ArticlePDF Available

An enhanced antioxidant strategy of astaxanthin encapsulated in ROS-responsive nanoparticles for combating cisplatin-induced ototoxicity

Springer Nature
Journal of Nanobiotechnology
Authors:

Abstract and Figures

Background Excessive accumulation of reactive oxygen species (ROS) has been documented as the crucial cellular mechanism of cisplatin-induced ototoxicity. However, numerous antioxidants have failed in clinical studies partly due to inefficient drug delivery to the cochlea. A drug delivery system is an attractive strategy to overcome this drawback. Methods and results In the present study, we proposed the combination of antioxidant astaxanthin (ATX) and ROS-responsive/consuming nanoparticles (PPS-NP) to combat cisplatin-induced ototoxicity. ATX-PPS-NP were constructed by the self-assembly of an amphiphilic hyperbranched polyphosphoester containing thioketal units, which scavenged ROS and disintegrate to release the encapsulated ATX. The ROS-sensitivity was confirmed by ¹H nuclear magnetic resonance spectroscopy, transmission electron microscopy and an H2O2 ON/OFF stimulated model. Enhanced release profiles stimulated by H2O2 were verified in artificial perilymph, the HEI-OC1 cell line and guinea pigs. In addition, ATX-PPS-NP efficiently inhibited cisplatin-induced HEI-OC1 cell cytotoxicity and apoptosis compared with ATX or PPS-NP alone, suggesting an enhanced effect of the combination of the natural active compound ATX and ROS-consuming PPS-NP. Moreover, ATX-PPS-NP attenuated outer hair cell losses in cultured organ of Corti. In guinea pigs, NiRe-PPS-NP verified a quick penetration across the round window membrane and ATX-PPS-NP showed protective effect on spiral ganglion neurons, which further attenuated cisplatin-induced moderate hearing loss. Further studies revealed that the protective mechanisms involved decreasing excessive ROS generation, reducing inflammatory chemokine (interleukin-6) release, increasing antioxidant glutathione expression and inhibiting the mitochondrial apoptotic pathway. Conclusions Thus, this ROS-responsive nanoparticle encapsulating ATX has favorable potential in the prevention of cisplatin-induced hearing loss. Graphical Abstract
This content is subject to copyright. Terms and conditions apply.
Guetal. Journal of Nanobiotechnology (2022) 20:268
https://doi.org/10.1186/s12951-022-01485-8
RESEARCH
An enhanced antioxidant strategy
ofastaxanthin encapsulated inROS-responsive
nanoparticles forcombating cisplatin-induced
ototoxicity
Jiayi Gu1,2,3†, Xueling Wang1,2,3†, Yuming Chen1,2,3, Ke Xu1,2,3, Dehong Yu1,2,3,4* and Hao Wu1,2,3*
Abstract
Background: Excessive accumulation of reactive oxygen species (ROS) has been documented as the crucial cellular
mechanism of cisplatin-induced ototoxicity. However, numerous antioxidants have failed in clinical studies partly due
to inefficient drug delivery to the cochlea. A drug delivery system is an attractive strategy to overcome this drawback.
Methods and results: In the present study, we proposed the combination of antioxidant astaxanthin (ATX) and ROS-
responsive/consuming nanoparticles (PPS-NP) to combat cisplatin-induced ototoxicity. ATX-PPS-NP were constructed
by the self-assembly of an amphiphilic hyperbranched polyphosphoester containing thioketal units, which scavenged
ROS and disintegrate to release the encapsulated ATX. The ROS-sensitivity was confirmed by 1H nuclear magnetic
resonance spectroscopy, transmission electron microscopy and an H2O2 ON/OFF stimulated model. Enhanced release
profiles stimulated by H2O2 were verified in artificial perilymph, the HEI-OC1 cell line and guinea pigs. In addition, ATX-
PPS-NP efficiently inhibited cisplatin-induced HEI-OC1 cell cytotoxicity and apoptosis compared with ATX or PPS-NP
alone, suggesting an enhanced effect of the combination of the natural active compound ATX and ROS-consuming
PPS-NP. Moreover, ATX-PPS-NP attenuated outer hair cell losses in cultured organ of Corti. In guinea pigs, NiRe-PPS-NP
verified a quick penetration across the round window membrane and ATX-PPS-NP showed protective effect on spiral
ganglion neurons, which further attenuated cisplatin-induced moderate hearing loss. Further studies revealed that
the protective mechanisms involved decreasing excessive ROS generation, reducing inflammatory chemokine (inter-
leukin-6) release, increasing antioxidant glutathione expression and inhibiting the mitochondrial apoptotic pathway.
Conclusions: Thus, this ROS-responsive nanoparticle encapsulating ATX has favorable potential in the prevention of
cisplatin-induced hearing loss.
Keywords: ROS-responsive, Poly(propylene sulfide), Astaxanthin, Cisplatin-induced ototoxicity
© The Author(s) 2022. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which
permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the
original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or
other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line
to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this
licence, visit http:// creat iveco mmons. org/ licen ses/ by/4. 0/. The Creative Commons Public Domain Dedication waiver (http:// creat iveco
mmons. org/ publi cdoma in/ zero/1. 0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
Open Access
Journal of Nanobiotechnology
Jiayi Gu and Xueling Wang are First authors
*Correspondence: dehongyu@126.com; wuhao@shsmu.edu.cn
1 Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth
People’s Hospital, Shanghai Jiao Tong University School of Medicine,
Shanghai, China
Full list of author information is available at the end of the article
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 2 of 17
Guetal. Journal of Nanobiotechnology (2022) 20:268
Introduction
To date, the precise mechanism underlying cisplatin-
induced ototoxicity is still unclear. However, accu-
mulation of reactive oxygen species (ROS) has been
documented as a critical mediator in cisplatin-induced
ototoxicity, involved in many pathological processes.
In the cochlea, cisplatin upregulates the expression of
NADPH oxidase 3 and xanthine oxidase, two major
sources of ROS [13]. ROS then facilitate the imbalance
between ROS and the antioxidant system by deplet-
ing antioxidant molecules, including superoxide dis-
mutase, glutathione peroxidase, and catalase [46],
which is capable of inducing cochlear lipid peroxidation
by increasing concentrations of toxic chemicals, such as
malondialdehyde or 4-hydroxynonenal (4-HNE) [79].
Besides, excessive ROS opens calcium-permeable chan-
nels in sarcoplasmic/endoplasmic reticulum membranes
and plasma membranes [10], leading to an increase in
cytosolic calcium levels and eventual apoptotic and
autophagic cell death [11].
A wide range of therapeutic molecules including anti-
oxidants (apocynin, vitamin C), anti-inflammatory
(curcumin, epigallocatechin-3-gallate), anti-apoptotic
(pifithrinα) compounds, have been applied in preclinical
studies due to their preventative and restorative effects.
Of these compounds, antioxidants exhibit application
prospects. Astaxanthin (ATX), a xanthophyll carot-
enoid, has strong antioxidant capacity by scavenging
free radicals, quenching singlet oxygen, enhancing anti-
oxidant enzymes and inhibiting lipid peroxidation [12].
Meanwhile, it is associated with maintaining the mito-
chondrial redox state and functional integrity against
oxidative stress [13]. However, due to severe restriction
of the blood-labyrinth barrier, lack of sufficient drug con-
centrations in the inner ear often exist after intravenous
injection or oral intake (systemic administration) [14].
Even after intratympanic injection (local administration),
ATX can hardly penetrate the round window membrane
(RWM) and enter the inner ear due to its hydrophobicity
and instability. In our previous study, ATX first demon-
strated its otoprotective effect against cisplatin-induced
ototoxicity with the aid of lipid-polymer hybrid nano-
particles (LPN) [15]. erefore, inner ear drug delivery
strategies have attracted much attention due to their
contribution in allowing drug access to the inner ear,
improving cochlear drug concentrations and ultimately
strengthening drug efficacy [16, 17].
Recently, increasing attention has been paid to the
development of stimuli-responsive nanoparticles,
which realize the accurate spatiotemporal control of
drug release and minimize toxicity by avoiding dam-
age to non-target sites [18]. e stimuli that activate the
Graphical Abstract
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 3 of 17
Guetal. Journal of Nanobiotechnology (2022) 20:268
responsiveness of nanomaterials can either be endog-
enous (redox, pH, enzyme) or exogenous (light, heat,
magnetic field and ultrasound) [19]. Notably, ROS-
responsive drug delivery systems in endogenous patho-
logical conditions have been extensively studied [2023].
According to the literatures, ROS-responsive systems
include polysulfides, polyselenides, polythioketals, poly-
oxalates, oligoproline- and catechol-based materials [24].
Poly(propylene sulfide) (PPS), as a type of polysulfide,
can be successively oxidized to sulfoxide by hydrogen
peroxide (H2O2) and to sulfone by hypochlorite [25].
e two-step reaction results in a large change in polar-
ity and increases water solubility, consequently lead-
ing to an extensive disintegration of PPS and control
of drug release. As reported, Poly(propylene sulfide)-
poly(ethylene glycol) (PPS-PEG) has been used for drug
delivery in a broad spectrum of diseases, including can-
cers, diabetes and inflammation-related injuries [2628].
Recently, ROS-responsive nanoparticles encapsulating
berberine have been developed and applied in noise-
induced hearing loss [29].
In this study, we fabricated a novel ROS-responsive/
consuming nanoparticle system loaded with the antioxi-
dant ATX (ATX-PPS-NP) and confirmed its ‘on demand’
spatiotemporal release invitro and in vivo. is system
combined the ROS-consuming effect of PPS-PEG and
the antioxidant efficacy of ATX. e protection against
cisplatin-induced ototoxicity was verified in HEI-OC1
cells, cochlear explants and guinea pigs, where antioxi-
dant/anti-inflammatory/anti-apoptotic processes were
involved. ATX-PPS-NP eventually reduced the loss of
spiral ganglion neuron (SGNs) and prevented hearing
loss in animal models of cisplatin-induced hearing loss.
us, providing a novel strategy for the prevention of cis-
platin-induced hearing loss.
Materials andmethods
Materials
PPS-PEG (molecular weight [MW] 54 kDa) and FITC-
conjugated PPS-PEG (FITC-PPS-PEG, MW 54 kDa)
were purchased from Xi’an Ruixi Biological Technol-
ogy Co., Ltd (Xi’an, China), and successful synthesis
was confirmed by 1H nuclear magnetic resonance (1H-
NMR) spectra. As shown in Additional file 1: Fig. S1,
FTIR spectra illustrated an absorption peak at 1024 cm1
after treating PPS-PEG with 30% H2O2 solution, indicat-
ing the chemical bond change into S = O according to
the literature. Astaxanthin and Nile red were purchased
from Sigma (St Louis, MO, USA). Cisplatin was provided
by Aladdin (Shanghai, China). Chlorpromazine (CPZ),
filipin, nystatin and nocodazole were purchased from
Sigma (St Louis, MO, USA), and EIPA was from Med-
ChemExpress (Shanghai, China). ROS probes, Amplex
UltraRed Reagent and 2ʹ, 7ʹ-dichlorofluorescein diacetate
(DCFHDA), were obtained from Invitrogen (Carlsbad,
CA, USA) and Sigma.Texas Red conjugated cisplatin was
supplied by Ursa Bioscience (Abingdon, MD, USA).e
FITC-Annexin V/PI double staining kit was obtained
from BD Biosciences (San Diego, CA, USA).e GSH-
Glo Glutathione Assay kit was obtained from Promega
(Madison, WI, USA).e IL-6 ELISA kit was provided
by R&D Systems (Minneapolis, MN, USA). Antibod-
ies, includingcleaved-caspase 3, cleaved-caspase 9, P53,
cytochrome-C andα-tubulin, were purchased from CST
(Danvers, MA, USA), and Myosin VIIa was from Pro-
teus Biosciences (Ramona, CA, USA).4-hydroxynonenal
(4-HNE) was purchased from Abcam (Cambridge, MA,
USA).
Cell culture
HEI-OC1 (generated at House Ear Institute, Los Angeles,
CA, USA), an inner ear cell line derived from the audi-
tory organ of the transgenic mouse Immortomouse, is
a potential tool for studying the mechanisms of ototoxic
drugs invitro [30]. HEI-OC1 cells were cultured in Dul-
becco’s Modified Eagle’s Medium (DMEM) and supple-
mented with 10% (v/v) fetal bovine serum (FBS) in a 10%
CO2 incubator at 33°C.
Animals
Healthy female adult guinea pigs (weighing 200–250g,
with no otitis media) and mouse pups (C57BL/6J, P0-3)
were used. roughout the study, all animals were main-
tained under standard laboratory conditions, housed
under standard animal facility conditions with adequate
food and water. All animal studies were approved and
conducted in accordance with the guidelines of the Ethics
Committee of the Shanghai Jiaotong University, School
of Medicine (Shanghai, China).
Preparation ofastaxanthin‑loaded ROS‑responsive
nanoparticles (ATX‑PPS‑NP)
ATX-PPS-NP was fabricated using a single emulsion and
solvent evaporation method as described previously [15].
irty milligrams of PPS-PEG and 2mg ATXwere dis-
solved in 1ml of dichloromethane solution, followed by
pouring 3ml of sodium cholate solution (3%, w/v) into
the organic solvent. e mixture was sonicated at 260W
for 4min (Scientz Biotechnology Co., Ltd., Ningbo City,
China). e resulting oil/water emulsion was further
diluted in 36 ml of 0.5% sodium cholate solution and
stirred overnight to remove the organic solvent. ATX-
PPS-NP were collected by centrifugation (11,000×g,
30min) and washed twice to remove excessive emulsifier
and unloaded ATX. FITC-PPS-NP were prepared in the
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 4 of 17
Guetal. Journal of Nanobiotechnology (2022) 20:268
same way except that PPS-PEG was replaced with FITC-
PPS-PEG. NiRe-PPS-NP were loaded with the fluores-
cent probe Nile red. e NP were stored at 4°C.
Characterization
Particle size, polydispersity index (PDI) and z-potential
values were measured by zeta potential measurement
(Nano ZS90 Zetasizer, Malvern Instruments Co., Ltd.,
UK) equipped with dynamic light scattering (DLS).
Encapsulation efficiency (EE %) and drug loading (DL
%) of the ATX-PPS-NP were determined using liquid
chromatography combined with mass spectrometry
(LC–MS) as described previously [16].
ATX stability
Absorbance measurements were performed to assess
the stability of ATX loaded in NP [31]. Natural ATX
in EtOH/DCM (% v/v; 60/40) was used as the stand-
ard for the ATX calibration curve (absorbance-con-
centration curve) based on Beer–Lambert law, under
the maximum absorption (λmax) at 490 nm. At vari-
ous time points (0, 7, 14 and 21 days), ATX-PPS-NP
or ATX were diluted and dissolved in EtOH/DCM for
the ATX concentrations measurements by microplate
reader.e remaining ATX was normalized by dividing
the concentration of ATX at day 0.
ROS‑responsive release prole invitro
e release kinetics of PPS-NP invitro was simulated
by the fluorescence quenching of loaded Nile red, a
special fluorescent probe which fluoresces strongly in a
hydrophobic environment, but quenches when released
into an aqueous phase where it is poorly soluble. Ten
microliters of NiRe-PPS-NP was added to H2O2 solu-
tion diluted in artificial perilymph (0, 5 and 500mM)
to each well of a 96-well plate which was stirred and
placed in a gas bath at 37°C. e Nile red fluorescence
was measured using a microplate reader (Tecan Spark,
Tecan Group Ltd.) at an excitation/emission wave-
length of 535nm/612nm at specific time points from
0 to 72h.
To evaluate ROS-responsive drug release, H2O2 solu-
tion was used to stimulate ROS release. Briefly, NiRe-
PPS-NPwere subjected to 0 or 500mM H2O2 solution at
each ON phase for 24h, and replaced byPBS for another
24h for the OFF phase,circulating for a maximum dura-
tion of 7days. At the end of each interval,Nile red fluo-
rescence was measured by a microplate reader (Tecan
Spark, Tecan Group Ltd.) at an excitation/emission wave-
length of 535nm/612nm.
To further evaluate disintegration of ATX-PPS-NP in
the ROS environment, transmission electron microscopy
(TEM) was used to observe the morphological changes in
PPS-NP. Briefly, ATX-PPS-NP were treated with 0.3% or
3% H2O2 solution for 24 or 48h.
ROS‑responsive release prole—in HEI‑OC1 cells
HEI-OC1 cells were seeded and treated with 60 μM
cisplatin for 24 h, followed by 24 h-culture in NiRe-
PPS-NP. After washing twice, nuclei were stained with
Hoechst 33,342. H2O2 solution (200μM) was used as a
positive control. Fluorescent images were captured with
a laser confocal microscope (LSM880, Zeiss, Germany).
Cellular uptake assay
HEI-OC1 cells (5 × 104 cells per well) were seeded in a
24-well plate and allowed to adhere overnight. e cells
were then incubated with FITC-PPS-NP for 1, 3, 6, 12
and 24h. e cells were washed with PBS, fixed with
4% paraformaldehyde (PFA) for 15 min and stained
with Hoechst 33,342. e FITC fluorescence was exam-
ined using a laser confocal microscope (LSM-880,
Zeiss, Germany). To quantify cellular uptake, cells were
prepared as previously described in a 6-well plate and
collected for flow cytometry (BD Biosciences, Fortessa,
CA, USA).
To examine the endocytosis mechanism of PPS-NP,
HEI-OC1 cells were pre-incubated with the following
inhibitors based on previous studies [32] at nontoxic
concentrations: 15 µg/ml of chlorpromazine (CPZ), to
inhibit clathrin-mediated endocytosis; 100μM of EIPA
(MCE), to block macropinocytosis and phagocytosis;
6µg/ml filipin and 50µg/ml of nystatin, to inhibit cav-
eolae-mediated endocytosis; and 2µg/ml nocodazole, to
disrupt microtubules. e inhibitors were added to the
cell culture medium or incubated for 45min at 4°C prior
to addition of the FITC-PPS-NP suspensions for another
3h. Subsequently, the cells were washed and collected for
flow cytometry (BD Biosciences, Fortessa, CA, USA).
ROS‑responsive accumulation ofPPS‑NP
In HEI‑OC1 cells
HEI-OC1 cells were cultured in dishes for confocal
images. e cells were pretreated with CDDP (60 μM)
or free medium for 24h, and additionally co-incubated
with Amplex UltraRed Reagent and FITC-PPS-NP.
Immediately, the cells wereimaged with a laser confocal
microscope (LSM880, Zeiss) in a time series, at 10min
intervals for up to 30min. H2O2 solution (200μM) was
used as a positive control.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 5 of 17
Guetal. Journal of Nanobiotechnology (2022) 20:268
In cultured Organ ofCorti
e organs of Corti were dissected from neonatal mice
(postnatal days 1–3) and cultured in the medium as
described previously [17]. Explants were co-incubated
with Texas Red conjugated cisplatin and FITC-PPS-NP
for 24h. e tissues were then stained with Myosin VIIa
and Hoechst 33,342. Fluorescent images were captured
with a laser confocal microscope (LSM880, Zeiss).
Protective eect inHEI‑OC1 cells
Cell viability assay
Cell viability in different patterns of cisplatin and drug
was examined using the CCK-8 assay (Dojindo, Japan). In
brief, HEI-OC1 cells (1 × 105 cells per well) were seeded
in 96-well plates and incubated overnight. e cells were
divided into six groups (ATX, PPS-NP, ATX-PPS-NP,
administrated at the same ATX concentration of 1μg/ml)
as follows: (1) Control, (2) drug alone for 24h, (3) cispl-
atin (60μM) for 24h, (4) pretreatment with drug for 4h
and withdrawal of the drug followed by treatment with
CDDP for 24 h (pre-4 h), (5) pretreatment with drug
for 4h followed by a co-treatment with CDDP for 24h
(pre-4h + co-24h) and (6) direct co-treatment with drug
and CDDP for 24h (co-24h). After washing three times,
100μl of dissolved CCK-8 solution was added to each
well and incubated for 2h at 33°C. e absorbance was
measured using a microplate reader (Tecan Spark, Tecan
Group Ltd.) at 450nm. All data were normalized based
on background intensity. Blank PPS-NP with the same
concentrations were also examined in all cases with nan-
oparticles involved.
Cell apoptosis study
Cellular apoptosis was quantified by flow cytometry
using an FITC-Annexin V/PI double staining kit (BD
Biosciences, San Diego, CA, USA). HEI-OC1 cells were
pretreated with ATX, PPS-NP and ATX-PPS-NP for
4h followed by co-treatment with CDDP for 24 h. e
cells were then rinsed, trypsinized and collected after
centrifugation. e cells were resuspended in 100μl of
1 × binding buffer containing 5μl FITC-Annexin V and
PI, incubated for 15min at room temperature in the dark
and then analyzed by flow cytometry (BD Biosciences,
Fortessa, CA, USA). Both Annexin V + /PI cells repre-
senting early-apoptotic cells and Annexin V + /PI + cells
representing late-apoptotic cells were considered apop-
totic. Blank PPS-NP at the same concentrations were also
examined in all cases with nanoparticles involved.
Molecular mechanism oftheprotective eect inHEI‑OC1
cells
HEI-OC1 cells were divided into six groups: (1) Con-
trol, (2) CDDP, 60μM, 24h, (3–5) pretreated with ATX,
PPS-NP and ATX-PPS-NP, respectively, for 4h followed
by co-treatment with CDDP for 24 h. Each group was
included in the following experiments.
Intracellular ROS level
e intracellular ROS level was detected using a fluo-
rescent dye, 2’,7’-dichlorofluorescein diacetate (DCF-
HDA). Non-fluorescent DCFH is converted into the
highly fluorescent DCF by an intracellular oxida-
tion. For the assay, cells were seeded in a 24-well
plate overnight and treated with drugs as above. After
rinsing twice, 10 μM DCFHDA was added and cul-
tured for 30 min at room temperature in the dark.
Images were acquired by laser confocal microscopy
(488nm/530nm) and fluorescence intensities of ran-
domly chosen cells were quantified.
Glutathione (GSH) andinterleukin‑6 (IL‑6) assay
HEI-OC1 cells (1 × 105 cells per well) were seeded in
96-well plates and incubated overnight, followed by
drug administration as above. After 24h, the medium
was removed and the content of GSH was calculated
using aGSH-Glo Glutathione Assay kit. e removed
medium was collected for IL-6 detection using an
ELISA kit,according to the manufacturer’s protocol.
Apoptosis‑associated proteins
e levels of apoptosis-associated proteins were evalu-
ated by western blot. HEI-OC1 cells were cultured
and treated with drugs as above. After washing twice
with ice cold PBS on ice and lysed in RIPA, the pro-
tein extracts were subjected to 12% SDS-PAGE and
electrotransferred to PVDF membranes. is was fol-
lowed by blocking and incubation with specific anti-
bodies to cleaved-caspase 3, cleaved-caspase 9, P53 and
cytochrome-C (α-tubulin served as the control stand-
ard). e relative intensity was quantified by ImageJ
software.
Protective eect incultured Organ ofCorti
e organs of Corti were cultured as above and divided
into five groups: (1) Control, (2) CDDP, 60 μM, 24 h,
(3–5) pretreated with ATX, PPS-NP and ATX-PPS-NP,
respectively, at the same ATX concentration of 1 μg/
ml, for 4h and followed by co-treatment with CDDP for
24h. After fixation, hair cells were labelled with Myosin
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 6 of 17
Guetal. Journal of Nanobiotechnology (2022) 20:268
VIIa and quantified in the apex, middle and basal turns.
Fluorescent images were captured with a laser confocal
microscope (LSM880, Zeiss). e numbers of surviv-
ing hair cells in 1μm tissue of Organ of Corti were cal-
culated, and the percentages of those were compared in
groups.
Protective eect inguinea pigs
Evaluation ofRWM penetration
NiRe-PPS-NP were used to investigate whether PPS-
NP could penetrate the RWM. Briefly, guinea pigs were
anaesthetized with ketamine (60mg/kg, IP) and xyla-
zine (10 mg/kg, IP). NiRe-PPS-NP in gelatin sponge
were placed on the RWM as previously reported [15],
for 0.5, 2 and 6h, respectively (n = 3). After fixation,
the RWM was microdissected and prepared for confo-
cal imaging according to theprocedures described in
previous studies [33].A series of images of the RWM
by the z-axis scanning model at 1-µm vertical intervals
from the middle ear side to the scala tympani side were
collected and mean fluorescence intensities (MFI) were
calculated. e outer epithelium layer (OE), connective
tissue layer (CT), and inner epithelium layer (IE) of the
RWM z-stack series images were differentiated by cell
morphology.
In vivo release prole ofATX
Guinea pigs were divided into the control and cisplatin-
treated groups and intraperitoneally injected with PBS
and cisplatin (12mg/kg), respectively. At designed time
points of 0.5, 2, 6 and 12 h after RWM application of
ATX-PPS-NP in the control group and 2h in the cispl-
atin group, the cochleae were dissected after euthanasia
and perilymph was collected by aspiration using a sharp
glass pipette inserted into the scala tympani from the
RWM. Samples were stored at 80°C. e concentra-
tions of ATX were quantified using LC–MS. After dilut-
ing the samples, a 30µL aliquot of the sample was added
with 150µL IS (diclofenac, 100ng/ml) in ACN. e mix-
ture was vortexed for 10min and centrifuged at 3200g
for 10min and then vortexed for 10min and centrifuged
at 5800rpm for 10min. A 5µL aliquot of the supernatant
was injected for LC–MS analysis. e minimum concen-
tration detected was 1ng/ml.
Protective eect—Auditory Brainstem Responses (ABR) tests
To evaluate auditory function, ABR tests were recorded
in response to pure tone stimuli (4, 5.6, 8, 11.2, 16, 22.6,
32 and 45kHz) between 0 and 90dB in 5dB increments
using a closed-field microphone. e acoustic signals
were generated with a RZ6 signal processor and BioSig
software (TDT, Alachua FL, USA). resholds were
defined as the lowest stimulus level at which a response
was observed.
Guinea pigs were administrated with ATX-PPS-NP
(0.36 mg/ml, 7μl), ATX (0.36 mg/ml,7μl) or normal
saline (N.S.) on the RWM. One hour later, the animals
were intraperitoneally injected with cisplatin (12mg/kg).
ABR tests were conducted before the operation and one
or three days after CDDP injection.
Mechanism oftheprotective eect inanimals
Guinea pig cochlea samples were prepared as reported
previously [34]. Briefly, mouse cochleae were quickly dis-
sected from the temporal bone following rapid decapi-
tation. After fixation, decalcification and progressive
dehydration, the samples were embedded in paraffin.
Mid-modiolar section samples were then cut at 5–7μm
thickness and mounted on glass slides. Paraffin sections
were deparaffinized in xylene, rehydrated through an
ethanol series, and stained with H&E.For immunostain-
ing of 4-HNE in SGNs,cochlear sections were prepared
as previously described [35] and imaged with an optical
microscope. R2, R3, R4 was defined as cochlear turns
from apex to base.ImageJ software was used to measure
the area of SGN regions. SGN density was defined by the
number of SGNs/area of the SGN region. e integrated
optical density (IOD) of 4-HNE staining in the SGN area
was measured by ImageJ software.
Statistics
All data are reported as mean ± standard deviation (SD).
Analysis of Variance (ANOVA) with Bonferroni correc-
tion for multiple comparisons was used to determine
treatment effects. p < 0.05 was considered significant.
Results
Characterization ofATX‑PPS‑NP
e physicochemical characteristics of nanoparticles
were validated (Fig. 1) by measuring particle size, PDI
and z-potential values. As shown in Fig.1B and Table1,
the particle size of PPS-NP and ATX-PPS-NP were
169.57 ± 7.87 nm and 182.27 ± 5.05 nm in diameter,
respectively, and fitted the Gaussian shape line. A low
PDI value (0.38 ± 0.02) also indicated a relatively uniform
distribution of nanoparticles. Encapsulation efficiency
(EE %) and drug loading (DL %) were 68.07% ± 3.02%
and 4.31% ± 0.22%, respectively. Moreover, PPS-NP and
ATX-PPS-NP were positively charged at 10.86 ± 3.27mV
and 14.73 ± 0.87mV, respectively.
We investigated the stability of ATX-PPS-NP by meas-
uring the remaining concentrations of ATX at different
time points (day 0, 7, 14, 21) under light or heat stimu-
lation. As shown in Fig. 1C, ATX-PPS-NP significantly
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 7 of 17
Guetal. Journal of Nanobiotechnology (2022) 20:268
improved the stability of ATX on days 7, 14, and 21
under 37°C with or without light (dark), characterized
by the remaining concentration of ATX which was nota-
bly higher in the ATX-PPS-NP group than in the ATX
group. Furthermore, on day 21, ATX-PPS-NP preserved
a greater amount of ATX, 78% and 71%, respectively, in
dark and light environments, compared with 46% and
32% in free ATX groups (p < 0.01, p < 0.001). Temperature
had no impact on ATX stability when encapsulated in
PPS-NP under light (Fig.1D).
ROS‑responsive release prole invitro
e ROS-responsive release from PPS-NP was assessed
by the loss of fluorescence of encapsulated Nile red
(NiRe) (Fig.1E, F). Specifically, at each time point, the
fluorescence value which was subtracted from that of
Fig. 1 Physicochemical characterization of ATX-PPS-NP and in vitro ROS-responsive release. A The preparation of ATX-PPS-NP. B Gaussian
distribution of the ATX-PPS-NP. CD In 4 °C or 37 °C environments, the remaining percentages of ATX alone or encapsulated in PPS-NP in dark
or light conditions within 21 days. *p < 0.05, **p < 0.01, ***p < 0.001 vs ATX dark, ###p < 0.001 vs ATX light. E In vitro Nile red (NiRe) release from
NiRe-PPS-NP in various concentrations of H2O2 solutions. F In vitro release of NiRe with intermittent exposure (OFF/ON cycles every other day) to
500 mM H2O2 solution. G Representative TEM images of ATX-PPS-NP in control and various oxidation groups
Table 1 Particle size, zeta potential, PDI values, drug loading (DL%) and encapsulation efficiency (EE%) of PPS-NP and ATX-PPS-NP
Nanoparticles (NP) Particle Size (nm) Zeta potential (mV) PDI EE % DL %
PPS-NP 169.57 ± 7.87 10.86 ± 3.27 0.29 ± 0.03 / /
ATX-PPS-NP 182.27 ± 5.05 14.73 ± 0.87 0.38 ± 0.02 68.07 ± 3.02 4.31 ± 0.22
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 8 of 17
Guetal. Journal of Nanobiotechnology (2022) 20:268
the sample prior to H2O2 addition was normalized by
the control. Under 5 mM H2O2 oxidation, approxi-
mately 45% of NiRe was released over 48h, double higher
than 20% released from the control group, while NiRe
released from NiRe-PPS-NP oxidized by 500mM H2O2
was approximately 100%. Moreover, NiRe release at each
time point was higher in the oxidation group than in the
control. ese results indicated that ROS could quickly
trigger the disintegration of NiRe-PPS-NP at an early
stage and the encapsulated drug was released in a dose-
dependent manner upon exposure to H2O2. To illustrate
the smart release of encapsulated drugs, NiRe-PPS-NP
wereintermittently exposed to 0 or 500mM H2O2 solu-
tion OFF/ON cycles every other day. e slope of the
release curve was steeper during the ON phases than
during the OFF phases, indicating that “on demand”
release could be achieved. Together, the release pro-
files demonstrated ROS concentration-dependent, on
demand release of the loaded drugs from PPS-NP.
To further evaluate disintegration of ATX-PPS-NP
in the ROS environment, TEM was used to observe the
morphological change in PPS-NP. As shown in Fig.1G,
PPS-NP exhibited a spherical structure with an average
diameter between 100–200 nm, in agreement with the
size measurement performed by DLS. After oxidation
with 0.3%-3% H2O2 solution for 24–48h mimicking ROS
stimulation, NP progressively disintegrated and fused,
suggesting ROS-responsiveness of PPS-NP.
Cellular uptake andendocytosis mechanism
To investigate the endocytosis of PPS-NP and the
underlying mechanism, we fabricated FITC-PPS-NP as
mentioned above, where FITC was used as a tracer flu-
orescein in the formulation of PPS-NP. Confocal laser
scanning microscopy and flow cytometry were per-
formed to observe and quantify the uptake profile in
HEI-OC1 cells, respectively. As shown in Fig.2A–C, cel-
lular uptake showed a time-dependent increase between
1–12h. After reaching the peak at 12h, the uptake activ-
ity decreased at 24h.
To elucidate the endocytic pathways of PPS-NP, we
studied the effects of selective inhibitors and low temper-
ature (4°C) on cellular uptake. Of the inhibitors studied,
treatment with CPZ led to significantly reduced fluores-
cence, suggesting that clathrin-mediated endocytosis
may contribute to the uptake of PPS-NP in HEI-OC1
cells (Fig.2D), in line with the size of particles (about
100nm in diameter) internalized inthe clathrin-medi-
ated pathway [36]. Furthermore, low temperature (4°C)
also decreased the internalization of FITC-PPS-NP, sug-
gesting energy-dependent endocytosis.
ROS‑responsive accumulation ofPPS‑NP inHEI‑OC1 cells
andcultured Organ ofCorti
Firstly, we screened the optimal dose and time for the
CDDP-treated HEI-OC1 cell model. e results showed
that 60 μM CDDP for 24 h induced approximately
70% cell viability and a significant generation of ROS
Fig. 2 In vitro cellular uptake of FITC-PPS-NP. A Confocal images demonstrated cellular uptake of FITC-PPS-NP for various periods of time (1, 3, 6, 12
and 24 h) in HEI-OC1. B, C Flow cytometry assay of cellular uptake and quantifications of MFI of FITC. D Endocytosis mechanism of PPS-NP. **p < 0.01
vs control, ***p < 0.001 vs control
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 9 of 17
Guetal. Journal of Nanobiotechnology (2022) 20:268
(Additional file1: Fig. S2). en we evaluated the ROS-
responsive accumulation of FITC-PPS-NP. In Fig.3A–C,
HEI-OC1 cells treated with CDDP (60μM, 24h) were co-
cultured with FITC-PPS-NP and Amplex Ultrared, which
respectively trafficked intracellular locations of NP and
labeled ROS generated, and H2O2 (200μM, 3h) was used
as a positive control. Serial images of live cells demon-
strated thatFITC-PPS-NP progressively entered cells and
approached where ROS were located, suggesting passive
ROS-responsive accumulation of FITC-PPS-NP invitro.
A stronger green fluorescence was observed in the CDDP
group compared with the control group, indicating that
ROS promoted the uptake ofFITC-PPS-NP.
Figure 3D showed the intracellular ROS-responsive-
ness of NiRe-PPS-NP. In HEI-OC1 cells, NiRe-PPS-
NP were quickly internalized. After exposure to CDDP
(60μM), the red fluorescence was quenched, indicating
ROS-triggered release of NiRe into the aqueous phase in
response to oxidative stress, while H2O2 (200μM) was
used as a positive control.
In Fig. 3E, Texas Red and FITC fluorescence traf-
ficked CDDP and NP, respectively, while Myosin VIIa
labeled hair cells. Compared with a fairly weak FITC sig-
nal in the control group,the whole and detailed images
of organotypic culture inthe cisplatin group showed a
stronger green fluorescence, mainly inthe spiral limbus
and hair cells, suggesting that cisplatin treatment pro-
moted the uptake of NP. e merging signal of Texas Red
and FITC indicated that FITC-PPS-NP targeted the spe-
cific sites damaged by cisplatin, where ROS seemed to be
generated.
Cytoprotective andanti‑apoptotic activity ofATX‑PPS‑NP
As illustrated in Fig. 4A, HEI-OC1 cells were treated
with CDDP and different drugs, including ATX (1μg/
ml), PPS-NP (drug-free) and ATX-PPS-NP (1 μg/
ml). Cell viability was significantly increased in the
ATX pre-4 h group (p < 0.001) and drug-free PPS-NP
pre-4h + co-24h group (p < 0.001), which was inhibited
by CDDP. ATX-PPS-NP resulted in higher viability after
Fig. 3 ROS-responsive accumulation and release profile in HEI-OC1 cells and cultured Organ of Corti. AC Live cell imaging of cellular uptake of
FITC-PPS-NP within 30 min under CDDP (60 μM, 24 h) stimulation, H2O2 (200 μM, 3 h) as a positive control. D Confocal images showed the Nile Red
fluorescence changed at various time points in control or CDDP (60 μM) treated HEI-OC1 cells, H2O2 (200 μM) as a positive control. E Representative
confocal images of cochlear explants. Texas Red (TR) and FITC fluorescence respectively labeled CDDP and NP, while Myosin VIIa labeled hair cells
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 10 of 17
Guetal. Journal of Nanobiotechnology (2022) 20:268
pre-4h + co-24h of administration, up to 1.5-fold com-
pared with that of the CDDP group. ese results suggest
that ATX-PPS-NP, combining ROS-consuming PPS-NP
and antioxidant ATX, demonstrated an enhanced protec-
tive effect against cisplatin-induced cytotoxicity.
Annexin V-FITC/PI double staining was used to label
apoptotic cells and the percentage of these cells was
analyzed by flow cytometry (Fig.4B, C). CDDP induced
18.43% of apoptosis, which was reduced by ATX-PPS-NP
(p < 0.01), while no significant difference occurred in the
ATX and drug-free PPS-NP group, confirming that ATX-
PPS-NP exhibited notable efficiency in attenuating cell
apoptosis.
As shown in Fig.4D, E, the intact arrangement of hair
cells was destroyed following CDDP administration and
was partially restored by ATX-PPS-NP. To evaluate the
protective effect of ATX-PPS-NP, hair cell counts were
calculated. e results revealed that the CDDP group
exhibited severe loss of hair cells and ATX-PPS-NP pre-
served hair cells in the basal turn ofthe organ of Corti
explants.
The mechanisms oftheantioxidant, anti‑inammatory
andanti‑apoptotic eects ofATX‑PPS‑NP
e underlying mechanisms of CDDP-induced ototox-
icity are complicated, which resulted in the multifacto-
rial protective effects of ATX-PPS-NP. We studied these
effects from three aspects. Firstly, we explored the anti-
oxidant effect. After screening the optimal drug treat-
ment to reduce ROS production, pre 4h + co 24 h was
Fig. 4 Cytoprotective and anti-apoptotic activity of ATX-PPS-NP against CDDP-induced toxicity in vitro. A A parallel comparison of cell viability
in CDDP-treated HEI-OC1 cells with administrations of ATX (1 μg/ml), PPS-NP (drug free) and ATX-PPS-NP (1 μg/ml) in three types. *** p < 0.001 vs
control, ### p < 0.001 vs CDDP, &&& p < 0.001 vs ATX-PPS-NP. B Flow cytometry showing the percentage of apoptotic cells labeled by Annexin V-FITC/PI
double staining in ATX (1 μg/ml), PPS-NP (drug free) and ATX-PPS-NP (1 μg/ml) pretreated HEI-OC1 cells, followed by CDDP (60 μM, 24 h) exposure.
C Quantifications of apoptotic cells. ** p < 0.01, *** p < 0.001 vs control, # p < 0.05, ## p < 0.01, ### p < 0.001 vs CDDP, &&& p < 0.001 vs ATX-PPS-NP. D
Immunostaining of Myosin VIIa labeling hair cells in cochlea explants in control, CDDP (60 μM), ATX (1 μg/ml), PPS-NP (drug free) and ATX-PPS-NP
(1 μg/ml) group. E Quantifications of hair cells in apex, middle and basal turn of cochlea explants. *, #, & p < 0.05 vs control, **, ##, && p < 0.01 vs control,
***, ##, &&& p < 0.001 vs control, $ p < 0.05 vs CDDP, $$ p < 0.01 vs CDDP
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 11 of 17
Guetal. Journal of Nanobiotechnology (2022) 20:268
selected for the following studies (Additional file1: Fig.
S3). In Fig. 5A–B, as demonstrated by a reduction in
MFI of DCFHDA (green fluorescence), the level of ROS
induced by CDDP decreased following treatment with
ATX-PPS-NP (p < 0.05), suggesting the highest antioxi-
dant efficacy of ATX-PPS-NP against CDDP-induced
accumulation of ROS among the three drugs stud-
ied. On the other hand, neither the ATX nor drug-free
PPS-NP groups displayed significant reduction in ROS,
confirming that a combination of ROS-consuming PPS-
NP and the ROS-scavenging compound ATX led to an
enhanced antioxidant effect.
It is acknowledged that the excessive generation of ROS
overwhelms the redox balance [5]. We then detected
the GSH level to investigate the antioxidant efficacy of
ATX-PPS-NP. As shown in Fig.5C, CDDPsignificantly
decreased GSH level, while ATX-PPS-NP restored GSH
level to approximately that of the control group when
Fig. 5 The mechanisms of antioxidant, anti-inflammatory and anti-apoptotic effects of ATX-PPS-NP.A Confocal images of intracellular ROS level
stained by DCFHDA (green fluorescence) in ATX (1 μg/ml), PPS-NP (drug free) and ATX-PPS-NP (1 μg/ml) pretreated HEI-OC1 cells, followed by
CDDP (60 μM, 24 h) administration. B Quantifications of MFI of DCFHDA. C GSH levels. D IL-6 levels. EF The expression levels and qualifications of
apoptosis-associated proteins evaluated by Western blot analyses. * p < 0.05, ** p < 0.01, *** p < 0.001 vs control, # p < 0.05, ## p < 0.01, ### p < 0.001 vs
CDDP, &&& p < 0.001 vs ATX-PPS-NP
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 12 of 17
Guetal. Journal of Nanobiotechnology (2022) 20:268
compared to the ATX or PPS-NP group. is indicated
that ATX-PPS-NP strengthened theantioxidant defense
system by reducing the depletion of GSH contents.
Secondly, the accumulation of ROS has also been iden-
tified as an important mediator of the inflammatory
response. To clarify the anti-inflammatory effect of ATX-
PPS-NP, a cytokine assay (IL-6) was performed. Com-
pared to the control group, a significantly higher level of
IL-6 was detected in the CDDP group. Treatment with
ATX-PPS-NP was effective in decreasing cochlear pro-
inflammatory cytokine levels (Fig.5D). By contrast, free
ATX alone or blank PPS-NP showed no significant sup-
pression of IL-6.
Lastly, in terms of protein expression involved in
the process of apoptosis, we evaluated the protein
level of cleaved-caspase 3, cleaved-caspase 9, P53 and
cytochrome-C (Fig.5E, F). e results demonstrated that
the expression of cytochrome-C and cleaved-caspase 3
was significantly elevated by CDDP treatment and sub-
sequently reduced by ATX-PPS-NP. However, cleaved-
caspase 9 and P53 were unchanged. Cytochrome-C is
an initial factor in the mitochondrial apoptotic pathway
and caspase-3 serves as a converging point in intrinsic
and extrinsic pathways. ese results suggest that exces-
sive ROS induced by CDDP triggered mitochondrial
depolarization and the cytochrome-C release, activating
caspase-3 and ultimately leading to intrinsic apoptosis,
which was suppressed by ATX-PPS-NP.
Overall, ATX-PPS-NP had potent antioxidant, anti-
inflammatory and anti-apoptotic effects on CDDP-
treated cells in vitro, eventually leading to increased
cellular viability and attenuation of cell apoptosis.
Evaluation ofRWM penetration andinvivo release prole
Previous studies on inner ear drug delivery systems
mostly focused on their pharmacokinetics or cochlear
distributions, but seldom on the interaction with the
RWM. Here, we adapted a novel method reported by
Chen etal. [33] to observe the RWM penetration pro-
cess by NP. In detail, fluorescent images were captured
in the three layers of the RWM (OE, outer epithelium
layer; CT, connective tissue; IE, inner epithelium layer) at
Fig. 6 Evaluation of RWM penetration and in vivo release profile. A Fluorescent images of NiRe-PPS-NP penetrating three layers (OE, CT and IE) of
round window membrane in control, 0.5, 2 and 6 h time points. B MFI of Nile Red along the RWM from middle ear side to scala tympani side. C ATX
concentrations in control cochlear perilymph in various time points (0.5, 2, 6 and 12 h) and CDDP-treated ones (2 h). *** p < 0.001
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 13 of 17
Guetal. Journal of Nanobiotechnology (2022) 20:268
various time points after administration of NiRe-PPS-NP
(Fig.6A). As illustrated in Fig.6B, the MFI of NiRe-PPS-
NP increased quickly in the 0.5h group, indicating the
successful and rapid migration of NP from the middle ear
side to the scala tympani side of the RWM. A significant
increase in MFI was observed in the 2h group, suggest-
ing stable transport of NP into the inner ear. MFI in the
6h group was reduced to a level lower than that in the
0.5h group, which indicated that amajority of NP had
penetrated theRWM into the cochlea.
To assess the penetrating capacity of ATX-PPS-NP
through the RWM, we examined the ATX concentration
in perilymph following the administration of a single dose
of ATX-PPS-NP into the RWM. As shown in Fig. 6C,
at 0.5h after administration, the concentration of ATX
which had diffused into the inner ear was 76.23 ± 9.74ng/
ml. is was sustained for almost 12h. In contrast, ATX
concentrations in the perilymph of CDDP-treated ani-
mals were much higher than those without CDDP dam-
age, 272.97 ± 58.18 ng/ml vs. 46.00 ± 24.77 ng/ml (***
p < 0.001), suggesting the ROS-triggered drug release of
ATX-PPS-NP in guinea pigs, consistent with the ROS-
responsive drug release invitro (Fig.1E–G, 3D).
Protective eects onauditory function inguinea pigs
andunderlying mechanisms
Encouraged by the efficient cytoprotection of ATX-
PPS-NP in vitro, the protective effect was then
studiedin vivo.Firstly, H&E stainingverified thatATX-
PPS-NP were well-tolerated,no inflammatory response
was observed in the RWM (Additional file1: Fig. S4). e
surgical procedures used for RWM administration were
further confirmed to be safe, based on no elevation in
ABR thresholds (Fig.7A). We then classified cisplatin-
induced hearing loss into moderate and severe types
according to the duration of CDDP treatment. It can be
seen in Fig. 7A that ATX-PPS-NP administration pro-
tected against CDDP-induced moderate hearing loss by
13.57, 15.00 and12.86dB SPL at 4.0, 5.6 and 8.0 kHz,
respectively, while no significant changes appeared in the
unencapsulated ATX group. Taken together, these results
verified that ATX-PPS-NP showed superior protection
in HEI-OC1 cells, cochlear explants and animal mod-
els compared with equivalent ATX alone, revealing the
enhanced antioxidant effect of ATX-PPS-NP. Addition-
ally, no improvement in hearing was observed in animals
with severe hearing loss, indicating that ATX-PPS-NP
was only effective in protection againstmoderate hearing
loss. We found that pretreatment ofATX-PPS-NP atten-
uated cisplatin-induced moderate hearing loss at low fre-
quencies, with a negligible effectat high frequencies.
e present studyrevealed thatfocal vacuolization and
nuclear degeneration were observed in cisplatin-treated
SGNs, which was alleviated in the ATX-PPS-NP group
(Fig.7D).e SGN density was determined as the num-
ber of SGNs per unit area in the Rosenthal canal. As
shown in Fig. 7B, ATX-PPS-NP preserved SGNs from
destruction by cisplatin in the apical turn of the coch-
lea,consistent with the structural site where the protec-
tion of auditory function occurred. Animals treated with
CDDP (Day 1) is an appropriate model for studying the
mechanisms of ATX-PPS-NP in SGNs alone.
However, the number or morphology of hair cells and
supporting cells were not influenced in CDDP-induced
moderate hearing loss, and the thickness of the stria
vascularis was not changed (Additional file1: Fig. S5).
e mismatching of protection targets between coch-
lear explants and guinea pigs could be the result of the
specific damaging regions by different administration
types. Direct exposure to culture medium containing
cisplatin led to outer hair cell (OHC) loss, while systemi-
cally administrated cisplatin firstly caused SGN loss. In
addition, functional impairments or subcellular changes
which could not be obtained without performing ultra-
structural level morphological analyses in OHCs may
occur, but OHC count was unchanged on day 1 after
CDDP administration.
To evaluate the in vivo antioxidant efficacy of ATX-
PPS-NP, 4-HNE (a cytotoxic end product of lipid per-
oxidation, one of the markers for oxidative stress) was
stained with 3, 3’-diaminobenzidine (DAB) for assess-
ment of ROS generation. Figure 7C, E showed higher
expression of 4-HNE in the cytoplasm of SGNs in the
CDDP (Day 1) group compared to the control. Weaker
expression of 4-HNE was observed in the ATX-PPS-NP
group, with no changes in the ATX group. e quantifi-
cations of IOD/area demonstrated a reduction in 4-HNE
in the apical and middle turns, which verified the strong
ROS-scavenging ability of ATX-PPS-NP.
Discussion
Up to now, cisplatin is an indispensable chemothera-
peutic compound for a broad spectrum of solid tumors.
However, its high therapeutic efficacy is often coupled
with a potential ototoxicity with an average incidence
over 60% [1]. With no pharmaceutical drugs approved by
FDA, astaxanthin is considered as a promising candidate
against cisplatin-induced ototoxicity due to its power-
ful antioxidant property, with ROS-scavenging capacity
6000 times higher than vitamin C, 550 times higher than
vitamin E, 75 times higher than a-lipoic acid [37]. How-
ever, its bioavailability in inner ear is limited by low coch-
lear ATX concentration on account of its hydrophobicity,
short half-life and susceptibility to chemical degrada-
tion under certainconditions (light, temperature, alkali,
oxidation and isomerization) [38]. To overcome these
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 14 of 17
Guetal. Journal of Nanobiotechnology (2022) 20:268
Fig. 7 Protective effects on auditory function in guinea pigs and underlying mechanisms. A ABR thresholds of various groups. *** p < 0.001 control
vs CDDP (Day 1) and CDDP (Day 3), # p < 0.05 ATX-PPS-NP + CDDP (Day 1) vs CDDP (Day 1), ## p < 0.01 ATX-PPS-NP + CDDP (Day 1) vs CDDP (Day 1), &
p < 0.05 ATX-PPS-NP + CDDP (Day 1) vs ATX + CDDP (Day 1) B & D. H&E staining of SGNs and calculations of SGN density in 2–4 turns of guinea pig
cochleae. * p < 0.05 vs control, ** p < 0.01 vs control, *** p < 0.001 vs control, # p < 0.05 vs CDDP. C & E. Immunohistochemistry of 4-HNE in SGNs and
quantifications of integrated optical density (IOD) of 4-HNE staining. *** p < 0.001 vs control, ## p < 0.01 vs CDDP
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 15 of 17
Guetal. Journal of Nanobiotechnology (2022) 20:268
limitations, PEGylation was applied in the fabrication
of nanoparticles due to its effect in prolonging circula-
tion half-life in serum bydecreasing protein absorption
and avoiding the formation of aggregates [39]. Here, we
verified that the encapsulation of PPS-NP preserved the
stability of ATX (Fig.1C, D),in consonance with meth-
ods using microencapsulation with chitosan, polymeric
nanospheres, emulsions and β-cyclodextrin in previous
studies [40]. Previous studiesindicated that size between
150 and 300nm with a positivesurface charge has advan-
tages in permeating the RWM andentering the cochlea
faster [41], which conferred ATX-PPS-NP advantages in
providing sufficient ATX concentration in inner ear.
As previous studies have shown that ROS-responsive
systems have the potential to achieve oxidation-specific
drug release in a series of disease models [2629], here,
ATX-PPS-NP demonstrated ROS-responsive accumula-
tion and enhanced drug release either in artificial peri-
lymph, HEI-OC1 cell line or guinea pigs. Besides, the
process of ROS-triggering disintegration of PPS-NP was
also ROS-consuming, which facilitated an enhanced anti-
oxidant effect when combining with ATX. Moreover,
evidences indicatedoxidative stress initiated the inflam-
matory process, resulting in the synthesis and secretion
of proinflammatory cytokines [42], and activated mito-
chondrial apoptotic machinery. e reduction of IL-6
levels and the protein expressions of cytochrome-C and
cleaved-caspase 3 in ATX-PPS-NP group suggested that
an enhanced antioxidant efficacy further led to enhanced
anti-inflammatory and anti-apoptotic effects on CDDP-
treated cells. Eventually, it was verified that ATX-PPS-NP
showed superior cellular protection in HEI-OC1 cell line,
cochlear explants and guinea pigs compared with equiva-
lent ATX. us, ATX-PPS-NP could reduce ATX doses
andpotentially minimize the side-effects caused by off-
target effects [43], with protective efficacy guaranteed.
Outer hair cells of the organ of Corti, SGNs and stria
vascularis are three major targets of cisplatin-induced
ototoxicity. Time sequence studies [44, 45] explor-
ing impairment processes at these 3 sites have verified
that theyrun in parallel, other than secondary injuries
dependent on another ototoxic process. A recent study
found thata delay in ABR wave 1 latency which probably
implied the damage to SGN mitochondria and myelina-
tion was the earliestfunctional and cellular changes after
cisplatin treatment,indicating SGN as an early and direct
damaging targetin cisplatin-induced ototoxicity [46]. In
the present study, the damaging target was determined
by the analysis of histological and immunohistochemi-
cal staining of the basilar membrane and mid-modiolar
sections. Notably, ATX-PPS-NPs increased the SGNs
survival and preserved the greater morphology of SGNs
in apical turns (Fig.7B, D), with nonoticeable changes of
OHCs and SCs in Organ of Corti (Fig. S5). However, the
lack of SGN cell lines and technological difficulty in the
culture of Organ of Corti containing intact SGNs con-
stricts the evaluation of the protection of ATX-PPS-NP
on SGNs invitro.
4-HNE is a cytotoxic end product of lipid peroxida-
tion and can be eliminated by conjugating to GSH [35].
Here, we found that ATX-PPS-NP facilitated a reduced
4-HNE generation (Fig.7C, E) and elevated level of GSH
(Fig.5C), suggesting the involvement of ATX-PPS-NP in
the mitigation of lipid peroxidation and the further cell
death induced by CDDP. As to the functional analysis,
the pretreatment ofATX-PPS-NPs attenuated cisplatin-
induced moderate hearing loss at low frequencies, with
negligible effectsat high frequencies (Fig.7A). e rea-
son why auditory function protection occurred in low
frequencies is that, on one hand, cisplatin induces more
severe hearing loss at high frequencies. On the other
hand, drug efficacy is likely to depend on the intrinsic
properties of the cochlea. For example,the capacity of
spiral ganglion cells to respond to ROS challenges may
vary along the tonotopic axis, with a higher SOD2immu-
nopositivity in SGNs located at the apex than those at the
base [47].
Conclusion
In the present study, we developed a novel drug delivery
system combining antioxidant ATX and ROS-responsive/
consuming nanoparticles (PPS-NP) to combat cisplatin-
induced ototoxicity. ATX-PPS-NP were proven to be ROS-
responsive in artificial perilymph, the HEI-OC1 cell line and
guinea pigs. In addition, ATX-PPS-NP accumulated in ROS-
specific sites and subsequently disintegrated for smart drug
release. In HEI-OC1 cells, ATX-PPS-NP mitigated ROS
level (DCFHDA) and inflammatory reaction (IL-6). Moreo-
ver,ATX-PPS-NP enhanced cell viability andalleviated cispl-
atin-induced mitochondrial apoptotic pathwayby reducing
the protein expression of cytochrome-C and cleaved cas-
pase-3. In guinea pigs, RWM evaluation and LC–MS analy-
sis verified the feasible penetration of ATX-PPS-NP into the
inner ear. Based on these findings, we administrated ATX-
PPS-NP on the RWM before CDDP injection and observed
partial recovery of auditory function from moderate hearing
loss. ATX-PPS-NPs increased the SGNs survival and miti-
gated the lipid peroxidation induced by CDDP.erefore,
this work provides an enhanced antioxidant therapy by com-
bining ROS-responsive/consuming nanoparticles and ATX
for cisplatin-induced ototoxicity.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 16 of 17
Guetal. Journal of Nanobiotechnology (2022) 20:268
Supplementary Information
The online version contains supplementary material available at https:// doi.
org/ 10. 1186/ s12951- 022- 01485-8.
Additional le1: Figure S1. Nuclear magnetic resonance 1H NMR
spectra of PPS-PEG and FITC-PPS-PEG. Figure S2. Screening the optimal
dose and time for CDDP administration in HEI-OC1. Figure S3. Screening
the optimal application of drugs (ATX, PPS-NP, ATX-PPS-NP) in cell models.
Figure S4. H&E staining of round window membrane (RWM). Figure S5.
Structural changes of cochlea in CDDP (day1)-treated mice.
Author contributions
JG executed experiments and drafted the manuscript. XW designed
experiments and analyzed data. YC and KX collected data. DY conceived the
study. HW revised the manuscript. All authors read and approved the final
manuscript.
Funding
The present study was supported by the National Natural Science Foun-
dation of China (No. 81970874), Natural Science Foundation of Shang-
hai (No. 19ZR1429400), Natural Science Foundation of Shanghai (No.
21ZR1437600), Shanghai Municipal Science and Technology Major Project (No.
2018SHZDZX05), Shanghai Key Laboratory of Translational Medicine on Ear
and Nose Diseases (No. 14DZ2260300), and Innovative research team of high-
level local universities in Shanghai.
Availability of data and materials
Date and material are available for any research.
Declarations
Ethics approval and consent to participate
All animal studies were approved and conducted in accordance with the
guidelines of the Ethics Committee of the Shanghai Jiaotong University,
School of Medicine (Shanghai, China).
Consent for publication
We agree for publication.
Competing interests
The authors declare that they have no known competing financial interests
or personal relationships that could have appeared to influence the work
reported in this paper.
Author details
1 Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth
People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China. 2 Ear Institute, Shanghai Jiao Tong University School of Medicine,
Shanghai, China. 3 Shanghai Key Laboratory of Translational Medicine on Ear
and Nose Diseases (14DZ2260300), Shanghai, China. 4 Materdicine Lab, School
of Life Sciences, Shanghai University, Shanghai, China.
Received: 14 March 2022 Accepted: 31 May 2022
References
1. Karasawa T, Steyger PS. An integrated view of cisplatin-induced nephro-
toxicity and ototoxicity. Toxicol Lett. 2015;237:219–27.
2. Banfi B, Malgrange B, Knisz J, Steger K, Dubois-Dauphin M, Krause KH.
NOX3, a superoxide-generating NADPH oxidase of the inner ear. J Biol
Chem. 2004;279:46065–72.
3. Rybak LP, Mukherjea D, Jajoo S, Kaur T, Ramkumar V. siRNA-mediated
knock-down of NOX3: therapy for hearing loss? Cell Mol Life Sci.
2012;69:2429–34.
4. Rybak LP, Husain K, Morris C, Whitworth C, Somani S. Effect of protective
agents against cisplatin ototoxicity. Am J Otol. 2000;21:513–20.
5. Sheth S, Mukherjea D, Rybak LP, Ramkumar V. Mechanisms of Cisplatin-
Induced Ototoxicity and Otoprotection. Front Cell Neurosci. 2017;11:338.
6. Lautermann J, Crann SA, McLaren J, Schacht J. Glutathione-dependent
antioxidant systems in the mammalian inner ear: effects of aging, oto-
toxic drugs and noise. Hear Res. 1997;114:75–82.
7. Rybak LP. Mechanisms of cisplatin ototoxicity and progress in otoprotec-
tion. Curr Opin Otolaryngol Head Neck Surg. 2007;15:364–9.
8. Lee JE, Nakagawa T, Kim TS, Endo T, Shiga A, Iguchi F, Lee SH, Ito J. Role of
reactive radicals in degeneration of the auditory system of mice follow-
ing cisplatin treatment. Acta Otolaryngol. 2004;124:1131–5.
9. Kopke R, Allen KA, Henderson D, Hoffer M, Frenz D, de Water T. A radical
demise. Toxins and trauma share common pathways in hair cell death.
Ann N Y Acad Sci. 1999;884:171–91.
10. Song MY, Makino A, Yuan JX. Role of reactive oxygen species and redox in
regulating the function of transient receptor potential channels. Antioxid
Redox Signal. 2011;15:1549–65.
11. Pinton P, Giorgi C, Siviero R, Zecchini E, Rizzuto R. Calcium and apoptosis:
ER-mitochondria Ca2+ transfer in the control of apoptosis. Oncogene.
2008;27:6407–18.
12. Dose J, Matsugo S, Yokokawa H, Koshida Y, Okazaki S, Seidel U, Eggers-
dorfer M, Rimbach G, Esatbeyoglu T. Free radical scavenging and cellular
antioxidant properties of astaxanthin. Int J Mol Sci. 2016;17:103.
13. Wolf AM, Asoh S, Hiranuma H, Ohsawa I, Iio K, Satou A, Ishikura M, Ohta S.
Astaxanthin protects mitochondrial redox state and functional integrity
against oxidative stress. J Nutr Biochem. 2010;21:381–9.
14. Zou J, Pyykko I, Hyttinen J. Inner ear barriers to nanomedicine-aug-
mented drug delivery and imaging. J Otol. 2016;11:165–77.
15. Gu J, Chen Y, Tong L, Wang X, Yu D, Wu H. Astaxanthin-loaded polymer-
lipid hybrid nanoparticles (ATX-LPN): assessment of potential otoprotec-
tive effects. J Nanobiotechnology. 2020;18:53.
16. Sun C, Wang X, Zheng Z, Chen D, Wang X, Shi F, Yu D, Wu H. A single
dose of dexamethasone encapsulated in polyethylene glycol-coated
polylactic acid nanoparticles attenuates cisplatin-induced hearing loss
following round window membrane administration. Int J Nanomedi-
cine. 2015;10:3567–79.
17. Chen Y, Gu J, Liu J, Tong L, Shi F, Wang X, Wang X, Yu D, Wu H.
Dexamethasone-loaded injectable silk-polyethylene glycol hydro-
gel alleviates cisplatin-induced ototoxicity. Int J Nanomedicine.
2019;14:4211–27.
18. Alejo T, Uson L, Arruebo M. Reversible stimuli-responsive nanomateri-
als with on-off switching ability for biomedical applications. J Control
Release. 2019;314:162–76.
19. Morachis JM, Mahmoud EA, Almutairi A. Physical and chemical
strategies for therapeutic delivery by using polymeric nanoparticles.
Pharmacol Rev. 2012;64:505–19.
20. Qiao C, Yang J, Shen Q, Liu R, Li Y, Shi Y, Chen J, Shen Y, Xiao Z, Weng
J, Zhang X. traceable nanoparticles with dual targeting and ROS
response for RNAi-based immunochemotherapy of intracranial glio-
blastoma treatment. Adv Mater. 2018;30: e1705054.
21. Liang X, Duan J, Li X, Zhu X, Chen Y, Wang X, Sun H, Kong D, Li C,
Yang J. Improved vaccine-induced immune responses via a ROS-
triggered nanoparticle-based antigen delivery system. Nanoscale.
2018;10:9489–503.
22. Xu X, Saw PE, Tao W, Li Y, Ji X, Bhasin S, Liu Y, Ayyash D, Rasmussen J, Huo
M, et al. ROS-Responsive polyprodrug nanoparticles for triggered drug
delivery and effective cancer therapy. Adv Mater. 2017;29:1700141.
23. Zhang Y, Guo Q, An S, Lu Y, Li J, He X, Liu L, Zhang Y, Sun T, Jiang C. ROS-
switchable polymeric nanoplatform with stimuli-responsive release for
active targeted drug delivery to breast cancer. ACS Appl Mater Interfaces.
2017;9:12227–40.
24. El-Mohtadi F, d’Arcy R, Tirelli N. Oxidation-responsive materials: biological
rationale, state of the art, multiple responsiveness, and open issues.
Macromol Rapid Commun. 2019;40: e1800699.
25. Jeanmaire D, Laliturai J, Almalik A, Carampin P, d’Arcy R, Lallana E, Evans R,
Winpenny REP, Tirelli N. Chemical specificity in REDOX-responsive materi-
als: the diverse effects of different Reactive Oxygen Species (ROS) on
polysulfide nanoparticles. Polym Chem. 2014;5:1393–404.
26. Kim K, Lee CS, Na K. Light-controlled reactive oxygen species (ROS)-
producible polymeric micelles with simultaneous drug-release triggering
and endo/lysosomal escape. Chem Commun (Camb). 2016;52:2839–42.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 17 of 17
Guetal. Journal of Nanobiotechnology (2022) 20:268
fast, convenient online submission
thorough peer review by experienced researchers in your field
rapid publication on acceptance
support for research data, including large and complex data types
gold Open Access which fosters wider collaboration and increased citations
maximum visibility for your research: over 100M website views per year
At BMC, research is always in progress.
Learn more biomedcentral.com/submissions
Ready to submit your research
Ready to submit your research
? Choose BMC and benefit from:
? Choose BMC and benefit from:
27. Poole KM, Nelson CE, Joshi RV, Martin JR, Gupta MK, Haws SC, Kavanaugh
TE, Skala MC, Duvall CL. ROS-responsive microspheres for on demand
antioxidant therapy in a model of diabetic peripheral arterial disease.
Biomaterials. 2015;41:166–75.
28. Tang M, Hu P, Zheng Q, Tirelli N, Yang X, Wang Z, Wang Y, Tang Q, He Y.
Polymeric micelles with dual thermal and reactive oxygen species (ROS)-
responsiveness for inflammatory cancer cell delivery. J Nanobiotechnol-
ogy. 2017;15:39.
29. Zhao Z, Han Z, Naveena K, Lei G, Qiu S, Li X, Li T, Shi X, Zhuang W, Li Y,
et al. ROS-Responsive nanoparticle as a berberine carrier for OHC-tar-
geted therapy of noise-induced hearing loss. ACS Appl Mater Interfaces.
2021;13:7102–14.
30. Kalinec G, Thein P, Park C, Kalinec F. HEI-OC1 cells as a model for investi-
gating drug cytotoxicity. Hear Res. 2016;335:105–17.
31. Rodriguez-Ruiz V, Salatti-Dorado JA, Barzegari A, Nicolas-Boluda A,
Houaoui A, Caballo C, Caballero-Casero N, Sicilia D, Bastias Venegas
J, Pauthe E, et al. Astaxanthin-loaded nanostructured lipid carriers for
preservation of antioxidant activity. Molecules. 2018;23:2601.
32. Chai GH, Hu FQ, Sun J, Du YZ, You J, Yuan H. Transport pathways of solid
lipid nanoparticles across Madin-Darby canine kidney epithelial cell
monolayer. Mol Pharm. 2014;11:3716–26.
33. Zhang L, Xu Y, Cao W, Xie S, Wen L, Chen G. Understanding the transloca-
tion mechanism of PLGA nanoparticles across round window membrane
into the inner ear: a guideline for inner ear drug delivery based on
nanomedicine. Int J Nanomedicine. 2018;13:479–92.
34. Whitlon DS, Szakaly R, Greiner MA. Cryoembedding and sectioning of
cochleas for immunocytochemistry and in situ hybridization. Brain Res
Brain Res Protoc. 2001;6:159–66.
35. Park HJ, Kim MJ, Rothenberger C, Kumar A, Sampson EM, Ding D, Han C,
White K, Boyd K, Manohar S, et al. GSTA4 mediates reduction of cisplatin
ototoxicity in female mice. Nat Commun. 2019;10:4150.
36. Kinnear C, Moore TL, Rodriguez-Lorenzo L, Rothen-Rutishauser B, Petri-
Fink A. Form follows function: nanoparticle shape and its implications for
nanomedicine. Chem Rev. 2017;117:11476–521.
37. Zhang ZW, Xu XC, Liu T, Yuan S. Mitochondrion-permeable antioxidants
to treat ROS-burst-mediated acute diseases. Oxid Med Cell Longev.
2016;2016:6859523.
38. Martínez-Delgado AA, Khandual S, Villanueva-Rodríguez SJ. Chemical
stability of astaxanthin integrated into a food matrix: effects of food
processing and methods for preservation. Food Chem. 2017;225:23–30.
39. Moore TL, Rodriguez-Lorenzo L, Hirsch V, Balog S, Urban D, Jud C, Rothen-
Rutishauser B, Lattuada M, Petri-Fink A. Nanoparticle colloidal stability
in cell culture media and impact on cellular interactions. Chem Soc Rev.
2015;44:6287–305.
40. Ambati RR, Phang SM, Ravi S, Aswathanarayana RG. Astaxanthin: sources,
extraction, stability, biological activities and its commercial applications–
a review. Mar Drugs. 2014;12:128–52.
41. Mittal R, Pena SA, Zhu A, Eshraghi N, Fesharaki A, Horesh EJ, Mittal J,
Eshraghi AA. Nanoparticle-based drug delivery in the inner ear: current
challenges, limitations and opportunities. Artif Cells Nanomed Biotech-
nol. 2019;47:1312–20.
42. Hussain T, Tan B, Yin Y, Blachier F, Tossou MC, Rahu N. Oxidative stress and
inflammation: what polyphenols can do for us? Oxid Med Cell Longev.
2016;2016:7432797.
43. Ballance WC, Qin EC, Chung HJ, Gillette MU, Kong H. Reactive oxygen
species-responsive drug delivery systems for the treatment of neurode-
generative diseases. Biomaterials. 2019;217: 119292.
44. van Ruijven MW, de Groot JC, Smoorenburg GF. Time sequence of degen-
eration pattern in the guinea pig cochlea during cisplatin administration.
A quantitative histological study. Hear Res. 2004;197:44–54.
45. van Ruijven MW, de Groot JC, Klis SF, Smoorenburg GF. The cochlear
targets of cisplatin: an electrophysiological and morphological time-
sequence study. Hear Res. 2005;205:241–8.
46. Chen Y, Bielefeld EC, Mellott JG, Wang W, Mafi AM, Yamoah EN, Bao J. Early
physiological and cellular indicators of cisplatin-induced ototoxicity. J
Assoc Res Otolaryngol. 2021;22:107–26.
47. Ying YL, Balaban CD. Regional distribution of manganese superoxide
dismutase 2 (Mn SOD2) expression in rodent and primate spiral ganglion
cells. Hear Res. 2009;253:116–24.
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in pub-
lished maps and institutional affiliations.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Potent loop diuretics can lead to temporary hearing loss, which usually recovers over time without causing permanent hearing damage. Loop diuretics (e.g., furosemide) [80][81][82] Block Na + -K + -2Cl − cotransporter in marginal cells [80] Stria vascularis edema; Disrupted K + recycling; temporary loss of EP [80] Reversible hearing loss hypoxia-induced EC swelling [80]; transient EP disappearance [80] Cisplatin [34,64,[83][84][85][86][87] ROS generation [88]; ZO-1/cx26/cx43 downregulation [34]; Increased paracellular permeability Permanent SNHL apoptosis [89] TNF-α upregulation in SV [84]; mitochondrial dysfunction [88] EP reduction (≥20 mV) ...
... Potent loop diuretics can lead to temporary hearing loss, which usually recovers over time without causing permanent hearing damage. Loop diuretics (e.g., furosemide) [80][81][82] Block Na + -K + -2Cl − cotransporter in marginal cells [80] Stria vascularis edema; Disrupted K + recycling; temporary loss of EP [80] Reversible hearing loss hypoxia-induced EC swelling [80]; transient EP disappearance [80] Cisplatin [34,64,[83][84][85][86][87] ROS generation [88]; ZO-1/cx26/cx43 downregulation [34]; Increased paracellular permeability Permanent SNHL apoptosis [89] TNF-α upregulation in SV [84]; mitochondrial dysfunction [88] EP reduction (≥20 mV) ...
Article
Full-text available
The inner ear is a relatively isolated organ, protected by the blood-labyrinth barrier (BLB). This barrier creates a unique lymphatic fluid environment within the inner ear, maintaining a stable physiological state essential for the mechano-electrical transduction process in the inner ear hair cells while simultaneously restricting most drugs from entering the lymphatic fluid. Under pathological conditions, dysfunction of the stria vascularis and disruption in barrier structure can lead to temporary or permanent hearing impairment. This review describes the structure and function of the BLB, along with recent advancements in modeling and protective studies related to the BLB. The review emphasizes some newly developed non-invasive inner ear drug delivery strategies, including ultrasound therapy assisted by microbubbles, inner ear-targeting peptides, sound therapy, and the route of administration of the cerebrospinal fluid conduit. We argue that some intrinsic properties of the BLB can be strategically utilized for effective inner ear drug delivery.
... 4) The presence of the blood-labyrinth barrier (BLB) in the inner ear, which regulates the ionic composition of the inner and outer lymph, protects the inner ear from blood-borne toxins and selectively allows ions, fluids, and nutrients to enter the cochlea. This may result in the inability of drugs to reach an effective concentration in the inner ear, thus reducing their protective effects after systemic administration Gu et al., 2022;Barbara et al., 2022). Identifying an effective mode of drug administration and drug delivery medium is an area that needs to be explored in depth. ...
Article
Full-text available
Cisplatin is widely used to treat various solid tumors. However, its toxicity to normal tissues limits its clinical application, particularly due to its ototoxic effects, which can result in hearing loss in patients undergoing chemotherapy. While significant progress has been made in preclinical studies to elucidate the cellular and molecular mechanisms underlying cisplatin-induced ototoxicity (CIO), the precise mechanisms remain unclear. Moreover, the optimal protective agent for preventing or mitigating cisplatin-induced ototoxicity has yet to be identified. This review summarizes the current understanding of the roles of apoptosis, autophagy, ferroptosis, pyroptosis, and protective agents in cisplatin-induced ototoxicity. A deeper understanding of these cell death mechanisms in the inner ear, along with the protective agents, could facilitate the translation of these agents into clinical therapeutics, help identify new therapeutic targets, and provide novel strategies for cisplatin-based cancer treatment.
... In this study, we developed a PEGDA-GDY/G@TGF-β hydrogel as a wound dressing and investigated its use in diabetic wound healing. The core-shell type is commonly included in smart materials to achieve the "ON/OFF" function of the stimulus-response [57]. In our study, the smart response was dependent on gelatin that turned from a gel state to a fluid state at the temperature above 30 °C. ...
Article
Full-text available
The number of people with hearing loss disorders is enormous, causing great physical and mental stress to patients, as well as a huge social burden. Among these patients, hearing loss caused by inner ear lesions accounts for a large proportion. Therefore, treatment of the inner ear is important. Inner ear drug delivery systems, which can reduce the side effects of systemic drug administration by delivering drugs directly to the inner ear, are important in sensorineural hearing loss. Here, the development of inner ear drug delivery systems is focused, including the complex physiological structure that they face, types of drugs delivered, routes of administration, and forms of drug delivery carrier platforms. Recent studies in this process are presented and it is concluded with a summary and outlook on the problems faced and possible solutions.
Article
Sensorineural hearing loss (SNHL) is a prevalent multifactorial condition affecting over 1.5 billion people worldwide. Its etiology includes genetic mutations, environmental factors, and age‐related degeneration, leading to irreversible damage to the cochlear hair cells and auditory neurons. Although conventional interventions, such as hearing aids and cochlear implants, provide partial relief, they do not restore the natural hearing function. Recent advances in the understanding of the molecular mechanisms underlying SNHL, including oxidative stress, autophagy, apoptosis, ferroptosis, and pyroptosis, have provided novel insights into potential therapeutic targets. Emerging treatment strategies include gene therapy, which aims to correct genetic defects through gene replacement and editing; exosome‐based therapy, which facilitates cellular repair and protection; stem cell therapy, which promotes the hair cells and auditory neurons regeneration; and nanomedicine, which enables targeted drug delivery and cochlear protection. Despite these promising developments, challenges remain in translating these therapies from preclinical research to clinical applications, owing to safety, delivery efficiency, and long‐term efficacy concerns. This review summarizes the latest progress in SNHL research, highlights the key pathophysiological mechanisms, and explores treatment strategies. Understanding these advances is essential to develop effective interventions to prevent, and potentially reverse SNHL, ultimately improving the quality of life of affected individuals.
Article
Full-text available
Cisplatin is an established component of treatment protocols for various solid malignancies but carries a significant potential for serious adverse effects. Ototoxicity from cisplatin treatment is an important dose-limiting toxicity that manifests as bilateral, progressive, irreversible, dose-dependent sensorineural hearing loss, ear pain, tinnitus, and vestibular dysfunction. Despite the recent approval of sodium thiosulphate for the prevention of cisplatin-induced hearing loss (CIHL) in pediatric patients, structured prevention programs are not routinely implemented in most hospitals, and reducing platinum-induced ototoxicity in adults remains an important clinical problem without established treatment options. Cochlear oxidative stress plays a fundamental role in CIHL. Here, we review the molecular mechanisms leading to oxidative stress in CIHL and the clinical and preclinical studies testing antioxidants in CIHL to guide future clinical trials in assessing the efficacy and safety of candidate antioxidant compounds in this clinical setting.
Article
Full-text available
Local intratympanic drug delivery to the inner ear possesses significant otological clinical promise as cisplatin‐induced hearing loss (CIHL) therapy, inducing significantly less side effects than systemic drug delivery. However, the multiple detoured barriers, round window membrane (RWM) and poorly controlled drug release hinder successful non‐invasive drug delivery through intratympanic administration (IT). Here, a novel near‐infrared (NIR) responsive nanocomposite functionalized with saponin, denoted gold nanorod@dexamethasone‐mesoporous silica‐saponin (AuNR@DEX‐MS‐saponin, NPs/DEX), is developed to enhance RWM permeation and to control the drug release spatiotemporally. First, the physiochemical properties and release profile of the synthesized nanocomposites are assessed, after which the biocompatibility of the nanocomposites and oto‐protection against CIHL are shown in vitro and in vivo. The findings demonstrated that DEX is delivered to the inner ear with high efficiency through IT, due to the permeation enhancement effect of the nanocomposite. Moreover, the nanocomposite with low dose of DEX is highly effective in recovering CIHL, attenuating hair cell loss, and alleviating synaptic ribbon damage. These findings provide insight into NIR‐responsive local delivery for inner ear illnesses.
Article
Full-text available
Multidrug combination therapy in the inner ear faces diverse challenges due to the distinct physicochemical properties of drugs and the difficulties of overcoming the oto‐biologic barrier. Although nanomedicine platforms offer potential solutions to multidrug delivery, the access of drugs to the inner ear remains limited. Micro/nanomachines, capable of delivering cargo actively, are promising tools for overcoming bio‐barriers. Herein, a novel microrobot‐based strategy to penetrate the round window membrane (RWM) is presented and multidrug in on‐demand manner is delivered. The tube‐type microrobot (TTMR) is constructed using the template‐assisted layer‐by‐layer (LbL) assembly of chitosan/ferroferric oxide/silicon dioxide (CS/Fe3O4/SiO2) and loaded with anti‐ototoxic drugs (curcumin, CUR and tanshinone IIA, TSA) and perfluorohexane (PFH). Fe3O4 provides magnetic actuation, while PFH ensures acoustic propulsion. Upon ultrasound stimulation, the vaporization of PFH enables a microshotgun‐like behavior, propelling the drugs through barriers and driving them into the inner ear. Notably, the proportion of drugs entering the inner ear can be precisely controlled by varying the feeding ratios. Furthermore, in vivo studies demonstrate that the drug‐loaded microrobot exhibits superior protective effects and excellent biosafety toward cisplatin (CDDP)‐induced hearing loss. Overall, the microrobot‐based strategy provides a promising direction for on‐demand multidrug delivery for ear diseases.
Article
Full-text available
Cisplatin chemotherapy often causes permanent hearing loss, which leads to a multifaceted decrease in quality of life. Identification of early cisplatin-induced cochlear damage would greatly improve clinical diagnosis and provide potential drug targets to prevent cisplatin’s ototoxicity. With improved functional and immunocytochemical assays, a recent seminal discovery revealed that synaptic loss between inner hair cells and spiral ganglion neurons is a major form of early cochlear damage induced by noise exposure or aging. This breakthrough discovery prompted the current study to determine early functional, cellular, and molecular changes for cisplatin-induced hearing loss, in part to determine if synapse injury is caused by cisplatin exposure. Cisplatin was delivered in one to three treatment cycles to both male and female mice. After the cisplatin treatment of three cycles, threshold shift was observed across frequencies tested like previous studies. After the treatment of two cycles, beside loss of outer hair cells and an increase in high-frequency hearing thresholds, a significant latency delay of auditory brainstem response wave 1 was observed, including at a frequency region where there were no changes in hearing thresholds. The wave 1 latency delay was detected as early cisplatin-induced ototoxicity after only one cycle of treatment, in which no significant threshold shift was found. In the same mice, mitochondrial loss in the base of the cochlea and declining mitochondrial morphometric health were observed. Thus, we have identified early spiral ganglion-associated functional and cellular changes after cisplatin treatment that precede significant threshold shift.
Article
Full-text available
Background: Ototoxicity is one of the major side effects of platinum-based chemotherapy, especially cisplatin therapy. To date, no FDA approved agents to alleviate or prevent this ototoxicity are available. However, ototoxicity is generally believed to be produced by excessive generation of reactive oxygen species (ROS) in the inner ear, thus leading to the development of various antioxidants, which act as otoprotective agents. Astaxanthin (ATX) is an interesting candidate in the development of new therapies for preventing and treating oxidative stress-related pathologies, owing to its unique antioxidant capacity. Methods and results: In this study, we aimed to evaluate the potential antioxidant properties of ATX in the inner ear by using the HEI-OC1 cell line, zebrafish, and guinea pigs. Because ATX has poor solubility and cannot pass through round window membranes (RWM), we established lipid-polymer hybrid nanoparticles (LPN) for loading ATX. The LPN enabled ATX to penetrate RWM and maintain concentrations in the perilymph in the inner ear for 24 h after a single injection. ATX-LPN were found to have favorable biocompatibility and to strongly affect cisplatin-induced generation of ROS, on the basis of DCFHDA staining in HEI-OC1 cells. JC-1 and MitoTracker Green staining suggested that ATX-LPN successfully reversed the decrease in mitochondrial membrane potential induced by cisplatin in vitro and rescued cells from early stages of apoptosis, as demonstrated by FACS stained with Annexin V-FITC/PI. Moreover, ATX-LPN successfully attenuated OHC losses in cultured organ of Corti and animal models (zebrafish and guinea pigs) in vivo. In investigating the protective mechanism of ATX-LPN, we found that ATX-LPN decreased the expression of pro-apoptotic proteins (caspase 3/9 and cytochrome-c) and increased expression of the anti-apoptotic protein Bcl-2. In addition, the activation of JNK induced by CDDP was up-regulated and then decreased after the administration of ATX-LPN, while P38 stayed unchanged. Conclusions: To best of our knowledge, this is first study concluded that ATX-LPN as a new therapeutic agent for the prevention of cisplatin-induced ototoxicity.
Article
Full-text available
Cisplatin is one of the most widely used chemotherapeutic drugs for the treatment of cancer. Unfortunately, one of its major side effects is permanent hearing loss. Here, we show that glutathione transferase α4 (GSTA4), a member of the Phase II detoxifying enzyme superfamily, mediates reduction of cisplatin ototoxicity by removing 4-hydroxynonenal (4-HNE) in the inner ears of female mice. Under cisplatin treatment, loss of Gsta4 results in more profound hearing loss in female mice compared to male mice. Cisplatin stimulates GSTA4 activity in the inner ear of female wild-type, but not male wild-type mice. In female Gsta4-/- mice, cisplatin treatment results in increased levels of 4-HNE in cochlear neurons compared to male Gsta4-/- mice. In CBA/CaJ mice, ovariectomy decreases mRNA expression of Gsta4, and the levels of GSTA4 protein in the inner ears. Thus, our findings suggest that GSTA4-dependent detoxification may play a role in estrogen-mediated neuroprotection.
Article
Full-text available
Background Cisplatin is an extensively used anti-neoplastic agent for the treatment of various solid tumors. However, a high incidence of severe ototoxicity is accompanied by its use in the clinic. Currently, no drugs or therapeutic strategies have been approved for the treatment of cisplatin-induced ototoxicity by the FDA. Purpose The purpose of this study was to investigate the otoprotective effects of dexamethasone (DEX)-loaded silk-polyethylene hydrogel (DEX-SILK) following round window membrane administration in the cisplatin-induced ototoxicity mouse model. Methods The morphology, gelation kinetics, viscosity and secondary structure of the DEX-SILK hydrogel were analyzed. DEX concentration in the perilymph was tested at different time points following hydrogel injection on the RWM niche. Cultured cells (HEI-OC1), organ of Corti explants (C57/BL6, P0-2), and cisplatin-induced hearing loss mice model (C57/BL6) were used as in vitro and in vivo models for investigating the otoprotective effects of DEX-SILK hydrogel against cisplatin. Results Encapsulation of DEX with a loading of 8% (w/v) did not significantly change the silk gelation time, and DEX was evenly distributed in the Silk-PEG hydrogel as visualized by scanning electron microscopy (SEM). The concentration of Silk majorly influenced DEX distribution, morphological characteristics, viscosity, and gelation time. The optimized DEX-SILK hydrogel (8% w/v loading, 15% silk concentration, 10 μl) was administered directly onto the RWM of the guinea pigs. The DEX concentration in the perilymph was maintained above 1 μg/ml for at least 21 days for the DEX-SILK, while it was maintained for less than 6 h in the control sample of free DEX. DEX-SILK (5-60 ng/ml) exhibited significant protective effects against cisplatin-induced cellular ototoxicity and notably reduced the production of reactive oxygen species (ROS). Eventually, pretreatment with DEX-SILK effectively preserved outer hair cells in the cultured organ of Corti explants and demonstrated significant hearing protection at 4, 8, and 16 kHz in the cisplatin-induced hearing loss mice as compared to the effects noted following pretreatment with DEX. Conclusion These results demonstrated the clinical value of DEX-SILK for the therapy of cisplatin-induced ototoxicity.
Article
Full-text available
Hearing loss is the most common neurosensory impairment worldwide. While conductive hearing loss can be managed by surgery, the management of sensorineural hearing loss (SNHL), related to the damage of sensory cells of the inner ear is more challenging to manage medically. Many causes of SNHL such as sudden idiopathic SNHL, Meniere’s disease, noise-induced hearing loss, autoimmune hearing loss or hearing loss from exposure to ototoxic substances can benefit from delivery of otoprotective drugs to the inner ear. However, systemic drug delivery through oral, intravenous and intramuscular methods leads to undesirable side effects due to the inner ear’s limited blood supply and the relatively poor penetration of the blood–inner ear barrier (BLB). Therefore, there has been an increased interest for the targeted drug delivery to the inner ear using nanoparticles. Drug delivery through nanoparticles offers several advantages including drug stabilization for controlled release and surface modification for specific targeting. Understanding the biocompatibility of nanoparticles with cochlea and developing novel non-invasive delivery methods will promote the translation of nanoparticle-mediated drug delivery for auditory disorders from bench to bedside.
Article
Full-text available
Astaxanthin is a xanthophyll carotenoid showing efficient scavenging ability and represents an interesting candidate in the development of new therapies for preventing and treating oxidative stress-related pathologies. However, its high lipophilicity and thermolability often limits its antioxidant efficacy in human applications. Here, we developed a formulation of lipid carriers to protect astaxanthin’s antioxidant activity. The synthesis of natural astaxanthin-loaded nanostructured lipid carriers using a green process with sunflower oil as liquid lipid is presented. Their antioxidant activity was measured by α-Tocopherol Equivalent Antioxidant Capacity assay and was compared to those of both natural astaxanthin and α-tocopherol. Characterizations by dynamic light scattering, atomic force microscopy, and scattering electron microscopy techniques were carried out and showed spherical and surface negative charged particles with z-average and polydispersity values of ~60 nm and ~0.3, respectively. Astaxanthin loading was also investigated showing an astaxanthin recovery of more than 90% after synthesis of nanostructured lipid carriers. These results demonstrate the capability of the formulation to stabilize astaxanthin molecule and preserve and enhance the antioxidant activity.
Article
Overproduction of reactive oxygen species (ROS) and inflammation are two key pathogeneses of noise-induced hearing loss (NIHL), which leads to outer hair cell (OHC) damage and hearing loss. In this work, we successfully developed ROS-responsive nanoparticles as berberine (BBR) carriers (PL-PPS/BBR) for OHC-targeted therapy of NIHL: Prestin-targeting peptide 2 (PrTP2)-modified nanoparticles (PL-PPS/BBR), which effectively accumulated in OHC areas, and poly(propylene sulfide)120 (PPS120), which scavenged ROS and converted to poly(propylene sulfoxide)120 in a ROS environment to disintegrate and provoke the rapid release of BBR with anti-inflammatory and antioxidant effects. In this study, satisfactory anti-inflammatory and antioxidant effects of PL-PPS/BBR were confirmed. Immunofluorescence and scanning electron microscopy (SEM) images showed that PL-PPS/BBR effectively accumulated in OHCs and protected the morphological integrity of OHCs. The auditory brainstem response (ABR) results demonstrated that PL-PPS/BBR significantly improved hearing in NIHL guinea pigs after noise exposure. This work suggested that PL-PPS/BBR may be a new potential treatment for noise-associated injury with clinical application.
Article
Many long-acting extended drug release systems can provide controlled or sustained release of therapeutic payloads. In the majority of those systems drug release cannot be stopped once it has started because they operate autonomously regardless of the evolution of the treatment and/or the patient´s needs. However, in several pathologies such as diabetes, hormonal disorders, pain management, etc. a pulsatile drug release is required to adjust the dose of drug release to the specific needs in a spatio-temporal manner. Additionally, in other pathologies such as cancer or antimicrobial therapy the release of the drug with spatio-temporal control to prevent unwanted side effects represents an unmet need. With this aim reversible stimuli-responsive nanomaterials with an on-off switching ability have been developed in order to provide a spatio-temporal control of the drug released. Those systems can be activated in response to exogenous (light, magnetic field, electrical fields, etc.) or endogenous triggers (pH, enzyme-substrate complex formation, protein-cell binding, etc.) thanks to the use of reversible phase-transition materials. In this review we compile in vitro and preclinical results in which those materials have been successfully used. The types of stimuli used to trigger drug release as well as the different nanomaterials used are reviewed in order to provide a general overview of the field. We anticipate that further studies in this field will be expanded towards the development of multimodal hybrid systems which combine therapy and imaging while reporting the evolution of the treatment in real time.
Article
Neurodegenerative diseases and disorders seriously impact memory and cognition and can become life-threatening. Current medical techniques attempt to combat these detrimental effects mainly through the administration of neuromedicine. However, drug efficacy is limited by rapid dispersal of the drugs to off-target sites while the site of administration is prone to overdose. Many neuropathological conditions are accompanied by excessive reactive oxygen species (ROS) due to the inflammatory response. Accordingly, ROS-responsive drug delivery systems have emerged as a promising solution. To guide intelligent and comprehensive design of ROS-responsive drug delivery systems, this review article discusses the two following topics: (1) the biology of ROS in both healthy and diseased nervous systems and (2) recent developments in ROS-responsive, drug delivery system design. Overall, this review article would assist efforts to make better decisions about designing ROS-responsive, neural drug delivery systems, including the selection of ROS-responsive functional groups.
Article
In this review, a general introduction to biological oxidants (focusing on reactive oxygen species, ROS) and the biomedical rationale behind the development of materials capable of responding to ROS is provided. The state of the art for preparative aspects and mechanistic responses of the most commonly used macromolecular ROS‐responsive systems, including polysulfides, polyselenides, polythioketals, polyoxalates, and also oligoproline‐ and catechol‐based materials, is subsequently given. The endowment of multiple responsiveness, with specific emphasis on the cases where a molecular logic gate behavior can be obtained, is focused on. Finally, fundamental open issues, which include implications of the “drug”‐like character of ROS‐responsive materials (inherent anti‐inflammatory behavior) and the poor quantitative understanding of ROS roles in biology, are discussed.