Gene transfection mediated by polyethyleneiminepolyethylene glycol nanocarrier prevents cisplatininduced spiral ganglion cell damage

2015-01-18 05:49
关键词:效度信度分析法

Department of Otorhinolaryngology, the Second Affliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China

Gene transfection mediated by polyethyleneiminepolyethylene glycol nanocarrier prevents cisplatininduced spiral ganglion cell damage

Guan-gui Chen*, Min Mao, Li-zi Qiu, Qi-ming Liu

Department of Otorhinolaryngology, the Second Affliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China

Polyethyleneimine-polyethylene glycol (PEI-PEG), a novel nanocarrier, has been used for transfection and gene therapy in a variety of cells. In our previous study, we successfully carried out PEI-PEG-mediated gene transfer in spiral ganglion cells. It remains unclear whether PEI-PEG could be used for gene therapy with X-linked inhibitor of apoptosis protein (XIAP) in the inner ear. In the present study, we performed PEI-PEG-mediatedXIAPgene transfection in the cochlea of Sprague-Dawley rats,viascala tympani fenestration, before daily cisplatin injections. Auditory brainstem refex tests demonstrated the protective effects ofXIAPgene therapy on auditory function. Immunohistochemical staining revealed XIAP protein expression in the cytoplasm of cells in the spiral ganglion, the organ of Corti and the stria vascularis. Reverse transcription-PCR detected high levels ofXIAPmRNA expression in the cochlea. The present fndings suggest that PEI-PEG nanocarrier-mediatedXIAPgene transfection results in XIAP expression in the cochlea, prevents damage to cochlear spiral ganglion cells, and protects hearing.

nerve regeneration; polyethyleneimine-polyethylene glycol; spiral ganglion cells; X-linked inhibitor of apoptosis protein; gene therapy; nanocarrier; cisplatin; neural regeneration; ototoxicity; cochlea

Funding:This study was supported by the Natural Science Foundation of Guangdong Province in China, No. S2011040003553.

Chen GG, Mao M, Qiu LZ, Liu QM (2015) Gene transfection mediated by polyethyleneimine-polyethylene glycol nanocarrier prevents cisplatin-induced spiral ganglion cell damage. Neural Regen Res. 10(3)∶425-431.

Introduction

Ototoxicity is a potentially serious side-effect of many drugs commonly used in the clinic, including aminoglycoside antibiotics, chemotherapy drugs and diuretics (Schacht et al., 2012). Indeed, drug-induced hearing loss is the primary cause of deafness in Chinese children, with 30,000 new cases diagnosed each year, of which more than 60% suffering from drug-induced deafness (Zheng et al., 2008). With increases in tumor incidence, hearing impairment due to chemotherapy, such as cisplatin, is increasingly common. Mammalian hair cells and spiral ganglion neurons have poor regeneration capacity, and damage to these cells can cause irreversible hearing loss. Therefore, there is an urgent need to protect spiral ganglion neurons, improve cell regeneration and promote functional reconstruction (Atkinson et al., 2012; Schacht et al., 2012).

Previous studies investigating the protection and repair of functional cells of the inner ear have proposed a number of viable treatments, including the use of various types of neurotrophic polypeptides, such as nerve growth factor, brain-derived neurotrophic factor (Liu et al., 2011), and neurotrophin-3 (Jiang et al., 2007; Atkinson et al., 2012; Rak et al., 2014); oxygen free radical scavenging and anti-apoptotic therapy (Rybak et al., 2000); and stem cell therapy (Nakagawa, 2014). Because of the high molecular weight and short half-life of neurotrophic factors and anti-apoptotic drugs, and the presence of the blood-labyrinth barrier (Boussif et al., 1995; Lin and Trune, 1997), direct administration into the inner ear is challenging. With systemic administration, it is difficult to maintain a therapeutically effective drug concentration in the inner ear. Local injection and fenestration surgery often require repeated treatments, limiting their application. Delivering therapeutic genes into the inner ear would be a better solution, with the potential to maintain lifelong expression of the gene. Anatomical and immunological studies of the inner ear have shown that the cochlea is a suitable target organ forin vivogene therapy (Xia and Yin, 2013; Nakagawa, 2014), with excellent therapeutic prospects, but a suitable gene vector remains to be found. Vectors can be viral or non-viral. Viral vector-mediated gene therapy in the inner ear faces safety issues and immune rejection (Sun and Wu, 2013; Xia and Yin, 2013). However, traditional non-viral vector liposomes also have some disadvantages such as poor expression effciency and duration (Sun and Wu, 2013). The transfection efficiency of viral vector-mediated genes into spiral ganglion cells ismore than 70% (Husseman and Raphael, 2009; Okada et al., 2012). Transfection effciency is notably lower in non-viral vectors (approximately 10%) than viral vectors, and is a major restricting factor in the application of non-viral vectors in gene therapy of the inner ear (Pyykkö et al., 2011). Therefore, it is necessary to fnd a non-viral vector with high effciency and good safety.

Polyethyleneimine-polyethylene glycol (PEI-PEG), a novel nanocarrier, has been successfully used for gene transfection in a variety of cells. However, the application of PEI-PEG to gene therapy for the inner ear has, to our knowledge, not yet been investigated. In our previous study (Chen and Xu, 2014), the non-viral vectors PEI-PEG and lipofectamine 2000 were transfected intoin vitrocultured spiral ganglion cells. PEI-PEG transfected spiral ganglion cells with an efficiency of 16.5%, higher than that of lipofectamine 2000 (11.2%). Moreover, the cytotoxicity to spiral ganglion cells was lower with the vector system than with lipofectamine 2000. After transfection, the morphology of spiral ganglion cells was not noticeably abnormal. These results indicate that PEI-PEG performs better than conventional cationic liposomes in several measures, in studies of spiral ganglion cells. X-linked inhibitor of apoptosis protein (XIAP) is frequently used in anti-apoptotic studies (Wang et al., 2011; Wu et al., 2014). In the present study, we sought to determine whether PEI-PEG would mediateXIAPgene expression in the inner ear, and if so, in which cell types and in which parts of the cochlea. Furthermore, we investigated whether PEI-PEG-mediatedXIAPgene expression would protect the cochlea against damage by cisplatin, to assess the potential of PEI-PEG in gene therapy of the inner ear.

Materials and Methods

Preparation of plasmid dNA (pdNA)

Plasmid pIRES2-EGFP/XIAP (constructed in-house) was amplifed byE. coli, extracted, and purifed using a plasmid extraction kit (Qiagen, Dusseldorf, Germany). The content of prepared plasmids was measured at 260 and 280 nm using an ultraviolet spectrometer (Youke, Shanghai, China). Electrophoresis was performed on 1.0% agarose gel at 100 V for 40 minutes. DNA purity was in accordance with the requirements of the experiment, according to the ratioA260/A280. Samples were stored at -20°C.

Preparation of PEI-PEG/XIAP composite

Preliminaryin vitrotests showed that good transfection efficiency was achieved in spiral ganglion cells when the molar ratio of amino groups in the polymer to DNA phosphate groups was equal to 15. Therefore, polymer to DNA phosphate groups = 15 was used in the present study. pIRES2-EGFP/XIAP plasmid (1 µg) was diluted in 50 µL of Dulbecco’s modifed Eagle’s medium and gently shaken. The corresponding amount of PEI-PEG, 4.6 at polymer to DNA phosphate groups = 15 (constructed in-house; Chen and Xu, 2014), was added to an Eppendorf tube containing 50 µL of Dulbecco’s modified Eagle’s medium. The composite was obtained after 5 minutes of shaking at room temperature.

Experimental animals

A total of 38 clean-grade male and female Sprague-Dawley rats, aged 2—3 months and weighing 250—300 g, were provided by the Guangdong Medical Experimental Animal Center in China (animal license No. SCXK (Yue) 2013-0002). The rats were housed in a dark, quiet environment at 18—26°C and at 40—70% relative humidity. The protocols were approved by the Animal Ethics Committee of the Second Affiliated Hospital of Guangzhou Medical University, China.

Upon examination of the ears, the external auditory canal of the rats was smooth, the tympanic membrane was normal, and pinna response was normal. All rats underwent auditory brainstem response (ABR) audiometry. Two rats with ABR thresholds above a decibel sound pressure level (dB SPL) of 40 dB were excluded. The remaining rats were randomly assigned to three groups (n= 12 per group): D-Hanks, XIAP, and control. Following 2 weeks of injection in the cochlea scala tympani in the three groups, drilling of the scala tympani was performed. A single dose of D-Hanks’ solution was injected into cochleas in D-Hanks group and PEI-PEG/XIAP composite was injected into the XIAP group. Nothing was injected into the control animals. 2 weeks after cochlea injection, 3 mg/kg cisplatin (Sigma-Aldrich, St. Louis, MO, USA) was daily intraperitoneally injected for 6 days.

drilling of the scala tympani and administration of composite component

In the XIAP group, rats were anesthetized with 1% sodium pentobarbital (45 mg/kg, i.p.), placed on the operating table, and the skin around the left ear was sterilized. The auditory vesicle of the left ear was exposed under a binocular dissecting microscope (Carl Zeiss, Oberkochen, Germany). A hole (0.1—0.2 mm in diameter) was made in the outer wall of the scala tympani in the cochlear substrate, using an electric surgical drill (Stryker, Kalamazoo, MI, USA) with a diamond head. PEI-PEG/XIAP composite (10 µL, 0.01 g/L was slowly injected over 5 minutes using a microsyringe (Hamilton, Bonaduz, Switzerland) to a maximum depth of 0.2 mm. After injection, the needle was maintained in place for a further 2 minutes to prevent liquid outflow. Upon removal of the needle, a small amount of sarcoplasm was used to block the drilled hole, and the wound was sutured. After the surgery, penicillin (200,000 U/d) was injected intramuscularly for 3 consecutive days to prevent infection. The rats in the D-Hanks group underwent the same procedure but 10 µL D-Hanks’solution was injected instead of PEI-PEG/XIAP composite. The rats in the control group underwent anesthesia and drilling of the scala tympani but were not injected.

detection of ABR thresholds

Figure 2 XIAP immunoreactivity in the cochlea after PEI-PEG-mediatedXIAPgene transfection into the cochlea of rats with cisplatin-induced ototoxicity (immunohistochemical staining, × 100).

Figure 3 Histopathological changes of the cochlea of rats with cisplatin-induced ototoxicity after PEI-PEG-mediatedXIAPgene transfection.

The rats were anesthetized with 1% sodium pentobarbital (45 mg/kg, i.p.). Reference electrodes (Fude Medical Electronics Co., Ltd., Xi’an, China) were placed in the auditory vesicle behind the ipsilateral (left) ear. Ground poles were placed in the papilla or nasal tip behind the contralateral (right) ear. Recording electrodes were placed in the midpoint of the bilateral external auditory canal on the scalp. Low-intensity white noise (< 20 dB SPL) was applied to the contralateral side. Short sound (click) stimulation was used at a frequency of 4,000 Hz, flter range 100—3,000 Hz, 1,000 times, at a repetition rate of 10 times/second and at an intensity of 5—100 dB SPL. ABR was measured and hearing thresholds were recorded 24 hours before cochlear injection, 24 hours after cochlear injection, and 24 hours after the fnal injection of cisplatin.

Preparation of cochlear sections

After the final ABR measurement, all rats were intraperitoneally anesthetized with 1% pentobarbital (50 mg/kg). The sternum was cut open to expose the heart, which was perfused with 4°C physiological saline and 4°C fxative (4% paraformaldehyde in 0.1 M PBS, pH 7.4). The rats were rapidly decapitated, and the left auditory vesicle was obtained. The cochlea was dissected in an ice bath, and postfxed in the same fxative at 4°C overnight. Samples were decalcifed in 10% sodium ethylenediaminetetraacetic acid solution (pH 7.4) for 14 days, embedded in paraffn, and sliced into 5-µmthick sections parallel to the cochlear axis.

Hematoxylin-eosin staining for spiral ganglion cell counts in the rat cochlea

Paraffin-embedded sections of rat cochlea were dewaxed with xylene, dehydrated through a graded alcohol series, stained with hematoxylin and eosin for 5 minutes, washed with running water and decolorated with 1% hydrochloric acid in ethanol for a few seconds. 0.5% ammonium hydroxide was then added to stain the hematoxylin. After rinsing under running tap water for 1 hour, samples were placed in distilled water, dehydrated with 70 and 90% alcohol (10 minutes each), and stained with ethanol eosin for 2—3 minutes. Finally, the sections were dehydrated with pure ethanol, permeabilized in xylene, and mounted with neutral resin. From the serial sections proximal to the cochlear axis, one section was obtained from every contiguous fve, resulting in a total of six sections from each rat. The density of spiral ganglion cells in Rosenthal’s canal was quantifed with Image Pro Plus 6.0 image analysis system (MediaCybernetics, Bethesda, MD, USA). Cell density (n/mm2) was equal to the number of spiral ganglion cells/section area of Rosenthal’s canal, and the mean of all sections was calculated.

Immunohistochemical staining for XIAP expression in the rat cochlea

Sections of rat cochlea were treated with 3% H2O2for 20 minutes to inactivate endogenous peroxidase, washed with distilled water, and antigen retrieval was performed using a microwave. Sections were then incubated with goat anti-XIAP polyclonal antibody (1:200; Boster, Wuhan, Hubei Province, China) at 4°C overnight, biotinylated rabbit anti-goat IgG (1:300; Boster) secondary antibody at 37°C for 30 minutes, and streptavidin-biotin complex at 37°C for 30 minutes. After washing, sections were visualized with 3,3′-diaminobenzidine and mounted with neutral resin. PBS, instead of primary antibody, was used as a blank control. Samples were observed under a light microscope (CX31; Olympus, Tokyo, Japan).

Reverse transcription forXIAPmRNA expression in the rat cochlea

The rats were sacrifced by overdose of anesthesia, their fur was soaked in 70% ethanol, and they were decapitated. Auditory vesicles were removed in an ice bath to obtain the inner ears. Both ears of each animal were considered a metering unit. Total RNA was extracted. Reverse transcription-PCR was performed using a one-step reverse transcription kit (Qiagen, Dusseldorf, Germany), according to the manufacturer’s instructions. β-Actin served as the internal reference. PCR conditions were as follows: reverse transcription at 50°C for 30 minutes; initial activation at 95°C for 15 minutes, 35 cycles of denaturation at 94°C for 50 seconds, annealing at 60°C for 50 seconds, extension at 72°C for 60 seconds; extension at 72°C for 10 minutes. XIAP primer sequences: upstream primer, 5′-AGG AAC CCT GCC ATG TAT TG-3′; downstream primer, 5′-TGT TGT TCC CAA GGG TCT TC-3′. β-Actin primer sequence: upstream primer, 5′-CGC ACC ACT GGC ATT GTC AT-3′; downstream primer, 5′-TIC TCC TTG ATG TCA CGC AC-3′. The product length was 494 bp. PCR products were electrophoresed on a 1.5% agarose gel. A Gel Imaging Analysis System (Tanon2020; Tanon, Shanghai, China) was employed to measure optical density. The expression of XIAP mRNA was calculated as the average optical density ratio of the target band to the internal reference band.

Statistical analysis

Measurement data were expressed as the mean ± SD and analyzed using SPSS 13.0 Statistical Software (SPSS Inc., Chicago, IL, USA). Two-way repeated measures analysis of variance was used to compare ABR thresholds. Intergroup differences of spiral ganglion cell number andXIAPmRNA expression were compared by one-way analysis of variance, followed by the Student-Newman-Keuls test. A value ofP<0.05 was considered statistically signifcant.

Results

Effect of PEI-PEG-mediatedXIAPgene transfection on ABR thresholds in rats with cisplatin-induced ototoxicityBaseline ABR thresholds were similar between rats from each group (P> 0.05), meeting the test requirements. Cochlear injection did not produce a significant difference in ABR thresholds (P> 0.05). However, after intraperitoneal injection of cisplatin for 6 days, ABR thresholds were signifcantly higher in the D-Hanks and XIAP groups than in the same groups after cochlear injection (P< 0.05), and also higher than in the control group (P< 0.05). However, ABR thresholds were lower in the XIAP group than in the D-Hanks group after the injection of cisplatin (P< 0.05), suggesting thatXIAPgene therapy reduced the damage of cisplatin to hearing (Figure 1).

Effect of PEI-PEG-mediatedXIAPgene transfection on cochlear tissue in rats with cisplatin-induced ototoxicity

Immunohistochemical staining revealed that a weak immunoreactivity of XIAP in the cytoplasm of cells in the spiral ganglion, organ of Corti, and stria vascularis border of the cochlea in the D-Hanks and control groups. XIAP immunoreactivity was strongest in rats in the XIAP group (Figure 2).

Hematoxylin-eosin staining revealed the complete structure of the cochlea. Spiral ganglion cells, organ of Corti and stria vascularis were prominent in the three groups. In the control group, spiral ganglion cells were elliptical, regular and densely arranged, with uniform staining in Rosenthal’s canal of the cochlea. In the D-Hanks and XIAP groups, spiral ganglion cells were sparsely arranged and disordered, the intercellular spaces were wider than in the control group, cytoplasm was lightly stained, and some cells exhibited vacuolar degeneration. There were fewer spiral ganglion cells in the experimental groups after treatment with cisplatin; spiral ganglion cell number, ranked in descending order, was as follows: control group > XIAP group > D-Hanks group (P<0.05; Figure 3).

Effect of PEI-PEG-mediatedXIAPgene transfection onXIAPmRNA expression in cochlear tissue of rats with cisplatin-induced ototoxicity

Reverse transcription-PCR showed thatXIAPmRNA expression was greater in the XIAP group than in the other two groups (P< 0.05; Figure 4), with no difference in expression between the D-Hanks and control groups (P> 0.05).

discussion

Cisplatin is widely used as a first-line anti-cancer drug. It has a strong broad-spectrum effect by inhibiting DNA replication in cancer cells. However, it has severe side effects, which include gastrointestinal reactions, bone marrow suppression, and kidney and inner ear damage. Of these, the side effects in the kidneys and the inner ear are the most serious (Dille et al., 2012). A large proportion (50—100%) of patients undergoing chemotherapy with cisplatin experience a degree of hearing loss (Rybak et al., 2007; Dille et al., 2012). Cisplatin damage to hearing has a complex multifactorial mechanism. Previous studies demonstrated that cisplatin ototoxicity was associated with drug-induced oxygen free radical damage, with oxidative stress ultimately resulting in the apoptosis of inner ear hair cells and spiral ganglion cellsviaa series of cascades (Deavall et al., 2012; Pereira et al., 2012; Fu et al., 2013). Human inner ear hair cells and spiral ganglion cells are nonrenewable, so it is important to prevent drug ototoxicity.

Hearing loss induced by noise, aging or ototoxicity is correlated with cochlear hair cell and spiral ganglion cell death. The final pathway is cell apoptosis. Anti-apoptotic therapy is therefore effective in ameliorating the damage of these harmful factors to the inner ear. Anti-apoptotic therapy using a caspase antagonist reduces age-related cochlear neuronal degeneration (Ruan et al., 2014), diminishes noise damage (Wang et al., 2011), and reduces cisplatin ototoxicity (Cooper et al., 2006; Chan et al., 2007; Waissbluth et al., 2012). Cooper et al. (2006) used adeno-associated virus vector to transfect XIAP into mouse cochlea, before injecting cisplatin intraperitoneally after 2 months of target gene expression.XIAPgene therapy was found to protect hearing and elevate the survival rate of cochlear outer hair cells by 45%. Chan et al. (2007) also used an adeno-associated viral vector to transfectXIAPand successfully prevent cisplatin ototoxicity, resulting in a 68% reduction in hearing impairment, and a 50% reduction in cochlear hair cell damage. Together, these results highlight XIAP as a promising new treatment for cisplatin ototoxicity. However, while several studies have examined viral vectorXIAPgene transfection in the cochlea, there have been no reports to date of similar studies with non-viral vectors. Boussif et al. (1995) frst reported the use of polyethylenimine as a gene vector; however, the success of polyethylenimine alone is limited (Choosakoonkriang et al., 2003). PEI-PEG has been successfully applied to a variety of cell transfection and gene therapies (Okada et al., 2012; Liu et al., 2013), but gene therapy in the inner ear has not, to our knowledge, been reported. Our previous study confirmed that PEI-PEG could transfect cultured spiral ganglion cellsin vitro, and transfection effciency and cytotoxicity were better than that obtained with conventional cationic liposomes (Chen and Xu, 2014). In the present study, we built on those fndings and successfully performed PEI-PEG transfection of theXIAPgene into the cochlea, resulting in elevated local expression ofXIAPmRNA. Immunohistochemical staining revealed thatXIAPgene expression was enhanced in the cochlear axis, organ of Corti and stria vascularis, and that it blocked cisplatin ototoxicity, protected spiral ganglion cells, and prevented hearing impairment.

Before inner ear gene transfer is used as a method to treat sensorineural deafness, it is necessary to establish whether the surgery and injection risk damage to the structure and function of the inner ear. Numerous studies have addressed the effects of various methods of gene transfer on the structure and function of the inner ear, including injection through the round window membrane (Wang et al., 2012), scala tympani fenestration surgery (Bogaerts et al., 2008), and semicircular canal fenestration surgery (Gassner et al., 2012). Scala tympani fenestration for viral and liposome vector injection had a small effect on cochlear function and structure (Praetorius et al., 2003). Stöver et al. (1999) injected adenovirus vector into the inner ear by scala tympani fenestration and round window membrane injection; 5 days later, ABR detection showed that the surgery had no notable effect on hearing. Gassner et al. (2012) injected adenovirus vector carrying green fluorescent protein into the cochlea by semicircular canal fenestration surgery, with a slight infuence on hearing. Fu et al. (2008) transplanted rat embryonic neural stem cells into the cochlea of naïve rats through cochlear fenestration using recombinant adenovirus carrying green fuorescent protein (Ad-GFP). ABR detection demonstrated that no significant differences in auditory threshold were detected in rats before or after the surgery. Scanning electron microscopy revealed that outer hair cells in the cochlea showed a scattered loss of less than 1%, not significantly different from the untreated group, further demonstrating that transplantation of neural stem cells infected with Ad-GFP into the normal rat cochlea does not affect auditory threshold or cochlear morphology. Our previous study suggested that ABR threshold was temporarily decreased (< 20 dB) but recovered to normal within 28 days after bone marrow mesenchymal stem cells were transplanted in the cochlea through fenestration of the scala tympani (Chen et al., 2010). In the present nanocarrier experiment, we also used scala tympani fenestration, and found no significant difference between ABR thresholds before and after cochlear injection, indicating that cochlear injection did not affect rat ABR thresholds, and further demonstrating that scala tympani fenestration is a safe method by which to administer the PEI-PEG nanocarrier complex.

In summary, we have demonstrated successful PEI-PEG-delivery of XIAP into cochlear tissue cells, which resulted in the prevention of cisplatin ototoxicity. Our results provide a promising new option in gene therapy of sensorineuraldeafness and highlight the possibilities for nanomaterial-mediated cochlear-targeted gene therapy.

Figure 4XIAPmRNA expression in cochlear tissue of rats with cisplatin-induced ototoxicity after PEI-PEG-mediatedXIAPgene transfection.

Figure 1 Changes in ABR thresholds after PEI-PEG-mediatedXIAPgene transfection into the cochlea of rats with cisplatin-induced ototoxicity.

Author contributions:GGC participated in study design and experiment implementation, wrote the paper and was in charge of paper authorization. MM conducted the experiment of tissue sections and immunohistochemistry. LZQ participated in audiology detection. QML ensured the integrity of the data and data statistics. All authors approved the final version of the paper.

Conficts of interest:None declared.

Atkinson PJ, Wise AK, Flynn BO, Nayagam BA, Hume CR, O’Leary SJ, Shepherd RK, Richardson RT (2012) Neurotrophin gene therapy for sustained neural preservation after deafness. PLoS One 7:e52338.

Bogaerts S, Douglas S, Corlette T, Pau H, Saunders D, McKay S, Oleskevich S (2008) Microsurgical access for cell injection into the mammalian cochlea. J Neurosci Methods 168:156-163.

Boussif O, Lezoualc’h F, Zanta MA, Mergny MD, Scherman D, Demeneix B, Behr JP (1995) A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: polyethylenimine. Proc Natl Acad Sci U S A 92:7297-7301.

Chan DK, Lieberman DM, Musatov S, Goldfein JA, Selesnick SH, Kaplitt MG (2007) Protection against cisplatin-induced ototoxicity by adeno-associated virus-mediated delivery of the X-linked inhibitor of apoptosis protein is not dependent on caspase inhibition. Otol Neurotol 28:417-425.

Chen GG, Xu YL (2014) Polyethyenimine-polyethylene glycol as a gene transfer vector for spiral ganglion cells in vitro. Zhongguo Zuzhi Gongcheng Yanjiu 18:3345-3349.

Chen GG, Xie DH, Liu QX, Tan ZQ (2010) Expression of brain-derived neurotrophic factor modifed bone marrow mesenchymal stem cells in the cochlea of drug deafened guinea pigs and its protection role. Zhonghua Er Bi Yan Hou Tou Jing Wai Ke Za Zhi 45:924-929.

Choosakoonkriang S, Lobo BA, Koe GS, Koe JG, Middaugh CR (2003) Biophysical characterization of PEI/DNA complexes. J Pharm Sci 92:1710-1722.

Cooper LB, Chan DK, Roediger FC, Shaffer BR, Fraser JF, Musatov S, Selesnick SH, Kaplitt MG (2006) AAV-mediated delivery of the caspase inhibitor XIAP protects against cisplatin ototoxicity. Otol Neurotol 27:484-490

Deavall DG, Martin EA, Horner JM, Roberts R (2012) Drug-induced oxidative stress and toxicity. J Toxicol 2012:645460.

Dille MF, Wilmington D, McMillan GP, Helt W, Fausti SA, Konrad Martin D (2012) Development and validation of a cisplatin dose-ototoxicity model. J Am Acad Audiol 23:510-521.

Fu Y, Ding D, Wei L, Jiang H, Salvi R (2013) Ouabain-induced apoptosis in cochlear hair cells and spiral ganglion neurons in vitro. Biomed Res Int 2013:628064.

Fu Y, Wang SQ, Wang JT, Wang GP, Xie J, Gong SS (2008) Experimental study on embryonic neural stem cells transplantation into natural rat cochlea via round window. Zhonghua Er Bi Yan Hou Tou Jing Wai Ke Za Zhi 43:944-949.

问卷经小范围发放并修改后,问题设置为30题,利用SPSS19.0处理数据。问卷信度分析采用了常用的Cronbach's Alpha值为指标。信度系数越大,表明测量可信度越大。分析显示,在选取的几项调查问题量表的Cronbach's Alpha值均高于0.7,说明本问卷调查数据信度较好。研究采用主成分分析法对各个量表进行分析,KMO值均大于0.6,Bartlett的球形度检验统计值的显著性概率为0.000,小于0.001,说明本问卷调查数据效度较好。如表1所示。

Gassner D, Durham D, Pfannenstiel SC, Brough DE, Staecker H (2012) Canalostomy as a surgical approach for cochlear gene therapy in the rat. Anat Rec (Hoboken) 295:1830-1836.

Husseman J, Raphael Y (2009) Gene therapy in the inner ear using adenovirus vectors. Adv Otorhinolaryngol 66:37-51.

Jiang M, Zhang YQ, He GX, Sun H (2007) Protective effect of NT-3 gene mediated by hydroxyapatite nanoparticle on the cochlea of guinea pigs injured by excitotoxicity. Zhong Nan Da Xue Xue Bao Yi Xue Ban 32:563-567.

Lin DW, Trune DR (1997) Breakdown of stria vascularis blood-labyrinth barrier in C3H/lpr autoimmune disease mice. Otolaryngol Head Neck Surg 117:530-534.

Liu C, Liu F, Feng L, Li M, Zhang J, Zhang N (2013) The targeted co-delivery of DNA and doxorubicin to tumor cells via multifunctional PEI-PEG based nanoparticles. Biomaterials 34:2547-2564.

Liu W, Kinnefors A, Boström M, Rask-Andersen H (2011) Expression of TrkB and BDNF in human cochlea-an immunohistochemical study. Cell Tissue Res 345:213-221.

Nakagawa T (2014) Strategies for developing novel therapeutics for sensorineural hearing loss. Front Pharmacol 5:206.

Okada H, Iizuka T, Mochizuki H, Nihira T, Kamiya K, Inoshita A, Kasagi H, Kasai M, Ikeda K (2012) Gene transfer targeting mouse vestibule using adenovirus and adeno-associated virus vectors. Otol Neurotol 33:655-659

Praetorius M, Baker K, Weich CM, Plinkert PK, Staecker H (2003) Hearing preservation after inner ear gene therapy: the effect of vector and surgical approach. ORL J Otorhinolaryngol Relat Spec 65:211-214.

Pyykkö I, Zou J, Zhang W, Zhang Y (2011) Nanoparticle-based delivery for the treatment of inner ear disorders. Curr Opin Otolaryngol Head Neck Surg 19:388-396

Rak K, Völker J, Jürgens L, Scherzad A, Schendzielorz P, Radeloff A, Jablonka S, Mlynski R, Hagen R (2014) Neurotrophic effects of taurine on spiral ganglion neurons in vitro. Neuroreport 25:1250-1254.

Ruan Q, Zeng S, Liu A, Chen Z, Yu Z, Zhang R, He J, Bance M, Robertson G, Yin S, Wang J (2014) Overexpression of X-Linked Inhibitor of Apoptotic Protein (XIAP) reduces age-related neuronal degeneration in the mouse cochlea. Gene Ther doi: 10.1038/gt. 2014.

Rybak LP, Husain K, Morris C, Whitworth C, Somani S (2000) Effect of protective agents against cisplatin ototoxicity. Am J Otol 21:513-520.

Rybak LP, Whitworth CA, Mukherjea D, Ramkumar V (2007) Mechanisms of cisplatin-induced ototoxicity and prevention. Hear Res 226:157-167.

Schacht J, Talaska AE, Rybak LP (2012) Cisplatin and aminoglycoside antibiotics: hearing loss and its prevention. Anat Rec (Hoboken) 295:1837-1850.

Stöver T, Yagi M, Raphael Y (1999) Cochlear gene transfer: round window versus cochleostomy inoculation. Hear Res 136:124-130.

Sun H, Wu X (2013) Current status and prospects of non-viral vector in inner ear gene therapy. Lin Chung Er Bi Yan Hou Tou Jing Wai Ke Za Zhi 27:1339-1342.

Waissbluth S, Pitaro J, Daniel SJ (2012) Gene therapy for cisplatin-induced ototoxicity: a systematic review of in vitro and experimental animal studies. Otol Neurotol 33:302-310.

Wang H, Murphy R, Taaffe D, Yin S, Xia L, Hauswirth WW, Bance M, Robertson GS, Wang J (2012) Effcient cochlear gene transfection in guinea-pigs with adeno-associated viral vectors by partial digestion of round window membrane. Gene Ther 19:255-263.

Wang J, Tymczyszyn N, Yu Z, Yin S, Bance M, Robertson GS (2011) Overexpression of X-linked inhibitor of apoptosis protein protects against noise-induced hearing loss in mice. Gene Ther 18:560-568.

Wu H, Che X, Zheng Q, Wu A, Pan K, Shao A, Wu Q, Zhang J, Hong Y (2014) Caspases: a molecular switch node in the crosstalk between autophagy and apoptosis. Int J Biol Sci 10:1072-1083.

Xia L, Yin S (2013) Local gene transfection in the cochlea (Review). Mol Med Rep 8:3-10.

Zheng XY, Zhang L, Chen G, Pei LJ, Song XM (2008) Prevalence of visual, hearing, speech, physical, intellectual and mental disabilities in China, 2006. Zhonghua Liu Xing Bing Xue Za Zhi 29:634-638.

Copyedited by Slone-Murphy J, Stow A, Yu J, Qiu Y, Li CH, Song LP, Zhao M

0.4103/1673-5374.153691

http://www.nrronline.org/

Accepted: 2014-11-07

猜你喜欢
效度信度分析法
异步机传统分析法之困难及其克服
效度验证模式系统整合与效度研究发展策略
《广东地区儿童中医体质辨识量表》的信度和效度研究
作为数学教育研究质量分析的信度
层次分析法在起重机械腐蚀评估中的应用
问卷是否可信
——基于体育核心期刊论文(2010—2018年)的系统分析
谈高效课堂下效度的提升策略
层次分析法在生态系统健康评价指标体系中的应用
基于时间重叠分析法的同车倒卡逃费探析
语言测试效度研究的另一视角:考试的因子结构研究