Next Article in Journal
ApoB100 and Atherosclerosis: What’s New in the 21st Century?
Previous Article in Journal
Intestinal Dysbiosis in Subjects with Obesity from Western Mexico and Its Association with a Proinflammatory Profile and Disturbances of Folate (B9) and Carbohydrate Metabolism
Previous Article in Special Issue
The Role of Indoor Microbiome and Metabolites in Shaping Children’s Nasal and Oral Microbiota: A Pilot Multi-Omic Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Effects of Astragaloside IV on Hearing, Inflammatory Factors, and Intestinal Flora in Mice Exposed to Noise

School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510310, China
*
Authors to whom correspondence should be addressed.
Metabolites 2024, 14(2), 122; https://doi.org/10.3390/metabo14020122
Submission received: 30 November 2023 / Revised: 22 January 2024 / Accepted: 29 January 2024 / Published: 11 February 2024
(This article belongs to the Special Issue Environmental Toxicology and Metabolism)

Abstract

:
Long-term exposure to noise can cause irreversible hearing loss. Considering that there is no effective drug treatment, it is important to seek preventive treatment for noise-induced hearing loss (NIHL). Although astragaloside IV (AS-IV) protects against NIHL by reducing serum inflammatory factors, there is scarce information on the regulation of inflammatory factors by AS-IV to prevent NIHL. We investigated the hearing thresholds and relationship between the serum levels of inflammatory cytokines and intestinal microbiota of c57bl/6j mice exposed to noise (103 dB SPL 4 h·d−1) for 7 days, treated with or without AS-IV. Our results revealed a lower hearing threshold and lower serum levels of TNF-α, TNF-γ, IL-6, IL-1β, and IFN-γ in the mice treated with AS-IV. Additionally, AS-IV increased the abundance levels of the phylum Firmicutes, class Bacillus, order Lactobacillus, and family Lactobacillus (p < 0.05), and decreased those of the phylum Bacteroidetes and order Bacteroidales (p < 0.05). Lactobacillus and Bacilli negatively correlated with TNF-α, TNF-γ, and IL-1β; Erysipelotrichaceae negatively correlated with INF-γ; and Clostridiales positively correlated with IL-1β. In conclusion, AS-IV reduces the elevation of hearing thresholds in mice, preventing hearing loss in mice exposed to noise, and under the intervention of AS-IV, changes in the levels of inflammatory factors correlate with intestinal flora. We suggest that AS-IV improves intestinal flora and reduces inflammation levels in c57bl/6j mice exposed to noise.

1. Introduction

Approximately 5% of the global population is affected by noise-induced hearing loss (NIHL), which imposes an enormous economic and health burden on individuals and society [1]. Furthermore, occupational noise-induced deafness is the second most common occupational disease and affects approximately 16% of workers [2]. According to a systematic review, the noise-induced permanent threshold shift after 10 years of exposure at Lex,8h = 100 dB of 3–6 kHz is 27 dB [3]. A recent study on manufacturing workers has shown that the mean prevalence of high-frequency NIHL is 41.64% [4]. Although current research on NIHL drug therapy includes anti-inflammatory (such as dexamethasone [5], methylprednisolone [6], and curcumin [7]), antioxidant (such as resveratrol [8] and salicylate Trolox [9]), and antiapoptotic drugs (such as all-trans retinoic acid [10] and AM-111 [11]), there are no specific drugs for NIHL [1]. In clinical practice, hearing aids [12] and cochlear implants [13] are generally supportive treatments for permanent hearing loss, but the patients can neither fundamentally repair damaged or missing hair cells (HCs), nor they can achieve complete reconstruction of hearing function. Because hearing cells are considered terminal cells, they cannot regenerate once severely damaged [14]. Therefore, it is important to clarify the pathological mechanism of NIHL and seek effective preventive drugs for NIHL. In this study, we used the noise intensity of production work (Lex,8h ≤ 100 dB(A)) [3] with broadband noise in which harm to the auditory system may be less than an octave band to construct a NIHL model [15]; the noise should cause NIHL or pathological manifestations of outer hair cells but not cause too serious damage to avoid the excessive use of high doses of AS-IV.
Inflammatory infiltration and HC deficiency are important contributors to the pathogenesis of NIHL [16,17]. In particular, at an early stage of the development of NIHL, the levels of IL-6, TNF-α, IL-1β, and other inflammatory factors are increased [18]. One study showed that the average loss of outer hair cells (OHCs) in mice exposed to 120 dB SPL for 1 h was 29.3% [19], and another study showed that the highest loss of OHCs was over 60% in mice exposed to noise at 105 dB SPL for 2 h [20]. Noise stimulates HCs, making them active, producing an increase in mitochondria [21], and leading to superoxide production and reactive oxygen species (ROS) production [22]. The destruction of cochlear hair cells can lead to permanent noise-induced hearing loss [23,24,25]. Therefore, in this study, we considered that hearing loss had occurred or was about to occur when the pathological changes of outer hair cell loss and shedding occurred, and then ABR was used to judge the hearing state of the mice at that time [26,27].
Recent studies have shown that oral administration of AS-IV at a dose of 200 mg/kg·d−1 may reduce the elevation of hearing thresholds by decreasing the expression of ROS and active-caspase-3 in a noise-exposed guinea pig model [28]. Furthermore, AS-IV has significant pharmacological effects, such as an anti-inflammatory effect on cartilage degeneration in patients with osteoarthritis [29], an antioxidant effect that inhibits the NLRP3/caspase-1 axis to inhibit NLRP3 inflammasome-mediated pyroptosis [30], and an immunomodulatory effect by regulating the T-cell receptor signaling pathway and Th17 cell differentiation [31]. AS-IV can alleviate E. coli-induced peritonitis by regulating neutrophil migration [32]. However, the specific mechanism by which AS-IV prevents noise-induced deafness is still unclear.
Although AS-IV (molecular formula, C41H68O14) is a saponin with poor absorption, and the absolute bioavailability of AS-IV in rats with oral administration is 3.66% [33], the abundance of gut microorganisms and fecal metabolites is altered by AS-IV, which may contribute to their antifibrotic and cardioprotective effects [34]. One study showed that 105 dB SPL noise exposure affected gut microbiota and metabolic disorders in rats [35]. Long-term low-intensity noise exposure can increase the abundance of Firmicutes and diminish that of Bacteroidetes in intestinal flora, and it can induce cognitive decline in mice [36]. Moreover, chronic noise exposure significantly reduces the abundance of Rikenellaceae, Ruminococcaceae, Anaerobia, Lachnospira, and Odoribacter in the microbial community and significantly increases the levels of IL-6, NF-κB, iNOS, and NGAL in the mouse intestine [37].
According to a previous study, AS-IV might reduce noise-induced increases in hearing threshold by reducing inflammatory factors [38,39], and AS-IV could improve disease prognosis by ameliorating intestinal flora with optimizing intestinal metabolites [40]. However, the correlation between inflammatory factors and intestinal flora of mice in which NIHL is protected by AS-IV remains poorly understood [39,41]. Therefore, the present study aimed to initially explore the mechanism by which AS-IV reduces NIHL damage through synergistic alterations in gut microbiota and inflammation.

2. Materials and Methods

2.1. Animals Groups

Eighteen 6-week-old specific-pathogen-free male c57bl/6j mice, weighing approximately 14–16 g, were purchased from the GuangDong Medical Laboratory Animal Center (License No. SCXK 2022-0002, GDMLAC, Foshan, China). The mice were housed in the Laboratory Animal Center, Guangdong Pharmaceutical University under the following conditions: temperature of 20 ± 0.5 °C, relative humidity of 55% ± 5%, and a light/dark period of 12 h. All experimental procedures were approved by the Animal Ethics Committee of Guangdong Pharmaceutical University (No. gdpulacspf2022124). The mice were randomly divided into three groups, namely the control group, the noise-exposed group, and the AS-IV group, with six mice in each group. AS-IV (>98%, Macklin, Shanghai, China) with 0.05% sodium carboxymethylcellulose was prepared as suspension and administered by gavage at 100 mg/kg body weight once a day until the end of experimental exposure [38,39]. The 6-week-old mice were purchased and subjected to a week of environmental acclimation, with the first ABR and the beginning of exposure to noise and AS-IV at 7 weeks of age, the end of noise exposure at 8 weeks of age, and the last ABR at 10 weeks of age.

2.2. Noise Exposure and Procedures

The mice were placed in a small metal cage (7 × 7 × 10 cm) in a self-built noise exposure box (50 × 50 × 40 cm). The noise was broadband noise with a frequency of 20 Hz–20 kHz [42]. A broad-spectrum loudspeaker (FEI L0 YD3-2001; Guangzhou, China) was positioned 10 cm above the metal cage. The loudspeaker received noise transmitted from a computer and through a power amplifier for cyclic playback. The noise-exposed group was exposed to 103 dB SPL noise for 7 days in a row for 4 h a day (LEX,8h = 100.0 dB) [3]. The control mice were kept in a quiet room with less than 40 dB SPL, while other conditions were the same as those in the noise-exposed group.

2.3. Auditory Brainstem Response (ABR) Audiometry

Aiming to determine the hearing level, the hearing thresholds of the c57 mice were measured before noise exposure and 14 days after the end of exposure. The c57 mice were anesthetized by intraperitoneal injection of 40 mg/kg of 1% sodium pentobarbital. ABR audiometry was conducted using the Neuro-Audio device from Neurosoft (Ivanovo, Russia) and was recorded using the supporting system (NEURO-AUDIO.NET). The temperature of the c57 mice was maintained using a heating pad. Left and right reference electrodes were inserted subcutaneously behind each tested ear, and the recording electrodes were inserted into the vertex. The tested ears of the mice were presented with click and tone burst stimuli (4, 8, and 16 kHz) through a pair of in-ear headphones. The signal started at 100 dB; the stride was 10 dB initially and 5 dB when approaching the threshold. The results were set as 512 repetitions of the stimulus superposition waveform, the stimulus frequency was 20 times per second, the low-frequency filter was 100 Hz, and the high-frequency filter was 2000 Hz. The minimum sound stimulus intensity that could cause wave III and a reproducible waveform was defined as the response threshold to evaluate the hearing impairment of mice in each group [43,44,45].

2.4. Observation of Cochlear Hair Cells

After the mice were sacrificed, the tissues around the acoustic bulla of the temporal bone were removed, and the cochlea was isolated, fixed in 4% paraformaldehyde, and decalcified with 0.5 M EDTA to suitable hardness. The basilar membrane was dissected under a stereoscopic microscope (SZ760; Cnoptec, Chongqing, China). The isolated basilar membrane was permeabilized with 0.1% Triton X-100 (Amresco, Washington, DC, USA) for 30 min, stained with bovine serum albumin to prepare Actin-Tracker Green (Beyotime, Shanghai, China) for 70 min, and washed with phosphate-buffered saline two to three times. Pictures were taken under a benchtop microscope (EVOSTM M5000; Thermo Fisher Scientific, Waltham, MA, USA).

2.5. Analysis of Inflammatory Indicators

Blood was collected using the orbital blood collection method [46], and mouse serum was obtained after centrifugation. An ELISA kit (MEIMIAN, Yancheng, China) was used to detect TNF-α, TNF-γ, IL-6, IL-1β, and IFN-γ content in mouse serum. We created a concentration absorbance standard curve based on the standard sample and calculated the concentration of the inflammatory substances in the sample according to the standard curve.

2.6. Analysis of the Microbiota

Mouse fecal samples were obtained by stimulating the tail and stored in sterile centrifuge tubes at −80 °C, which were used to identify the 16S rRNA gene sequence. Then, the DNA of the corresponding genome was extracted following the instructions of the kit manufacturer. Integrity, purity, and concentration of DNA were determined by 1% agarose gel electrophoresis and an ultramicrophotometer. Polymerase chain reaction was used to amplify genomic DNA. A database was constructed and sequenced using the previous methods [47]. Then, based on valid sequencing data, Operational Taxonomic Uni clustering and species analysis were conducted, and annotation was performed to obtain species information and distribution of the microbiota in each group. The microbiota histogram was used to analyze the composition of the microbiota at the phylum, class, order, family, and other levels.

2.7. Statistical Analysis

All data were analyzed using GraphPad Prism version 9.0.0 for Windows (GraphPad Software, La Jolla, CA, USA) and expressed as the mean ± standard deviation. Analysis of single-factor differences between groups was achieved using one-way analysis of variance, Brown–Forsythe and Welch ANOVA tests. Multiple pairwise comparisons were based on the least-significant-difference method. Correlation analysis was conducted using the Spearman test. The significance level was set at α = 0.05.

3. Results

3.1. AS-IV Improves Hearing in Noise-Exposed Mice

We assessed the level of hearing in the mice using ABR. The hearing thresholds of the mice increased after noise exposure (click: 21.3 dB vs. 42.5 dB, p < 0.0001; 4 kHz: 26.9 dB vs. 54.4 dB, p < 0.0001; 8 kHz: 23.8 dB vs. 37.7 dB, p < 0.0001; and 16 kHz: 27.1 dB vs. 43.3 dB, p = 0.0002, indicating increases of 21.2 dB, 27.5 dB, 13.9 dB, and 16.2 dB, respectively). After taking AS-IV, the hearing threshold of the mice was lower than that of the noise-exposed group at click, 4 kHz, 8 kHz, and 16 kHz.
The hearing thresholds of the mice in the AS-IV group at click, 4 kHz, 8 kHz, and 16 kHz were 13.1 dB, 16.4 dB, 11.7 dB, and 10.8 dB, respectively, which were lower than those in the noise-exposed group (p < 0.05) (Table 1).

3.2. AS-IV Reduces the Loss of OHCs in Noise-Exposed Mice

The OHCs of the control group mice were arranged neatly without any cell loss. After 14 days of noise exposure, the cochlear OHCs of the noise-exposed group showed significant deformation; the damage to the OHCs (Figure 1) was more serious in the noise group, whereas the damage to OHCs giving AS-IV was small or absent. The OHC loss in the mice from the AS-IV group was lower than that in the noise-exposed group. Combined with the ABR results, our findings indicate that AS-IV can alleviate hearing loss.

3.3. AS-IV Reduces the Levels of Inflammatory Factors in Noise-Exposed Mice

The levels of TNF-α, TNF-γ, IL-6, IL-1β, and IFN- γ were significantly increased in the noise-exposed group compared with the control group (by 158.7%, 78.5%, 45.8%, 42.1%, and 38.8%, respectively; p < 0.05). After treatment with AS-IV, the levels of TNF-α, TNF-γ, and IL-1β were significantly decreased compared with those in the noise-exposed group (p < 0.05) (Table 2).

3.4. AS-IV Alleviates the Disorder of Intestinal Flora in Noise-Exposed Mice

The dominant microbiota in the mice were Firmicutes (50.1%), Bacteroidetes (40.7%), Verrucomicrobia (5.1%), Epsilonbacteraeota (2.5%), and Proteobacteria (1.3%). Noise exposure resulted in decreases in the abundance of phylum Firmicutes, phylum Bacteroidetes, class Bacilli, order Lactobacillales, order Bacteroidales, and family Lactobacillaceae (p < 0.05). However, administration of AS-IV increased the abundance levels of phylum Firmicutes, class Bacilli, order Lactobacillales, and family Lactobacillaceae in the intestine of the noise-exposed mice (p < 0.05) (Figure 2A–D).
Using principal coordinate analysis (PCoA) to evaluate the overall differences in the gut microbiota structure of the mice caused by noise exposure, principal coordinates 1 and 2 explained 54.1% and 22.8% of the Bray–Curtis differences, respectively. The difference in the microbiota structure was statistically significant between the noise-exposed group and control group and between the noise group and the noise + AS-IV group. The difference between the control group and the noise + AS-IV group was not significant (Figure 2E).
The results of linear discriminant analysis (LDA > 2) showed that in the control group, the main enriched bacterial class was Deferribacteria, and the main bacterial species was Mucispirillum_Schaedeleri. The main enriched bacterial phyla in the noise group were Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria. The main enriched bacterial species in the noise group were Ruminococcaceae_bacterium_Marseille, Alistipes_sp, and gut_metagenome. The species Lachnospiraceae_Bacterium_A4 of the phylum Bacteroidetes and the species Butyricimonas_synergistica of the phylum Firmicutes were enriched in the noise + AS-IV group (Figure 2F).

3.5. Correlation Analysis between Gut Microbiota and Inflammatory Factors

We generated a correlation matrix through the Spearman correlation coefficient and selected the top five bacterial communities in class, order, family, genus, and species for correlation analysis with mouse serum inflammatory indicators. Among them, the order Lactobacillales, family Lactobacillaceae, genus Lactobacillus, and class Bacilli were associated with serum TNF-α, TNF-γ, and IL-1β. There was a strong negative correlation of Erysipelotrichia, Erysipelotrichales, and Erysipelotrichaceae with IFN-γ and a strong positive correlation of Clostridia and Clostridiales with IL-1β (Figure 3).

4. Discussion

We showed that AS-IV had beneficial effects on hearing, which has rarely been studied before. Namely, not only did AS-IV reduce the inflammatory levels in the noise-exposed mice and regulate their intestinal flora, but it also reduced the noise-induced loss of outer hair cells and slowed noise-induced hearing loss.
We established an NIHL mouse model by exposing the mice to 103 dB SPL of white noise for 4 h per day for 7 consecutive days and testing for relevant indicators. Furthermore, we examined whether oral administration of AS-IV altered the hearing level of the mice exposed to noise. Before noise exposure, the baseline hearing levels were determined through ABR. The average hearing thresholds of the mice at click, 4 kHz, 8 kHz, and 16 kHz were 19.7 ± 6.0 dB, 25.6 ± 6.8 dB, 16.2 ± 3.6 dB, and 16.7 ± 5.0 dB, respectively, consistent with the existing studies [48]. After noise exposure, there was a significant increase in the hearing threshold (p < 0.05). To explore the effect of noise exposure on mice HCs, we detected the cochlear HCs of the mice using immunofluorescence with phalloidin, which can bind specifically to the cytoskeleton of HCs. The results showed that the noise of this intensity caused the loss of OHCs, while there was less damage to OHCs following AS-IV treatment (Figure 1). This finding indicates that AS-IV prevents noise-induced OHC outright loss and plays a protective role in hearing [49,50].
In addition, we analyzed whether oral administration of AS-IV altered the serum levels of inflammatory factors in the mice exposed to high-intensity noise. The results showed that TNF-α, TNF-γ, IL-6, sIL-1β, and IFN-γ content in the serum increased (p < 0.05), consistent with previous research [51,52,53,54,55]. The increase in the levels of TNF-α and IL-1β reflects the activation of monocytes and macrophages, which transform into macrophages and migrate below the basilar membrane, promoting inflammation and damage in the cochlear HCs [56]. Moreover, strong noise enhances the increased production of Ca2+ through cytokine TNF-α-mediated TRPV1 induction, while Ca2+ promotes the inflammation and apoptosis of inner ear tissue and cells [55]. Moon S K et al. [57] found that IFN-γ increased the sensitivity of HCs to lower-concentration Ca2+ cytotoxicity through the JAK1/1-STAT2 signaling pathway, which further promoted apoptosis of HCs. Marshall et al. [58] studied TNF-α as a proinflammatory cytokine and its receptor, TNF-α receptor, and showed that they play a key role in cell death mechanisms, including necrotic apoptosis and apoptosis. In addition, our study showed that the increased expression levels of the proinflammatory cytokines IL-1β and IL-6 in the mice exposed to noise were attenuated after administration of AS-IV. IL-6 is a key inflammatory marker in many mouse models, including the age-related hearing loss mouse model [59] and the COPD (chronic obstructive pulmonary disease)-like inflammatory mouse model [60]. IL-6 also induces excessive production of vascular endothelial growth factor, resulting in enhanced angiogenesis and increased vascular permeability [61], which is associated with progressive hearing loss in Meniere’s disease [62]. A study of pediatric otitis patients found a large amount of inflammatory factor IL-1β in the ear effusion [63]. IL-1β is a proinflammatory cytokine mainly produced by monocytes and macrophages, and it promotes cell apoptosis [64]. Similar to this research, Sai et al. [65] found that IL-1β content in HCs was significantly increased when the miniature pigs were exposed to 120 dB. In addition, Zhang et al.’s [66] research on Meniere’s disease indicated that the significantly increased content of IL-1β was due to serum/glucocorticoid-inducible kinase-1 depletion, which led to the damage to ear OHCs and the vestibular nerve. However, AS-IV decreased the protein levels of TNF-α and IL-6 in adipocytes through the miR-21/PTEN/PI3K/AKT signaling pathway [67]. Other studies have shown that AS-IV can interact with the gut microbiota and be decomposed and used by the gut microbiota, inducing the production of various beneficial short-chain fatty acids, which can reduce the degree of systemic inflammation [39,68].
The dominant groups in the mice intestinal flora included Firmicutes (50.1%) and Bacteroidetes (40.7%). We showed that the proportions of Firmicutes, Bacilli, and Lactobacillales decreased (p < 0.05) in mice after noise exposure, while the proportions of Firmicutes, Bacilli, and Lactobacillales increased (p < 0.05) and the proportions of Bacteroidetes and Bacteroidales decreased (p < 0.05) in the noise-exposed mice treated with AS-IV. Therefore, the preventive effect of AS-IV was partially achieved by reversing the structure of the intestinal microbiota. The gut microbiota of healthy humans mainly consists of Firmicutes (49%) and Bacteroidetes (23%), where Firmicutes mainly include Clostridia, Bacilli, and Mollicutes, and Bacteroidetes mainly include Bacteroidales and Flavobacteriaceae [69]. It has been shown that dietary factors have a significant impact on Firmicutes, and Firmicutes can also produce butyrate salts, which reduce the level of inflammation in the body [70]. Bacilli contain Bacillales and Lactobacillales, and Lactobacillales were shown to play an important role in the microbial community in this study. Our experiment showed that the relative abundance of Lactobacillales decreased after noise exposure. Lactobacillales are probiotics that mediate the metabolism of tryptophan into norharman and inhibit M1-type macrophages [71]. Qiulan et al. [72] found an increased abundance of AI-IV and increased content of butyric acid and valeric acid in the intestine to improve slow transit constipation. Our research results showed that Lactobacillales returned to the level of the control group without any abnormal increase when taking AS-IV, and did not cause microbial community disorders [73]. Bacteroidales is one of the most abundant members of the mammalian gut microbiota and an important microbiota for synthesizing sphingolipids in the intestine [74]. Moreover, sphingolipids mediate metabolic and immune signaling events related to chronic inflammatory diseases such as autoimmune and chronic enteritis [75]. Bacteroidales mainly colonize the colon and use dietary oligosaccharides such as arabinose and high galacturonic acid [76]. Their survival and reproduction in the intestine depend on the monosaccharides they use and their interactions with butyrate salts [77]. The changes in Bacteroidales abundance in this study may be related to the alteration in intestinal butyrate content caused by AS-IV administration [72].
Furthermore, to explore the specific mechanisms, Spearman correlation coefficients were used to investigate whether there was a correlation between inflammatory factors in the serum of the noise-exposed mice and their gut microbiota. Our results indicated that Lactobacillus negatively correlated with the mouse serum inflammatory factors TNF-α, TNF-γ, and IL-1β (R = −0.59, p = 0.012; R = −0.66, p = 0.004; and R = −0.63, p = 0.006). TNF-α and TNF-γ are proinflammatory cytokines secreted by macrophages, monocytes, and T lymphocytes, which have the effects of promoting cell apoptosis and inflammation through the activation of transcription factor NF-κB [78]. Lactobacillales are probiotics that can inhibit the release of proinflammatory cytokines (such as TNF-α) and promote the release of anti-inflammatory factors such as IL-10 [79]. TNF-α positively correlates with sensorineural hearing loss [59]. A retrospective cohort study suggested that inhibiting treatment with AS-IV can improve or even restore patient hearing [80]. This is because TNF-α blockers can reduce cellular inflammatory response, reduce cochlear HC apoptosis, and alleviate hearing loss [81]. After the use of AS-IV in this study, the abundance of Lactobacillales increased, and inflammatory mediators decreased, providing a protective effect on the body. It has been speculated that AS-IV may exert systemic anti-inflammatory and antioxidant effects by increasing intestinal volume, thereby protecting hearing [82]. Our results also showed a positive correlation between Bacteroidales and serum IL-6 (R = 0.57, p = 0.015), consistent with some previous studies [83]. On the one hand, Bacteroidales cause intestinal T cells and macrophages to produce IL-6 through the MyD88 pathway [84], inducing systemic and local inflammation [85,86]. On the other hand, Bacteroidales recruit colonic epithelial lymphocytes to maintain intestinal epithelial barrier function [84]. Our findings suggest that noise can cause an increase in harmful gut microbiota. The use of AS-IV in this experiment significantly reduced the increasing trend of Bacteroidetes. It could be speculated that AS-IV may reduce the level of IL-6 in the body by reducing the abundance of Bacteroidetes, thereby protecting the hearing system from damage.

5. Conclusions

Our study showed that AS-IV intervention in the intestinal microbiota of the noise-exposed mice correlated with inflammatory factors, showing a positive correlation of Bacteroidales with IL-6 (R = 0.57, p = 0.015) and a negative correlation with TNF-α and IL-1β (R = −0.59, p = 0.012, and R = −0.63, p = 0.006). We confirmed that AS-IV protected the noise-exposed mice against hearing loss (p < 0.05) by reducing the inflammatory indicators in the circulatory system, which was correlated with gut microbiota. Therefore, we hypothesize that AS-IV relies on changes in gut microbiota abundance to reduce inflammation levels in the body and protect the hearing system from noise damage, which provides a reference for the application of AS-IV in the prevention and treatment of some noise-induced diseases.

Author Contributions

J.L.: Methodology, Writing—original draft. J.Y.: Methodology, Writing—original draft. Y.X. (Yun Xia): Writing—original draft. J.W.: Conceptualization, Supervision. Y.X. (Yuan Xia): Conceptualization, Supervision, Writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the Health Commission of Guangdong Province in China (No. A2021195 to YX).

Institutional Review Board Statement

All mice work was approved by the Ethical Review Committee of Guangdong Pharmaceutical University, China (No. gdpulacspf2022124).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in the article.

Conflicts of Interest

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.

References

  1. Natarajan, N.; Batts, S.; Stankovic, K.M. Noise-Induced Hearing Loss. J. Clin. Med. 2023, 12, 2347. [Google Scholar] [CrossRef]
  2. Daniel, E. Noise and hearing loss: A review. J. Sch. Health 2007, 77, 225–231. [Google Scholar] [CrossRef]
  3. Lie, A.; Skogstad, M.; Johannessen, H.A.; Tynes, T.; Mehlum, I.S.; Nordby, K.C.; Engdahl, B.; Tambs, K. Occupational noise exposure and hearing: A systematic review. Int. Arch. Occup. Environ. Health 2016, 89, 351–372. [Google Scholar] [CrossRef]
  4. Liu, S.; Zou, H.; Lei, S.; Xin, J.; Qian, P.; Liu, Y.; Chen, Y.; Yu, K.; Zhang, M. The role of kurtosis and kurtosis-adjusted energy metric in occupational noise-induced hearing loss among metal manufacturing workers. Front. Public Health 2023, 11, 1159348. [Google Scholar] [CrossRef]
  5. Mutlu, A.; Ocal, F.C.A.; Erbek, S.; Ozluoglu, L. The protective effect of adrenocorticotropic hormone treatment against noise-induced hearing loss. Auris Nasus Larynx 2018, 45, 929–935. [Google Scholar] [CrossRef]
  6. Müller, M.; Tisch, M.; Maier, H.; Löwenheim, H. Reduction of permanent hearing loss by local glucocorticoid application : Guinea pigs with acute acoustic trauma. Hno 2017, 65 (Suppl. S1), 59–67. [Google Scholar] [CrossRef]
  7. Soyaliç, H.; Gevrek, F.; Karaman, S. Curcumin protects against acoustic trauma in the rat cochlea. Int. J. Pediatr. Otorhinolaryngol. 2017, 99, 100–106. [Google Scholar] [CrossRef]
  8. Seidman, M.D.; Tang, W.; Bai, V.U.; Ahmad, N.; Jiang, H.; Media, J.; Patel, N.; Rubin, C.J.; Standring, R.T. Resveratrol decreases noise-induced cyclooxygenase-2 expression in the rat cochlea. Otolaryngol. Head Neck Surg. 2013, 148, 827–833. [Google Scholar] [CrossRef]
  9. Yamashita, D.; Jiang, H.-Y.; Le Prell, C.; Schacht, J.; Miller, J. Post-exposure treatment attenuates noise-induced hearing loss. Neuroscience 2005, 134, 633–642. [Google Scholar] [CrossRef]
  10. Ahn, J.H.; Shin, J.E.; Chung, B.Y.; Lee, H.M.; Kang, H.H.; Chung, J.W.; Pak, J.H. Involvement of retinoic acid-induced peroxiredoxin 6 expression in recovery of noise-induced temporary hearing threshold shifts. Environ. Toxicol. Pharmacol. 2013, 36, 463–471. [Google Scholar] [CrossRef]
  11. Staecker, H.; Jokovic, G.; Karpishchenko, S.; Kienle-Gogolok, A.; Krzyzaniak, A.; Lin, C.D.; Navratil, P.; Tzvetkov, V.; Wright, N.; Meyer, T. Efficacy and Safety of AM-111 in the Treatment of Acute Unilateral Sudden Deafness-A Double-blind, Randomized, Placebo-controlled Phase 3 Study. Otol. Neurotol. 2019, 40, 584–594. [Google Scholar] [CrossRef]
  12. Koch, M.; Eßinger, T.M.; Stoppe, T.; Lasurashvili, N.; Bornitz, M.; Zahnert, T. Fully implantable hearing aid in the incudostapedial joint gap. Hear. Res. 2016, 340, 169–178. [Google Scholar] [CrossRef]
  13. Frosolini, A.; Badin, G.; Sorrentino, F.; Brotto, D.; Pessot, N.; Fantin, F.; Ceschin, F.; Lovato, A.; Coppola, N.; Mancuso, A.; et al. Application of Patient Reported Outcome Measures in Cochlear Implant Patients: Implications for the Design of Specific Rehabilitation Programs. Sensors 2022, 22, 8770. [Google Scholar] [CrossRef]
  14. Groves, A.K. The challenge of hair cell regeneration. Exp. Biol. Med. 2010, 235, 434–446. [Google Scholar] [CrossRef]
  15. Gittleman, S.N.; Le Prell, C.G.; Hammill, T.L. Octave band noise exposure: Laboratory models and otoprotection efforts. J. Acoust. Soc. Am. 2019, 146, 3800. [Google Scholar] [CrossRef]
  16. Kujawa, S.G.; Liberman, M.C. Synaptopathy in the noise-exposed and aging cochlea: Primary neural degeneration in acquired sensorineural hearing loss. Hear. Res. 2015, 330 Pt B, 191–199. [Google Scholar] [CrossRef]
  17. Kujawa, S.G.; Liberman, M.C. Adding insult to injury: Cochlear nerve degeneration after “temporary” noise-induced hearing loss. J. Neurosci. 2009, 29, 14077–14085. [Google Scholar] [CrossRef]
  18. Mizushima, Y.; Fujimoto, C.; Kashio, A.; Kondo, K.; Yamasoba, T. Macrophage recruitment, but not interleukin 1 beta activation, enhances noise-induced hearing damage. Biochem. Biophys. Res. Commun. 2017, 493, 894–900. [Google Scholar] [CrossRef]
  19. Möhrle, D.; Reimann, K.; Wolter, S.; Wolters, M.; Varakina, K.; Mergia, E.; Eichert, N.; Geisler, H.S.; Sandner, P.; Ruth, P.; et al. NO-Sensitive Guanylate Cyclase Isoforms NO-GC1 and NO-GC2 Contribute to Noise-Induced Inner Hair Cell Synaptopathy. Mol. Pharmacol. 2017, 92, 375–388. [Google Scholar] [CrossRef]
  20. Fan, B.; Lu, F.; Du, W.J.; Chen, J.; An, X.G.; Wang, R.F.; Li, W.; Song, Y.L.; Zha, D.J.; Chen, F.Q. PTEN inhibitor bisperoxovanadium protects against noise-induced hearing loss. Neural Regen. Res. 2023, 18, 1601–1606. [Google Scholar]
  21. Wang, X.; Zhu, Y.; Long, H.; Pan, S.; Xiong, H.; Fang, Q.; Hill, K.; Lai, R.; Yuan, H.; Sha, S.H. Mitochondrial Calcium Transporters Mediate Sensitivity to Noise-Induced Losses of Hair Cells and Cochlear Synapses. Front. Mol. Neurosci. 2018, 11, 469. [Google Scholar] [CrossRef] [PubMed]
  22. Wu, F.; Xiong, H.; Sha, S. Noise-induced loss of sensory hair cells is mediated by ROS/AMPKα pathway. Redox Biol. 2020, 29, 101406. [Google Scholar] [CrossRef]
  23. Liberman, M.C.; Dodds, L.W. Single-neuron labeling and chronic cochlear pathology. III. Stereocilia damage and alterations of threshold tuning curves. Hear. Res. 1984, 16, 55–74. [Google Scholar] [CrossRef] [PubMed]
  24. Liberman, M.C.; Beil, D.G. Hair cell condition and auditory nerve response in normal and noise-damaged cochleas. Acta Oto-Laryngol. 1979, 88, 161–176. [Google Scholar] [CrossRef]
  25. Nordmann, A.S.; Bohne, B.A.; Harding, G.W. Histopathological differences between temporary and permanent threshold shift. Hear. Res. 2000, 139, 13–30. [Google Scholar] [CrossRef]
  26. Liberman, M.C.; Dodds, L.W. Single-neuron labeling and chronic cochlear pathology. II. Stereocilia damage and alterations of spontaneous discharge rates. Hear. Res. 1984, 16, 43–53. [Google Scholar] [CrossRef] [PubMed]
  27. Kujawa, S.G.; Liberman, M.C. Translating animal models to human therapeutics in noise-induced and age-related hearing loss. Hear. Res. 2019, 377, 44–52. [Google Scholar] [CrossRef] [PubMed]
  28. Xiong, M.; He, Q.; Lai, H.; Wang, J. Astragaloside IV inhibits apoptotic cell death in the guinea pig cochlea exposed to impulse noise. Acta Oto-Laryngol. 2012, 132, 467–474. [Google Scholar] [CrossRef]
  29. Yang, K.; Xie, Q.; Tang, T.; Zhao, N.; Liang, J.; Shen, Y.; Li, Z.; Liu, B.; Chen, J.; Cheng, W.; et al. Astragaloside IV as a novel CXCR4 antagonist alleviates osteoarthritis in the knee of monosodium iodoacetate-induced rats. Phytomedicine 2023, 108, 154506. [Google Scholar] [CrossRef]
  30. Huang, D.; Shi, S.; Wang, Y.; Wang, X.; Shen, Z.; Wang, M.; Pei, C.; Wu, Y.; He, Y.; Wang, Z. Astragaloside IV alleviates PM2.5-caused lung toxicity by inhibiting inflammasome-mediated pyroptosis via NLRP3/caspase-1 axis inhibition in mice. Biomed. Pharmacother. 2022, 150, 112978. [Google Scholar] [CrossRef]
  31. Weng, S.; Huang, L.; Cai, B.; He, L.; Wen, S.; Li, J.; Zhong, Z.; Zhang, H.; Huang, C.; Yang, Y.; et al. Astragaloside IV ameliorates experimental autoimmune myasthenia gravis by regulating CD4+ T cells and altering gut microbiota. Chin. Med. 2023, 18, 97. [Google Scholar] [CrossRef]
  32. Huang, P.; Lu, X.; Yuan, B.; Liu, T.; Dai, L.; Liu, Y.; Yin, H. Astragaloside IV alleviates E. coli-caused peritonitis via upregulation of neutrophil influx to the site of infection. Int. Immunopharmacol. 2016, 39, 377–382. [Google Scholar] [CrossRef]
  33. Zhang, W.; Zhang, C.; Liu, R.; Li, H.; Zhang, J.; Mao, C.; Chen, C. Quantitative determination of Astragaloside IV, a natural product with cardioprotective activity, in plasma, urine and other biological samples by HPLC coupled with tandem mass spectrometry. J. Chromatogr. B 2005, 822, 170–177. [Google Scholar] [CrossRef]
  34. Du, X.Q.; Shi, L.P.; Chen, Z.W.; Hu, J.Y.; Zuo, B.; Xiong, Y.; Cao, W.F. Astragaloside IV Ameliorates Isoprenaline-Induced Cardiac Fibrosis in Mice via Modulating Gut Microbiota and Fecal Metabolites. Front. Cell. Infect. Microbiol. 2022, 12, 836150. [Google Scholar] [CrossRef]
  35. Li, N.; Zhang, X.; Cui, Y.; Wu, H.; Yu, Y.; Yu, S. Dysregulations of metabolites and gut microbes and their associations in rats with noise induced hearing loss. Front. Microbiol. 2023, 14, 1229407. [Google Scholar] [CrossRef]
  36. Cui, B.; Su, D.; Li, W.; She, X.; Zhang, M.; Wang, R.; Zhai, Q. Effects of chronic noise exposure on the microbiome-gut-brain axis in senescence-accelerated prone mice: Implications for Alzheimer’s disease. J. Neuroinflamm. 2018, 15, 190. [Google Scholar] [CrossRef]
  37. Chi, H.; Cao, W.; Zhang, M.; Su, D.; Yang, H.; Li, Z.; Li, C.; She, X.; Wang, K.; Gao, X.; et al. Environmental noise stress disturbs commensal microbiota homeostasis and induces oxi-inflammmation and AD-like neuropathology through epithelial barrier disruption in the EOAD mouse model. J. Neuroinflamm. 2021, 18, 9. [Google Scholar] [CrossRef]
  38. Xiong, M.; Lai, H.; He, Q.; Wang, J. Astragaloside IV attenuates impulse noise-induced trauma in guinea pig. Acta Oto-Laryngol. 2011, 131, 809–816. [Google Scholar] [CrossRef]
  39. Li, Z.; Hu, E.; Zheng, F.; Wang, S.; Zhang, W.; Luo, J.; Tang, T.; Huang, Q.; Wang, Y. The effects of astragaloside IV on gut microbiota and serum metabolism in a mice model of intracerebral hemorrhage. Phytomedicine 2023, 121, 155086. [Google Scholar] [CrossRef]
  40. Wu, S.; Wen, F.; Zhong, X.; Du, W.; Chen, M.; Wang, J. Astragaloside IV ameliorate acute alcohol-induced liver injury in mice via modulating gut microbiota and regulating NLRP3/caspase-1 signaling pathway. Ann. Med. 2023, 55, 2216942. [Google Scholar] [CrossRef]
  41. Liang, Y.; Chen, B.; Liang, D.; Quan, X.; Gu, R.; Meng, Z.; Gan, H.; Wu, Z.; Sun, Y.; Liu, S.; et al. Pharmacological Effects of Astragaloside IV: A Review. Molecules 2023, 28, 6118. [Google Scholar] [CrossRef]
  42. Lai, R.; Fang, Q.; Wu, F.; Pan, S.; Haque, K.; Sha, S.H. Prevention of noise-induced hearing loss by calpain inhibitor MDL-28170 is associated with upregulation of PI3K/Akt survival signaling pathway. Front. Cell. Neurosci. 2023, 17, 1199656. [Google Scholar] [CrossRef]
  43. Fulian, W.; Xuying, J.; Shuang, F.; Changzhi, S. The Changes of the Electrophysiological Characteristics in the Pathway from Auditory Nerve to Cochlear Nucleus after Noise-exposure in Mouse. J. Audiol. Speech Pathol. 2023, 31, 349–354. [Google Scholar]
  44. Duque, D.; Pais, R.; Malmierca, M.S. Stimulus-specific adaptation in the anesthetized mouse revealed by brainstem auditory evoked potentials. Hear. Res. 2018, 370, 294–301. [Google Scholar] [CrossRef]
  45. Nieto-Diego, J.; Malmierca, M.S. Topographic Distribution of Stimulus-Specific Adaptation across Auditory Cortical Fields in the Anesthetized Rat. PLoS Biol. 2016, 14, e1002397. [Google Scholar] [CrossRef]
  46. Whittaker, A.L.; Barker, T.H. The Impact of Common Recovery Blood Sampling Methods, in Mice (Mus Musculus), on Well-Being and Sample Quality: A Systematic Review. Animals 2020, 10, 989. [Google Scholar] [CrossRef]
  47. Xia, Y.; Wang, C.; Zhang, X.; Li, J.; Li, Z.; Zhu, J.; Zhou, Q.; Yang, J.; Chen, Q.; Meng, X. Combined effects of lead and manganese on locomotor activity and microbiota in zebrafish. Ecotoxicol. Environ. Saf. 2023, 263, 115260. [Google Scholar] [CrossRef]
  48. Kennedy, C.L.; Shuster, B.; Amanipour, R.; Milon, B.; Patel, P.; Elkon, R.; Hertzano, R. Metformin Protects Against Noise-Induced Hearing Loss in Male Mice. Otol. Neurotol. 2023, 44, 956–963. [Google Scholar] [CrossRef] [PubMed]
  49. Yoshida, N.; Hequembourg, S.J.; Atencio, C.A.; Rosowski, J.J.; Liberman, M.C. Acoustic injury in mice: 129/SvEv is exceptionally resistant to noise-induced hearing loss. Hear. Res. 2000, 141, 97–106. [Google Scholar] [CrossRef] [PubMed]
  50. Suzuki, J.; Hemmi, T.; Maekawa, M.; Watanabe, M.; Inada, H.; Ikushima, H.; Oishi, T.; Ikeda, R.; Honkura, Y.; Kagawa, Y.; et al. Fatty acid binding protein type 7 deficiency preserves auditory function in noise-exposed mice. Sci. Rep. 2023, 13, 21494. [Google Scholar] [CrossRef] [PubMed]
  51. Paciello, F.; Pisani, A.; Rolesi, R.; Escarrat, V.; Galli, J.; Paludetti, G.; Grassi, C.; Troiani, D.; Fetoni, A.R. Noise-Induced Cochlear Damage Involves PPAR Down-Regulation through the Interplay between Oxidative Stress and Inflammation. Antioxidants 2021, 10, 1188. [Google Scholar] [CrossRef] [PubMed]
  52. Li, S.; Zheng, H.; Xing, Z.; Liu, Y.; Han, L.; Wang, Z.; Yu, L. The circadian timing of noise exposure influences noise-induced inflammatory responses in the mouse cochlea. Braz. J. Otorhinolaryngol. 2022, 88 (Suppl. S3), S1–S8. [Google Scholar] [CrossRef] [PubMed]
  53. Paciello, F.; Di Pino, A.; Rolesi, R.; Troiani, D.; Paludetti, G.; Grassi, C.; Fetoni, A.R. Anti-oxidant and anti-inflammatory effects of caffeic acid: In vivo evidences in a model of noise-induced hearing loss. Food Chem. Toxicol. 2020, 143, 111555. [Google Scholar] [CrossRef] [PubMed]
  54. Shin, S.A.; Lyu, A.R.; Jeong, S.H.; Kim, T.H.; Park, M.J.; Park, Y.H. Acoustic Trauma Modulates Cochlear Blood Flow and Vasoactive Factors in a Rodent Model of Noise-Induced Hearing Loss. Int. J. Mol. Sci. 2019, 20, 5316. [Google Scholar] [CrossRef] [PubMed]
  55. Dhukhwa, A.; Bhatta, P.; Sheth, S.; Korrapati, K.; Tieu, C.; Mamillapalli, C.; Ramkumar, V.; Mukherjea, D. Targeting Inflammatory Processes Mediated by TRPVI and TNF-α for Treating Noise-Induced Hearing Loss. Front. Cell. Neurosci. 2019, 13, 444. [Google Scholar] [CrossRef] [PubMed]
  56. Bae, S.H.; Yoo, J.E.; Hong, J.W.; Park, H.R.; Noh, B.; Kim, H.; Kang, M.; Hyun, Y.M.; Gee, H.Y.; Choi, J.Y.; et al. LCCL peptide cleavage after noise exposure exacerbates hearing loss and is associated with the monocyte infiltration in the cochlea. Hear. Res. 2021, 412, 108378. [Google Scholar] [CrossRef] [PubMed]
  57. Moon, S.K.; Woo, J.I.; Lim, D.J. Involvement of TNF-α and IFN-γ in Inflammation-Mediated Cochlear Injury. Ann. Otol. Rhinol. Laryngol. 2019, 128 (Suppl. S6), 8s–15s. [Google Scholar] [CrossRef]
  58. Marshall, K.D.; Baines, C.P. Necroptosis: Is there a role for mitochondria? Front. Physiol. 2014, 5, 323. [Google Scholar] [CrossRef]
  59. Liu, J.; Chen, H.; Lin, X.; Zhu, X.; Huang, J.; Xu, W.; Tan, M.; Su, J. Melatonin Suppresses Cyclic GMP-AMP Synthase-Stimulator of Interferon Genes Signaling and Delays the Development of Hearing Loss in the C57BL/6J Presbycusis Mouse Model. Neuroscience 2023, 517, 84–95. [Google Scholar] [CrossRef]
  60. Ochoa, C.E.; Mirabolfathinejad, S.G.; Ruiz, V.A.; Evans, S.E.; Gagea, M.; Evans, C.M.; Dickey, B.F.; Moghaddam, S.J. Interleukin 6, but not T helper 2 cytokines, promotes lung carcinogenesis. Cancer Prev. Res. 2011, 4, 51–64. [Google Scholar] [CrossRef]
  61. Hashizume, M.; Hayakawa, N.; Suzuki, M.; Mihara, M. IL-6/sIL-6R trans-signalling, but not TNF-alpha induced angiogenesis in a HUVEC and synovial cell co-culture system. Rheumatol. Int. 2009, 29, 1449–1454. [Google Scholar] [CrossRef] [PubMed]
  62. Huang, C.; Wang, Q.; Pan, X.; Li, W.; Liu, W.; Jiang, W.; Huang, L.; Peng, A.; Zhang, Z. Up-Regulated Expression of Interferon-Gamma, Interleukin-6 and Tumor Necrosis Factor-Alpha in the Endolymphatic Sac of Meniere’s Disease Suggesting the Local Inflammatory Response Underlies the Mechanism of This Disease. Front. Neurol. 2022, 13, 781031. [Google Scholar] [CrossRef] [PubMed]
  63. Liu, S.; Guo, L.; Chen, M.; Liu, W.; Li, Y.; Wang, X.; Li, S.; Zhang, J.; Ni, X. Evaluation of Caspase-1, Interleukin-1β, and Interleukin-18, in the Middle Ear Effusion in Children with Otitis Media with Effusion. Front. Pediatr. 2021, 9, 732973. [Google Scholar] [CrossRef]
  64. Wang, P.; Qian, H.; Xiao, M.; Lv, J. Role of signal transduction pathways in IL-1β-induced apoptosis: Pathological and therapeutic aspects. Immun. Inflamm. Dis. 2023, 11, e762. [Google Scholar] [CrossRef] [PubMed]
  65. Sai, N.; Yang, Y.Y.; Ma, L.; Liu, D.; Jiang, Q.Q.; Guo, W.W.; Han, W.J. Involvement of NLRP3-inflammasome pathway in noise-induced hearing loss. Neural Regen. Res. 2022, 17, 2750–2754. [Google Scholar] [PubMed]
  66. Zhang, D.G.; Yu, W.Q.; Liu, J.H.; Kong, L.G.; Song, Y.D.; Li, X.F.; Fan, Z.M.; Lyu, Y.F.; Li, N.; Wang, H.-B. Serum/glucocorticoid-inducible kinase 1 deficiency induces NLRP3 inflammasome activation and autoinflammation of macrophages in a murine endolymphatic hydrops model. Nat. Commun. 2023, 14, 1249. [Google Scholar] [CrossRef] [PubMed]
  67. Guo, X.; Yin, T.; Chen, D.; Xu, S.; Ye, R.; Zhang, Y. Astragaloside IV Regulates Insulin Resistance and Inflammatory Response of Adipocytes via Modulating MIR-21/PTEN/PI3K/AKT Signaling. Endocr. Metab. Immune Disord. Drug Targets 2023, 23, 1538–1547. [Google Scholar] [CrossRef]
  68. Zhao, Z.X.; Tang, X.H.; Jiang, S.L.; Pang, J.Q.; Xu, Y.B.; Yuan, D.D.; Zhang, L.L.; Liu, H.M.; Fan, Q. Astragaloside IV improves the pharmacokinetics of febuxostat in rats with hyperuricemic nephropathy by regulating urea metabolism in gut microbiota. Front. Pharmacol. 2022, 13, 1031509. [Google Scholar] [CrossRef]
  69. Jandhyala, S.M.; Talukdar, R.; Subramanyam, C.; Vuyyuru, H.; Sasikala, M.; Nageshwar Reddy, D. Role of the normal gut microbiota. World J. Gastroenterol. 2015, 21, 8787–8803. [Google Scholar] [CrossRef]
  70. Long, X.; Mu, S.; Zhang, J.; Xiang, H.; Wei, W.; Sun, J.; Kuang, Z.; Yang, Y.; Chen, Y.; Zhao, H.; et al. Global signatures of the microbiome and metabolome during hospitalization of septic patients. Shock 2023, 59, 716–724. [Google Scholar] [CrossRef]
  71. Zhou, Q.; Tao, X.; Guo, F.; Wu, Y.; Deng, D.; Lv, L.; Dong, D.; Shang, D.; Xiang, H. Tryptophan metabolite norharman secreted by cultivated Lactobacillus attenuates acute pancreatitis as an antagonist of histone deacetylases. BMC Med. 2023, 21, 329. [Google Scholar] [CrossRef] [PubMed]
  72. He, Q.; Han, C.; Huang, L.; Yang, H.; Hu, J.; Chen, H.; Dou, R.; Ren, D.; Lin, H. Astragaloside IV alleviates mouse slow transit constipation by modulating gut microbiota profile and promoting butyric acid generation. J. Cell. Mol. Med. 2020, 24, 9349–9361. [Google Scholar] [CrossRef]
  73. Liang, X.; Li, Y.; Cheng, L.; Wu, Y.; Wu, T.; Wen, J.; Huang, D.; Liao, Z.; Tan, C.; Luo, Y.; et al. Gut microbiota dysbiosis characterized by abnormal elevation of Lactobacillus in patients with immune-mediated necrotizing myopathy. Front. Cell. Infect. Microbiol. 2023, 13, 1243512. [Google Scholar] [CrossRef] [PubMed]
  74. Brown, E.M.; Ke, X.; Hitchcock, D.; Jeanfavre, S.; Avila-Pacheco, J.; Nakata, T.; Arthur, T.D.; Fornelos, N.; Heim, C.; Franzosa, E.A.; et al. Bacteroides-Derived Sphingolipids Are Critical for Maintaining Intestinal Homeostasis and Symbiosis. Cell Host Microbe 2019, 25, 668–680.e7. [Google Scholar] [CrossRef] [PubMed]
  75. Maceyka, M.; Spiegel, S. Sphingolipid metabolites in inflammatory disease. Nature 2014, 510, 58–67. [Google Scholar] [CrossRef] [PubMed]
  76. Patnode, M.L.; Beller, Z.W.; Han, N.D.; Cheng, J.; Peters, S.L.; Terrapon, N.; Henrissat, B.; Le Gall, S.; Saulnier, L.; Hayashi, D.K.; et al. Interspecies Competition Impacts Targeted Manipulation of Human Gut Bacteria by Fiber-Derived Glycans. Cell 2019, 179, 59–73.e13. [Google Scholar] [CrossRef] [PubMed]
  77. Park, S.-Y.; Rao, C.; Coyte, K.Z.; Kuziel, G.A.; Zhang, Y.; Huang, W.; Franzosa, E.A.; Weng, J.-K.; Huttenhower, C.; Rakoff-Nahoum, S. Strain-level fitness in the gut microbiome is an emergent property of glycans and a single metabolite. Cell 2022, 185, 513–529.e21. [Google Scholar] [CrossRef]
  78. Aggarwal, B.B.; Gupta, S.C.; Kim, J.H. Historical perspectives on tumor necrosis factor and its superfamily: 25 years later, a golden journey. Blood 2012, 119, 651–665. [Google Scholar] [CrossRef]
  79. Sarin, S.K.; Pande, A.; Schnabl, B. Microbiome as a therapeutic target in alcohol-related liver disease. J. Hepatol. 2019, 70, 260–272. [Google Scholar] [CrossRef]
  80. Wu, B.; Luo, Y.; Wu, D.; Wang, Y.; Shen, M. Phenotypic and genotypic characterization of Chinese adult patients with NLRP3-associated autoinflammatory disease with hearing loss. Rheumatology 2023, 1–9. [Google Scholar] [CrossRef]
  81. Koçdor, P.; Özkan, E.; Akpunar, F.; Hızal, E.; Özdemir, Y.G. Protective Effects of Infliximab Against Kanamycin-Induced Ototoxicity in Rats. Otol. Neurotol. 2023, 44, e463–e470. [Google Scholar] [CrossRef] [PubMed]
  82. Gu, J.; Sun, R.; Wang, Q.; Liu, F.; Tang, D.; Chang, X. Standardized Astragalus Mongholicus Bunge-Curcuma Aromatica Salisb. Extract Efficiently Suppresses Colon Cancer Progression Through Gut Microbiota Modification in CT26-Bearing Mice. Front. Pharmacol. 2021, 12, 714322. [Google Scholar] [CrossRef] [PubMed]
  83. Smith, R.P.; Easson, C.; Lyle, S.M.; Kapoor, R.; Donnelly, C.P.; Davidson, E.J.; Parikh, E.; Lopez, J.V.; Tartar, J.L. Gut microbiome diversity is associated with sleep physiology in humans. PLoS ONE 2019, 14, e0222394. [Google Scholar] [CrossRef] [PubMed]
  84. Kuhn, K.A.; Schulz, H.M.; Regner, E.H.; Severs, E.L.; Hendrickson, J.D.; Mehta, G.; Whitney, A.K.; Ir, D.; Ohri, N.; Robertson, C.E.; et al. Bacteroidales recruit IL-6-producing intraepithelial lymphocytes in the colon to promote barrier integrity. Mucosal Immunol. 2018, 11, 357–368. [Google Scholar] [CrossRef]
  85. Zhu, M.Z.; Xu, H.M.; Liang, Y.J.; Xu, J.; Yue, N.N.; Zhang, Y.; Tian, C.M.; Yao, J.; Wang, L.S.; Nie, Y.Q.; et al. Edible exosome-like nanoparticles from portulaca oleracea L mitigate DSS-induced colitis via facilitating double-positive CD4(+)CD8(+)T cells expansion. J. Nanobiotechnol. 2023, 21, 309. [Google Scholar] [CrossRef]
  86. Hidalgo-García, L.; Ruiz-Malagon, A.J.; Huertas, F.; Rodríguez-Sojo, M.J.; Molina-Tijeras, J.A.; Diez-Echave, P.; Becerra, P.; Mirón, B.; Morón, R.; Rodríguez-Nogales, A.; et al. Administration of intestinal mesenchymal stromal cells reduces colitis-associated cancer in C57BL/6J mice modulating the immune response and gut dysbiosis. Pharmacol. Res. 2023, 195, 106891. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Fluorescence image of middle turns of mouse hair cells (scale bar, 50 μm; the arrows point to the missing outer hair cells).
Figure 1. Fluorescence image of middle turns of mouse hair cells (scale bar, 50 μm; the arrows point to the missing outer hair cells).
Metabolites 14 00122 g001
Figure 2. Effect of AS-IV on intestinal flora in noise-exposed mice. (AD) The distribution of the main dominant microbiota under phyla, class, order, and family in each group. (E) Principal coordinate analysis of the three groups based on Bray–Curtis algorithm. (F) Only the taxa meeting an LDA threshold > 2 are visualized. (G) LEfSe cladogram shows the most differentially abundant taxa for the three groups.
Figure 2. Effect of AS-IV on intestinal flora in noise-exposed mice. (AD) The distribution of the main dominant microbiota under phyla, class, order, and family in each group. (E) Principal coordinate analysis of the three groups based on Bray–Curtis algorithm. (F) Only the taxa meeting an LDA threshold > 2 are visualized. (G) LEfSe cladogram shows the most differentially abundant taxa for the three groups.
Metabolites 14 00122 g002
Figure 3. Correlation between main gut microbiota and inflammatory factors. Black boxes indicate strong correlation (|r| > 0.50, p < 0.05); * means significance at p < 0.05; and ** means significance at p < 0.01.
Figure 3. Correlation between main gut microbiota and inflammatory factors. Black boxes indicate strong correlation (|r| > 0.50, p < 0.05); * means significance at p < 0.05; and ** means significance at p < 0.01.
Metabolites 14 00122 g003
Table 1. Hearing threshold results of mice in the control, noise-exposed, and noise + AS-IV groups before and after noise exposure ( x - ± s, dB SPL).
Table 1. Hearing threshold results of mice in the control, noise-exposed, and noise + AS-IV groups before and after noise exposure ( x - ± s, dB SPL).
GroupNo. of
Ears
Pre-ExposurePost-Exposure
Click4 kHz8 kHz16 kHzClick4 kHz8 kHz16 kHz
Control1225.3 ± 5.531.1 ± 2.920.8 ± 12.522.1 ± 8.521.3 ± 5.726.9 ± 6.823.8 ± 7.927.1 ± 8.7
Noise1219.7 ± 6.025.6 ± 6.816.2 ± 3.616.7 ± 5.042.5 ± 6.1 a54.4 ± 11.6 a37.7 ± 7.1 a43.3 ± 9.1 a
Noise + AS-IV1223.3 ± 5.527.4 ± 6.714.9 ± 6.818.5 ± 5.229.4 ± 10.1 bc38.1 ± 10.7 bc26.0 ± 8.3 b32.5 ± 10.6 b
F 3.032.821.632.1524.1823.2611.079.11
p value 0.0620.0740.2120.133<0.001<0.001<0.001<0.001
a p < 0.05, control vs. noise. b p < 0.05, noise + AS-IV vs. noise. c p < 0.05, control vs. noise + AS-IV.
Table 2. TNF-α, TNF-γ, IL-6, IL-1β, and IFN-γ content of each group of mice ( x - ± s, pg/mL).
Table 2. TNF-α, TNF-γ, IL-6, IL-1β, and IFN-γ content of each group of mice ( x - ± s, pg/mL).
GroupNo. of MiceTNF-αTNF-γIL-6IL-1βIFN-γ
Control6152.2 ± 15.733.1 ± 10.877.3 ± 15.771.9 ± 6.8585.3 ± 50.0
Noise6393.8 ± 51.3 a59.1 ± 8.3 a112.7 ± 13.6 a102.2 ± 13.0 a812.4 ± 93.3 a
Noise + AS-IV 6277.8 ± 86.7 bc46.6 ± 6.9 bc96.4 ± 17.483.5 ± 6.2 bc820.4 ± 22.8 c
F 25.3113.027.7116.5927.38
p value <0.001<0.0010.005<0.001<0.001
a p < 0.05, control vs. noise. b p < 0.05, noise + AS-IV vs. noise. c p < 0.05, control vs. noise + AS-IV.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Li, J.; Yang, J.; Xia, Y.; Wang, J.; Xia, Y. Effects of Astragaloside IV on Hearing, Inflammatory Factors, and Intestinal Flora in Mice Exposed to Noise. Metabolites 2024, 14, 122. https://doi.org/10.3390/metabo14020122

AMA Style

Li J, Yang J, Xia Y, Wang J, Xia Y. Effects of Astragaloside IV on Hearing, Inflammatory Factors, and Intestinal Flora in Mice Exposed to Noise. Metabolites. 2024; 14(2):122. https://doi.org/10.3390/metabo14020122

Chicago/Turabian Style

Li, Junyi, Jian Yang, Yun Xia, Junyi Wang, and Yuan Xia. 2024. "Effects of Astragaloside IV on Hearing, Inflammatory Factors, and Intestinal Flora in Mice Exposed to Noise" Metabolites 14, no. 2: 122. https://doi.org/10.3390/metabo14020122

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop