Nanocapsules For Programmed Neurotransmitter Release Toward Artificial Extracellular Synaptic

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

review

Drug Delivery www.small-journal.com

Nanocapsules for Programmed Neurotransmitter Release:


Toward Artificial Extracellular Synaptic Vesicles
Keitaro Sou,* Duc Long Le, and Hirotaka Sato

The human nervous system has a


Nanocapsules present a promising platform for delivering chemicals and complicated signal transduction network
biomolecules to a site of action in a living organism. Because the biological in which excitatory or inhibitory signal
action of the encapsulated molecules is blocked until they are released from molecules called neurotransmitters are
the nanocapsules, the encapsulation structure enables triggering of the essential for nerve excitation and inhibi-
tion.[1,6,7] Based on evidence that injec-
topical and timely action of the molecules at the target site. A similar mecha-
tion of external excitatory and inhibitory
nism seems promising for the spatiotemporal control of signal transduction neurotransmitter molecule solutions acts
triggered by the release of signal molecules in neuronal, metabolic, and transiently on the neuronal system,[8,9]
immune systems. From this perspective, nanocapsules can be regarded as the development of neurotransmitter-
practical tools to apply signal molecules such as neurotransmitters to inter- delivery systems is key to the practical
application of the neurotransmitter mole­
vene in signal transduction. However, spatiotemporal control of the payload
cules. Organic electronic devices have
release from nanocapsules persists as a key technical issue. Stimulus- been developed to support such delivery
responsive nanocapsules that release payloads in response to external input systems.[10–12] Such electronic devices
of physical stimuli are promising platforms to enable programmed payload capable of releasing neurotransmitters
release. These programmable nanocapsules encapsulating neurotransmit- are attractive to modify signal transduc-
ters are expected to lead to new insights and perspectives related to artificial tion in the nervous system experimentally
and therapeutically. However, the input
extracellular synaptic vesicles that might provide an experimental and thera-
of an electrical signal might present diffi-
peutic strategy for neuromodulation and nervous system disorders. culties in the nervous system because the
electronic signal influences the neuronal
function.[13,14] Moreover, the insertion and
1. Introduction implantation of invasive devices in neuronal tissues involve a
high risk of damaging the nervous system mechanically. Cur-
Molecules such as neurotransmitters, cytokines, lipids, pep- rent strategies for clinical neurostimulation are limited either
tides, proteins, RNAs, nitric oxide, and carbon monoxide play by high invasiveness, low precision, or poor depth of penetra-
important roles in communication between cells through tion. Consequently, further technical efforts are being aimed at
signal transduction in living organisms.[1–5] These signal mole­ the development of non-invasive systems that enable precise
cules are fundamentally reserved in intracellular vesicles and neurotransmitter release in deep tissues. Nanoparticles are
are released to extracellular space as vesicles (exosomes) or anticipated for use as potent tools to achieve this goal.[15,16]
molecules (signal molecules) as necessary. Once the target cells From a broader perspective, a drug-delivery system using
sense the vesicles or signal molecules, the signal is introduced nanoparticles can be expected to provide useful knowledge
into intracellular space to induce biological events through a for the development of neurotransmitter-delivery systems. For
signal transduction pathway. Such signal transduction using drug-delivery applications, lipid vesicles called liposomes are
exosomes and signal molecules enables remote communica- the most popular nanocapsules.[17–19] Since liposomes were first
tion between cells. reported in the 1960s, there has been a considerable number
of research featuring liposomes as a drug-delivery method.
The main target has been the delivery of conventional anti-
Dr. K. Sou cancer drugs safely and effectively to tumors. For this purpose,
Research Institute for Science and Engineering
Waseda University poly(ethylene glycol)-modified liposomes encapsulating doxo-
3-4-1 Ohkubo, Shinjuku, Tokyo 169-8555, Japan rubicin (DOXIL) have been approved for clinical application.[20]
E-mail: soukei@aoni.waseda.jp Furthermore, liposomes have been anticipated for use as bio-
D. L. Le, Prof. H. Sato compatible and biodegradable capsules to encapsulate proteins,
School of Mechanical and Aerospace Engineering nucleic acids, and biomolecules for biomedical applications,
Nanyang Technological University
50 Nanyang Avenue, Singapore 639798, Singapore
such as artificial blood cells and gene delivery.[21–25]
Recently, we reported for the first time our demonstration
The ORCID identification number(s) for the author(s) of this article
can be found under https://doi.org/10.1002/smll.201900132. of neurotransmitter-loaded thermosensitive liposomes trig-
gering on-demand muscle relaxation in a living organism.[26]
DOI: 10.1002/smll.201900132 The neurotransmitter-loaded liposomes, which have structures

Small 2019, 15, 1900132 1900132  (1 of 12) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.small-journal.com

resembling synaptic vesicles encapsulating a neurotransmitter


in a lipid bilayer membrane, can be regarded as artificial extra- Keitaro Sou is an adjunct
cellular synaptic vesicles that enable modification of the signal associate professor at Waseda
transduction by replacing the function of synaptic vesicles. As University. He received his
described herein, we specifically examine the nanocapsules for Ph.D. in chemistry from
programmed neurotransmitter releases for application as artifi- Waseda University in 2000.
cial extracellular synaptic vesicles. He was research associate,
assistant professor, and
associate professor at ARISE
(1999–2009), and associate
2. Neurotransmitters and Synaptic Vesicles
professor at WABIOS
Neurons communicate information via a structure called a syn- (2014–2017), Waseda
apse. Actually, “excitatory synapses” excite neurons receiving University, and a visiting
signal and “suppressive synapses” suppress the excitement. The researcher at University of Texas Health Science Center
signal transduction is triggered by excitatory or inhibitory signal at San Antonio (2001, 2003). His research focuses on the
molecules called neurotransmitters. Numerous neurotransmit- development of self-assembled functional materials for
ters have been identified,[27] a major of which are typically water- bioscience and biomedical applications.
soluble molecules having a dissociable group, as presented in
Table  1. These molecules act as excitatory or inhibitory neuro- Duc Long Le is a Ph.D. student
transmitters to neurons. They are secreted mainly from neurons at the School of Mechanical
in the central nervous system (CNS), consisting of the brain and and Aerospace Engineering,
spinal cord, the peripheral nervous system (PNS) consisting of Nanyang Technological
nerves and ganglia outside of the brain and spinal cord in bilat- University. He received his
eral animals, and the invertebrate neuromuscular junction. B.E. degree at the School of
Acetylcholine, the first neurotransmitter discovered, was Mechanical and Aerospace
identified in 1914. Biogenic amines such as norepineph- Engineering from Nanyang
rine, dopamine, and serotonin, and amino acids such as Technological University in
γ-aminobutyric acid (GABA), glycine, glutamate (glutamic acid), 2017. His current research
and aspartate (aspartic acid) are known neurotransmitters. Pep- focuses on thermosensitive
tides such as substance P, met-enkephalin (endorphin), and nanovesicles for drug delivery
oxytocin are known neuropeptides. in cyborg insects.
Generally, the neurotransmitter molecules are decomposed
rapidly by enzymes in biological environments. For example, Hirotaka Sato is an asso-
acetylcholine released in biological fluids is decomposed rap- ciate professor at Nanyang
idly by acetylcholine esterase.[28] It is noteworthy that, by some Technological University. He
process, the neurons can store such biodegradable neurotrans- received his Ph.D. in chem-
mitters stably. Actually, the neurotransmitters are encapsu- istry from Waseda University
lated within a synaptic vesicle that is an organelle with a lipid in 2005. He was a research
bilayer membrane (Figure  1A). The neurotransmitters are associate at Waseda University
released as necessary to the extracellular space between axon (2005–2006), postdoctoral
terminal and dendrite (20–40 nm synaptic cleft) from the syn- fellow at the University of
aptic vesicles. The release of the neurotransmitters is induced Michigan (2007) and at the
by fusion of synaptic vesicles with neuron cell membrane (exo- University of California at
cytosis).[29] The fusion of synaptic vesicles with a neuronal cell Berkeley (2008–2011),
membrane is triggered in a Ca2+-dependent manner where the and assistant professor at Nanyang Technological
voltage-dependent calcium channel on the neuronal membrane University (2011–2017). His research focuses on MEMS,
controls the influx of Ca2+ into the axon terminal.[30] The neu- insect–machine hybrid system, and electrochemistry.
rotransmitters released to the synaptic cleft bind with receptors
on the dendrite to induce excitation or inhibition of neuronal
systems. The neurotransmitter action is transient because of
its short biological half-life. Consequently, the encapsulation 3. Brain Delivery Using Stimuli-Responsive
structure of neurotransmitters is regarded as a sophisticated Nanoparticles
strategy to escape from biological decomposition in biological
environments and to induce the transient excitation or inhibi- Brain is one of the major target organs for drug delivery using
tion to neurons as necessary. For understanding of this bio- nanoparticles.[16,31] The blood capillary provides an access route
logical mechanism, neurotransmitter-loaded nanocapsule is a to brain for endogenous and drug-delivery molecules. However,
reasonable structure as an artificial synaptic vesicle to deliver the brain capillary has a tight junction between endothelial cells
neurotransmitters stably to extracellular space around a syn- called blood–brain barrier (BBB) creating a restrictive barrier
apse (Figure 1B). between CNS and blood circulation. Consequently, an issue

Small 2019, 15, 1900132 1900132  (2 of 12) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.small-journal.com

Table 1.  Major neurotransmitters.

Neurotransmitter Structure Functions Secretion sites


Acetylcholine O Excitatory to vertebrate skeletal muscles; CNS; PNS; invertebrate neuromuscular
CH3 excitatory or inhibitory at other sites junction
N
H3C O CH3
CH3
Biogenic amines
Norepinephrine HO Excitatory or inhibitory CNS; PNS

NH2
HO
OH
Dopamine HO Generally excitatory CNS; PNS

NH2
HO

Serotonin HO Generally inhibitory CNS

NH2

N
H
Histamine HN Generally excitatory CNS

N NH2
Amino acids
γ-Aminobutyric acid (GABA) Inhibitory CNS; invertebrate neuromuscular junction
H2N COOH
Glycine Inhibitory CNS
H2N COOH
Glutamate, glutamic acid COOH Excitatory CNS; invertebrate neuromuscular junction

H2 N COOH
Aspartate, aspartic acid COOH Excitatory CNS

COOH
H2N
Neuropeptides
Substance P Arg-Pro-Lys-Pro-Gln-Gln-Phe-Phe-Gly-Leu-Met Excitatory CNS; PNS
Met-enkephalin (endorphin) Tyr-Gly-Gly-Phe-Met Generally inhibitory CNS
Oxytocin Cys-Tyr-Ile-Gln-Asn-Cys-Pro-Leu-Gly Generally inhibitory CNS

to be verified is whether the nanoparticles can cross the BBB. affinity ligand molecules to the overexpressing receptors (i.e.,
In fact, nanoparticles reach CNS during systemic circulation LDL receptor,[34,35] transferrin receptor[36,37]) on endothelial cells
through intracellular and/or intercellular pathways (Figure 2A). composing BBB facilitate the nanoparticle delivery into CNS
The BBB plays a role to transport endogenous nanoparticles across the BBB. Another intracellular route is adsorptive tran-
into CNS from blood circulation. Lipoproteins (i.e., low-density scytosis. Positively charged substances adsorb on the negatively
lipoprotein, LDL), endogenous nanoparticles for the systemic charged endothelial cell surface in an electrostatic manner. In
delivery of lipids to cells, could cross the BBB via receptor- adsorptive transcytosis pathway, the endothelial cells uptake the
mediated transcytosis through intracellular pathway.[32,33] The adsorbed cationic substances by endocytosis and release them
receptor-mediated transcytosis mechanism provides a feasible into the brain interstitial fluid by exocytosis. Polycationic pro-
strategy for active delivery of drug-delivery nanoparticles across teins and cationic peptides have been investigated to develop
the BBB. To this end, surface-modified nanoparticles with drug-delivery systems based on adsorptive transcytosis.[38]

Small 2019, 15, 1900132 1900132  (3 of 12) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.small-journal.com

Figure 1.  Signal transduction at synapse mediated by neurotransmitter molecules released from A) native synaptic vesicles and B) neurotransmitter-
loaded nanocapsules (artificial extracellular synaptic vesicles).

Nanoparticles modified with cationic proteins, cationic pep- Nanocapsules of various types such as liposomes, micelles,
tides, cationic polymers, and cationic amino acids are possible polymeric nanoparticles, silica nanoparticles, and magnetic
nanovectors to deliver drugs and genes to brain cells in CNS nanoparticles have been developed for drug-delivery applica-
across the BBB.[39,40] Some small molecules such as lipophilic tions.[46–48] Particularly, stimulus-responsive nanocapsules
molecules, amino acids, and glucose are known to cross the which release their cargos in response to external physical
BBB by diffusion or transport protein pathway. These specific and chemical stimuli are promising platforms to enable on-
molecules which cross the BBB would be delivered into CNS demand release of encapsulated drugs from the nanocapsules
by release from circulating nanoparticles in the brain capillary. (Figure 2B).[49–51]
For passive delivery, the circulation time of nanoparticles Stimuli-responsive nanoparticles to endogenous stimuli such
is an important factor to access the CNS. Generally, optimal as pH, redox, and hypoxia have been studied for drug delivery
nanoparticle size for prolonged circulation is in the range less and stimuli-responsive drug release for cancer therapy.[48,52–54]
than 200 nm. For brain delivery across the BBB, nanoparticles Since these endogenous stimuli systems depend on physiolog-
ranging 20–100 nm are used for prolonged circulation.[41,42] It ical conditions, precise spatiotemporal control of the payload
is recognized that passive nanoparticle delivery through tight release would be difficult. On the other hand, exogenous stimuli
junctions between endothelial cells is still a challenge in normal such as heat, light, ultrasound, and magnetic field enable pro-
brain. Exceptionally, the capillaries in circumventricular organs grammed payload release from stimuli-responsive nanoparticles,
such as pineal body have fenestrations which allow high perme- as these external signals can be applied with controlled time,
ability unlike the BBB. Electron microscopic studies have dem- intensity, and region in an intervening manner. Among the exog-
onstrated the presence of transendothelial pores (30–80 nm in enous stimuli, heat is an attractive option thanks to its versatility
diameter) in the circumventricular organs.[43–45] These fenes- and minimal invasion to biological tissues. Thermosensitive
trated capillaries in the circumventricular organs are remark- liposomes are therefore promising biocompatible platforms that
able pathways to access CNS by passive delivery. In addition, can be programmed for release of cargo by heat stimulus.[55–57]
the evidence that brain injury such as cerebral ischemia and
brain trauma, and neurodegenerative diseases, including Par-
kinson’s disease, Alzheimer disease, and epilepsy, cause leaky 4. Thermosensitive Liposomes
compromised BBB due to disruption of tight junction may
open passive delivery pathway to the site of action.[16,31] Nanocapsules with lipid bilayer membranes enable liposome
After delivering the neurotransmitters using nan­ocapsules encapsulation of neurotransmitters. The neurotransmitter-
into the nervous system, such as brain, a key technical issue loaded liposomes are simply prepared by the hydration of
is how to control the release of neurotransmitters, precisely lipids in an aqueous neurotransmitter solution.[26] The size of
and on demand, from the nanocapsules at a region of interest. liposomes is controllable to be 50–200 nm by using conventional
A salient consideration in the release of signal mole­ cules techniques such as an extrusion method by passing through a
including neurotransmitters in living cells is that these signal membrane filter with nanopores.[58] Surface modification of
molecules are released from the cells as necessary in response liposomes with poly(ethylene glycol) chain achieves prolonged
to biological and environmental stimuli. The released mole­ circulation time for passive delivery.[18–20] Also, the conjugation
cules therefore act as signal molecules to trigger biological chemistry to attach the specific ligand molecules onto liposome
events through a signal transduction pathway. Such a concept surfaces for active delivery has been well established so far.[59,60]
of release and subsequent action with the system has been Liposomes are formed by self-assembly through non-covalent
introduced into the development of drug-delivery systems. intramolecular interaction between lipid molecules in aqueous

Small 2019, 15, 1900132 1900132  (4 of 12) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.small-journal.com

Figure 2.  Brain delivery with stimulus-responsive nanoparticles. A) Possible routes for nanoparticle brain delivery across the blood–brain barrier (BBB).
Normal brain capillary has a tight junction between the endothelial cells called BBB, creating a restrictive barrier between central nervous system
(CNS) and blood circulation. Nanoparticles cross the BBB through intracellular pathway as receptor-mediated transcytosis or adsorptive transcytosis.
Small molecules such as lipophilic molecules, amino acids, and glucose released from nanoparticles in the brain capillary can cross the BBB through
diffusion or transport protein pathway. Brain injury and neurodegenerative diseases possibly cause leaky capillaries by BBB dysfunction (compromised
BBB). Circumventricular organs have fenestrated capillaries with transendothelial pores (30–80 nm in diameter).[43–45] These leaky capillaries provide
intercellular route for passive delivery of nanoparticles and drug-delivery molecules into CNS. B) On-demand payload release from stimulus-responsive
nanoparticles responding to external input. The release of the payload triggers the biological actions of the payload.

phase. Therefore, the strategy to control the payload release The thermotropic phase transition of the lipid bilayer mem-
from liposomes is based on the regulation of the molecular brane is a dynamic phenomenon accompanying the marked
assembly state of the lipid bilayer membrane. Liposomes of change in molecular assembling state. At Tm, the conformation
which the molecular assembling state is responsive to heat of carboncarbon bonds in hydrocarbon chains of the lipid
stimulus are typically called temperature-sensitive, tempera- molecule changes from a trans form to a gauche form, dras-
ture-responsive, or thermosensitive liposomes.[55–57] A typical tically altering the molecular packing state of the lipid mole­
bilayer membrane composed of saturated diacyl phospho- cule.[62,63] It is noteworthy that the membrane permeability of
cholines show a gel to liquid-crystalline phase transition. The the lipid bilayer membrane is increased at Tm by the formation
temperature triggering the phase transition is called the gel to of a grain boundary defect between the solid and liquid phases
liquid-crystalline phase transition temperature (Tm). Figure 3 A (Figure 3B).[55] As the formation of the grain boundary defect
shows that Tm is correlated linearly with the reciprocal of hydro- in lipid bilayer membrane is facilitated by mixing lipids with
carbon chain length in terms of the number of carbon atoms different Tm, liposomes composed of 1,2-dipalmitoyl-sn-glycero-
(n) in a range of n = 9–24 (R2 = 0.999).[61] Tm is approximated by 3-phosphocholine (DPPC, Tm  = 41 °C) and 1,2-distearoyl-sn-
the following Equation (1): glycero-3-phosphocholine (DSPC, Tm  = 55 °C) have been
investigated in an early study.[64] Although the drug release is
Tm = − 1948.9 (1/n ) + 162.8 (1) enhanced, it is generally understood that the release efficiency

Small 2019, 15, 1900132 1900132  (5 of 12) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.small-journal.com

Figure 3.  Temperature-dependent payload release from thermosensitive liposomes with different gel to liquid-crystalline phase transition temperatures
(Tm). A) Correlation of the main phase transition temperature of bilayer membrane composed of fully hydrated, saturated diacyl phosphocholine and
the reciprocal of hydrocarbon chain length in the number of carbon atoms, n. The inset portrays the chemical structure of the saturated diacyl phospho-
cholines. The presented data were collected from published data.[61] B) Change of membrane permeability on thermosensitive liposomes at Tm. Mixed
state of solid and liquid phases at around Tm leads to the grain boundary defect in the bilayer membrane that permits payload release. C) Schematics
showing payload release from thermosensitive liposomes by heat stimulus. Encapsulation of hypertonic solution accelerates the payload release. D)
Representative profiles of payload release from thermosensitive liposomes composed by saturated diacyl phosphocholine with different hydrocarbon
chain length by heat stimulus. Adapted with permission.[68] Copyright 2016, American Chemical Society.

is not high. This would be caused by the instability of the grain 45 °C (approximately 1.6% min−1). The disordered liposome
boundary defect. Later, Needham et al. found that the incor- membrane above Tm allowed water to flow into the liposomes
poration of lysolecithin, monoacyl phospholipid, enhances the to compensate for the osmotic pressure gap between the inside
payload release by stabilizing the defect formed by phase transi- and the outside of the liposomes, which rapidly pushed the
tion.[65] By mixing 10 mol% of 1-stearoyl-2-hydroxy-sn-glycero- encapsulated GABA out. When the osmotic pressure gap is
3-phosphocholine (MSPC) to liposomes composed of DPPC and cancelled by releasing the GABA, the release rate slows down to
1,2-distealoyl-sn-glycero-3-phosphoethnolamine with covalently a great extent. The threshold temperature that triggers the pay-
attached poly(ethylene glycol) (PEG-DSPE), the permeability of load release corresponding to Tm of the phosphocholines is pro-
doxorubicin through the membrane is increased tenfold com- grammable as a factor of the hydrocarbon chain length of phos-
pared with the liposomes without MSPC.[66] The mechanism is phocholines (Figure 3D).[68] Mixtures of lipids with different Tm
that the MSPC accumulates in the grain boundary defect and can be regarded as varying Tm at a narrower temperature scale
forms stable nanopore (≈10 nm).[55,67] Although the efficiency depending on the mixing ratio of the lipids.[69] It is desirable
of payload release from thermosensitive liposomes without for the thermosensitive liposomes used for therapeutic applica-
grain boundary defect stabilizer by heating above Tm is not so tions to retain the neurotransmitters in liposomes at tempera-
high, increasing the osmotic pressure of the inner aqueous tures lower than the human body temperature (around 37 °C).
phase of liposomes markedly enhances the payload release When the temperature is elevated above the body temperature
from liposomes (Figure 3C). Thermosensitive liposomes encap- by externally applying thermal energy, the neurotransmitter is
sulating hypertonic solution (550 mOsm L−1) rapidly release the released at a threshold temperature. It should be noted that the
cargo above the phase transition temperature in physiological survival rate of the normal cells decreases when the tempera-
osmotic pressure (300 mOsm L−1).[57] In our recent investiga- ture exceeds 43 °C.[70] Thus, the appropriate target temperature
tion, approximately 45% of GABA was released from DPPC to release neurotransmitters from liposomes is in the range of
liposomes encapsulating hypertonic 550 mm GABA solution 37 to 43 °C. The threshold temperature at 39–40 °C on DPPC
for the first 10 s during incubation at 45 °C (4.5% s−1).[26] Sub- liposomes (n = 16 in Figure 3D) is in an optimum temperature
sequently, there was a minor release of GABA for 5 min at range to release neurotransmitters for therapeutic applications.

Small 2019, 15, 1900132 1900132  (6 of 12) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.small-journal.com

Because most neurotransmitters are water-soluble small mole- stimulus, thermosensitive nanomaterials enable the on-demand
cules, as shown in Table 1, neurotransmitters of many kinds can payload release by laser irradiation (Figure 4B).[57,82,85–88,90]
be encapsulated stably in the inner aqueous phase of liposomes. Radio frequency,[79] alternative magnetic fields,[81] and ultra-
Moreover, the small molecules are ideal cargo for efficient sound[83,84,89] also allow non-invasive heating that can trigger
release through the small grain boundary defect in the bilayer on-demand drug release (Figure 4C). These nanomaterials
membrane. Consequently, these thermosensitive liposomes developed for drug-delivery systems are potent platforms for
can be a common platform for programmed neurotransmitter on-demand neurotransmitter release in nervous tissues in
release with heat stimulation. Noteworthy exceptions are hydro- combination with non-invasive focal heating of tissues in a con-
phobic or amphipathic molecules embedded in lipid membranes trolled fashion.
and macromolecules with low membrane permeability.

6. Toward Artificial Extracellular Synaptic Vesicles


5. Programmed Payload Release from
Despite the importance of neurotransmitter delivery to mod-
Nanocapsules
ulate the nervous system for development of therapeutic
Typical payload release profiles from conventional liposomes strategies for nervous system disorders, reports of studies
are in the category of sustained release, which is a slow release elucidating the on-demand release of neurotransmitters from
of contents over an extended period of time.[71,72] For example, nanoparticles are still scarce among the relevant literature. An
the drug release from liposomes composed of DSPC and PEG- early study using mesoporous silica nanospheres proposed a
DSPE is below 15% per day for at least 4 days in plasma at stimulus-responsive controlled release of neurotransmitters
37 °C.[71] An example of a system requiring sustained release and drug molecules with chemically removable surface-derivat-
is insulin delivery. Insulin is a peptide hormone that is secreted ized cadmium sulfide nanoparticle caps.[91] In this system, the
naturally from β-cells on islets of Langerhans. Once the func- release of drug molecules from mesoporous silica nanospheres
tion of pancreatic islets to secrete the insulin is disordered, is triggered by adding disulfide bond-reducing molecules such
the blood glucose level becomes elevated, resulting in diabetes as dithiothreitol and mercaptoethanol in vitro. Nanoparticles
mellitus. Although the injection of insulin is effective to treat have been considered for encapsulation of neurotransmitters of
diabetes, a salient difficulty is maintaining an effective insulin several kinds such as dopamine,[92–101] leucine-enkephalin,[102]
level in blood. Pharmaceutical devices for “sustained release” glutamate,[103] neuropeptide substance P,[104] acetylcholine,[105]
of insulin have been developed to overcome this difficulty.[46,73] adenosine,[106] and GABA.[26,107–109] Nevertheless, few in vitro
Further advanced glucose-responsive insulin delivery systems studies have succeeded in the on-demand release of neuro-
have been proposed to treat diabetes.[74,75] transmitters.[103] A recent study finally demonstrated for the
Stimulus-sensitive liposomes enable burst release and first time that neurotransmitter-loaded nanocapsules composed
multiple release responding to the input of physical stimuli of thermosensitive liposomes can trigger on-demand muscle
(Figure 4A). Burst release is an attractive profile to treat cancer relaxation in living organisms by heat stimulus.[26] Although the
cells with anti-cancer drugs.[57,76,77] A higher dose of anti-cancer study was limited to the effect of a single stimulus on muscle
drugs is effective to kill the cancer cells, but dosages have been relaxation by the burst release of inhibitory neurotransmitter
constrained by the side effects of the drugs in conventional (GABA) from thermosensitive liposomes, the report described
systemic delivery. Moreover, when weak drug treatment is extension to a stepwise payload release system by controlling
repeated, cancer cells acquire drug resistance and become more the heating temperature and time.
difficult to deal with. However, using stimuli-sensitive nanocap- GABA plays an important role in the mechanism and
sules, the drug release site can be restricted by local heating at treatment of epilepsy.[110] Experimental and clinical evidence
the target site. Consequently, the local drug concentration can indicates that drugs that increase synaptic GABA are potent
be increased rapidly at the target tumor site. This local therapy anticonvulsants. Actually, GABA-loaded nanocapsules can
enables minimization of the systemic side effects and maximi- provide an invasive GABA release system for experimenta-
zation of the drug action at the tumor site. For development tion and for the treatment of epileptic seizures. Introduction
of the signal molecule release system in neurons, on-demand of a further precise and rapid heating system is expected to
stepwise release is a practical profile. Organic electronic devices be necessary to enable on-demand stepwise payload release
have achieved on-demand precise release of neurotransmit- in target tissues such as the brain. Photothermal stimulus by
ters responding to the external electronic input. Further irradiation of laser to light to heat conversion materials is a
combination with biosensors for neurotransmitters enables potent approach to achieving rapid on-demand cargo release
auto-regulated neuromodulation as an organic electronic bio- from thermosensitive nanoparticles. Near infrared dyes,
mimetic neuron.[78] porphyrin-derivatives, and gold nanoparticles are combined
On-demand release of payload from a nanoplatform is a with thermosensitive nanocapsules as light-to-heat conver-
recent advanced technology intended for development of drug- sion materials.
delivery systems in which stimulus-responsive nanomaterials Moreover, because signal transduction is regulated by the
are applicable for on-demand control of the drug release.[79–90] release of inhibitory neurotransmitters and excitatory neu-
The light source energy is useful for the remote and rapid con- rotransmitters, selective release of inhibitory and excitatory
trol of physical stimulus input at a target region.[57,80,82,85–88,90] neurotransmitters from nanocapsules remains as a technical
As the photothermal materials convert the light energy to heat challenge. Chitgupi et al. reported an advanced system for

Small 2019, 15, 1900132 1900132  (7 of 12) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.small-journal.com

Figure 4.  Programmed payload release from nanocapsules by physical stimulus. A) Representative profiles of payload release responding to the
stimulus input. Arrows indicate the time points at which the stimulus is applied. B) On-demand payload release from thermosensitive liposomes by
irradiation of near infrared (NIR) light (980 nm wavelength) in HeLa cells labeled with a temperature-sensitive fluorescent molecule (ER thermo yellow).
Right: Representative fluorescence images of ER thermo yellow (upper panel) and calcein (lower panel) in HeLa cells during heating experiments.
Fluorescence intensity of ER thermo yellow decreases concomitantly with increasing temperature, that of calcein is increased when calcein is released
from the liposomes. The elapsed time is presented at the top left of each image. Reproduced with permission.[57] Copyright 2015, Royal Society of
Chemistry. C) On-demand payload release from thermosensitive liposomes by irradiation of high-intensity focused ultrasound (HIFU). Irradiation of
HIFU produces heating at the topical region. Reproduced with permission.[83] Copyright 2015, American Chemical Society.

selective and selectable light-triggered cargo release from chromophores.[82,114–119] In fact, IR825, a photothermal dye,
liposomes containing purpurin-phospholipid (Pur-P) or pyroph- absorbs near-infrared light of around 825 nm and converts the
eophorbide-phospholipid (Pyr-P) by irradiation of lasers having light energy to heat. Thermosensitive liposomes embedding
different wavelengths (Figure  5A).[111] In this system, Pur-P is IR825 in their bilayer membrane release the payload by irradia-
excited to produce heat by the irradiation of a 655 nm laser. tion using a 808 nm near-infrared laser (Figure 5C).[115] Simi-
However, the excitation wavelength for Pyr-P is 690 nm. There- larly, cyanine dyes such as indocyanine green (ICG, 805 nm),[82]
fore, by irradiation with a laser of 655 or 690 nm wavelength, IR820 (820 nm),[116] cypate (788 nm),[117] IR780 (780 nm),[118]
selective cargo release from liposomes containing Pur-P or Pyr-P DiR (748 nm),[119] and DiD (644 nm)[120] have been used to
can be achieved (Figure 5B). Light at wavelengths in the near- release payloads from thermosensitive liposomes using near-
infrared region (650–950 nm wavelength) presents benefits as infrared laser irradiation (Figure 5D). The absorption wave-
an input signal to allow for deeper penetration into tissues than length of cyanine dyes is controllable by their chemical structure,
that which can be achieved using light in the ultraviolet-visible for example, the absorption shifts to longer wavelengths
range.[112,113] Photothermal payload release from thermosensi- with extension of the polymethine chain comparing DiR and
tive liposomes can be achieved by encapsulated near-infrared DiD. Water-soluble ICG and IR820 are encapsulated into an
dyes without using chemically modified special lipids with aqueous core of liposomes; others are embedded in a lipid

Small 2019, 15, 1900132 1900132  (8 of 12) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.small-journal.com

Figure 5.  Selective payload release from nanocapsules by irradiation of near-infrared laser light of different wavelengths. A) Structure of porphyrin–
phospholipids and absorbance of liposomes containing 2 mol% porphyrin–phospholipids in PBS. Laser irradiation wavelengths used to trigger the
payload release are indicated by arrows. Reproduced with permission.[111] Copyright 2018, American Chemical Society. B) Selective, selectable light-
triggered release of doxorubicin (Dox) or basic orange (BO) from liposomes containing porphyrin–phospholipids. Indicated liposome mixtures were
subjected to laser irradiation to release liposomal cargo with a 665 nm laser (upper) or 690 nm laser (lower). Reproduced with permission.[111] Copyright
2018, American Chemical Society. C) Schematic illustration showing the structure of thermosensitive stealth liposome containing IR825 and release of
Dox from the liposomes by irradiation of near-infrared light. Reproduced with permission.[115] Copyright 2015, John Wiley & Sons Inc. D) Commercially
available cyanine dyes with different absorption wavelengths that are used to trigger photothermal payload release from liposomes by near-infrared
laser irradiation.

bilayer membrane similarly to IR825. This knowledge can be human health. The cause of Parkinson’s disease is generally
extended to the selective release of neurotransmitters from unknown, but the dopamine level in the corpus striatum is
thermosensitive liposomes embedded near infrared dyes with lowered because of progressive substantia nigra degenera-
different excitation wavelength in a lipid bilayer membrane. tion.[121] Although a further understanding of the mecha-
Such a selective release system would enable experimental and nisms for nanoparticle uptake across the BBB is expected,
therapeutic neuromodulation by the release of excitatory or systemic intravenous administration is a feasible approach
inhibitory neurotransmitters from nanocapsules substituting to deliver nanoparticles into the nervous system, such as the
the function of synaptic vesicles as artificial extracellular artifi- brain.[16,31–42] For example, systemic intravenous administra-
cial vesicles (Figure 6). tion of the neurotransmitter dopamine-loaded PLGA nano-
particles causes markedly increased levels of dopamine and
its metabolites in the striatum of parkinsonian rats.[97] A more
7. Outlook and Perspective recent study has demonstrated an inhibitory neurotransmitter
GABA-loaded thermosensitive liposome injected intramuscu-
Neurodegenerative diseases such as Parkinson’s disease larly triggers on-demand muscle relaxation responding to heat
and Alzheimer’s disease are increasingly problematic for stimulation.[26]

Small 2019, 15, 1900132 1900132  (9 of 12) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.small-journal.com

Figure 6.  Conceptual illustration of artificial extracellular synaptic vesicles used to modify the nervous system by on-demand selective release of excita-
tory or inhibitory neurotransmitter from nanocapsules.

This evidence underscores the fact that nanoparticles encap- Keywords


sulating excitatory and inhibitory neurotransmitters function
drug delivery, liposomes, nanocapsules, neurotransmitters, synaptic
as artificial extracellular synaptic vesicles in living organisms.
vesicles
Further development of programmable nanocapsules for on-
demand selective and stepwise release of neurotransmitters can Received: January 8, 2019
be expected to lead to breakthroughs for experimental and ther- Revised: February 20, 2019
apeutic approaches to nervous system disorders. In parallel, Published online: March 19, 2019
developing a noninvasive technique to apply a stimulus accu-
rately to the nanocapsules in deep tissues is important. Because
nanocapsules can co-encapsulate the photothermal conversion
agents with neurotransmitters, heat stimulus with a focus on [1] J. M. Lauder, Trends Neurosci. 1993, 16, 233.
the nanocapsules is feasible using light energy. Taken together, [2] T. Kishimoto, T. Taga, S. Akira, Cell 1994, 76, 253.
[3] A. K. Mustafa, M. M. Gadalla, S. H. Snyder, Sci. Signal. 2009,
evidence from earlier studies shows that efficient and noninva-
2, re2.
sive experimental and therapeutic application of neurotransmit- [4] M. E. Dinger, T. R. Mercer, J. S. Mattick, J. Mol. Endocrinol. 2008,
ters for nervous system disorders can be achieved by further 40, 151.
advancement of nanocapsules that enable precise programmed [5] K. Lindsey, S. Casson, P. Chilley, Trends Plant Sci. 2002, 7, 78.
neurotransmitter release on demand. [6] K. Ganguly, A. F. Schinder, S. T. Wong, M. Poo, Cell 2001, 105, 521.
[7] U. N. Ramírez-Jarquín, R. Tapia, ACS Chem. Neurosci. 2018, 9, 211.
[8] P. N. Usherwood, H. Grundfest, J. Neurophysiol. 1965, 28, 497.
[9] R. S. Sloviter, D. W. Dempster, Brain Res. Bull. 1985, 15, 39.
Acknowledgements [10] P. M. George, D. A. LaVan, J. A. Burdick, C.-Y. Chen, E. Liang,
R. Langer, Adv. Mater. 2006, 18, 577.
This work was partly supported by JSPS KAKENHI (JP16H03844) and the
[11] D. T. Simon, S. Kurup, K. C , Larsson, R. Hori, K. Tybrandt, M. Goiny,
Singapore Ministry of Education (MOE2017-T2-2-067, RG143/16 (S)).
E. W. H. Jager, M. Berggren, B. Canlon, A. Richter-Dahlfors,
Nat. Mater. 2009, 8, 742.
[12] S. Löffler, S. Seyock, R. Nybom, G. B. Jacobson, A. Richter-Dahlfors,
Conflict of Interest J. Controlled Release 2016, 243, 283.
[13] P. H. Peckham, J. S. Knutson, Annu. Rev. Biomed. Eng. 2005,
The authors declare no conflict of interest. 7, 327.

Small 2019, 15, 1900132 1900132  (10 of 12) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.small-journal.com

[14] K. T. Ragnarsson, Spinal Cord 2008, 46, 255. [50] J. S. Park, T. H. Han, K. Y. Lee, S. S. Han, J. J. Hwang, D. H. Moon,
[15] R. Airan, Science 2017, 357, 465. S. Y. Kim, Y. W. Cho, J. Controlled Release 2006, 115, 37.
[16] V. N. Bharadwaj, D. T. Nguyen, V. D. Kodibagkar, S. E. Stabenfeldt, [51] L. Chen, W. Wang, B. Su, Y. Wen, C. Li, Y. Zhou, M. Li, X. Shi,
Adv. Healthcare Mater. 2018, 7, 1700668. H. Du, Y. Song, L. Jiang, ACS Nano 2014, 8, 744.
[17] A. D. Bangham, R. W. Horne, J. Mol. Biol. 1964, 8, 660. [52] J. Z. Du, X. J. Du, C. Q. Mao, J. Wang, J. Am. Chem. Soc. 2011, 133,
[18] V. P. Torchilin, Nat. Rev. Drug Discovery 2005, 4, 145. 17560.
[19] D. Papahadjopoulos, T. M. Allen, A. Gabizon, E. Mayhew, [53] H. Y. Yang, M. S. Jang, Y. Li, J. H. Lee, D. S. Lee, ACS Appl. Mater.
K. Matthay, S. K. Huang, K. D. Lee, M. C. Woodle, D. D. Lasic, Interfaces 2017, 9, 19184.
C. Redemann, F. J. Martin, Proc. Natl. Acad. Sci. U. S. A. 1991, 88, [54] T. Thambi, V. G. Deepagan, H. Y. Yoon, H. S. Han, S. H. Kim,
11460. S. Son, D. G. Jo, C. H. Ahn, Y. D. Suh, K. Kim, I. Chan Kwon,
[20] A. Gabizon, R. Catane, B. Uziely, B. Kaufman, T. Safra, R. Cohen, D. S. Lee, J. H. Park, Biomaterials 2014, 35, 1735.
F. Martin, A. Huang, Y. Barenholz, Cancer Res. 1994, 54, 987. [55] Z. Al-Ahmady, K. Kostarelos, Chem. Rev. 2016, 116, 3883.
[21] E. Tsuchida, K. Sou, A. Nakagawa, H. Sakai, T. Komatsu, [56] K. Kono, H. Hayashi, T. Takagishi, J. Controlled Release 1994, 30,
K. Kobayashi, Bioconjugate Chem. 2009, 20, 1419. 69.
[22] T. Sato, H. Sakai, K. Sou, M. Medebach, O. Glatter, E. Tsuchida, [57] S. Arai, C.-L. K. Lee, Y.-T. Chang, H. Sato, K. Sou, RSC Adv. 2015,
J. Phys. Chem. B 2009, 113, 8418. 5, 93530.
[23] Y. Okamura, S. Katsuno, H. Suzuki, H. Maruyama, M. Handa, [58] F. Olson, C. A. Hunt, F. C. Szoka, W. J. Vail, D. Papahadjopoulos,
Y. Ikeda, S. Takeoka, J. Controlled Release 2010, 148, 373. Biochim. Biophys. Acta, Biomembr. 1979, 557, 9.
[24] P. L. Felgner, T. R. Gadek, M. Holm, R. Roman, H. W. Chan, [59] T. Li, T. Amari, K. Semba, T. Yamamoto, S. Takeoka, Nanomed.:
M. Wenz, J. P. Northrop, G. M. Ringold, M. Danielsen, Proc. Natl. Nanotechnol. Biol. Med. 2017, 13, 1219.
Acad. Sci. U. S. A. 1987, 84, 7413. [60] S. Dong, J. D. W. Teo, L. Y. Chan, C.-L. K. Lee, K. Sou, ACS Appl.
[25] S. R. Sarker, Y. Aoshima, R. Hokama, T. Inoue, K. Sou, S. Takeoka, Nano Mater. 2018, 1, 1009.
Nanomedicine 2013, 8, 1361. [61] R. Koynova, M. Caffrey, Biochim. Biophys. Acta, Rev. Biomembr.
[26] D. L. Le, Ferdinandus, C. K. Tnee, T. T. V. Doan, S. Arai, M. Suzuki, 1998, 1376, 91.
K. Sou, H. Sato, ACS Appl. Mater. Interfaces 2018, 10, 37812. [62] J. F. Nagle, S. Tristram-Nagle, Biochim. Biophys. Acta, Rev.
[27] B. Si, E. Song, Chemosensors 2018, 6, 1. Biomembr. 2000, 1469, 159.
[28] P. Taylor, Z. Radić, Annu. Annu. Rev. Pharmacol. Toxicol. 1994, 34, [63] Z. V. Leonenko, E. Finot, H. Ma, T. E. Dahms, D. T. Cramb,
281. Biophys. J. 2004, 86, 3783.
[29] R. Jahn, D. Fasshauer, Nature 2012, 490, 201. [64] M. B. Yatvin, J. N. Weinstein, W. H. Dennis, R. Blumenthal, Science
[30] O. D. Uchitel, D. A. Protti, V. Sanchez, B. D. Cherksey, 1978, 202, 1290.
M. Sugimori, R. Llinás, Proc. Natl. Acad. Sci. U. S. A. 1992, 89, [65] G. R. Anyarambhatla, D. Needham, J. Liposome Res. 1999, 9, 491.
3330. [66] J. K. Mills, D. Needham. Biochim. Biophys. Acta, Biomembr. 2005,
[31] C. Saraiva, C. Praça, R. Ferreira, T. Santos, L. Ferreira, 1716, 77.
L. Bernardino, J. Controlled Release 2016, 235, 34. [67] D. Needham, J. Y. Park, A. M. Wright, J. Tong. Faraday Discuss.
[32] B. Dehouck, L. Fenart, M.-P. Dehouck, A. Pierce, G. Torpier, 2013, 161, 515.
R. Cecchelli, J. Cell Biol. 1997, 138, 877. [68] K. Sou, L. Y. Chan, C.-L. K. Lee, ACS Sens. 2016, 1, 650.
[33] N. J. Abbott, A. A. K. Patabendige, D. E. M. Dolman, S. R. Yusof, [69] J. Ehrig, E. P. Petrov1, P. Schwille, New J. Phys. 2011, 13, 045019.
D. J. Begley, Neurobiol. Dis. 2010, 37, 13. [70] B. Hildebrandt, P. Wust, O. Ahlers, A. Dieing, G. Sreenivasa,
[34] J. Kreuter, D. Shamenkov, V. Petrov, P. Ramge, K. Cychutek, T. Kerner, R. Felix, H. Riess, Crit. Rev. Oncol./Hematol. 2002, 43, 33.
C. Koch-Brandt, R. Alyautdin, J. Drug Targeting 2002, 10, 317. [71] G. Blume, G. Cevc, Biochim. Biophys. Acta, Biomembr. 1990,
[35] M. Demeule, J. C. Currie, Y. Bertrand, C. Ché, T. Nguyen, 1029, 91.
A. Régina, R. Gabathuler, J. P. Castaigne, R. Béliveau, J. Neuro- [72] M. Matbou Riahi, A. Sahebkar, K. Sadri, S. Nikoofal-Sahlabadi,
chem. 2008, 106, 1534. M. R. Jaafari, Int. J. Pharm. 2018, 540, 89.
[36] R. Qiao, Q. Jia, S. Hüwel, R. Xia, T. Liu, F. Gao, H. J. Galla, M. Gao, [73] D. P. Huynh, M. K. Nguyen, B. S. Pi, M. S. Kim, S. Y. Chae, K. C. Lee,
ACS Nano 2012, 6, 3304. B. S. Kim, S. W. Kim, D. S. Lee, Biomaterials 2008, 29, 2527.
[37] S. Ruan, L. Qin, W. Xiao, C. Hu, Y. Zhou, R. Wang, X. Sun, W. Yu, [74] P. Díez, B. Esteban-Fernández de Ávila, D. E. Ramírez-Herrera,
Q. He, H. Gao, Adv. Funct. Mater. 2018, 28, 1802227. R. Villalonga, J. Wang, Nanoscale 2017, 9, 14307.
[38] F. Hervé, N. Ghinea, J.-M. Scherrmann, AAPS J. 2008, 10, 455. [75] L. Hou, Y. Zheng, Y. Wang, Y. Hu, J. Shi, Q. Liu, H. Zhang,
[39] J. V. Georgieva, D. Kalicharan, P. O. Couraud, I. A. Romero, Z. Zhang, ACS Appl. Mater. Interfaces 2018, 10, 21927.
B. Weksler, D. Hoekstra, I. S. Zuhorn, Mol. Ther. 2011, 19, 318. [76] K. Kono, T. Ozawa, T. Yoshida, F. Ozaki, Y. Ishizaka, K. Maruyama,
[40] Y. Cheng, Q. Dai, R. A. Morshed, X. Fan, M. L. Wegscheid, C. Kojima, A. Harada, S. Aoshima, Biomaterials 2010, 31, 7096.
D. A. Wainwright, Y. Han, L. Zhang, B. Auffinger, A. L. Tobias, [77] P. A. Yaroslavov, D. I. Panova, D. A. Sybachin, D. V. Spiridonov,
E. Rincón, B. Thaci, A. U. Ahmed, P. C. Warnke, C. He, M. S. Lesniak, P. A. Zezin, O. Mergel, A. Gelissen, D. R. Tiwari, D. F. Plamper,
Small 2014, 10, 5137. P. W. Richtering, P. F. Menger, Nanomed.: Nanotechnol. Biol. Med.
[41] G. Sonavane, K. Tomoda, K. Makino, Colloids Surf. B 2008, 66, 274. 2017, 13, 1491.
[42] V. N. Bharadwaj, J. Lifshitz, P. D. Adelson, V. D. Kodibagkar, S. [78] D. T. Simon, K. C. Larsson, D. Nilsson, G. Burström, D. Galter,
E. Stabenfeldt, Sci. Rep. 2016, 6, 1. M. Berggren, A. Richter-Dahlfors, Biosens. Bioelectron. 2015, 71,
[43] H. D. Dellmann, Microsc. Res. Tech. 1998, 41, 85. 359.
[44] C. L. Willis, C. J. Garwood, D. E. Ray. Neuroscience 2007, 150, 498. [79] S. D. Kong, W. Zhang, J. H. Lee, K. Brammer, R. Lal, M. Karin,
[45] S. Miyata, Front. Neurosci. 2015, 9, 390. S. Jin, Nano Lett. 2010, 10, 5088.
[46] R. Mo, T. Jiang, J. Di, W. Tai, Z. Gu, Chem. Soc. Rev. 2014, 43, 3595. [80] Y. Zhang, Q. Yin, L. Yin, L. Ma, L. Tang, J. Cheng, Angew. Chem.,
[47] K. Ulbrich, K. Holá, V. Šubr, A. Bakandritsos, J. Tuček, R. Zbořil, Int. Ed. 2013, 52, 6435.
Chem. Rev. 2016, 116, 5338. [81] J. H. Lee, K. J. Chen, S. H. Noh, M. A. Garcia, H. Wang, W. Y. Lin,
[48] H. Y. Yang, Y. Li, D. S. Lee, Adv. Ther. 2018, 1, 1800011. H. Jeong, B. J. Kong, D. B. Stout, J. Cheon, H. R. Tseng, Angew.
[49] S. Mura, J. Nicolas, P. Couvreur. Nat. Mater. 2013, 12, 991. Chem., Int. Ed. 2013, 52, 4384.

Small 2019, 15, 1900132 1900132  (11 of 12) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.small-journal.com

[82] P. Zhao, M. Zheng, Z. Luo, P. Gong, G. Gao, Z. Sheng, C. Zheng, [102] M. Kumar, R. S. Pandey, K. C. Patra, S. K. Jain, M. L. Soni,
Y. Ma, L. Cai, Sci. Rep. 2015, 5, 14258. J. S. Dangi, J. Madan, Int. J. Biol. Macromol. 2013, 61, 189.
[83] X. Liang, J. Gao, J. Jiang, J. Luo, L. Jing, X. Li, Y. Jin, Z. Dai, [103] W. Li, R. Luo, X. Lin, A. D. Jadhav, Z. Zhang, L. Yan, C. Y. Chan,
ACS Nano 2015, 9, 1280. X. Chen, J. He, C. H. Chen, P. Shi, Biomaterials 2015, 65, 76.
[84] S. Zhang, X. Qian, L. Zhang, W. Peng, Y. Chen, Nanoscale 2015, 7, [104] Y. Z. Zhao, R. R. Jin, W. Yang, Q. Xiang, W. Z. Yu, Q. Lin, F. R. Tian,
7632. K. L. Mao, C. Z. Lv, Y. X. Wáng, C. T. Lu, PLoS One 2016, 11, e0148848.
[85] L. Deng, Q. Li, S. Al-Rehili, H. Omar, A. Almalik, A. Alshamsan, [105] L. Fan, J. Wang, F. Meng, Y. Luo, X. Sui, B. Zhao, W. Li, D. Quan,
J. Zhang, N. M. Khashab, ACS Appl. Mater. Interfaces 2016, 8, J. Yang, Y. Wang, J. Biomed. Nanotechnol. 2018, 14, 2066.
6859. [106] A. Gaudin, M. Yemisci, H. Eroglu, S. Lepetre-Mouelhi, O. F. Turkoglu,
[86] Q. Feng, Y. Zhang, W. Zhang, Y. Hao, Y. Wang, H. Zhang, L. Hou, B. Dönmez-Demir, S. Caban, M. F. Sargon, S. Garcia-Argote,
Z. Zhang, Acta Biomater. 2017, 49, 402. G. Pieters, O. Loreau, B. Rousseau, O. Tagit, N. Hildebrandt,
[87] G. Gao, Y. W. Jiang, H. R. Jia, F. G. Wu, Biomaterials 2019, 188, 83. Y. Le Dantec, J. Mougin, S. Valetti, H. Chacun, V. Nicolas,
[88] G. Jalani, V. Tam, F. Vetrone, M. Cerruti, J. Am. Chem. Soc. 2018, D. Desmaële, K. Andrieux, Y. Capan, T. Dalkara, P. Couvreur, Nat.
140, 10923. Nanotechnol. 2014, 9, 1054.
[89] Y. Cao, Y. Chen, T. Yu, Y. Guo, F. Liu, Y. Yao, P. Li, D. Wang, [107] G. C. Vaz, A. P. Bahia, F. C. de Figueiredo Müller-Ribeiro,
Z. Wang, Y. Chen, H. Ran, Theranostics 2018, 8, 1327. C. H. Xavier, K. P. Patel, R. A. Santos, F. A. Moreira, F. Frézard,
[90] S. Lee, G. Hwang, T. H. Kim, S. J. Kwon, J. U. Kim, K. Koh, B. Park, M. A. Fontes, Neuroscience 2015, 285, 60.
H. Hong, K. J. Yu, H. Chae, Y. Jung, J. Lee, T. I. Kim, ACS Nano [108] G. C. Vaz, N. M. Sharma, H. Zheng, M. C. Zimmerman, R. S. Santos,
2018, 12, 6756. F. Frezard, M. A. P. Fontes, K. P. Patel, J. Neurosci. Methods 2016,
[91] C. Y. Lai, B. G. Trewyn, D. M. Jeftinija, K. Jeftinija, S. Xu, S. Jeftinija, 273, 55.
V. S. Lin, J. Am. Chem. Soc. 2003, 125, 4451. [109] M. A. P. Fontes, G. C. Vaz, T. Z. D. Cardoso, M. F. de Oliveira,
[92] M. J. During, A. Freese, A. Y. Deutch, P. G. Kibat, B. A. Sabel, M. J. Campagnole-Santos, R. A. S. dos Santos, N. M. Sharma,
R. Langer, R. H. Rot, Exp. Neurol. 1992, 115, 193. K. P. Patel, F. Frézard, Nanomed.: Nanotechnol. Biol. Med. 2018,
[93] M. A. Malvindi, R. Di Corato, A. Curcio, D. Melisi, M. G. Rimoli, 14, 781.
C. Tortiglione, A. Tino, C. George, V. Brunetti, R. Cingolani, [110] D. M. Treiman, Epilepsia 2001, 42, 8.
T. Pellegrino, A. Ragusa, Nanoscale 2011, 3, 5110. [111] U. Chitgupi, S. Shao, K. A. Carter, W. C. Huang, J. F. Lovell, Nano
[94] A. Trapani, E. De Giglio, D. Cafagna, N. Denora, G. Agrimi, Lett. 2018, 18, 1331.
T. Cassano, S. Gaetani, V. Cuomo, G. Trapani, Int. J. Pharm. 2011, [112] K. Gu, Y. Xu, H. Li, Z. Guo, S. Zhu, S. Zhu, P. Shi, T. D. James,
419, 296. H. Tian, W. H. Zhu, J. Am. Chem. Soc. 2016, 138, 5334.
[95] C. Spuch, C. Navarro, J. Drug Delivery 2011, 2011, 1. [113] A. Shao, Y. Xie, S. Zhu, Z. Guo, S. Zhu, J. Guo, P. Shi, T. D. James,
[96] S. Di Gioia, A. Trapani, D. Mandracchia, E. De Giglio, S. Cometa, H. Tian, W. H. Zhu, Angew. Chem., Int. Ed. 2015, 54, 7275.
V. Mangini, F. Arnesano, G. Belgiovine, S. Castellani, L. Pace, [114] S. J. Leung, M. Romanowski, Theranostics 2012, 2, 1020.
M. A. Lavecchia, G. Trapani, M. Conese, G. Puglisi, T. Cassano, [115] M. Li, C. Teh, C. Y. Ang, S. Y. Tan, Z. Luo, Q. Qu, Y. Zhang,
Eur. J. Pharm. Biopharm. 2015, 94, 180. V. Korzh, Y. Zhao, Adv. Funct. Mater. 2015, 25, 5602.
[97] R. Pahuja, K. Seth, A. Shukla, R. K. Shukla, P. Bhatnagar, [116] B. Wu, B. Wan, S. T. Lu, K. Deng, X. Q. Li, B. L. Wu, Y. S. Li,
L. K. Chauhan, P. N. Saxena, J. Arun, B. P. Chaudhari, D. K. Patel, R. F. Liao, S. W. Huang, H. B. Xu, Int. J. Nanomed. 2017, 12, 4467.
S. P. Singh, R. Shukla, V. K. Khanna, P. Kumar, R. K. Chaturvedi, [117] Y. Zhao, X. Dai, X. Wei, Y. Yu, X. Chen, X. Zhang, C. Li, ACS Appl.
K. C. Gupta, ACS Nano 2015, 9, 4850. Mater. Interfaces 2018, 10, 14426.
[98] Y. S. Kang, H. J. Jung, J. S. Oh, D. Y. Song, CNS Neurosci. Ther. [118] S. Guha, S. K. Shaw, G. T. Spence, F. M. Roland, B. D. Smith,
2016, 22, 817. Langmuir 2015, 31, 7826.
[99] A. Ragusa, P. Priore, A. M. Giudetti, G. Ciccarella, A. Gaballo, [119] J. H. Su, H. P. Sun, Q. S. Meng, Q. Yin, P. C. Zhang, Z. W. Zhang,
Appl. Sci. 2018, 8, 474. H. J. Yu, Y. P. Li, Adv. Funct. Mater. 2016, 26, 7495.
[100] M. Qu, Q. Lin, S. He, L. Wang, Y. Fu, Z. Zhang, L. Zhang, [120] K. A. D. Gregersen, Z. B. Hill, J. C. Gadd, B. S. Fujimoto, D. J. Maly,
J. Controlled Release 2018, 277, 173. D. T. Chiu, ACS Nano 2010, 4, 7603.
[101] A. Lopalco, A. Cutrignelli, N. Denora, A. Lopedota, M. Franco, [121] H. Bernheimer, W. Birkmayer, O. Hornykiewicz, K. Jellinger,
V. Laquintana, Nanomaterials 2018, 8, 178. F. Seitelberger, J. Neurol. Sci. 1973, 20, 415.

Small 2019, 15, 1900132 1900132  (12 of 12) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim

You might also like