Skip to main content

Sox2 overexpression alleviates noise-induced hearing loss by inhibiting inflammation-related hair cell apoptosis

Abstract

Background

The transcription factor Sox2 plays important roles in the developmental processes of multiple organs and tissues. However, whether Sox2 can protect mature or terminally differentiated cells against injury is still unknown.

Methods

We investigated the roles of Sox2 in cochlear hair cells, which are terminally differentiated cells, using conditional transgenic mice and several hearing loss models.

Results

Sox2 overexpression dramatically mitigated the degree of cochlear hair cell loss when exposed to ototoxic drugs. Noise-induced apoptosis of cochlear hair cells and hearing loss were also significantly alleviated by Sox2 overexpression. Notably, noise-induced upregulation of pro-inflammatory factors such as TNF-α and IL6 was inhibited by Sox2 overexpression. Then we used lipopolysaccharide to clarify the effect of Sox2 on cochlear inflammation, and Sox2 overexpression significantly inhibited lipopolysaccharide-induced upregulation of pro-inflammatory factors and alleviated inflammation-related cochlear hair cell death.

Conclusions

These results demonstrate a novel protective role of Sox2 in mature and terminally differentiated cochlear hair cells by inhibiting inflammation.

Background

In mammals, cochlear mechanosensory hair cells (HCs) convert sound waves into electrical signals that are transmitted to the brain’s auditory centers to produce hearing. Aging, noise trauma, ototoxic drugs, and infections can lead to HC loss or degeneration and thus result in the onset of sensorineural hearing loss [1,2,3,4,5,6,7]. Because of the inability of HCs to regenerate after injury in the mature mammalian cochlea, protecting HCs against injury is extremely important for hearing preservation [8,9,10].

Sox2 (HMG-box transcription factor sex-determining region Y-box 2), a well-studied pluripotent transcription factor, is critical for the embryonic development of many organs [11,12,13]. For example, Sox2 serves as a regulator in the process of taste bud sensory cell differentiation from endodermal progenitor cells [12], and the expression of Sox2 has a strong influence on the branching process in the respiratory system [13]. Deficiency of Sox2 leads to abnormal cartilage development in the tracheal mesenchyme and to abnormal cellular composition [14], and Sox2 has been shown to play a significant role in the development of the inner ear [15]. In addition to its essential role in stem cell self-renewal and reprogramming, Sox2 also plays a role in the survival of stem cells and cancer cells [16]. Sox2 directly up-regulates the expression of Survivin, which inhibits the mitochondria-dependent apoptotic pathway in neural stem cells [17]. Although the roles of Sox2 in stem cells and cancer cells have been clarified in many studies, so far no studies about the possible role of Sox2 in mature or terminally differentiated cells have been reported.

In the development process of the inner ear, Sox2 is expressed in pro-sensory cells in the embryonic pro-sensory domain. When the terminal differentiation of cochlear progenitors is initiated, Sox2 expression is maintained in the supporting cells but gradually decreases in auditory HCs. Once the cochlea becomes fully mature and hearing function has been developed, the expression of Sox2 in HCs becomes undetectable [18, 19]. During the maturation of the vestibular sensory epithelium, Sox2 expression is maintained in supporting cells and type 2 vestibular HCs, but is gradually diminished in type 1 vestibular HCs. Sox2 is required for the maintenance of HCs during embryonic development of the zebrafish inner ear and for HC regeneration in zebrafish embryos after injury [20]. Our recent study reported that Sox2-negative type 1 vestibular HCs are more vulnerable than Sox2-positive type 2 vestibular HCs to 3,3'-iminodipropionitrile toxicity in adult mice [21], suggesting that Sox2 might exert a positive effect on the survival of sensory HCs. To explore the possibility that Sox2 might exert a positive effect on the survival of auditory HCs, we used conditional Sox2-upregulated transgenic mice and several cochlear HC injury models to investigate the possible roles of Sox2 in HC damage and survival.

Materials and methods

Animals and genotyping

The Rosa26-CAG-LSL (loxP-stop-loxP)-Sox2 transgenic mice, referred to as Sox2OE mice, were designed by our group and Beijing Biocytogen Co., Ltd. The Atoh1CreER and PrestinCreER transgenic mice, which have been described in detail in previous studies [22, 23], were the kind gifts of Prof. Zuo Jian. For the Atoh1CreER line, tamoxifen was injected subcutaneously once daily at 75 mg/kg body weight at postnatal day (P) 0–1. For the PrestinCreER line, tamoxifen was injected intraperitoneally once daily at 225 mg/kg body weight at P21–22. Mice of either sex were used for all experiments. Animals were bred under specific and opportunistic pathogen-free conditions, and tail DNA genotyping was performed for genetic identification. The primers used in the genotyping protocols are listed in Additional file 1: Table S2.

Auditory brainstem response (ABR) measurement

Mice were injected with a solution of Zoletil (70 g/kg) and dexmedetomidine hydrochloride (1000 mg/kg) intraperitoneally to induce anesthesia. The efficacy of anesthesia was periodically ascertained through foot pinch response. The body temperature was maintained at 37 °C to avoid any influence on the auditory physiology test. ABR measurements were used to assess the mouse auditory function with a System 3 ABR workstation (Tucker Davis Technology, Alachua, FL, USA). Hearing function at 8, 16, 24, and 32 kHz was evaluated through an open field speaker, and acoustic thresholds of sound pressure level were determined using the BioSigRP software (Tucker Davis Technology, Alachua, FL, USA).

Cochlear HC injury models

To construct the ototoxic drug-induced HC injury model in the neonatal stage, cochlear epithelia samples were obtained for explant culture from P2 transgenic mice that had received tamoxifen (Sigma, T5648-5G) administration at P 0–1. The tissues were treated with 1 mM neomycin (Sigma) for 6 h and then harvested after an additional 24 h in the absence of neomycin.

To construct the noise-induced HC injury model, the hearing function of P30 mice that had received tamoxifen administration at P21–P22 were examined by ABR test 24 h before noise exposure. The mice were then exposed to broadband noise at 116 dB for 2 h. Hearing function was evaluated again by ABR test 7 days after noise exposure, and the mice were killed and cochlear samples were harvested for morphological analysis.

To construct the lipopolysaccharide (LPS)-induced HC injury model, mice were given 10 μl LPS (Sigma-Aldrich, L2880) at 1, 2, or 4 mg/kg by tympanum injection and killed 3 days later.

Immunofluorescence and morphological analysis

Immunofluorescence staining was performed as previously reported [32]. Rabbit polyclonal anti-Myosin7A (Proteus Biosciences, 1: 1000 dilution, 25-6790), goat polyclonal anti-SRY (sex-determining region Y)-box 2 (Sox2) (Santa Cruz Biotechnology, 1:1000 dilution, sc-17320), mouse monoclonal anti- Parvalbumin (Sigma, 1:1000 dilution, P3088), mouse monoclonal anti-Myosin7a (Santa Cruz Biotechnology, 1:500 dilution, sc-74516), rabbit mAb anti-TNF-α (Cell Signaling Technology, 1:400 dilution, 11948), and rat monoclonal anti-IL6 (ThermoFisher Scientific, 1:200 dilution, 11-7061-82) were used as primary antibodies, and control incubations were routinely processed without primary antibody treatments.

Propidium iodide (PI) staining was used to detect the apoptotic and necrotic HC nuclei after noise exposure as previously reported 23. Fixed cochleae were permeabilized in 1% Triton X-100 solution for 30 min and then stained with 5 mg/ml PI dissolved in PBS for 1 h at room temperature. After the final washes with PBS (10 min each), the tissue was dissected in PBS by removing the modiolus.

TUNEL staining was used to detect apoptotic HC death. Tissues were fixed with 4% PFA for 1 h, treated with 1% Triton X-100 37 °C for 40 s, and incubated in TUNEL staining solution (Roche) according to the manufacturer’s instructions.

Confocal imaging and cell counting

Specimens were examined by confocal fluorescence microscopy (Leica SP8). To quantify the immunostaining-positive cells, nine separate segments along the entire cochlea were selected from the apex to the base.

Real-time PCR

RNA was extracted from six cochleae of P30 adult mice from each group and was subsequently reverse transcribed into cDNA using Superscript III reverse transcriptase (Invitrogen) according to the manufacturer's instructions. Real-time PCR was performed on an ABI 7500 real-time PCR system (Applied Biosystems) using the TB Green™ PrimeScript™ RT-PCR Kit (Takara). The Actb gene was used as the endogenous control. Primer sets are listed in Additional file 1: Table S1. The quantification of relative gene expression, compared to Actb, was analyzed by the 2−ΔΔCT method.

Western blotting

Proteins were extracted from isolated cochlear epithelia with lysis buffer containing proteinase inhibitors, separated by SDS-PAGE gel, and transferred onto PVDF membranes (Immobilon-P; Millipore, Bedford, MA, USA). Membranes were blocked in 5% nonfat dried milk in TBST (20 mM Tris–HCl, 500 mM NaCl, and 0.1% Tween-20) for 1 h and later incubated with the primary antibodies overnight at 4 °C. The following antibodies were used for Western blot analysis: mouse monoclonal anti-beta-Actin (Thermo Fisher Scientific, 1:5000 dilution, MA515739), rabbit mAb anti-TNF-α (Cell Signaling Technology, 1:3000 dilution, 11948), and rat monoclonal anti-IL6 (ThermoFisher Scientific, 1:2000 dilution, 11-7061-82). The next day, the blots were washed three times with TBST and subsequently incubated with corresponding HRP-conjugated secondary antibodies for 1 h at room temperature. The immunoreactive bands were detected using an ECL kit (Pierce).

Statistical analysis

Data were analyzed using GraphPad Prism 8. Student’s 2-tailed t-test was used to analyze the differences between two groups, and one-way ANOVA followed by the Bonferroni post-test was used for comparisons of differences among three or four groups. Data are presented as the means ± S.E.M, and p < 0.05 was considered significant.

Study approval

This study followed the “Guiding Directive for Humane Treatment of Laboratory Animals” reporting guidelines enacted by the Chinese National Ministry of Science and Technology in 2006. All experiments were approved by the Shanghai Medical Experimental Animal Administrative Committee (Permit Number: 2009-0082) and were in accordance with animal welfare principles.

Results

Sox2 abundance is correlated with HC tolerance to ototoxic drugs in neonatal HCs

First, we examined the Sox2 expression pattern in the cochleae of wild type mice. Myosin7a was used as the marker of cochlear HCs. The cochlear epithelium is a delicate tissue containing region-specific HCs, which can be roughly classified into two categories, namely inner hair cells (IHCs) and outer hair cells (OHCs), which have different functions, gene expression patterns, and morphological characteristics [24] (Fig. 1A). Immunofluorescence results revealed that Sox2 was strongly expressed in all cochlear supporting cells, including inner border cells, inner phalangeal cells, inner pillar cells, outer pillar cells, Deiters’ cells, and Hensen’s cells (Fig. 1C). In the cochleae of adult control (PrestinCreER+/−/ Sox2OE−/−) mice, Sox2 was not observed in the cochlear HCs (Fig. 1C). However, in the cochleae of neonatal mice, Sox2 was also observed in some IHCs and OHCs, but the Sox2 abundance in HCs decreased from the apical to the basal turns (Additional file 1: Fig. S1A, note the Sox2 + HCs indicated by arrows in the apical turns). Similar to what was observed previously [25, 26], we found that the HC sensitivity to ototoxic drugs increased from the apical to basal turns because more HCs were lost in the basal turns compared to the apical turns in neonatal mice (Additional file 1: Fig. S1B).

Fig. 1
figure 1

The cochlear morphology and hearing function were not affected by Sox2 overexpression. A The cross-section of the cochlear sensory epithelium. The IHCs and OHCs are located on the top layer, and most supporting cells (including BC, IphC, IPC, OPC, DC, and HeC) are located on the under layer. Some HCs can be observed in the top layer. IPC inner pillar cell, OPC outer pillar cell, DC Deiters’ cell, IPhC inner phalangeal cell, BC inner border cell, HeC Hensen’s cell, IS inner sulcus, CC Claudius cell. B Schematic for creating the transgenic mouse models. The Rosa26-CAG-LSL(loxP-stop-loxP)-Sox2 transgenic mice, referred to as the Sox2OE mice, were designed using the CRISPR/Cas9 method. C Images of Sox2 immunofluorescence (whole mount, HC layer) showed that there was no detectable Sox2 expression in the cochlear HCs of the control group, while in the PrestinCreER+/–/Sox2OE+/– group Sox2 expression was clearly observed in OHCs. D Experimental protocol for E–F. Tamoxifen was injected once daily during P21–P22, and the cochlear morphology and hearing function were assessed by immunofluorescence and ABR at P30. E Representative Myosin7a immunofluorescence showed no difference in HC morphology between the PrestinCreER+/–/Sox2OE+/– and control groups. F Quantification of IHCs and OHCs in the PrestinCreER+/–/Sox2OE+/– and control groups. G The ABR thresholds of adult PrestinCreER+/–/Sox2OE+/– and control mice. Data are presented as the mean ± SEM. n = 5 for each group. Scale bar = 20 µm

Sox2 overexpression in HCs did not alter HC morphology or auditory function under physiological conditions

To overexpress Sox2 in the cochlear HCs, we generated the inducible Rosa26-CAG-LSL-Sox2 mouse line in which forced expression of Sox2 was driven by the CAG promoter in a Cre-mediated manner. To examine the effects of Sox2 overexpression (Sox2OE) on cochlear HC survival in the neonatal and adult stages, respectively, we used two Cre mouse lines, namely, the Atoh1CreER line that targets the HCs including both IHCs and OHCs in the neonatal stage and the PrestinCreER line that targets OHCs in the adult stage (Fig. 1B). We generated PrestinCreER+/–/Sox2OE+/– mice in which constitutive expression of Sox2 in Prestin+ cells were obtained by tamoxifen administration, and these are referred to as Prestin-Sox2OE mice. PrestinCreER+/–/Sox2OE–/– mice served as controls. To evaluate the expression of Sox2 in OHCs and to determine the effect of Sox2OE on HC function under physiological conditions, Prestin-Sox2OE and control mice were injected intraperitoneally (i.p.) with tamoxifen from P21 to P22, and their hearing function and cochlear morphological features were assessed at P30 (Fig. 1D). In the control group, no Sox2 staining was observed in OHCs from mature cochleae, while nuclear staining of Sox2 was clearly observed in OHCs of Prestin-Sox2OE mice, confirming the successful induction of Cre activity and Sox2 overexpression in the Prestin+ cells (Fig. 1C). Immunofluorescence results showed that the morphological features and quantities of cochlear HCs in the Prestin-Sox2OE group were similar to those of controls (Fig. 1E, F). In addition, the ABR results showed that the hearing function of adult Prestin-Sox2OE mice was comparable to controls at 8, 16, 24, and 32 kHz (Fig. 1G). These results suggested that Sox2 overexpression in OHCs did not affect the maintenance of HC function.

Sox2 overexpression protected HCs against aminoglycoside-induced injury in the neonatal stage

To explore the function of Sox2 in HC survival, we first used an in vitro aminoglycoside antibiotic injury model. We generated Atoh1CreER+/–/Sox2OE+/– mice in which constitutive expression of Sox2 in Atoh1+ HCs was obtained by tamoxifen administration, and these are referred to as the Atoh1-Sox2OE group.

The cochlear epithelium from P2 Atoh1-Sox2OE mice that received tamoxifen injections at P 0-1 were cultured and treated with 1 mM neomycin for 6 h and then allowed to recover without neomycin for an additional 24 h prior to harvesting (Fig. 2A). Atoh1CreER+/–/Sox2OE–/– mice served as controls. Myosin7a immunofluorescence was used to determine the number of remaining HCs after neomycin treatment. In the control group, neomycin treatment led to massive HC loss, and the average numbers of remaining Myosin7a+ HCs were 57.0 ± 0.577, 31.5 ± 2.51, and 16.5 ± 1.52 cells/100 µm in the apical, middle, and basal turns, respectively (Fig. 2B–D). In the Atoh1-Sox2OE group, the average numbers of Myosin7a+ HCs were 56.8 ± 0.601, 54.2 ± 1.08, and 43.3 ± 2.33 cells/100 µm in the apical, middle, and basal turns, respectively. The average numbers of remaining HCs in the middle and basal turns from the Atoh1-Sox2OE groups were significantly larger than those from control groups (Fig. 2D, p < 0.001). Thus it appears that there is a protective effect of Sox2 overexpression against neomycin injury.

Fig. 2
figure 2

Sox2 overexpression protected HCs from aminoglycoside-induced injury in the neonatal stage A Experimental protocol for C-D. Cochlear epithelia from Atoh1CreER+/–/Sox2OE+/– and control mice that received tamoxifen injection at P0–P1 were dissected and cultured at P2. Cochlear explants were treated with 1 mM neomycin for 6 h and harvested after an additional 24 h. B The scheme for cell counting. C Representative images of Myosin7a+ cells in Atoh1CreER+/–/Sox2OE+/– and control mice treated with or without neomycin. D Quantification of the cochlear HCs per 100 µm in the Atoh1CreER+/–/Sox2OE+/– and control groups. Data are presented as means ± SEM; n = 6 for each group; ***P < 0.001. Scale bar = 20 µm

Sox2 overexpression attenuated noise-induced HC damage and hearing loss in adult mice

To determine whether the susceptibility of HCs to noise exposure changed after Sox2 overexpression, P31 Prestin-Sox2OE mice that received tamoxifen at P21–22 were exposed to 116 dB broadband noise for 2 h (Fig. 3A). The hearing thresholds of the mice were determined using ABR tests at P38, and the cochlear specimens were harvested afterwards. PrestinCreER+/–/Sox2OE–/– mice served as controls. In the control group, noise exposure led to significant OHC loss in the middle and basal turns of the cochlea (Fig. 3B–D). However, in the Prestin-Sox2OE cochlear epithelium, the noise-induced loss of OHCs was significantly attenuated in the middle and basal turns compared with the control mice (p < 0.01, Fig. 3B–D), indicating that Sox2-overexpressing cochlear OHCs were less sensitive to noise-induced injury.

Fig. 3
figure 3

Sox2 overexpression alleviated noise-induced hearing loss and HC damage. A Experimental protocol for B–F. Adult PrestinCreER+/–/Sox2OE+/– and control mice that received tamoxifen at P 21–22 were exposed to 116 dB broadband noise for 2 h. Cochlear morphology and hearing function were evaluated 7 days after noise exposure. B Representative images of Myosin7a+ cells in PrestinCreER+/–/Sox2OE+/– and control mice treated with or without noise exposure. C, D Cochleograms showed noise-induced loss of IHCs and OHCs in the PrestinCreER+/–/Sox2OE+/– and control groups. E, F Pure-tone ABR thresholds and threshold shifts in the PrestinCreER+/–/Sox2OE+/– and control group 7 days after the noise exposure. Scale bar = 20 µm. Data are presented as means ± SEM; n = 6 for each group; *P < 0.05, **P < 0.01, ***P < 0.001, indicated the significant difference between the PrestinCreER+/–/Sox2OE+/– with noise treatment and the PrestinCreER+/–/Sox2OE–/– with noise treatment groups

Hearing thresholds before noise exposure (baseline values) showed no significant difference between the Prestin-Sox2OE and the control groups (data not shown). At 7 days after noise exposure, mice in the control group had significant hearing loss, as demonstrated by the increased hearing thresholds at 8, 16, 24, and 32 kHz (Fig. 3E). The noise-induced threshold shifts in Prestin-Sox2OE mice were significantly lower at all frequencies compared with those of the control mice (p < 0.05, Fig. 3F), suggesting that Sox2 overexpression in OHCs protects against noise-induced hearing loss.

Sox2 overexpression protected OHCs by inhibiting the noise-induced apoptosis pathway

To determine the mechanism through which Sox2 overexpression protects HCs against injury, we examined the apoptotic and necrotic changes in HCs after noise exposure using PI staining based on previous research showing the morphological changes that occur in OHCs after noise exposure [27]. In the control group, noise exposure induced morphological nuclear changes that included both apoptotic (condensed nuclei) and necrotic (swollen nuclei) changes in OHCs at 6 h after noise exposure (Fig. 4A, B). However, the number of apoptotic nuclei was significantly decreased in the Prestin-Sox2OE group when compared with the control group (Fig. 4A, B). Meanwhile, no significant differences in the numbers of necrotic nuclei were observed between the Prestin-Sox2OE and control groups. TUNEL staining was used to further determine the differences in apoptotic HCs between the two groups (Fig. 4C). Significantly fewer Myosin7a+/TUNEL+ cells were observed in Prestin-Sox2OE mice compared to control mice (Fig. 4D), suggesting that Sox2 overexpression suppressed noise-induced apoptosis of OHCs.

Fig. 4
figure 4

Sox2 overexpression suppressed noise-induced HC apoptosis. A Representative images of PI-stained OHC nuclei in the cochlear epithelium. Both swollen and condensed OHC nuclei were observed in mice treated with noise exposure. Arrowheads point to swollen necrotic nuclei, and arrows point to condensed apoptotic nuclei. B The numbers of swollen or condensed OHC nuclei and missing OHCs in the cochlear epithelium at 6 h after noise exposure. C Representative images of TUNEL staining and Myosin7a immunofluorescence of the cochlear epithelium. TUNEL+/Myosin7a+ OHCs were observed in the mouse cochlea at 6 h after noise exposure. D Quantification of TUNEL+/Myosin7a+ HCs in the cochlear epithelium. Data are presented as means ± SEM.; n = 6. *P < 0.05, **P < 0.01. Scale bar = 20 μm

We next assessed the relative mRNA expression level of apoptosis and necrosis-related genes at 6 h after noise exposure. In the control group, noise exposure induced the increased expression of both apoptosis-related genes (Aif, EndoG, and Bax) and necrosis-related genes (RipK1 and RipK3) (Fig. 5A, B). In the Prestin-Sox2OE group, the expression levels of Aif, Bax, and EndoG were significantly decreased (Fig. 5A, p < 0.05), while the expression levels of RipK1 and RipK3 showed no significant difference when compared to the control group after noise exposure (Fig. 5B), further confirming that Sox2 overexpression protected OHCs by inhibiting the noise-induced apoptosis pathway.

Fig. 5
figure 5

Sox2 overexpression suppressed noise-induced upregulation of inflammatory cytokines. A The mRNA expression levels of apoptosis-related genes (Aif, Bax, EndoG, Casp3, Casp8, and Casp9) were elevated 6 h after the completion of noise exposure. B The mRNA expression levels of necrosis-related genes (Prkaα1, Ripk1, and Ripk3) and pro-inflammatory cytokines (Tnf, Il-1β, and Il-6) were elevated 6 h after the completion of noise exposure. C TNF-α immunofluorescence in HCs after noise exposure. The TNF-α expression level was elevated after traumatic noise exposure in HCs, while it was inhibited in the Sox2 overexpression group. Parvalbumin is used as a marker of cochlear HCs. Data are presented as means ± SEM.; n = 6. *P < 0.05, **P < 0.01. Scale bar = 20 μm

Sox2 overexpression alleviated HC damage by inhibiting inflammation

In consideration of the important role of inflammation in HC death, the mRNA expression of pro-inflammation mediators was measured. In the control group, noise exposure induced significantly increased expression of Tnf, Il-1β, and Il-6 at 6 h after noise exposure (Fig. 5B). In the Prestin-Sox2OE group, the expression level of these pro-inflammation mediators declined significantly to a level comparable to that in the group without noise exposure (Fig. 5B, p < 0.05), indicating that Sox2 overexpression dramatically attenuated the noise-induced inflammation process. To further evaluate the protein expression level of pro-inflammatory mediators in cochlear epithelia, we performed Western blotting analysis of TNF-α and IL6. These results showed that noise exposure led to significant increases in TNF-α and IL6 protein levels in the control group, but Sox2 overexpression significantly inhibited the noise-induced increase of TNF-α and IL6 protein (Fig. 6). We also measured the expression of TNF-α and IL6 protein in the organ of Corti using anti-TNF-α and anti-IL6 antibodies, and we found that noise-induced TNF-α and IL6 expression in cochlear HCs was relatively lower in the Prestin-Sox2OE group compared to the control group (Figs. 5C, 6).

Fig. 6
figure 6

Sox2 overexpression suppressed noise-induced upregulation of IL6 and TNF-α. A IL6 immunofluorescence in cochlear cross-section after noise treatment. The IL6 expression level was elevated after traumatic noise exposure in HCs, while it was inhibited in the Sox2 overexpression group. Myosin7a is used as a marker of cochlear HCs. B Western blot analysis of sensory epithelia shows increased expression levels of TNF-α and IL6 in control mice after noise injury, but it was significantly inhibited in Prestin-Sox2OE mice. β-Actin served as the sample loading control. Data are presented as means ± SEM.; n = 3. *P < 0.05, **P < 0.01. Scale bar = 20 μm

To determine the role of Sox2 upregulation in the inflammatory process, we constructed the lipopolysaccharide (LPS) challenge model. It has been previously reported that inflammation-related mRNA expression is upregulated in the cochlea within 6 h after LPS challenge [23]. To determine the optimal concentration of LPS in inflammation-mediated HC loss, 10 μl LPS at different concentrations (1 mg/kg, 2 mg/kg, and 4 mg/kg) was injected into the tympanic cavity of wild type mice and the cochleae were dissected for analysis 72 h later. LPS induced no obvious cochlear HC loss in the 1 mg/kg LPS group, mild HC loss in the basal turns in the 2 mg/kg LPS group, and severe HC loss in the middle and basal turns in the 4 mg/kg LPS group. Thus, 4 mg/kg of LPS was considered the optimal concentration for inducing significant HC loss in both the middle and basal turns.

We compared LPS-induced HC damage between the Prestin-Sox2OE and control groups. Significantly more cochlear HCs were observed in the middle and basal turns in the Prestin-Sox2OE group than in the control group, indicating that Sox2 overexpression largely alleviated LPS-induced HC damage (Fig. 7A–C). We next evaluated the mRNA expression level of pro-inflammatory cytokines induced by LPS. qPCR results showed that Tnf, Il-6, and Il-1β increased substantially after LPS treatment in the control group, but their expression was relatively lower in the Prestin-Sox2OE group, which was a sign that the inflammation reaction was limited (Fig. 7E). Western blot analysis also showed very high expression levels of TNF-α and IL6 protein in the control mice and limited expression in the Prestin-Sox2OE mice after LPS treatment (Fig. 8). The immunofluorescence results also showed that the LPS-induced upregulation of TNF-α and IL6 in HCs was attenuated by Sox2 overexpression (Figs. 7D, 8). Collectively, these results showed that Sox2 upregulation inhibited the cochlear inflammatory response and subsequent HC death.

Fig. 7
figure 7

Sox2 upregulation mitigated inflammation-induced HC loss. A Experimental protocol for the LPS-induced HC loss model. B Myosin7a immunofluorescence in cochlear HCs of adult PrestinCreER+/–/Sox2OE+/– mice and control mice with or without LPS treatment. C Cell counting results of cochlear HCs after 4 mg/kg of LPS administration. D TNF-α immunofluorescence in cochlear cross-section after LPS challenge. E The mRNA expression levels of pro-inflammatory cytokines. Sox2 overexpression prevented the LPS-induced increase in pro-inflammatory cytokines. Data are presented as means ± SEM; n = 6; **P < 0.01, and ***P < 0.001. Scale bar = 20 μm

Fig. 8
figure 8

Sox2 overexpression suppressed LPS-induced upregulation of IL6 and TNF-α. A IL6 immunofluorescence in cochlear cross-section after LPS treatment. Myosin7a is used as a marker of cochlear HCs. (B) Western blot analysis of sensory epithelia shows increased expression levels of TNF-α and IL6 in control mice after LPS treatment, but the LPS-induced increase of the TNF-α and IL6 protein was significantly inhibited in Prestin-Sox2OE mice. β-Actin served as the sample loading control. Data are presented as means ± SEM.; n = 3. *P < 0.05, **P < 0.01. Scale bar = 20 μm

Discussion

An estimated 466 million people are living with hearing loss worldwide. Although the exact mechanisms are not fully understood, oxidative stress, inflammation, autophagy, apoptosis, and necrosis have been reported to be involved in the onset and progression of sensorineural hearing loss [27, 28], and various strategies have been explored to try to protect sensory HCs and hearing. Some anti-apoptosis genes, like Bcl2 and Xiap, have been reported to have protective roles in cochlear HCs and hearing [29, 30]. Antioxidants such as N-acetyl-L-cysteine, coenzyme Q10, and vitamin E have been reported to be effective in protecting against ototoxic drugs and noise exposure [31,32,33,34]. Some genes, like Bmi1, G6PD, adenosine A1 receptor, and FoxG1, protect HCs by inhibiting oxidative stress or by suppressing inflammatory pathways that are induced by ototoxic drugs or that are related to aging-related damage [28, 35,36,37]. Besides classic anti-apoptosis, anti-necrosis, and anti-inflammation factors, some development-related factors, such as Wnt/beta-catenin and Islet-1, have been shown to be capable of protecting HCs against injury and thus preserving hearing [35, 38].

In the cochlea, apical HCs are responsible for sensing sound at lower frequencies, and basal HCs are responsible for sensing sound at higher frequencies. Hearing loss in patients is generally more severe at higher frequencies than lower frequencies, and our experimental data showed that the sensitivity of HCs to injury increased from the apical to basal turns. The three different treatments—aminoglycosides (Fig. 2), broadband noise (Fig. 3), and LPS (Fig. 7)—all led to an apical-to-basal gradient of damage in cochlear HCs, and severe HC loss occurred in the basal turn while the mildest HC loss occurred in the apical turn. Previous studies showed that exposure to low-decibel broad band noise (8–16 kHz) results in HC damage mainly in the middle turns, but the HC damage extends to the basal turn when the intensity of the noise is greater than 100 dB [39,40,41]. The mechanism behind the difference in HC injury sensitivity along the cochlear turns remains unclear. In this work, we found that Sox2 was expressed in the neonatal cochlear HCs, which are less sensitive to injury (Additional file 1: Fig. S1), and Sox2 overexpression alleviated HC loss induced by aminoglycosides in the neonatal stage (Fig. 2), suggesting that deficiency of Sox2 in HCs in the basal turn might be one of the reasons for their vulnerability to ototoxic drugs.

The influence of noise exposure on people’s physiological and mental state has been the focus of increasing attention in the past decade [42, 43]. The impact of noise-induced hearing loss is broad and profound, including the loss of the ability to communicate with others and subsequent social isolation. Up to now, few medicines approved by the Food and Drug Administration have been confirmed to be effective in preventing or treating noise-induced hearing loss in clinical trials. Consistent with previous studies [44,45,46], our data further confirmed that apoptosis, necrosis, and inflammation are involved in noise-induced HC death (Figs. 4, 5). Aif and Endo G, which act as apoptotic mediators, translocate into the nucleus during HC injury and lead to HC apoptosis [47,48,49]. Prkaa1 (also known as AMPK) and receptor-interacting protein kinase (RipK) have been reported be associated with necrotic HC death [27, 50]. Although the endogenous expression of Sox2 is very low in mature cochlear HCs, forced Sox2 overexpression in Prestin+ OHCs has a strong protective effect against noise-induced hearing injury (Fig. 3). PI staining results showed that Sox2 upregulation decreased the number of HCs that underwent apoptosis (Fig. 4), and this was accompanied by a decrease in noise-induced expression of pro-apoptosis genes (Aif, EndoG, and Bax) and the number of TUNEL+ HCs (Fig. 5). In contrast, Sox2 overexpression had no obvious effect on the number of HCs that underwent necrosis, as demonstrated by the PI staining results and the mRNA expression levels of pro-necrosis factors (Figs. 4, 5). Together, these results suggest that Sox2 overexpression protects HCs and hearing against noise-induced injury by inhibiting HC apoptosis.

The inflammatory response activated by extrinsic attack and infection is also a strictly regulated process, and an appropriate inflammatory response can eliminate the pathogen, repair damaged tissue, and maintain cell homeostasis [51, 52]. However, dysregulation of the inflammatory response is pivotal in the initiation and progression of multiple diseases, such as age-related diseases [53] and neurodegeneration diseases [54]. In the auditory system, LPS can penetrate into the labyrinth through the round window membrane, where it stimulates the production and release of cytokines and mediates the initiation and development of inflammatory reactions in the media and labyrinth, which in turn leads to auditory epithelial injury and subsequent sensorineural hearing loss [55]. In our experiments, LPS administration caused a significant increase in the expression of pro-inflammatory factors and a significant loss of OHCs, but this inflammation-induced HC loss was alleviated by Sox2 overexpression (Fig. 7). Previous studies have shown that many pro-inflammatory cytokines and inflammatory mediators can lead to cell apoptosis [56,57,58]. For example, increased interleukin levels mediate cell apoptosis by regulating the Bax/Bcl-2 ratio in the cardiovascular system [56], and the TNF-α signaling pathway manipulates cell apoptosis through the Bax/Bak pathway or through a caspase-dependent pathway [57,58,59,60,61]. Our previous study showed that inhibition of the TNF-α pathway was an effective means of protecting HCs [26]. In this paper, we found that Sox2 overexpression dramatically suppressed LPS-induced and noise-induced upregulation of inflammatory cytokines and apoptosis-related genes (Figs. 5, 6, 7, 8), indicating that Sox2 overexpression inhibited the cascade of inflammatory cytokine release and subsequent inflammation-mediated HC apoptosis and hearing loss.

Conclusions

We report for the first time that Sox2 protects mature and terminally differentiated cells against injury. Sox2 overexpression preserved hearing against noise exposure by inhibiting HC apoptosis, and this was correlated with the decreased expression of pro-inflammatory factors. These results thus suggest the protective role of Sox2 overexpression in hearing and sensory HCs and provide novel targets for strategies for the prevention and treatment of hearing loss.

Availability of data and materials

All data generated or analyzed during this study are included in this published article and its supplementary information files.

Abbreviations

Aif:

Apoptosis inducing factor

Endo G:

Endonuclease G

Prkaa1:

Phosphorylated-AMP kinase α

RIPK:

Receptor interacting protein kinase

PARP-1:

Poly(ADP-ribose) polymerase 1

ROS:

Reactive oxygen species

Sox2:

HMG-box transcription factor sex-determining region Y-box 2

TNF-α:

Tumor necrosis factor

Il-6:

Interleukin 6

Il-1β:

Interleukin 1β

References

  1. Yamasoba T, Lin FR, Someya S, Kashio A, Sakamoto T, Kondo K. Current concepts in age-related hearing loss: epidemiology and mechanistic pathways. Hear Res. 2013;303:30–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Fridberger A, Flock A, Ulfendahl M, Flock B. Acoustic overstimulation increases outer hair cell Ca2+ concentrations and causes dynamic contractions of the hearing organ. Proc Natl Acad Sci U S A. 1998;95:7127–32.

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Davis A, McMahon CM, Pichora-Fuller KM, Russ S, Lin F, Olusanya BO, Chadha S, Tremblay KL. Aging and hearing health: the life-course approach. Gerontologist. 2016;56:S256–67.

    PubMed  PubMed Central  Google Scholar 

  4. Desa DE, Nichols MG, Smith HJ. Aminoglycosides rapidly inhibit NAD(P)H metabolism increasing reactive oxygen species and cochlear cell demise. J Biomed Opt. 2018;24:1–14.

    PubMed  Google Scholar 

  5. Azadegan-Dehkordi F, Bahrami T, Shirzad M, Karbasi G, Yazdanpanahi N, Farrokhi E, Koohiyan M, Tabatabaiefar MA, Hashemzadeh-Chaleshtori M. Mutations in GJB2 as major causes of autosomal recessive non-syndromic hearing loss: first report of c.299–300delAT mutation in Kurdish population of Iran. J Audiol Otol. 2019;23:20–6.

    PubMed  Google Scholar 

  6. Nist-Lund CA, Pan B, Patterson A, Asai Y, Chen T, Zhou W, Zhu H, Romero S, Resnik J, Polley DB, et al. Improved TMC1 gene therapy restores hearing and balance in mice with genetic inner ear disorders. Nat Commun. 2019;10:236.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Ficenec SC, Schieffelin JS, Emmett SD. A review of hearing loss associated with Zika, Ebola, and Lassa Fever. Am J Trop Med Hyg. 2019;101:484–90.

    PubMed  PubMed Central  Google Scholar 

  8. Wagner EL, Shin J-B. Mechanisms of hair cell damage and repair. Trends Neurosci. 2019;42:414–24.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Waqas M, Gao S, Iram-Us-Salam, Ali MK, Ma Y, Li W. Inner ear hair cell protection in mammals against the noise-induced cochlear damage. Neural Plast. 2018; 2018:3170801.

  10. Dinh CT, Goncalves S, Bas E, Van De Water TR, Zine A. Molecular regulation of auditory hair cell death and approaches to protect sensory receptor cells and/or stimulate repair following acoustic trauma. Front Cell Neurosci. 2015;9:96.

    PubMed  PubMed Central  Google Scholar 

  11. Novak D, Huser L, Elton JJ, Umansky V, Altevogt P, Utikal J. SOX2 in development and cancer biology. Semin Cancer Biol. 2020;67:74–82.

    CAS  PubMed  Google Scholar 

  12. Okubo T, Pevny LH, Hogan BLM. Sox2 is required for development of taste bud sensory cells. Genes Dev. 2006;20:2654–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Gontan C, de Munck A, Vermeij M, Grosveld F, Tibboel D, Rottier R. Sox2 is important for two crucial processes in lung development: branching morphogenesis and epithelial cell differentiation. Dev Biol. 2008;317:296–309.

    CAS  PubMed  Google Scholar 

  14. Que JW, Luo XY, Schwartz RJ, Hogan BLM. Multiple roles for Sox2 in the developing and adult mouse trachea. Development. 2009;136:1899–907.

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Ahmed M, Wong EYM, Sun J, Xu J, Wang F, Xu P-X. Eya1-Six1 interaction is sufficient to induce hair cell fate in the cochlea by activating Atoh1 expression in cooperation with Sox2. Dev Cell. 2012;22:377–90.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Zhang S, Xiong X, Sun Y. Functional characterization of SOX2 as an anticancer target. Signal Transduct Target Therapy. 2020;5:135.

    CAS  Google Scholar 

  17. Feng R, Zhou S, Liu Y, Song D, Luan Z, Dai X, Li Y, Tang N, Wen J, Li L. Sox2 protects neural stem cells from apoptosis via up-regulating survivin expression. Biochem J. 2013;450:459–68.

    CAS  PubMed  Google Scholar 

  18. Dabdoub A, Puligilla C, Jones JM, Fritzsch B, Kathryn SEC, Pevny LH, Kelley MW. Sox2 signaling in prosensory domain specification and subsequent hair cell differentiation in the developing cochlea. Proc Natl Acad Sci USA. 2008;105:18396–401.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Oesterle EC, Campbell S, Taylor RR, Forge A, Hume CR. Sox2 and Jagged1 expression in normal and drug-damaged adult mouse inner ear. J Assoc Res Otolaryngol. 2008;9:65–89.

    PubMed  Google Scholar 

  20. Millimaki BB, Sweet EM, Riley BB. Sox2 is required for maintenance and regeneration, but not initial development, of hair cells in the zebrafish inner ear. Dev Biol. 2010;338:262–9.

    CAS  PubMed  Google Scholar 

  21. Zeng S, Ni W, Jiang H, You D, Wang J, Lu X, Liu L, Yu H, Wu J, Chen F, et al. Toxic effects of 3,3’-iminodipropionitrile on vestibular system in adult C57BL/6J mice in vivo. Neural Plast. 2020;2020:1823454.

    PubMed  PubMed Central  Google Scholar 

  22. Fang J, Zhang W-C, Yamashita T, Gao J, Zhu M-S, Zuo J. Outer hair cell-specific prestin-CreERT2 knockin mouse lines. Genesis. 2012;50:124–31.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Cox BC, Liu ZY, Lagarde MMM, Zuo J. Conditional gene expression in the mouse inner ear using Cre-IoxP. JARO J Assoc Res Otolaryngol. 2012;13:295–322.

    PubMed  Google Scholar 

  24. Liu H, Pecka JL, Zhang Q, Soukup GA, Beisel KW, He DZ. Characterization of transcriptomes of cochlear inner and outer hair cells. J Neurosci. 2014;34:11085–95.

    PubMed  PubMed Central  Google Scholar 

  25. Chen Y, Li L, Ni W, Zhang Y, Sun S, Miao D, Chai R, Li H. Bmi1 regulates auditory hair cell survival by maintaining redox balance. Cell Death Dis. 2015;6: e1605.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Zhang Y, Chen D, Zhao L, Li W, Ni Y, Chen Y, Li H. Nfatc4 deficiency attenuates ototoxicity by suppressing Tnf-mediated hair cell apoptosis in the mouse cochlea. Front Immunol. 2019;10:1660.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Zheng HW, Chen J, Sha SH. Receptor-interacting protein kinases modulate noise-induced sensory hair cell death. Cell Death Dis. 2014;5: e1262.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Bermudez-Munoz JM, Celaya AM, Hijazo-Pechero S, Wang J, Serrano M, Varela-Nieto I. G6PD overexpression protects from oxidative stress and age-related hearing loss. Aging Cell. 2020;19: e13275.

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Kim YJ, Choo OS, Lee JS, Jang JH, Woo HG, Choung YH. BCL2 interacting protein 3-like/NIX-mediated mitophagy plays an important role in the process of age-related hearing loss. Neuroscience. 2020;455:39–51.

    PubMed  Google Scholar 

  30. Sun S, Sun M, Zhang Y, Cheng C, Waqas M, Yu H, He Y, Xu B, Wang L, Wang J, et al. In vivo overexpression of X-linked inhibitor of apoptosis protein protects against neomycin-induced hair cell loss in the apical turn of the cochlea during the ototoxic-sensitive period. Front Cell Neurosci. 2014;8:248.

    PubMed  PubMed Central  Google Scholar 

  31. Kranzer K, Elamin WF, Cox H, Seddon JA, Ford N, Drobniewski F. A systematic review and meta-analysis of the efficacy and safety of N-acetylcysteine in preventing aminoglycoside-induced ototoxicity: implications for the treatment of multidrug-resistant TB. Thorax. 2015;70:1070–7.

    PubMed  Google Scholar 

  32. Kopke R, Slade MD, Jackson R, Hammill T, Fausti S, Lonsbury-Martin B, Sanderson A, Dreisbach L, Rabinowitz P, Torre P III, Balough B. Efficacy and safety of N-acetylcysteine in prevention of noise induced hearing loss: a randomized clinical trial. Hear Res. 2015;323:40–50.

    CAS  PubMed  Google Scholar 

  33. Fetoni AR, De Bartolo P, Eramo SLM, Rolesi R, Paciello F, Bergamini C, Fato R, Paludetti G, Petrosini L, Troiani D. Noise-induced hearing loss (NIHL) as a target of oxidative stress-mediated damage: cochlear and cortical responses after an increase in antioxidant defense. J Neurosci. 2013;33:4011–23.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Le Prell CG, Gagnon PM, Bennett DC, Ohlemiller KK. Nutrient-enhanced diet reduces noise-induced damage to the inner ear and hearing loss. Transl Res. 2011;158:38–53.

    PubMed  PubMed Central  Google Scholar 

  35. Liu L, Chen Y, Qi J, Zhang Y, He Y, Ni W, Li W, Zhang S, Sun S, Taketo MM, et al. Wnt activation protects against neomycin-induced hair cell damage in the mouse cochlea. Cell Death Dis. 2016;7:e2136–e2136.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. He ZH, Zou SY, Li M, Liao FL, Wu X, Sun HY, Zhao XY, Hu YJ, Li D, Xu XX, et al. The nuclear transcription factor FoxG1 affects the sensitivity of mimetic aging hair cells to inflammation by regulating autophagy pathways. Redox Biol. 2020;28: 101364.

    CAS  PubMed  Google Scholar 

  37. Kaur T, Borse V, Sheth S, Sheehan K, Ghosh S, Tupal S, Jajoo S, Mukherjea D, Rybak LP, Ramkumar V. Adenosine A1 receptor protects against cisplatin ototoxicity by suppressing the NOX3/stat1 inflammatory pathway in the cochlea. J Neurosci. 2016;36:3962–77.

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Huang M, Kantardzhieva A, Scheffer D, Liberman MC, Chen ZY. Hair cell overexpression of islet1 reduces age-related and noise-induced hearing loss. J Neurosci. 2013;33:15086–94.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Jensen JB, Lysaght AC, Liberman MC, Qvortrup K, Stankovic KM. Immediate and delayed cochlear neuropathy after noise exposure in pubescent mice. Plos One. 2015, 10.

  40. Muthaiah VPK, Chen G-D, Ding D, Salvi R, Roth JA. Effect of manganese and manganese plus noise on auditory function and cochlear structures. Neurotoxicology. 2016;55:65–73.

    CAS  PubMed  Google Scholar 

  41. Hirose K, Liberman MC. Lateral wall histopathology and endocochlear potential in the noise-damaged mouse cochlea. JARO J Assoc Res Otolaryngol. 2003;4:339–52.

    PubMed  Google Scholar 

  42. Basner M, Babisch W, Davis A, Brink M, Clark C, Janssen S, Stansfeld S. Auditory and non-auditory effects of noise on health. Lancet. 2014;383:1325–32.

    PubMed  Google Scholar 

  43. Agrawal Y, Platz EA, Niparko JK. Prevalence of hearing loss and differences by demographic characteristics among US adults—data from the National Health and Nutrition Examination Survey, 1999–2004. Arch Intern Med. 2008;168:1522–30.

    PubMed  Google Scholar 

  44. Sheth S, Mukherjea D, Rybak LP, Ramkumar V. Mechanisms of cisplatin-induced ototoxicity and otoprotection. Front Cell Neurosci. 2017;11:12.

    Google Scholar 

  45. Jiang MY, Karasawa T, Steyger PS. Aminoglycoside-induced cochleotoxicity: a review. Front Cell Neurosci. 2017;11:14.

    Google Scholar 

  46. Kurabi A, Keithley EM, Housley GD, Ryan AF, Wong ACY. Cellular mechanisms of noise-induced hearing loss. Hear Res. 2017;349:129–37.

    CAS  PubMed  Google Scholar 

  47. Jiang H, Sha SH, Forge A, Schacht J. Caspase-independent pathways of hair cell death induced by kanamycin in vivo. Cell Death Differ. 2006;13:20–30.

    CAS  PubMed  Google Scholar 

  48. Yamashita D, Miller JM, Jiang H-Y, Minami SB, Schacht J. AIF and EndoG in noise-induced hearing loss. NeuroReport. 2004;15:2719–22.

    PubMed  Google Scholar 

  49. Han W, Shi X, Nuttall AL. AIF and endoG translocation in noise exposure induced hair cell death. Hear Res. 2006;211:85–95.

    CAS  PubMed  Google Scholar 

  50. Chen FQ, Zheng HW, Hill K, Sha SH. Traumatic noise activates Rho-family GTPases through transient cellular energy depletion. J Neurosci. 2012;32:12421–30.

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Koh TJ, DiPietro LA. Inflammation and wound healing: the role of the macrophage. Expert Rev Mol Med. 2011. https://doi.org/10.1017/S1462399411001943.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Soehnlein O, Lindbom L. Phagocyte partnership during the onset and resolution of inflammation. Nat Rev Immunol. 2010;10:427–39.

    CAS  PubMed  Google Scholar 

  53. Green DR, Galluzzi L, Kroemer G. Mitochondria and the autophagy-inflammation-cell death axis in organismal aging. Science. 2011;333:1109–12.

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Vodret S, Bortolussi G, Iaconcig A, Martinelli E, Tiribelli C, Muro AF. Attenuation of neuro-inflammation improves survival and neurodegeneration in a mouse model of severe neonatal hyperbilirubinemia. Brain Behav Immun. 2018;70:166–78.

    CAS  PubMed  Google Scholar 

  55. Tateossian H, Morse S, Parker A, Mburu P, Warr N, Acevedo-Arozena A, Cheeseman M, Wells S, Brown SDM. Otitis media in the Tgif knockout mouse implicates TGF signalling in chronic middle ear inflammatory disease. Hum Mol Genet. 2013;22:2553–65.

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Liao YH, Xia N, Zhou SF, Tang TT, Yan XX, Lv BJ, Nie SF, Wang J, Iwakura Y, Xiao H, et al. Interleukin-17A contributes to myocardial ischemia/reperfusion injury by regulating cardiomyocyte apoptosis and neutrophil infiltration. J Am Coll Cardiol. 2012;59:420–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Moujalled DM, Cook WD, Okamoto T, Murphy J, Lawlor KE, Vince JE, Vaux DL. TNF can activate RIPK3 and cause programmed necrosis in the absence of RIPK1. Cell Death Dis. 2013;4:8.

    Google Scholar 

  58. Maianski NA, Roos D, Kuijpers TW. Tumor necrosis factor alpha induces a caspase-independent death pathway in human neutrophils. Blood. 2003;101:1987–95.

    CAS  PubMed  Google Scholar 

  59. Deng Y, Lin Y, Wu X. TRAIL-induced apoptosis requires Bax-dependent mitochondrial release of Smac/DIABLO. Genes Dev. 2002;16:33–45.

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Ding WX, Yin XM. Dissection of the multiple mechanisms of TNF-alpha-induced apoptosis in liver injury. J Cell Mol Med. 2004;8:445–54.

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Vince JE, Wong WWL, Khan N, Feltham R, Chau D, Ahmed AU, Benetatos CA, Chunduru SK, Condon SM, McKinlay M, et al. IAP antagonists target cIAP1 to induce TNF alpha- dependent apoptosis. Cell. 2007;131:682–93.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Natural Science Foundation of China (82071049, 81570911), and the Nature Science Foundation (No. 17ZR1404600) from Shanghai Science and Technology Committee.

Author information

Authors and Affiliations

Authors

Contributions

YN, YC, and WL designed experiments and revised the manuscript. DC, YZ, and GJ conducted the experiments, analyzed the data, and wrote the manuscript. HM and LZ conducted the experiments. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Wenyan Li, Yan Chen or Yusu Ni.

Ethics declarations

Ethics approval and consent to participate

We performed all animal procedures according to protocols that were approved by the Animal Care and Use Committee of Fudan University and that were consistent with the National Institute of Health’s Guide for the Care and Use of Laboratory Animals. We made all efforts to minimize the number of animals used and to prevent their suffering.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Table S1.

Primers used in qPCR. Table S2. Primers used in genotyping. Fig. S1. Cochlear HC sensitivity to ototoxic drugs increased from the apical to basal turns. (A) Images of cochlear cross-sections (orthogonal views) showing endogenous Sox2 expression in cochlear HCs and supporting cells in P2 mice. (B) Cochlear epithelia from P2 mice (wild type) were cultured and treated with 1 mM neomycin for 6 h, allowed to recover for 24 h, then fixed for Myosin7a immunofluorescence analysis. Scale bars: 20 μm.

Rights and permissions

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://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, D., Jia, G., Zhang, Y. et al. Sox2 overexpression alleviates noise-induced hearing loss by inhibiting inflammation-related hair cell apoptosis. J Neuroinflammation 19, 59 (2022). https://doi.org/10.1186/s12974-022-02414-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12974-022-02414-0

Keywords