hSearch in Google Scholar PubMed
[207] Luke GP, Hannah AS, Emelianov SY. Super-resolution ultrasound Imaging in vivo with Transient laser-activated nanodroplets. Nano Lett. 2016;16(4):2556–9.10.1021/acs.nanolett.6b00108Search in Google Scholar PubMed PubMed Central
[208] Brea D, Agulla J, Staes A, Gevaert K, Campos F, Sobrino T, et al. Study of protein expression in peri-infarct tissue after cerebral Ischemia. Sci Rep. 2015;5:12030.10.1038/srep12030Search in Google Scholar PubMed PubMed Central
[209] Hoyte LC, Brooks KJ, Nagel S, Akhtar A, Chen R, Mardiguian S, et al. Molecular magnetic resonance imaging of acute vascular cell adhesion molecule-1 expression in a mouse model of cerebral ischemia. J Cereb Blood Flow Metab. 2010;30(6):1178–87.10.1038/jcbfm.2009.287Search in Google Scholar PubMed PubMed Central
[210] Lin KY, Kwong GA, Warren AD, Wood DK, Bhatia SN. Nanoparticles that sense thrombin activity as synthetic urinary biomarkers of thrombosis. ACS Nano. 2013;7(10):9001–9.10.1021/nn403550cSearch in Google Scholar PubMed PubMed Central
[211] Sun Y, Gao W, Zhao Y, Cao W, Liu Z, Cui G, et al. Visualization and inhibition of mitochondria-nuclear translocation of apoptosis inducing factor by a graphene oxide-DNA nanosensor. Anal Chem. 2017;89(8):4642–7.10.1021/acs.analchem.7b00221Search in Google Scholar PubMed
[212] Gong T, Hong ZY, Chen CH, Tsai CY, Liao LD, Kong KV, et al. Optical interference-free surface-enhanced raman scattering CO-nanotags for logical multiplex detection of vascular disease-related biomarkers. ACS Nano. 2017;11(3):3365–75.10.1021/acsnano.7b00733Search in Google Scholar PubMed
[213] Hu Y, Cheng H, Zhao X, Wu J, Muhammad F, Lin S, et al. Surface-enhanced raman scattering active gold nanoparticles with enzyme-mimicking activities for measuring glucose and lactate in living tissues. ACS Nano. 2017;11(6):5558–66.10.1021/acsnano.7b00905Search in Google Scholar PubMed
[214] Molina CA, Barreto AD, Tsivgoulis G, Sierzenski P, Malkoff MD, Rubiera M, et al. Transcranial ultrasound in clinical sonothrombolysis (TUCSON) trial. Ann Neurol. 2009;66(1):28–38.10.1002/ana.21723Search in Google Scholar PubMed
[215] Li M, Liu Y, Chen J, Liu T, Gu Z, Zhang J, et al. Platelet bio-nanobubbles as microvascular recanalization nanoformulation for acute ischemic stroke lesion theranostics. Theranostics. 2018;8(18):4870–83.10.7150/thno.27466Search in Google Scholar PubMed PubMed Central
[216] Kim JY, Ryu JH, Schellingerhout D, Sun IC, Lee SK, Jeon S, et al. Direct imaging of cerebral thromboemboli using computed tomography and fibrin-targeted gold nanoparticles. Theranostics. 2015;5(10):1098–114.10.7150/thno.11679Search in Google Scholar PubMed PubMed Central
[217] Kim JY, Kim JK, Park JS, Byun Y, Kim CK. The use of PEGylated liposomes to prolong circulation lifetimes of tissue plasminogen activator. Biomaterials. 2009;30(29):5751–6.10.1016/j.biomaterials.2009.07.021Search in Google Scholar PubMed
[218] Chung TW, Wang SS, Tsai WJ. Accelerating thrombolysis with chitosan-coated plasminogen activators encapsulated in poly-(lactide-co-glycolide) (PLGA) nanoparticles. Biomaterials. 2008;29(2):228–37.10.1016/j.biomaterials.2007.09.027Search in Google Scholar PubMed
[219] Huang Y, Yu L, Ren J, Gu B, Longstaff C, Hughes AD, et al. An activated-platelet-sensitive nanocarrier enables targeted delivery of tissue plasminogen activator for effective thrombolytic therapy. J Control Release. 2019;300:1–12.10.1016/j.jconrel.2019.02.033Search in Google Scholar PubMed
[220] Chen JP, Yang PC, Ma YH, Tu SJ, Lu YJ. Targeted delivery of tissue plasminogen activator by binding to silica-coated magnetic nanoparticle. Int J Nanomedicine. 2012;7:5137–49.10.2147/IJN.S36197Search in Google Scholar PubMed PubMed Central
[221] Fukuta T, Asai T, Yanagida Y, Namba M, Koide H, Shimizu K, et al. Combination therapy with liposomal neuroprotectants and tissue plasminogen activator for treatment of ischemic stroke. FASEB J. 2017;31(5):1879–90.10.1096/fj.201601209RSearch in Google Scholar PubMed
[222] Fukuta T, Yanagida Y, Asai T, Oku N. Co-administration of liposomal fasudil and tissue plasminogen activator ameliorated ischemic brain damage in occlusion model rats prepared by photochemically induced thrombosis. Biochem Biophys Res Commun. 2018;495(1):873–7.10.1016/j.bbrc.2017.11.107Search in Google Scholar PubMed
[223] Bitner BR, Marcano DC, Berlin JM, Fabian RH, Cherian L, Culver JC, et al. Antioxidant carbon particles improve cerebrovascular dysfunction following traumatic brain injury. ACS Nano. 2012;6(9):8007–14.10.1021/nn302615fSearch in Google Scholar PubMed PubMed Central
[224] Petro M, Jaffer H, Yang J, Kabu S, Morris VB, Labhasetwar V. Tissue plasminogen activator followed by antioxidant-loaded nanoparticle delivery promotes activation/mobilization of progenitor cells in infarcted rat brain. Biomaterials. 2016;81:169–80.10.1016/j.biomaterials.2015.12.009Search in Google Scholar PubMed PubMed Central
[225] Lv W, Xu J, Wang X, Li X, Xu Q, Xin H. Bioengineered boronic ester modified dextran polymer nanoparticles as reactive oxygen species responsive nanocarrier for ischemic stroke treatment. ACS Nano. 2018;12(6):5417–26.10.1021/acsnano.8b00477Search in Google Scholar PubMed
[226] Shen Y, Cao B, Snyder NR, Woeppel KM, Eles JR, Cui XT. ROS responsive resveratrol delivery from LDLR peptide conjugated PLA-coated mesoporous silica nanoparticles across the blood-brain barrier. J Nanobiotechnol. 2018;16(1):13.10.1186/s12951-018-0340-7Search in Google Scholar PubMed PubMed Central
[227] Kaviarasi S, Yuba E, Harada A, Krishnan UM. Emerging paradigms in nanotechnology for imaging and treatment of cerebral ischemia. J Control Release. 2019;300:22–45.10.1016/j.jconrel.2019.02.031Search in Google Scholar PubMed
[228] Wang Y, Li SY, Shen S, Wang J. Protecting neurons from cerebral ischemia/reperfusion injury via nanoparticle-mediated delivery of an siRNA to inhibit microglial neurotoxicity. Biomaterials. 2018;161:95–105.10.1016/j.biomaterials.2018.01.039Search in Google Scholar PubMed
[229] Wang H, Agarwal P, Xiao Y, Peng H, Zhao S, Liu X, et al. A nano-in-micro system for enhanced stem cell therapy of ischemic diseases. ACS Cent Sci. 2017;3(8):875–85.10.1021/acscentsci.7b00213Search in Google Scholar PubMed PubMed Central
[230] Hers HG. Inborn lysosomal diseases. Gastroenterology. 1965;48:625–33.10.1016/S0016-5085(65)80041-5Search in Google Scholar
[231] Winchester B, Vellodi A, Young E. The molecular basis of lysosomal storage diseases and their treatment. Biochem Soc Trans. 2000;28(2):150–4.10.1042/bst0280150Search in Google Scholar PubMed
[232] Meikle PJ, Hopwood JJ, Clague AE, Carey WF. Prevalence of lysosomal storage disorders. JAMA. 1999;281(3):249–54.10.1001/jama.281.3.249Search in Google Scholar PubMed
[233] Neufeld EF. Lysosomal storage diseases. Annu Rev Biochem. 1991;60:257–80.10.1146/annurev.bi.60.070191.001353Search in Google Scholar PubMed
[234] Platt FM, d’Azzo A, Davidson BL, Neufeld EF, Tifft CJ. Lysosomal storage diseases. Nat Rev Dis Primers. 2018;4(1):27.10.1038/s41572-018-0025-4Search in Google Scholar PubMed
[235] Rigon L, Salvalaio M, Pederzoli F, Legnini E, Duskey JT, D’Avanzo F, et al. Targeting brain disease in mpsii: preclinical evaluation of ids-loaded plga nanoparticles. Int J Mol Sci. 2019;20(8):2014.10.3390/ijms20082014Search in Google Scholar PubMed PubMed Central
[236] Kim JY, Choi WI, Kim YH, Tae G. Brain-targeted delivery of protein using chitosan- and RVG peptide-conjugated, pluronic-based nano-carrier. Biomaterials. 2013;34(4):1170–8.10.1016/j.biomaterials.2012.09.047Search in Google Scholar PubMed
[237] MacBrinn M, Okada S, Woollacott M, Patel V, Ho MW, Tappel AL, et al. Beta-galactosidase deficiency in the hurler syndrome. New Engl J Med. 1969;281(7):338–43.10.1056/NEJM196908142810702Search in Google Scholar PubMed
[238] Suzuki K, Suzuki Y. Globoid cell leucodystrophy (Krabbe’s disease): deficiency of galactocerebroside beta-galactosidase. Proc Natl Acad Sci U S A. 1970;66(2):302–9.10.1073/pnas.66.2.302Search in Google Scholar PubMed PubMed Central
[239] Muhlstein A, Gelperina S, Kreuter J. Development of nanoparticle-bound arylsulfatase B for enzyme replacement therapy of mucopolysaccharidosis VI. Pharmazie. 2013;68(7):549–54.Search in Google Scholar
[240] Schuster T, Muhlstein A, Yaghootfam C, Maksimenko O, Shipulo E, Gelperina S, et al. Potential of surfactant-coated nanoparticles to improve brain delivery of arylsulfatase A. J Control Release. 2017;253:1–10.10.1016/j.jconrel.2017.02.016Search in Google Scholar PubMed
[241] Aldape K, Brindle KM, Chesler L, Chopra R, Gajjar A, Gilbert MR, et al. Challenges to curing primary brain tumours. Nat Rev Clin Oncol. 2019;16(8):509–20.10.1038/s41571-019-0177-5Search in Google Scholar PubMed PubMed Central
[242] Ricard D, Idbaih A, Ducray F, Lahutte M, Hoang-Xuan K, Delattre JY. Primary brain tumours in adults. Lancet. 2012;392(10145):432–46.10.1016/S0140-6736(18)30990-5Search in Google Scholar PubMed
[243] Zhang C, Nance EA, Mastorakos P, Chisholm J, Berry S, Eberhart C, et al. Convection enhanced delivery of cisplatin-loaded brain penetrating nanoparticles cures malignant glioma in rats. J Control Release. 2017;263:112–9.10.1016/j.jconrel.2017.03.007Search in Google Scholar PubMed PubMed Central
[244] Householder KT, DiPerna DM, Chung EP, Wohlleb GM, Dhruv HD, Berens ME, et al. Intravenous delivery of camptothecin-loaded PLGA nanoparticles for the treatment of intracranial glioma. Int J Pharm. 2015 20;479(2):374–80.10.1016/j.ijpharm.2015.01.002Search in Google Scholar PubMed
[245] Ruan SB, Yuan MQ, Zhang L, Hu GL, Chen JT, Cun XL, et al. Tumor microenvironment sensitive doxorubicin delivery and release to glioma using angiopep-2 decorated gold nanoparticles. Biomaterials. 2015;37:425–35.10.1016/j.biomaterials.2014.10.007Search in Google Scholar PubMed
[246] Haney MJ, Zhao Y, Li S, Higginbotham SM, Booth SL, Han HY, et al. Cell-mediated transfer of catalase nanoparticles from macrophages to brain endothelial, glial and neuronal cells. Nanomedicine. 2011;6(7):1215–30.10.2217/nnm.11.32Search in Google Scholar PubMed PubMed Central
[247] Hudome S, Palmer C, Roberts RL, Mauger D, Housman C, Towfighi J. The role of neutrophils in the production of hypoxic-ischemic brain injury in the neonatal rat. Pediatrires. 1997;41(5):607–16.10.1203/00006450-199705000-00002Search in Google Scholar PubMed
[248] Xue J, Zhao Z, Zhang L, Xue L, Shen S, Wen Y, et al. Neutrophil-mediated anticancer drug delivery for suppression of postoperative malignant glioma recurrence. Nat Nanotechnol. 2017;12(7):692–700.10.1038/nnano.2017.54Search in Google Scholar PubMed
[249] Lee HY, Choi YJ, Jung EJ, Yin HQ, Kwon JT, Kim JE, et al. Genomics-based screening of differentially expressed genes in the brains of mice exposed to silver nanoparticles via inhalation. J Nanopart Res. 2010;12:1567–78.10.1007/s11051-009-9666-2Search in Google Scholar
[250] Wu W, Lee SY, Wu X, Tyler JY, Wang H, Ouyang Z, et al. Neuroprotective ferulic acid (FA)-glycol chitosan (GC) nanoparticles for functional restoration of traumatically injured spinal cord. Biomaterials. 2014;35(7):2355–64.10.1016/j.biomaterials.2013.11.074Search in Google Scholar PubMed PubMed Central
[251] Wu J, Ding T, Sun J. Neurotoxic potential of iron oxide nanoparticles in the rat brain striatum and hippocampus. Neurotoxicology. 2013;34:243–53.10.1016/j.neuro.2012.09.006Search in Google Scholar PubMed
[252] Yan F, Wang Y, He S, Ku S, Gu W, Ye L. Transferrin-conjugated, fluorescein-loaded magnetic nanoparticles for targeted delivery across the blood-brain barrier. J Mater Sci Mater Med. 2013;24(10):2371–9.10.1007/s10856-013-4993-3Search in Google Scholar PubMed
[253] Raliya R, Saha D, Chadha TS, Raman B, Biswas P. Non-invasive aerosol delivery and transport of gold nanoparticles to the brain. Sci Rep. 2017;7:44718.10.1038/srep44718Search in Google Scholar PubMed PubMed Central
[254] Kwon EJ, Skalak M, Lo Bu R, Bhatia SN. Neuron-targeted nanoparticle for sirna delivery to traumatic brain injuries. ACS Nano. 2016;10(8):7926–33.10.1021/acsnano.6b03858Search in Google Scholar PubMed PubMed Central
[255] Maya-Vetencourt JF, Manfredi G, Mete M, Colombo E, Bramini M, Di Marco S, et al. Subretinally injected semiconducting polymer nanoparticles rescue vision in a rat model of retinal dystrophy. Nat Nanotechnol. 2020;15(8):698–708.10.1038/s41565-020-0696-3Search in Google Scholar PubMed
[256] Yoo J, Lee E, Kim HY, Youn DH, Jung J, Kim H, et al. Electromagnetized gold nanoparticles mediate direct lineage reprogramming into induced dopamine neurons in vivo for Parkinson’s disease therapy. Nat Nanotechnol. 2017;12(10):1006–14.10.1038/nnano.2017.133Search in Google Scholar PubMed
[257] Jeong SJ, Cooper JG, Ifergan I, McGuire TL, Xu D, Hunter Z, et al. Intravenous immune-modifying nanoparticles as a therapy for spinal cord injury in mice. Neurobiol Dis. 2017;108:73–82.10.1016/j.nbd.2017.08.006Search in Google Scholar PubMed PubMed Central
[258] Zhang L, Liu XG, Liu DQ, Yu XL, Zhang LX, Zhu J, et al. A conditionally releasable “do not eat me” cd47 signal facilitates microglia-targeted drug delivery for the treatment of Alzheimer’s Disease. Adv Funct Mater. 2020;30(24):1910691.10.1002/adfm.201910691Search in Google Scholar
[259] Tafoya MA, Madi S, Sillerud LO. Superparamagnetic nanoparticle-enhanced MRI of Alzheimer’s disease plaques and activated microglia in 3X transgenic mouse brains: Contrast optimization. J Magn Reson Imaging. 2017;46(2):574–88.10.1002/jmri.25563Search in Google Scholar PubMed
[260] Ribot EJ, Miraux S, Konsman JP, Bouchaud V, Pourtau L, Delville MH, et al. In vivo MR tracking of therapeutic microglia to a human glioma model. NMR Biomed. 2011;24(10):1361–8.10.1002/nbm.1699Search in Google Scholar PubMed
[261] Tang T, Valenzuela A, Petit F, Chow S, Leung K, Gorin F, et al. In vivo MRI of functionalized iron oxide nanoparticles for brain inflammation. Contrast Media Mol Imaging. 2018;3476476.10.1155/2018/3476476Search in Google Scholar PubMed PubMed Central
[262] Sillerud LO, Yang Y, Yang LY, Duval KB, Thompson J, Yang Y. Longitudinal monitoring of microglial/macrophage activation in ischemic rat brain using Iba-1-specific nanoparticle-enhanced magnetic resonance imaging. J Cereb Blood Flow Metab. 2020;40(1_Suppl):S117–33.10.1177/0271678X20953913Search in Google Scholar PubMed PubMed Central
[263] Ganbold T, Bao Q, Zandan J, Hasi A, Baigude H. Modulation of microglia polarization through silencing of nf-kappab p65 by functionalized curdlan nanoparticle-mediated RNAi. ACS Appl Mater Inter. 2020;12(10):11363–74.10.1021/acsami.9b23004Search in Google Scholar PubMed
[264] Naldini L. Gene therapy returns to centre stage. Nature. 2015;526(7573):351–60.10.1038/nature15818Search in Google Scholar PubMed
[265] Sheridan C. Why gene therapies must go virus-free. Nat Biotechnol. 2023;41(6):737–9.10.1038/s41587-023-01824-6Search in Google Scholar PubMed
[266] Han X, Mitchell MJ, Nie G. Nanomaterials for therapeutic RNA delivery. Matter. 2020;3(6):1948–75.10.1016/j.matt.2020.09.020Search in Google Scholar
[267] Huang X, Kon E, Han X, Zhang X, Kong N, Mitchell MJ, et al. Nanotechnology-based strategies against SARS-CoV-2 variants. Nat Nanotechnol. 2022;17(10):1027–37.10.1038/s41565-022-01174-5Search in Google Scholar PubMed
[268] Sanchez-Ramos J, Song SJ, Kong XY, Foroutan P, Martinez G, Dominguez-Viqueria W, et al. Chitosan-Mangafodipir nanoparticles designed for intranasal delivery of siRNA and DNA to brain. J Drug Deliv Sci Technol. 2018;43:453–60.10.1016/j.jddst.2017.11.013Search in Google Scholar PubMed PubMed Central
[269] Alvarez-Erviti L, Seow YQ, Yin HF, Betts C, Lakhal S, Wood MJA. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29(4):341–5.10.1038/nbt.1807Search in Google Scholar PubMed
[270] Yadav S, Gandham SK, Panicucci R, Amiji MM. Intranasal brain delivery of cationic nanoemulsion-encapsulated TNFalpha siRNA in prevention of experimental neuroinflammation. Nanomedicine-UK. 2016;12(4):987–1002.10.1016/j.nano.2015.12.374Search in Google Scholar PubMed PubMed Central
[271] Wang PZ, Zheng XY, Guo Q, Yang P, Pang XY, Qian K, et al. Systemic delivery of BACE1 siRNA through neuron-targeted nanocomplexes for treatment of Alzheimer’s disease. J Control Release. 2018;279:220–33.10.1016/j.jconrel.2018.04.034Search in Google Scholar PubMed
[272] Cooper JM, Wiklander PBO, Nordin JZ, Al-Shawi R, Wood MJ, Vithlani M, et al. Systemic exosomal siRNA delivery reduced alpha-synuclein aggregates in brains of transgenic mice. Mov Disord. 2014;27(12):2111–22.10.1002/mds.25978Search in Google Scholar PubMed PubMed Central
[273] Wei L, Guo XY, Yang T, Yu MZ, Chen DW, Wang JC. Brain tumor-targeted therapy by systemic delivery of siRNA with Transferrin receptor-mediated core–shell nanoparticles. Int J Pharm. 2016;510(1):394–405.10.1016/j.ijpharm.2016.06.127Search in Google Scholar PubMed
[274] Hu KK, Chen XH, Chen WY, Zhang LK, Li J, Ye JL, et al. Neuroprotective effect of gold nanoparticles composites in Parkinson’s disease model. Nanomed Nanotechnol. 2018;14(4):1123–36.10.1016/j.nano.2018.01.020Search in Google Scholar PubMed
[275] Chen L, Watson C, Morsch M, Cole NJ, Chung RS, Saunders DN, et al. Improving the delivery of SOD1 antisense oligonucleotides to motor neurons using calcium phosphate-lipid nanoparticles. Front Neurosci. 2017;11:476.10.3389/fnins.2017.00476Search in Google Scholar PubMed PubMed Central
[276] Cost PM, Cardoso AL, Custodia C, Cunha P, Pereira de Almeida L, Pedroso de Lima MC. MiRNA-21 silencing mediated by tumor-targeted nanoparticles combined with sunitinib: A new multimodal gene therapy approach for glioblastoma. J Control Release. 2015;207:31–9.10.1016/j.jconrel.2015.04.002Search in Google Scholar PubMed
[277] Riccardi C, D’Aria F, Digilio FA, Carillo MR, Amato J, Fasano D, et al. Fighting the Huntington’s Disease with a G-quadruplex-forming aptamer specifically binding to mutant huntingtin protein: Biophysical characterization, in vitro and in vivo studies. Int J Mol Sci. 2022;23(9):4804.10.3390/ijms23094804Search in Google Scholar PubMed PubMed Central
[278] Riccardi C, D’Aria F, Fasano D, Digilio FA, Carilo MR, Amato J, et al. Truncated analogues of a G-quadruplex-forming aptamer targeting mutant huntingtin: Shorter Is Better! Int J Mol Sci. 2022;23(20):12412.10.3390/ijms232012412Search in Google Scholar PubMed PubMed Central
[279] Dos Santos Rodrigues B, Oue H, Banerjee A, Kanekiyo T, Singh J. Dual functionalized liposome-mediated gene delivery across triple co-culture blood brain barrier model and specific in vivo neuronal transfection. J Control Release. 2018;286:264–78.10.1016/j.jconrel.2018.07.043Search in Google Scholar PubMed PubMed Central
[280] Aly AE, Harmon B, Padegimas L, Sesenoglu-Laird O, Cooper MJ, Yurek DM, et al. Intranasal delivery of hGDNF plasmid DNA nanoparticles results in long-term and widespread transfection of perivascular cells in rat brain. Nanomedicine-UK. 2019;16:20–33.10.1016/j.nano.2018.11.006Search in Google Scholar PubMed
[281] Liu Y, An S, Li JF, Kuang YY, He X, Guo YB, et al. Brain-targeted co-delivery of therapeutic gene and peptide by multifunctional nanoparticles in Alzheimer’s disease mice. Biomaterials. 2016;80:33–45.10.1016/j.biomaterials.2015.11.060Search in Google Scholar PubMed
[282] Israel LL, Galstyan A, Cox A, Shatalova ES, Sun T, Rashid MH, et al. Signature effects of vector-guided systemic nano bioconjugate delivery across Blood-Brain Barrier of normal, Alzheimer’s, and tumor mouse models. ACS Nano. 2022;16(8):11815–32.10.1021/acsnano.1c10034Search in Google Scholar PubMed PubMed Central
[283] Goss AL, Samudralwar RD, Das RR, Nath A. ANA Investigates: Neurological complications of COVID-19 vaccines. Ann Neurol. 2021;89(5):856–57.10.1002/ana.26065Search in Google Scholar PubMed PubMed Central
[284] High-dose AAV gene therapy deaths. Nat Biotechnol. 2020;38(8):910.10.1038/s41587-020-0642-9Search in Google Scholar PubMed
[285] Naqvi S, Panghal A, Flora SJS. Nanotechnology: a promising approach for delivery of neuroprotective drugs. Front Neurosci. 2020;14:494.10.3389/fnins.2020.00494Search in Google Scholar PubMed PubMed Central
[286] Pinheiro RGR, Coutinho AJ, Pinheiro M, Neves AR. Nanoparticles for targeted brain drug delivery: What Do We Know? Int J Mol Sci. 2021;22(21):11654.10.3390/ijms222111654Search in Google Scholar PubMed PubMed Central
[287] Verry C, Dufort S, Villa J, Gavard M, Iriart C, Grand S, et al. Theranostic AGuIX nanoparticles as radiosensitizer: A phase I, dose-escalation study in patients with multiple brain metastases (NANO-RAD trial). Radiother Oncol. 2021;160:159–65.10.1016/j.radonc.2021.04.021Search in Google Scholar PubMed
[288] Aheget H, Tristan-Manzano M, Mazini L, Cortijo-Gutierrez M, Galindo-Moreno P, Herrera C, et al. Exosome: a new player in translational nanomedicine. J Clin Med. 2020;9(8):2380.10.3390/jcm9082380Search in Google Scholar PubMed PubMed Central
[289] Kumthekar P, Ko CH, Paunesku T, Dixit K, Sonabend AM, Bloch O, et al. A first-in-human phase 0 clinical study of RNA interference-based spherical nucleic acids in patients with recurrent glioblastoma. Sci Transl Med. 2021;13(584):eabb3945.10.1126/scitranslmed.abb3945Search in Google Scholar PubMed PubMed Central
[290] Vashist A, Kaushik A, Vashist A, Bala J, Nikkhah-Moshaie R, Sagar V, et al. Nanogels as potential drug nanocarriers for CNS drug delivery. Drug Discov Today. 2018;23(7):1436–43.10.1016/j.drudis.2018.05.018Search in Google Scholar PubMed PubMed Central
[291] Kadakia E, Harpude P, Parayath N, Bottino D, Amiji M. Challenging the CNS targeting potential of systemically administered nanoemulsion delivery systems: a case study with rapamycin-containing fish oil nanoemulsions in mice. Pharm Res. 2019;36(9):134.10.1007/s11095-019-2667-7Search in Google Scholar PubMed
[292] Vong LB, Trinh NT, Nagasaki Y. Design of amino acid-based self-assembled nano-drugs for therapeutic applications. J Control Release. 2020;326:140–9.10.1016/j.jconrel.2020.06.009Search in Google Scholar PubMed
[293] Vong LB, Sato Y, Chonpathompikunlert P, Tanasawet S, Hutamekalin P, Nagasaki Y. Self-assembled polydopamine nanoparticles improve treatment in Parkinson’s disease model mice and suppress dopamine-induced dyskinesia. Acta Biomater. 2020;109:220–8.10.1016/j.actbio.2020.03.021Search in Google Scholar PubMed
[294] Khan AR, Liu MR, Khan MW, Zhai GX. Progress in brain targeting drug delivery system by nasal route. J Control Release. 2017;268:364–89.10.1016/j.jconrel.2017.09.001Search in Google Scholar PubMed
[295] De Virgilio A, Greco A, Fabbrini G, Inghilleri M, Rizzo MI, Gallo A, et al. Parkinson’s disease: Autoimmunity and neuroinflammation. Autoimmun Rev. 2016;15:1005–11.10.1016/j.autrev.2016.07.022Search in Google Scholar PubMed
[296] Campbell P, Morris H, Schapira A. Chaperone-mediated autophagy as a therapeutic target for Parkinson disease. Expert Opin Ther Tar. 2018;22(10):823–32.10.1080/14728222.2018.1517156Search in Google Scholar PubMed
[297] Gadhave K, Bolshette N, Ahire A, Pardeshi R, Thakur K, Trandafir C, et al. The ubiquitin proteasomal system: a potential target for the management of Alzheimer’s disease. J Cell Mol Med. 2016;20(7):1392–407.10.1111/jcmm.12817Search in Google Scholar PubMed PubMed Central
[298] Kwon HJ, Cha MY, Kim D, Kim DK, Soh M, Shin K, et al. Mitochondria-targeting ceria nanoparticles as antioxidants for Alzheimer’s Disease. ACS Nano. 2016;10(2):2860–70.10.1021/acsnano.5b08045Search in Google Scholar PubMed
[299] Wes PD, Sayed FA, Bard F, Gan L. Targeting microglia for the treatment of Alzheimer’s Disease. Glia. 2016;64(10):1710–32.10.1002/glia.22988Search in Google Scholar PubMed
[300] Tang MX, Taghibiglou C. The mechanisms of action of curcumin in Alzheimer’s Disease. J Alzheimers Dis. 2017;58(4):1003–16.10.3233/JAD-170188Search in Google Scholar PubMed
[301] Knudsen KB, Northeved H, Kumar PE, Permin A, Gjetting T, Andresen TL, et al. In vivo toxicity of cationic micelles and liposomes. Nanomedicine-UK. 2015;11(2):467–77.10.1016/j.nano.2014.08.004Search in Google Scholar PubMed
[302] Luong D, Kesharwani P, Deshmukh R, Mohd Amin MCI, Gupta U, Greish K, et al. PEGylated PAMAM dendrimers: Enhancing efficacy and mitigating toxicity for effective anticancer drug and gene delivery. Acta Biomater. 2016;43:14–29.10.1016/j.actbio.2016.07.015Search in Google Scholar PubMed
[303] Kim IY, Joachim E, Choi H, Kim K. Toxicity of silica nanoparticles depends on size, dose, and cell type. Nanomedicine-UK. 2015;11(6):1407–16.10.1016/j.nano.2015.03.004Search in Google Scholar PubMed
[304] Wu J, Wang C, Sun J, Xue Y. Neurotoxicity of silica nanoparticles: brain localization and dopaminergic neurons damage pathways. ACS Nano. 2011;5(6):4476–89.10.1021/nn103530bSearch in Google Scholar PubMed
[305] Wang DL, Lin ZF, Wang T, Yao ZF, Qin MN, Zheng SR, et al. Where does the toxicity of metal oxide nanoparticles come from: The nanoparticles, the ions, or a combination of both? J Hazard Mater. 2016;308:328–34.10.1016/j.jhazmat.2016.01.066Search in Google Scholar PubMed
[306] Jia YP, Ma BY, Wei XW, Qian ZY. The in vitro and in vivo toxicity of gold nanoparticles. Chinese Chem Lett. 2017;28:691–702.10.1016/j.cclet.2017.01.021Search in Google Scholar
[307] Larner SF, Wang J, Goodman J, Altman MBO, Xin M, Wang KKW. In vitro neurotoxicity resulting from exposure of cultured neural cells to several types of nanoparticles. J Cell Death. 2017;10:1179670717694523.10.1177/1179670717694523Search in Google Scholar PubMed PubMed Central
[308] Cai R, Chen C. The crown and the scepter: roles of the protein corona in nanomedicine. Adv Mater. 2019;31(45):1805740.10.1002/adma.201805740Search in Google Scholar PubMed
[309] Ren J, Andrikopoulos N, Velonia K, Tang H, Cai R, Ding F, et al. Chemical and biophysical signatures of the protein corona in nanomedicine. J Am Chem Soc. 2022;144(21):9184–205.10.1021/jacs.2c02277Search in Google Scholar PubMed
[310] Liu T, Xie Q, Dong Z, Peng Q. Nanoparticles-based delivery system and its potentials in treating central nervous system disorders. Nanotechnology. 2022;33(45):452001.10.1088/1361-6528/ac85f3Search in Google Scholar PubMed
[311] Tang Y, Gao J, Wang T, Zhang Q, Wang A, Huang M, et al. The effect of drug loading and multiple administration on the protein corona formation and brain delivery property of PEG-PLA nanoparticles. Acta Pharm Sin B. 2022;12(4):2043–56.10.1016/j.apsb.2021.09.029Search in Google Scholar PubMed PubMed Central
[312] Zhang Z, Guan J, Jiang Z, Yang Y, Liu J, Hua W, et al. Brain-targeted drug delivery by manipulating protein corona functions. Nat Commun. 2019;10(1):3561.10.1038/s41467-019-11593-zSearch in Google Scholar PubMed PubMed Central
[313] Cai R, Ren J, Guo M, Wei T, Liu Y, Xie C, et al. Dynamic intracellular exchange of nanomaterials’ protein corona perturbs proteostasis and remodels cell metabolism. Proc Natl Acad Sci U S A. 2022;119(23):e2200363119.10.1073/pnas.2200363119Search in Google Scholar PubMed PubMed Central
[314] Cai R, Ren J, Ji Y, Wang Y, Liu Y, Chen Z, et al. Corona of thorns: the surface chemistry-mediated protein corona perturbs the recognition and immune response of macrophages. ACS Appl Mater Inter. 2019;12(2):1997–2008.10.1021/acsami.9b15910Search in Google Scholar PubMed
[315] Grenier P, de Oliveira Viana IM, Lima EM, Bertrand N. Anti-polyethylene glycol antibodies alter the protein corona deposited on nanoparticles and the physiological pathways regulating their fate in vivo. J Control Release. 2018;287:121–31.10.1016/j.jconrel.2018.08.022Search in Google Scholar PubMed
[316] Tao C, Chuah YJ, Xu C, Wang DA. Albumin conjugates and assemblies as versatile bio-functional additives and carriers for biomedical applications. J Mater Chem B. 2019;7(3):357–67.10.1039/C8TB02477DSearch in Google Scholar PubMed
[317] Lacroix AL, Edwardson TG, Hancock MA, Dore MD, Sleiman HF. Development of DNA nanostructures for high-affinity binding to human serum albumin. J Am Chem Soc. 2017;139(21):7355–62.10.1021/jacs.7b02917Search in Google Scholar PubMed
[318] Peng Q, Zhang S, Yang Q, Zhang T, Wei X-Q, Jiang L, et al. Preformed albumin corona, a protective coating for nanoparticles based drug delivery system. Biomaterials. 2013;34(33):8521–30.10.1016/j.biomaterials.2013.07.102Search in Google Scholar PubMed
[319] Aoyama M, Hata K, Higashisaka K, Nagano K, Yoshioka Y, Tsutsumi Y. Clusterin in the protein corona plays a key role in the stealth effect of nanoparticles against phagocytes. Biochem Biophys Res Commun. 2016;480(4):690–5.10.1016/j.bbrc.2016.10.121Search in Google Scholar PubMed
[320] Schöttler S, Becker G, Winzen S, Steinbach T, Mohr K, Landfester K, et al. Protein adsorption is required for stealth effect of poly (ethylene glycol)-and poly (phosphoester)-coated nanocarriers. Nat Nanotechnol. 2016;11(4):372–7.10.1038/nnano.2015.330Search in Google Scholar PubMed
[321] Michaelis K, Hoffmann MM, Dreis S, Herbert E, Alyautdin RN, Michaelis M, et al. Covalent linkage of apolipoprotein e to albumin nanoparticles strongly enhances drug transport into the brain. J Pharmacol Exp Ther. 2006;317(3):1246–53.10.1124/jpet.105.097139Search in Google Scholar PubMed
[322] Wang Y, Zhang H, Xiao W, Liu Y, Zhou Y, He X, et al. Unmasking CSF protein corona: Effect on targeting capacity of nanoparticles. J Control Release. 2021;333:352–61.10.1016/j.jconrel.2021.04.001Search in Google Scholar PubMed
[323] Pinals RL, Yang D, Rosenberg DJ, Chaudhary T, Crothers AR, Iavarone AT, et al. Quantitative protein corona composition and dynamics on carbon nanotubes in biological environments. Angew Chem Int Ed. 2020;59(52):23668–77.10.1002/anie.202008175Search in Google Scholar PubMed PubMed Central
© 2023 the author(s), published by De Gruyter
This work is licensed under the Creative Commons Attribution 4.0 International License.
Articles in the same Issue
- Research Articles
- Preparation of CdS–Ag2S nanocomposites by ultrasound-assisted UV photolysis treatment and its visible light photocatalysis activity
- Significance of nanoparticle radius and inter-particle spacing toward the radiative water-based alumina nanofluid flow over a rotating disk
- Aptamer-based detection of serotonin based on the rapid in situ synthesis of colorimetric gold nanoparticles
- Investigation of the nucleation and growth behavior of Ti2AlC and Ti3AlC nano-precipitates in TiAl alloys
- Dynamic recrystallization behavior and nucleation mechanism of dual-scale SiCp/A356 composites processed by P/M method
- High mechanical performance of 3-aminopropyl triethoxy silane/epoxy cured in a sandwich construction of 3D carbon felts foam and woven basalt fibers
- Applying solution of spray polyurea elastomer in asphalt binder: Feasibility analysis and DSR study based on the MSCR and LAS tests
- Study on the chronic toxicity and carcinogenicity of iron-based bioabsorbable stents
- Influence of microalloying with B on the microstructure and properties of brazed joints with Ag–Cu–Zn–Sn filler metal
- Thermohydraulic performance of thermal system integrated with twisted turbulator inserts using ternary hybrid nanofluids
- Study of mechanical properties of epoxy/graphene and epoxy/halloysite nanocomposites
- Effects of CaO addition on the CuW composite containing micro- and nano-sized tungsten particles synthesized via aluminothermic coupling with silicothermic reduction
- Cu and Al2O3-based hybrid nanofluid flow through a porous cavity
- Design of functional vancomycin-embedded bio-derived extracellular matrix hydrogels for repairing infectious bone defects
- Study on nanocrystalline coating prepared by electro-spraying 316L metal wire and its corrosion performance
- Axial compression performance of CFST columns reinforced by ultra-high-performance nano-concrete under long-term loading
- Tungsten trioxide nanocomposite for conventional soliton and noise-like pulse generation in anomalous dispersion laser cavity
- Microstructure and electrical contact behavior of the nano-yttria-modified Cu-Al2O3/30Mo/3SiC composite
- Melting rheology in thermally stratified graphene-mineral oil reservoir (third-grade nanofluid) with slip condition
- Re-examination of nonlinear vibration and nonlinear bending of porous sandwich cylindrical panels reinforced by graphene platelets
- Parametric simulation of hybrid nanofluid flow consisting of cobalt ferrite nanoparticles with second-order slip and variable viscosity over an extending surface
- Chitosan-capped silver nanoparticles with potent and selective intrinsic activity against the breast cancer cells
- Multi-core/shell SiO2@Al2O3 nanostructures deposited on Ti3AlC2 to enhance high-temperature stability and microwave absorption properties
- Solution-processed Bi2S3/BiVO4/TiO2 ternary heterojunction photoanode with enhanced photoelectrochemical performance
- Electroporation effect of ZnO nanoarrays under low voltage for water disinfection
- NIR-II window absorbing graphene oxide-coated gold nanorods and graphene quantum dot-coupled gold nanorods for photothermal cancer therapy
- Nonlinear three-dimensional stability characteristics of geometrically imperfect nanoshells under axial compression and surface residual stress
- Investigation of different nanoparticles properties on the thermal conductivity and viscosity of nanofluids by molecular dynamics simulation
- Optimized Cu2O-{100} facet for generation of different reactive oxidative species via peroxymonosulfate activation at specific pH values to efficient acetaminophen removal
- Brownian and thermal diffusivity impact due to the Maxwell nanofluid (graphene/engine oil) flow with motile microorganisms and Joule heating
- Appraising the dielectric properties and the effectiveness of electromagnetic shielding of graphene reinforced silicone rubber nanocomposite
- Synthesis of Ag and Cu nanoparticles by plasma discharge in inorganic salt solutions
- Low-cost and large-scale preparation of ultrafine TiO2@C hybrids for high-performance degradation of methyl orange and formaldehyde under visible light
- Utilization of waste glass with natural pozzolan in the production of self-glazed glass-ceramic materials
- Mechanical performance of date palm fiber-reinforced concrete modified with nano-activated carbon
- Melting point of dried gold nanoparticles prepared with ultrasonic spray pyrolysis and lyophilisation
- Graphene nanofibers: A modern approach towards tailored gypsum composites
- Role of localized magnetic field in vortex generation in tri-hybrid nanofluid flow: A numerical approach
- Intelligent computing for the double-diffusive peristaltic rheology of magneto couple stress nanomaterials
- Bioconvection transport of upper convected Maxwell nanoliquid with gyrotactic microorganism, nonlinear thermal radiation, and chemical reaction
- 3D printing of porous Ti6Al4V bone tissue engineering scaffold and surface anodization preparation of nanotubes to enhance its biological property
- Bioinspired ferromagnetic CoFe2O4 nanoparticles: Potential pharmaceutical and medical applications
- Significance of gyrotactic microorganisms on the MHD tangent hyperbolic nanofluid flow across an elastic slender surface: Numerical analysis
- Performance of polycarboxylate superplasticisers in seawater-blended cement: Effect from chemical structure and nano modification
- Entropy minimization of GO–Ag/KO cross-hybrid nanofluid over a convectively heated surface
- Oxygen plasma assisted room temperature bonding for manufacturing SU-8 polymer micro/nanoscale nozzle
- Performance and mechanism of CO2 reduction by DBD-coupled mesoporous SiO2
- Polyarylene ether nitrile dielectric films modified by HNTs@PDA hybrids for high-temperature resistant organic electronics field
- Exploration of generalized two-phase free convection magnetohydrodynamic flow of dusty tetra-hybrid Casson nanofluid between parallel microplates
- Hygrothermal bending analysis of sandwich nanoplates with FG porous core and piezomagnetic faces via nonlocal strain gradient theory
- Design and optimization of a TiO2/RGO-supported epoxy multilayer microwave absorber by the modified local best particle swarm optimization algorithm
- Mechanical properties and frost resistance of recycled brick aggregate concrete modified by nano-SiO2
- Self-template synthesis of hollow flower-like NiCo2O4 nanoparticles as an efficient bifunctional catalyst for oxygen reduction and oxygen evolution in alkaline media
- High-performance wearable flexible strain sensors based on an AgNWs/rGO/TPU electrospun nanofiber film for monitoring human activities
- High-performance lithium–selenium batteries enabled by nitrogen-doped porous carbon from peanut meal
- Investigating effects of Lorentz forces and convective heating on ternary hybrid nanofluid flow over a curved surface using homotopy analysis method
- Exploring the potential of biogenic magnesium oxide nanoparticles for cytotoxicity: In vitro and in silico studies on HCT116 and HT29 cells and DPPH radical scavenging
- Enhanced visible-light-driven photocatalytic degradation of azo dyes by heteroatom-doped nickel tungstate nanoparticles
- A facile method to synthesize nZVI-doped polypyrrole-based carbon nanotube for Ag(i) removal
- Improved osseointegration of dental titanium implants by TiO2 nanotube arrays with self-assembled recombinant IGF-1 in type 2 diabetes mellitus rat model
- Functionalized SWCNTs@Ag–TiO2 nanocomposites induce ROS-mediated apoptosis and autophagy in liver cancer cells
- Triboelectric nanogenerator based on a water droplet spring with a concave spherical surface for harvesting wave energy and detecting pressure
- A mathematical approach for modeling the blood flow containing nanoparticles by employing the Buongiorno’s model
- Molecular dynamics study on dynamic interlayer friction of graphene and its strain effect
- Induction of apoptosis and autophagy via regulation of AKT and JNK mitogen-activated protein kinase pathways in breast cancer cell lines exposed to gold nanoparticles loaded with TNF-α and combined with doxorubicin
- Effect of PVA fibers on durability of nano-SiO2-reinforced cement-based composites subjected to wet-thermal and chloride salt-coupled environment
- Effect of polyvinyl alcohol fibers on mechanical properties of nano-SiO2-reinforced geopolymer composites under a complex environment
- In vitro studies of titanium dioxide nanoparticles modified with glutathione as a potential drug delivery system
- Comparative investigations of Ag/H2O nanofluid and Ag-CuO/H2O hybrid nanofluid with Darcy-Forchheimer flow over a curved surface
- Study on deformation characteristics of multi-pass continuous drawing of micro copper wire based on crystal plasticity finite element method
- Properties of ultra-high-performance self-compacting fiber-reinforced concrete modified with nanomaterials
- Prediction of lap shear strength of GNP and TiO2/epoxy nanocomposite adhesives
- A novel exploration of how localized magnetic field affects vortex generation of trihybrid nanofluids
- Fabrication and physicochemical characterization of copper oxide–pyrrhotite nanocomposites for the cytotoxic effects on HepG2 cells and the mechanism
- Thermal radiative flow of cross nanofluid due to a stretched cylinder containing microorganisms
- In vitro study of the biphasic calcium phosphate/chitosan hybrid biomaterial scaffold fabricated via solvent casting and evaporation technique for bone regeneration
- Insights into the thermal characteristics and dynamics of stagnant blood conveying titanium oxide, alumina, and silver nanoparticles subject to Lorentz force and internal heating over a curved surface
- Effects of nano-SiO2 additives on carbon fiber-reinforced fly ash–slag geopolymer composites performance: Workability, mechanical properties, and microstructure
- Energy bandgap and thermal characteristics of non-Darcian MHD rotating hybridity nanofluid thin film flow: Nanotechnology application
- Green synthesis and characterization of ginger-extract-based oxali-palladium nanoparticles for colorectal cancer: Downregulation of REG4 and apoptosis induction
- Abnormal evolution of resistivity and microstructure of annealed Ag nanoparticles/Ag–Mo films
- Preparation of water-based dextran-coated Fe3O4 magnetic fluid for magnetic hyperthermia
- Statistical investigations and morphological aspects of cross-rheological material suspended in transportation of alumina, silica, titanium, and ethylene glycol via the Galerkin algorithm
- Effect of CNT film interleaves on the flexural properties and strength after impact of CFRP composites
- Self-assembled nanoscale entities: Preparative process optimization, payload release, and enhanced bioavailability of thymoquinone natural product
- Structure–mechanical property relationships of 3D-printed porous polydimethylsiloxane films
- Nonlinear thermal radiation and the slip effect on a 3D bioconvection flow of the Casson nanofluid in a rotating frame via a homotopy analysis mechanism
- Residual mechanical properties of concrete incorporated with nano supplementary cementitious materials exposed to elevated temperature
- Time-independent three-dimensional flow of a water-based hybrid nanofluid past a Riga plate with slips and convective conditions: A homotopic solution
- Lightweight and high-strength polyarylene ether nitrile-based composites for efficient electromagnetic interference shielding
- Review Articles
- Recycling waste sources into nanocomposites of graphene materials: Overview from an energy-focused perspective
- Hybrid nanofiller reinforcement in thermoset and biothermoset applications: A review
- Current state-of-the-art review of nanotechnology-based therapeutics for viral pandemics: Special attention to COVID-19
- Solid lipid nanoparticles for targeted natural and synthetic drugs delivery in high-incidence cancers, and other diseases: Roles of preparation methods, lipid composition, transitional stability, and release profiles in nanocarriers’ development
- Critical review on experimental and theoretical studies of elastic properties of wurtzite-structured ZnO nanowires
- Polyurea micro-/nano-capsule applications in construction industry: A review
- A comprehensive review and clinical guide to molecular and serological diagnostic tests and future development: In vitro diagnostic testing for COVID-19
- Recent advances in electrocatalytic oxidation of 5-hydroxymethylfurfural to 2,5-furandicarboxylic acid: Mechanism, catalyst, coupling system
- Research progress and prospect of silica-based polymer nanofluids in enhanced oil recovery
- Review of the pharmacokinetics of nanodrugs
- Engineered nanoflowers, nanotrees, nanostars, nanodendrites, and nanoleaves for biomedical applications
- Research progress of biopolymers combined with stem cells in the repair of intrauterine adhesions
- Progress in FEM modeling on mechanical and electromechanical properties of carbon nanotube cement-based composites
- Antifouling induced by surface wettability of poly(dimethyl siloxane) and its nanocomposites
- TiO2 aerogel composite high-efficiency photocatalysts for environmental treatment and hydrogen energy production
- Structural properties of alumina surfaces and their roles in the synthesis of environmentally persistent free radicals (EPFRs)
- Nanoparticles for the potential treatment of Alzheimer’s disease: A physiopathological approach
- Current status of synthesis and consolidation strategies for thermo-resistant nanoalloys and their general applications
- Recent research progress on the stimuli-responsive smart membrane: A review
- Dispersion of carbon nanotubes in aqueous cementitious materials: A review
- Applications of DNA tetrahedron nanostructure in cancer diagnosis and anticancer drugs delivery
- Magnetic nanoparticles in 3D-printed scaffolds for biomedical applications
- An overview of the synthesis of silicon carbide–boron carbide composite powders
- Organolead halide perovskites: Synthetic routes, structural features, and their potential in the development of photovoltaic
- Recent advancements in nanotechnology application on wood and bamboo materials: A review
- Application of aptamer-functionalized nanomaterials in molecular imaging of tumors
- Recent progress on corrosion mechanisms of graphene-reinforced metal matrix composites
- Research progress on preparation, modification, and application of phenolic aerogel
- Application of nanomaterials in early diagnosis of cancer
- Plant mediated-green synthesis of zinc oxide nanoparticles: An insight into biomedical applications
- Recent developments in terahertz quantum cascade lasers for practical applications
- Recent progress in dielectric/metal/dielectric electrodes for foldable light-emitting devices
- Nanocoatings for ballistic applications: A review
- A mini-review on MoS2 membrane for water desalination: Recent development and challenges
- Recent updates in nanotechnological advances for wound healing: A narrative review
- Recent advances in DNA nanomaterials for cancer diagnosis and treatment
- Electrochemical micro- and nanobiosensors for in vivo reactive oxygen/nitrogen species measurement in the brain
- Advances in organic–inorganic nanocomposites for cancer imaging and therapy
- Advancements in aluminum matrix composites reinforced with carbides and graphene: A comprehensive review
- Modification effects of nanosilica on asphalt binders: A review
- Decellularized extracellular matrix as a promising biomaterial for musculoskeletal tissue regeneration
- Review of the sol–gel method in preparing nano TiO2 for advanced oxidation process
- Micro/nano manufacturing aircraft surface with anti-icing and deicing performances: An overview
- Cell type-targeting nanoparticles in treating central nervous system diseases: Challenges and hopes
- An overview of hydrogen production from Al-based materials
- A review of application, modification, and prospect of melamine foam
- A review of the performance of fibre-reinforced composite laminates with carbon nanotubes
- Research on AFM tip-related nanofabrication of two-dimensional materials
- Advances in phase change building materials: An overview
- Development of graphene and graphene quantum dots toward biomedical engineering applications: A review
- Nanoremediation approaches for the mitigation of heavy metal contamination in vegetables: An overview
- Photodynamic therapy empowered by nanotechnology for oral and dental science: Progress and perspectives
- Biosynthesis of metal nanoparticles: Bioreduction and biomineralization
- Current diagnostic and therapeutic approaches for severe acute respiratory syndrome coronavirus-2 (SARS-COV-2) and the role of nanomaterial-based theragnosis in combating the pandemic
- Application of two-dimensional black phosphorus material in wound healing
- Special Issue on Advanced Nanomaterials and Composites for Energy Conversion and Storage - Part I
- Helical fluorinated carbon nanotubes/iron(iii) fluoride hybrid with multilevel transportation channels and rich active sites for lithium/fluorinated carbon primary battery
- The progress of cathode materials in aqueous zinc-ion batteries
- Special Issue on Advanced Nanomaterials for Carbon Capture, Environment and Utilization for Energy Sustainability - Part I
- Effect of polypropylene fiber and nano-silica on the compressive strength and frost resistance of recycled brick aggregate concrete
- Mechanochemical design of nanomaterials for catalytic applications with a benign-by-design focus
Articles in the same Issue
- Research Articles
- Preparation of CdS–Ag2S nanocomposites by ultrasound-assisted UV photolysis treatment and its visible light photocatalysis activity
- Significance of nanoparticle radius and inter-particle spacing toward the radiative water-based alumina nanofluid flow over a rotating disk
- Aptamer-based detection of serotonin based on the rapid in situ synthesis of colorimetric gold nanoparticles
- Investigation of the nucleation and growth behavior of Ti2AlC and Ti3AlC nano-precipitates in TiAl alloys
- Dynamic recrystallization behavior and nucleation mechanism of dual-scale SiCp/A356 composites processed by P/M method
- High mechanical performance of 3-aminopropyl triethoxy silane/epoxy cured in a sandwich construction of 3D carbon felts foam and woven basalt fibers
- Applying solution of spray polyurea elastomer in asphalt binder: Feasibility analysis and DSR study based on the MSCR and LAS tests
- Study on the chronic toxicity and carcinogenicity of iron-based bioabsorbable stents
- Influence of microalloying with B on the microstructure and properties of brazed joints with Ag–Cu–Zn–Sn filler metal
- Thermohydraulic performance of thermal system integrated with twisted turbulator inserts using ternary hybrid nanofluids
- Study of mechanical properties of epoxy/graphene and epoxy/halloysite nanocomposites
- Effects of CaO addition on the CuW composite containing micro- and nano-sized tungsten particles synthesized via aluminothermic coupling with silicothermic reduction
- Cu and Al2O3-based hybrid nanofluid flow through a porous cavity
- Design of functional vancomycin-embedded bio-derived extracellular matrix hydrogels for repairing infectious bone defects
- Study on nanocrystalline coating prepared by electro-spraying 316L metal wire and its corrosion performance
- Axial compression performance of CFST columns reinforced by ultra-high-performance nano-concrete under long-term loading
- Tungsten trioxide nanocomposite for conventional soliton and noise-like pulse generation in anomalous dispersion laser cavity
- Microstructure and electrical contact behavior of the nano-yttria-modified Cu-Al2O3/30Mo/3SiC composite
- Melting rheology in thermally stratified graphene-mineral oil reservoir (third-grade nanofluid) with slip condition
- Re-examination of nonlinear vibration and nonlinear bending of porous sandwich cylindrical panels reinforced by graphene platelets
- Parametric simulation of hybrid nanofluid flow consisting of cobalt ferrite nanoparticles with second-order slip and variable viscosity over an extending surface
- Chitosan-capped silver nanoparticles with potent and selective intrinsic activity against the breast cancer cells
- Multi-core/shell SiO2@Al2O3 nanostructures deposited on Ti3AlC2 to enhance high-temperature stability and microwave absorption properties
- Solution-processed Bi2S3/BiVO4/TiO2 ternary heterojunction photoanode with enhanced photoelectrochemical performance
- Electroporation effect of ZnO nanoarrays under low voltage for water disinfection
- NIR-II window absorbing graphene oxide-coated gold nanorods and graphene quantum dot-coupled gold nanorods for photothermal cancer therapy
- Nonlinear three-dimensional stability characteristics of geometrically imperfect nanoshells under axial compression and surface residual stress
- Investigation of different nanoparticles properties on the thermal conductivity and viscosity of nanofluids by molecular dynamics simulation
- Optimized Cu2O-{100} facet for generation of different reactive oxidative species via peroxymonosulfate activation at specific pH values to efficient acetaminophen removal
- Brownian and thermal diffusivity impact due to the Maxwell nanofluid (graphene/engine oil) flow with motile microorganisms and Joule heating
- Appraising the dielectric properties and the effectiveness of electromagnetic shielding of graphene reinforced silicone rubber nanocomposite
- Synthesis of Ag and Cu nanoparticles by plasma discharge in inorganic salt solutions
- Low-cost and large-scale preparation of ultrafine TiO2@C hybrids for high-performance degradation of methyl orange and formaldehyde under visible light
- Utilization of waste glass with natural pozzolan in the production of self-glazed glass-ceramic materials
- Mechanical performance of date palm fiber-reinforced concrete modified with nano-activated carbon
- Melting point of dried gold nanoparticles prepared with ultrasonic spray pyrolysis and lyophilisation
- Graphene nanofibers: A modern approach towards tailored gypsum composites
- Role of localized magnetic field in vortex generation in tri-hybrid nanofluid flow: A numerical approach
- Intelligent computing for the double-diffusive peristaltic rheology of magneto couple stress nanomaterials
- Bioconvection transport of upper convected Maxwell nanoliquid with gyrotactic microorganism, nonlinear thermal radiation, and chemical reaction
- 3D printing of porous Ti6Al4V bone tissue engineering scaffold and surface anodization preparation of nanotubes to enhance its biological property
- Bioinspired ferromagnetic CoFe2O4 nanoparticles: Potential pharmaceutical and medical applications
- Significance of gyrotactic microorganisms on the MHD tangent hyperbolic nanofluid flow across an elastic slender surface: Numerical analysis
- Performance of polycarboxylate superplasticisers in seawater-blended cement: Effect from chemical structure and nano modification
- Entropy minimization of GO–Ag/KO cross-hybrid nanofluid over a convectively heated surface
- Oxygen plasma assisted room temperature bonding for manufacturing SU-8 polymer micro/nanoscale nozzle
- Performance and mechanism of CO2 reduction by DBD-coupled mesoporous SiO2
- Polyarylene ether nitrile dielectric films modified by HNTs@PDA hybrids for high-temperature resistant organic electronics field
- Exploration of generalized two-phase free convection magnetohydrodynamic flow of dusty tetra-hybrid Casson nanofluid between parallel microplates
- Hygrothermal bending analysis of sandwich nanoplates with FG porous core and piezomagnetic faces via nonlocal strain gradient theory
- Design and optimization of a TiO2/RGO-supported epoxy multilayer microwave absorber by the modified local best particle swarm optimization algorithm
- Mechanical properties and frost resistance of recycled brick aggregate concrete modified by nano-SiO2
- Self-template synthesis of hollow flower-like NiCo2O4 nanoparticles as an efficient bifunctional catalyst for oxygen reduction and oxygen evolution in alkaline media
- High-performance wearable flexible strain sensors based on an AgNWs/rGO/TPU electrospun nanofiber film for monitoring human activities
- High-performance lithium–selenium batteries enabled by nitrogen-doped porous carbon from peanut meal
- Investigating effects of Lorentz forces and convective heating on ternary hybrid nanofluid flow over a curved surface using homotopy analysis method
- Exploring the potential of biogenic magnesium oxide nanoparticles for cytotoxicity: In vitro and in silico studies on HCT116 and HT29 cells and DPPH radical scavenging
- Enhanced visible-light-driven photocatalytic degradation of azo dyes by heteroatom-doped nickel tungstate nanoparticles
- A facile method to synthesize nZVI-doped polypyrrole-based carbon nanotube for Ag(i) removal
- Improved osseointegration of dental titanium implants by TiO2 nanotube arrays with self-assembled recombinant IGF-1 in type 2 diabetes mellitus rat model
- Functionalized SWCNTs@Ag–TiO2 nanocomposites induce ROS-mediated apoptosis and autophagy in liver cancer cells
- Triboelectric nanogenerator based on a water droplet spring with a concave spherical surface for harvesting wave energy and detecting pressure
- A mathematical approach for modeling the blood flow containing nanoparticles by employing the Buongiorno’s model
- Molecular dynamics study on dynamic interlayer friction of graphene and its strain effect
- Induction of apoptosis and autophagy via regulation of AKT and JNK mitogen-activated protein kinase pathways in breast cancer cell lines exposed to gold nanoparticles loaded with TNF-α and combined with doxorubicin
- Effect of PVA fibers on durability of nano-SiO2-reinforced cement-based composites subjected to wet-thermal and chloride salt-coupled environment
- Effect of polyvinyl alcohol fibers on mechanical properties of nano-SiO2-reinforced geopolymer composites under a complex environment
- In vitro studies of titanium dioxide nanoparticles modified with glutathione as a potential drug delivery system
- Comparative investigations of Ag/H2O nanofluid and Ag-CuO/H2O hybrid nanofluid with Darcy-Forchheimer flow over a curved surface
- Study on deformation characteristics of multi-pass continuous drawing of micro copper wire based on crystal plasticity finite element method
- Properties of ultra-high-performance self-compacting fiber-reinforced concrete modified with nanomaterials
- Prediction of lap shear strength of GNP and TiO2/epoxy nanocomposite adhesives
- A novel exploration of how localized magnetic field affects vortex generation of trihybrid nanofluids
- Fabrication and physicochemical characterization of copper oxide–pyrrhotite nanocomposites for the cytotoxic effects on HepG2 cells and the mechanism
- Thermal radiative flow of cross nanofluid due to a stretched cylinder containing microorganisms
- In vitro study of the biphasic calcium phosphate/chitosan hybrid biomaterial scaffold fabricated via solvent casting and evaporation technique for bone regeneration
- Insights into the thermal characteristics and dynamics of stagnant blood conveying titanium oxide, alumina, and silver nanoparticles subject to Lorentz force and internal heating over a curved surface
- Effects of nano-SiO2 additives on carbon fiber-reinforced fly ash–slag geopolymer composites performance: Workability, mechanical properties, and microstructure
- Energy bandgap and thermal characteristics of non-Darcian MHD rotating hybridity nanofluid thin film flow: Nanotechnology application
- Green synthesis and characterization of ginger-extract-based oxali-palladium nanoparticles for colorectal cancer: Downregulation of REG4 and apoptosis induction
- Abnormal evolution of resistivity and microstructure of annealed Ag nanoparticles/Ag–Mo films
- Preparation of water-based dextran-coated Fe3O4 magnetic fluid for magnetic hyperthermia
- Statistical investigations and morphological aspects of cross-rheological material suspended in transportation of alumina, silica, titanium, and ethylene glycol via the Galerkin algorithm
- Effect of CNT film interleaves on the flexural properties and strength after impact of CFRP composites
- Self-assembled nanoscale entities: Preparative process optimization, payload release, and enhanced bioavailability of thymoquinone natural product
- Structure–mechanical property relationships of 3D-printed porous polydimethylsiloxane films
- Nonlinear thermal radiation and the slip effect on a 3D bioconvection flow of the Casson nanofluid in a rotating frame via a homotopy analysis mechanism
- Residual mechanical properties of concrete incorporated with nano supplementary cementitious materials exposed to elevated temperature
- Time-independent three-dimensional flow of a water-based hybrid nanofluid past a Riga plate with slips and convective conditions: A homotopic solution
- Lightweight and high-strength polyarylene ether nitrile-based composites for efficient electromagnetic interference shielding
- Review Articles
- Recycling waste sources into nanocomposites of graphene materials: Overview from an energy-focused perspective
- Hybrid nanofiller reinforcement in thermoset and biothermoset applications: A review
- Current state-of-the-art review of nanotechnology-based therapeutics for viral pandemics: Special attention to COVID-19
- Solid lipid nanoparticles for targeted natural and synthetic drugs delivery in high-incidence cancers, and other diseases: Roles of preparation methods, lipid composition, transitional stability, and release profiles in nanocarriers’ development
- Critical review on experimental and theoretical studies of elastic properties of wurtzite-structured ZnO nanowires
- Polyurea micro-/nano-capsule applications in construction industry: A review
- A comprehensive review and clinical guide to molecular and serological diagnostic tests and future development: In vitro diagnostic testing for COVID-19
- Recent advances in electrocatalytic oxidation of 5-hydroxymethylfurfural to 2,5-furandicarboxylic acid: Mechanism, catalyst, coupling system
- Research progress and prospect of silica-based polymer nanofluids in enhanced oil recovery
- Review of the pharmacokinetics of nanodrugs
- Engineered nanoflowers, nanotrees, nanostars, nanodendrites, and nanoleaves for biomedical applications
- Research progress of biopolymers combined with stem cells in the repair of intrauterine adhesions
- Progress in FEM modeling on mechanical and electromechanical properties of carbon nanotube cement-based composites
- Antifouling induced by surface wettability of poly(dimethyl siloxane) and its nanocomposites
- TiO2 aerogel composite high-efficiency photocatalysts for environmental treatment and hydrogen energy production
- Structural properties of alumina surfaces and their roles in the synthesis of environmentally persistent free radicals (EPFRs)
- Nanoparticles for the potential treatment of Alzheimer’s disease: A physiopathological approach
- Current status of synthesis and consolidation strategies for thermo-resistant nanoalloys and their general applications
- Recent research progress on the stimuli-responsive smart membrane: A review
- Dispersion of carbon nanotubes in aqueous cementitious materials: A review
- Applications of DNA tetrahedron nanostructure in cancer diagnosis and anticancer drugs delivery
- Magnetic nanoparticles in 3D-printed scaffolds for biomedical applications
- An overview of the synthesis of silicon carbide–boron carbide composite powders
- Organolead halide perovskites: Synthetic routes, structural features, and their potential in the development of photovoltaic
- Recent advancements in nanotechnology application on wood and bamboo materials: A review
- Application of aptamer-functionalized nanomaterials in molecular imaging of tumors
- Recent progress on corrosion mechanisms of graphene-reinforced metal matrix composites
- Research progress on preparation, modification, and application of phenolic aerogel
- Application of nanomaterials in early diagnosis of cancer
- Plant mediated-green synthesis of zinc oxide nanoparticles: An insight into biomedical applications
- Recent developments in terahertz quantum cascade lasers for practical applications
- Recent progress in dielectric/metal/dielectric electrodes for foldable light-emitting devices
- Nanocoatings for ballistic applications: A review
- A mini-review on MoS2 membrane for water desalination: Recent development and challenges
- Recent updates in nanotechnological advances for wound healing: A narrative review
- Recent advances in DNA nanomaterials for cancer diagnosis and treatment
- Electrochemical micro- and nanobiosensors for in vivo reactive oxygen/nitrogen species measurement in the brain
- Advances in organic–inorganic nanocomposites for cancer imaging and therapy
- Advancements in aluminum matrix composites reinforced with carbides and graphene: A comprehensive review
- Modification effects of nanosilica on asphalt binders: A review
- Decellularized extracellular matrix as a promising biomaterial for musculoskeletal tissue regeneration
- Review of the sol–gel method in preparing nano TiO2 for advanced oxidation process
- Micro/nano manufacturing aircraft surface with anti-icing and deicing performances: An overview
- Cell type-targeting nanoparticles in treating central nervous system diseases: Challenges and hopes
- An overview of hydrogen production from Al-based materials
- A review of application, modification, and prospect of melamine foam
- A review of the performance of fibre-reinforced composite laminates with carbon nanotubes
- Research on AFM tip-related nanofabrication of two-dimensional materials
- Advances in phase change building materials: An overview
- Development of graphene and graphene quantum dots toward biomedical engineering applications: A review
- Nanoremediation approaches for the mitigation of heavy metal contamination in vegetables: An overview
- Photodynamic therapy empowered by nanotechnology for oral and dental science: Progress and perspectives
- Biosynthesis of metal nanoparticles: Bioreduction and biomineralization
- Current diagnostic and therapeutic approaches for severe acute respiratory syndrome coronavirus-2 (SARS-COV-2) and the role of nanomaterial-based theragnosis in combating the pandemic
- Application of two-dimensional black phosphorus material in wound healing
- Special Issue on Advanced Nanomaterials and Composites for Energy Conversion and Storage - Part I
- Helical fluorinated carbon nanotubes/iron(iii) fluoride hybrid with multilevel transportation channels and rich active sites for lithium/fluorinated carbon primary battery
- The progress of cathode materials in aqueous zinc-ion batteries
- Special Issue on Advanced Nanomaterials for Carbon Capture, Environment and Utilization for Energy Sustainability - Part I
- Effect of polypropylene fiber and nano-silica on the compressive strength and frost resistance of recycled brick aggregate concrete
- Mechanochemical design of nanomaterials for catalytic applications with a benign-by-design focus