山东大学耳鼻喉眼学报 ›› 2022, Vol. 36 ›› Issue (3): 84-91.doi: 10.6040/j.issn.1673-3770.0.2021.562

• 研究进展 • 上一篇    下一篇

表观遗传学在慢性鼻窦炎伴鼻息肉发病机制中的作用

李佳倪1,朱冬冬1,2,孟粹达1,2   

  1. 1.吉林大学中日联谊医院 耳鼻咽喉头颈外科, 吉林 长春 130033;
    2.吉林省上气道过敏性疾病精准诊疗实验室, 吉林 长春 130033
  • 发布日期:2022-06-15
  • 通讯作者: 孟粹达. E-mail:mengcd@jlu.edu.cn
  • 基金资助:
    国家自然科学基金项目(81870701,82071016);吉林省自然科学基金项目(20200201411JC,20200201600JC,20200201200JC);吉林省重点实验室建设项目(20190901003JC);吉林省卫生科研人才专项项目(2019scz004,2020scz24);吴阶平医学基金会临床科研专项(320675018380)

The role of epigenetics in the pathogenesis of chronic rhinosinusitis with nasal polyps

LI Jiani1, ZHU Dongdong1,2Overview,MENG Cuida1,2   

  1. 1. Department of Otorhinolaryngology & Head and Neck Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, China;
    2. Jilin provincial Key Laboratory of Precise Diagnosis and Treatment of Upper Airway Allergic, Changchun 130033, Jilin, China
  • Published:2022-06-15

摘要: 慢性鼻窦炎伴鼻息肉其确切的发病机制尚未完全明确。表观遗传学(Epigenetics)主要指基于非基因序列改变所致基因表达水平的变化,包括DNA甲基化、组蛋白修饰和非编码RNA调控等。就相关基因的表观遗传学修饰及其引起的相应细胞因子和蛋白质等的表达改变在CRS发病机制中的作用作一综述,从全新的角度认识CRS,为明确其发病过程、探究治疗方法提供新的方向。

关键词: 慢性鼻窦炎, 鼻息肉, 表观遗传学, 甲基化, 发病机制

Abstract: The pathogenesis of chronic rhinosinusitis(CRS)with nasal polyps has not been fully elucidated. Epigenetics mainly refers to changes in gene expression levels based on non-gene sequence changes, such as DNA methylation, histone modification, and microRNA regulation. In this review, the role of epigenetic modification of CRS-related genes and changes in the expression levels of corresponding cytokines and other related proteins in the pathogenesis of CRS are discussed. The aim of this review is to provide new directions for research on the pathogenesis of CRS and the development of therapeutic methods.

Key words: Chronic Rhinosinusitis, Nasal Polyps, Epigenetics, Methylation, Pathogenesis

中图分类号: 

  • R765.4
[1] 李华斌, 赖玉婷, 姜文秀. 慢性鼻窦炎的内表型研究进展及精准治疗[J]. 山东大学耳鼻喉眼学报, 2019, 33(3): 9-13. doi:10.6040/j.issn.1673-3770.1.2018.043. LI Huabin, LAI Yuting, JIANG Wenxiu. Endotypes and precision medicine in chronic sinusitis treatment[J]. Journal of Otolaryngology and Ophthalmology of Shandong University, 2019, 33(3): 9-13. doi:10.6040/j.issn.1673-3770.1.2018.043.
[2] Dykewicz MS, Hamilos DL. Rhinitis and sinusitis[J]. J Allergy Clin Immunol, 2010, 125(2): 103-115. doi: 10.1016/j.jaci.2009.12.989.
[3] Chaaban MR, Walsh EM, Woodworth BA. Epidemiology and differential diagnosis of nasal polyps[J]. Am J Rhinol Allergy, 2013, 27(6): 473-478. doi:10.2500/ajra.2013.27.3981.
[4] North ML, Ellis AK. The role of epigenetics in the developmental origins of allergic disease[J]. Ann Allergy Asthma Immunol, 2011, 106(5): 355-361; quiz362. doi:10.1016/j.anai.2011.02.008.
[5] Ng HH, Adrian B. DNA methylation and chromatin modification[J]. Curr Opin Genet Dev, 1999, 9(2): 158-163. doi:10.1016/S0959-437X(99)80024-0.
[6] Zheng YB, Zhao Y, Yue LY, et al. Pilot study of DNA methylation in the pathogenesis of chronic rhinosinusitis with nasal polyps[J]. Rhinology, 2015, 53(4): 345-352. doi:10.4193/Rhino14.086.
[7] Hew KM, Walker AI, Kohli A, et al. Childhood exposure to ambient polycyclic aromatic hydrocarbons is linked to epigenetic modifications and impaired systemic immunity in T cells[J]. Clin Exp Allergy, 2015, 45(1): 238-248. doi:10.1111/cea.12377.
[8] White GP, Hollams EM, Yerkovich ST, et al. CpG methylation patterns in the IFN-γ promoter in naive T cells: variations during Th1 and Th2 differentiation and between atopics and non-atopics[J]. Pediatr Allergy Immunol, 2006, 17(8): 557-564. doi:10.1111/j.1399-3038.2006.00465.x.
[9] Jundi K, Greene C. Transcription of interleukin-8: how altered regulation can affect cystic fibrosis lung disease[J]. Biomolecules, 2015, 5(3): 1386-1398. doi:10.3390/biom5031386.
[10] van Drunen CM, Reinartz S, Wigman J, et al. Inflammation in chronic rhinosinusitis and nasal polyposis[J]. Immunol Allergy Clin North Am, 2009, 29(4): 621-629. doi:10.1016/j.iac.2009.07.003.
[11] Li JY, Jiao J, Wang M, et al. Hypomethylation of the IL8 promoter in nasal epithelial cells of patients with chronic rhinosinusitis with nasal polyps[J]. J Allergy Clin Immunol, 2019, 144(4): 993-1003.e12. doi:10.1016/j.jaci.2019.06.042.
[12] Soumelis V, Reche PA, Kanzler H, et al. Human epithelial cells trigger dendritic cell mediated allergic inflammation by producing TSLP[J]. Nat Immunol, 2002, 3(7): 673-680. doi:10.1038/ni805.
[13] Nagarkar DR, Poposki JA, Tan BK, et al. Thymic stromal lymphopoietin activity is increased in nasal polyps of patients with chronic rhinosinusitis[J]. J Allergy Clin Immunol, 2013, 132(3): 593-600.e12. doi:10.1016/j.jaci.2013.04.005.
[14] Kimura S, Pawankar R, Mori S, et al. Increased expression and role of thymic stromal lymphopoietin in nasal polyposis[J]. Allergy Asthma Immunol Res, 2011, 3(3): 186-193. doi:10.4168/aair.2011.3.3.186.
[15] Li JY, Jiao J, Gao YB, et al. Association between methylation in nasal epithelial TSLP gene and chronic rhinosinusitis with nasal polyps[J]. Allergy Asthma Clin Immunol Off J Can Soc Allergy Clin Immunol, 2019, 15(1): 71. doi:10.1186/s13223-019-0389-3.
[16] Heljasvaara R, Nyberg P, Luostarinen J, et al. Generation of biologically active endostatin fragments from human collagen XVIII by distinct matrix metalloproteases[J]. Exp Cell Res, 2005, 307(2): 292-304. doi:10.1016/j.yexcr.2005.03.021.
[17] Ergün S, Kilic N, Wurmbach JH, et al. Endostatin inhibits angiogenesis by stabilization of newly formed endothelial tubes[J]. Angiogenesis, 2001, 4(3): 193-206. doi:10.1023/a: 1014027218980.
[18] Zhang Y, Qu ZH, Cui M, et al. Combined endostatin and TRAIL gene transfer suppresses human hepatocellular carcinoma growth and angiogenesis in nude mice[J]. Cancer Biol Ther, 2009, 8(5): 466-473. doi:10.4161/cbt.8.5.7687.
[19] Suzaki Y, Hamada K, Sho M, et al. A potent antiangiogenic factor, endostatin prevents the development of asthma in a murine model[J]. J Allergy Clin Immunol, 2005, 116(6): 1220-1227. doi:10.1016/j.jaci.2005.08.052.
[20] Kidoguchi M, Noguchi E, Nakamura T, et al. DNA methylation of proximal PLAT promoter in chronic rhinosinusitis with nasal polyps[J]. Am J Rhinol Allergy, 2018, 32(5): 374-379. doi:10.1177/1945892418782236.
[21] Takabayashi T, Kato A, Peters AT, et al. Excessive fibrin deposition in nasal polyps caused by fibrinolytic impairment through reduction of tissue plasminogen activator expression[J]. Am J Respir Crit Care Med, 2013, 187(1): 49-57. doi:10.1164/rccm.201207-1292OC.
[22] Kim DY, Cho SH, Takabayashi T, et al. Chronic rhinosinusitis and the coagulation system[J]. Allergy Asthma Immunol Res, 2015, 7(5): 421-430. doi:10.4168/aair.2015.7.5.421.
[23] Kim JY, Kim DK, Yu MS, et al. Role of epigenetics in the pathogenesis of chronic rhinosinusitis with nasal polyps[J]. Mol Med Rep, 2018, 17(1): 1219-1227. doi:10.3892/mmr.2017.8001.
[24] Velimir GM, Hrvoje C, Livije K, et al. Surgical treatment for nasal polyposis: predictors of outcome[J]. Eur Arch Oto Rhino Laryngol Off J Eur Fed Oto Rhino Laryngol Soc EUFOS Affil Ger Soc Oto Rhino Laryngol Head Neck Surg, 2015, 272(12): 3735-3743. doi:10.1007/s00405-015-3519-7.
[25] Sreeparvathi A, Kalyanikuttyamma LK, Kumar M, et al. Significance of blood eosinophil count in patients with chronic rhinosinusitis with nasal polyposis[J]. J Clin Diagn Res, 2017, 11(2): MC08-MC11. doi:10.7860/JCDR/2017/25320.9445.
[26] Ueno S, Weidinger G, Osugi T, et al. Biphasic role for Wnt/beta-catenin signaling in cardiac specification in zebrafish and embryonic stem cells[J]. Proc Natl Acad Sci USA, 2007, 104(23): 9685-9690. doi:10.1073/pnas.0702859104.
[27] Malbon CC. Frizzleds: new members of the superfamily of G-protein-coupled receptors[J]. Front Biosci, 2004, 9: 1048-1058. doi:10.2741/1308.
[28] Lin LZ, Cui L, Zhou WL, et al. Β-Catenin directly regulates Islet1 expression in cardiovascular progenitors and is required for multiple aspects of cardiogenesis[J]. PNAS, 2007, 104(22): 9313-9318. doi:10.1073/pnas.0700923104.
[29] Kim JY, Cha MJ, Park YS, et al. Upregulation of FZD5 in eosinophilic chronic rhinosinusitis with nasal polyps by epigenetic modification[J]. Mol Cells, 2019, 42(4): 345-355. doi:10.14348/molcells.2019.2418.
[30] Benard A, Goossens-Beumer IJ, van Hoesel AQ, et al. Histone trimethylation at H3K4, H3K9 and H4K20 correlates with patient survival and tumor recurrence in early-stage colon cancer[J]. BMC Cancer, 2014, 14: 531. doi:10.1186/1471-2407-14-531.
[31] Snowden AW, Gregory PD, Case CC, et al. Gene-specific targeting of H3K9 methylation is sufficient for initiating repression in vivo[J]. Curr Biol, 2002, 12(24): 2159-2166. doi:10.1016/s0960-9822(02)01391-x.
[32] Bartke T, Vermeulen M, Xhemalce B, et al. Nucleosome-interacting proteins regulated by DNA and histone methylation[J]. Cell, 2010, 143(3): 470-484. doi:10.1016/j.cell.2010.10.012.
[33] Fuks F. DNA methylation and histone modifications: teaming up to silence genes[J]. Curr Opin Genet Dev, 2005, 15(5): 490-495. doi:10.1016/j.gde.2005.08.002.
[34] Andrew J, Tony B, Kouzarides. Regulation of chromatin by histone modifications[J]. Cell Research, 2011, 21(3): 381-395.
[35] Lal G, Bromberg JS. Epigenetic mechanisms of regulation of Foxp3 expression[J]. Blood, 2009, 114(18): 3727-3735. doi:10.1182/blood-2009-05-219584.
[36] Thomas LR, Miyashita H, Cobb RM, et al. Functional analysis of histone methyltransferase g9a in B and T lymphocytes[J]. J Immunol, 2008, 181(1): 485-493. doi:10.4049/jimmunol.181.1.485.
[37] Han SY, Lu J, Zhang Y, et al. Recruitment of histone deacetylase 4 by transcription factors represses interleukin-5 transcription[J]. Biochem J, 2006, 400(3): 439-448. doi:10.1042/BJ20061085.
[38] Lee JH, Chung SW, Park IH, et al. Expression of extracellular matrix metalloproteinase inducer in nasal polyps[J]. Am J Rhinol Allergy, 2010, 24(6): 127-131. doi:10.2500/ajra.2010.24.3503.
[39] Cho JS, Moon YM, Park IH, et al. Epigenetic regulation of myofibroblast differentiation and extracellular matrix production in nasal polyp-derived fibroblasts[J]. Clin Exp Allergy, 2012, 42(6): 872-882. doi:10.1111/j.1365-2222.2011.03931.x.
[40] Cho JS, Moon YM, Park IH, et al. Effects of histone deacetylase inhibitor on extracellular matrix production in human nasal polyp organ cultures[J]. Am J Rhinol Allergy, 2013, 27(1): 18-23. doi:10.2500/ajra.2013.27.3827.
[41] Yang WW, Ernst P. SET/MLL family proteins in hematopoiesis and leukemia[J]. Int J Hematol, 2017, 105(1): 7-16. doi:10.1007/s12185-016-2118-8.
[42] Theocharisa S, Margeli A, Kouraklis G. Peroxisome proliferator activated receptor-gamma ligands as potent antineoplastic agents[J]. Curr Med Chem Anticancer Agents, 2003, 3(3): 239-251. doi:10.2174/1568011033482431.
[43] Wei JJ, Wu XY, Peng Y, et al. Regulation of HMGA1 expression by microRNA-296 affects prostate cancer growth and invasion[J]. Clin Cancer Res, 2011, 17(6): 1297-1305. doi:10.1158/1078-0432.CCR-10-0993.
[44] Bartel DP. Metazoan microRNAs[J]. Cell, 2018, 173(1): 20-51. doi:10.1016/j.cell.2018.03.006.
[45] Turner ML, Schnorfeil FM, Brocker T. microRNAs regulate dendritic cell differentiation and function[J]. J Immunol, 2011, 187(8): 3911-3917. doi:10.4049/jimmunol.1101137.
[46] Luo X, Han MM, Liu JQ, et al. Epithelial cell-derived micro RNA-146a generates interleukin-10-producing monocytes to inhibit nasal allergy[J]. Sci Rep, 2015, 5: 15937. doi:10.1038/srep15937.
[47] Elbehidy RM, Youssef DM, El-Shal AS, et al. microRNA-21 as a novel biomarker in diagnosis and response to therapy in asthmatic children[J]. Mol Immunol, 2016, 71: 107-114. doi:10.1016/j.molimm.2015.12.015.
[48] Zhang XH, Zhang YN, Li HB, et al. Overexpression of miR-125b, a novel regulator of innate immunity, in eosinophilic chronic rhinosinusitis with nasal polyps[J]. Am J Respir Crit Care Med, 2012, 185(2): 140-151. doi:10.1164/rccm.201103-0456OC.
[49] Xuan LJ, Luan G, Wang Y, et al. microRNAs regulating mucin type O-glycan biosynthesis and transforming growth factor β signaling pathways in nasal mucosa of patients with chronic rhinosinusitis with nasal polyps in Northern China[J]. Int Forum Allergy Rhinol, 2019, 9(1): 106-113. doi:10.1002/alr.22230.
[50] Luo XQ, Shao JB, Xie RD, et al. Micro RNA-19a interferes with IL-10 expression in peripheral dendritic cells of patients with nasal polyposis[J]. Oncotarget, 2017, 8(30): 48915-48921. doi:10.18632/oncotarget.16555.
[51] Qing X, Zhang YQ, Peng Y, et al. miR-142-3p regulates inflammatory response by contributing to increased TNF-α in chronic rhinosinusitis with nasal polyposis[J]. Ear Nose Throat J, 2021, 100(1): NP50-NP56. doi:10.1177/0145561319847972.
[52] Mitoma H, Horiuchi T, Tsukamoto H, et al. Molecular mechanisms of action of anti-TNF-α agents - Comparison among therapeutic TNF-α antagonists[J]. Cytokine, 2018, 101: 56-63. doi:10.1016/j.cyto.2016.08.014.
[53] Thorley AJ, Ford PA, Giembycz MA, et al. Differential regulation of cytokine release and leukocyte migration by lipopolysaccharide-stimulated primary human lung alveolar type II epithelial cells and macrophages[J]. J Immunol, 2007, 178(1): 463-473. doi:10.4049/jimmunol.178.1.463.
[54] Ma ZX, Shen Y, Zeng Q, et al. miR-150-5p regulates EGR2 to promote the development of chronic rhinosinusitis via the DC-Th axis[J]. Int Immunopharmacol, 2018, 54: 188-197. doi:10.1016/j.intimp.2017.11.011.
[55] Doncel-Pérez E, Mateos-Hernández L, Pareja E, et al. Expression of early growth response gene-2 and regulated cytokines correlates with recovery from guillain-Barré syndrome[J]. J Immunol, 2016, 196(3): 1102-1107. doi:10.4049/jimmunol.1502100.
[56] Kim TD, Jung HR, Seo SH, et al. microRNA-150 modulates intracellular Ca2+ levels in naïve CD8+ T cells by targeting TMEM20[J]. Sci Rep, 2017, 7(1): 2623. doi:10.1038/s41598-017-02697-x.
[57] Yu HL, Ju JB, Liu JD, et al. Aberrant expression of miR-663 and transforming growth factor-β1 in nasal polyposis in children[J]. Exp Ther Med, 2018, 15(5): 4550-4556. doi:10.3892/etm.2018.5927.
[58] Park IH, Um JY, Hong SM, et al. Metformin reduces TGF-β1-induced extracellular matrix production in nasal polyp-derived fibroblasts[J]. Otolaryngol Head Neck Surg, 2014, 150(1): 148-153. doi:10.1177/0194599813513880.
[59] Luo Q, Zhang ZY, Liu D, et al. Human neutrophil elastase induces MUC5AC overexpression in chronic rhinosinusitis through tumour necrosis factor-α converting enzyme[J]. Acta Otolaryngol, 2016, 136(6): 641-648. doi:10.3109/00016489.2016.1144145.
[60] Wang J, Zhu MC, Wang LL, et al. Amphiregulin potentiates airway inflammation and mucus hypersecretion induced by urban particulate matter via the EGFR-PI3Kα-AKT/ERK pathway[J]. Cell Signal, 2019, 53: 122-131. doi:10.1016/j.cellsig.2018.10.002.
[61] Xu R, Li Q, Zhou J, et al. Secretoneurin induces airway mucus hypersecretion by enhancing the binding of EGF to NRP1[J]. Cell Physiol Biochem, 2014, 33(2): 446-456. doi:10.1159/000358625.
[62] Yan DQ, Ye Y, Zhang J, et al. Human neutrophil elastase induces MUC5AC overexpression in chronic rhinosinusitis through miR-146a[J]. Am J Rhinol Allergy, 2020, 34(1): 59-69. doi:10.1177/1945892419871798.
[63] Choksi SP, Lauter G, Swoboda P, et al. Switching on Cilia: transcriptional networks regulating ciliogenesis[J]. Development, 2014, 141(7): 1427-1441. doi:10.1242/dev.074666.
[64] Spassky N, Meunier A. The development and functions of multiciliated epithelia[J]. Nat Rev Mol Cell Biol, 2017, 18(7): 423-436. doi:10.1038/nrm.2017.21.
[65] Callejas-Díaz B, Fernandez G, Fuentes M, et al. Integrated mRNA and microRNA transcriptome profiling during differentiation of human nasal polyp epithelium reveals an altered ciliogenesis[J]. Allergy, 2020, 75(10): 2548-2561. doi:10.1111/all.14307.
[66] Diesch J, Zwick A, Garz AK, et al. A clinical-molecular update on azanucleoside-based therapy for the treatment of hematologic cancers[J]. Clin Epigenetics, 2016, 8: 71. doi:10.1186/s13148-016-0237-y.
[67] Fenaux P, Mufti GJ, Hellström-Lindberg E, et al. Azacitidine prolongs overall survival compared with conventional care regimens in elderly patients with low bone marrow blast count acute myeloid leukemia[J]. J Clin Oncol, 2010, 28(4): 562-569. doi:10.1200/JCO.2009.23.8329.
[68] Yeo NK, Park WJ, Eom DW, et al. Effects of azathioprine and its metabolites on inflammatory cytokines in human nasal polyp organ cultures[J]. Int Forum Allergy Rhinol, 2019, 9(6): 648-655. doi:10.1002/alr.22303.
[69] Yoon S, Eom GH. HDAC and HDAC inhibitor: from cancer to cardiovascular diseases[J]. Chonnam Med J, 2016, 52(1): 1-11. doi:10.4068/cmj.2016.52.1.1.
[1] 敖天, 程雷. 慢性鼻窦炎伴鼻息肉的内型研究及其指导下的精准控制与治疗[J]. 山东大学耳鼻喉眼学报, 2022, 36(3): 7-14.
[2] 熊攀辉,沈暘,杨玉成. 基于表型和内在型的慢性鼻窦炎诊治进展[J]. 山东大学耳鼻喉眼学报, 2022, 36(3): 15-19.
[3] 姚爽,娄鸿飞. 慢性鼻窦炎的内在型研究进展及精准医疗[J]. 山东大学耳鼻喉眼学报, 2022, 36(3): 20-29.
[4] 梁旭,史丽. 慢性鼻窦炎生物靶向药物治疗的研究进展[J]. 山东大学耳鼻喉眼学报, 2022, 36(3): 30-35.
[5] 石帅,郑泉,程雷. 度普利尤单抗在慢性鼻窦炎伴鼻息肉治疗中的研究进展[J]. 山东大学耳鼻喉眼学报, 2022, 36(3): 36-42.
[6] 王欢,胡俐,余洪猛. 慢性鼻窦炎相关嗅觉功能障碍研究进展[J]. 山东大学耳鼻喉眼学报, 2022, 36(3): 43-49.
[7] 宜若男,陈福权. 嗜酸性粒细胞与嗅觉功能障碍[J]. 山东大学耳鼻喉眼学报, 2022, 36(3): 50-55.
[8] 谷钰,万鑫,肖自安. 中性粒细胞和嗜酸性粒细胞在慢性鼻窦炎中的相互影响及临床治疗思考[J]. 山东大学耳鼻喉眼学报, 2022, 36(3): 56-63.
[9] 林海,朱莹,张维天. 慢性鼻窦炎发病中离子通道作用研究进展[J]. 山东大学耳鼻喉眼学报, 2022, 36(3): 64-70.
[10] 乔新杰,赵玉林. 慢性鼻窦炎中上皮间质转化信号转导通路及其他相关因子的研究进展[J]. 山东大学耳鼻喉眼学报, 2022, 36(3): 71-77.
[11] 黄丹怡,张婷,陈静,张薇. 上皮屏障在慢性鼻窦炎伴鼻息肉中的研究进展[J]. 山东大学耳鼻喉眼学报, 2022, 36(3): 78-83.
[12] 于龙刚,姜彦. 鼻细菌微生物组与慢性鼻窦炎伴鼻息肉相关性的研究进展[J]. 山东大学耳鼻喉眼学报, 2022, 36(3): 92-97.
[13] 资昊坤,肖旭平,李云秋. 口服糖皮质激素在慢性鼻窦炎伴鼻息肉围手术期的应用现状[J]. 山东大学耳鼻喉眼学报, 2022, 36(3): 98-103.
[14] 曹轩,肖旭平,李云秋. 透明质酸在慢性鼻窦炎中的研究进展[J]. 山东大学耳鼻喉眼学报, 2022, 36(3): 104-109.
[15] 刘一潼,周穗子,邱前辉. NLRP3炎症小体在慢性鼻窦炎和变应性鼻炎中的研究进展[J]. 山东大学耳鼻喉眼学报, 2022, 36(3): 142-146.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed   
[1] 马宝峰,党光福 . 折叠式人工晶状体经巩膜缝线固定术16例[J]. 山东大学耳鼻喉眼学报, 2008, 22(4): 376 -378 .
[2] 封新荣,张红霞,许贞姬,曲君君 . 射频消融术治疗慢性肥厚性鼻炎[J]. 山东大学耳鼻喉眼学报, 2006, 20(3): 225 -226 .
[3] 张秋贵,何海燕 . 青岛地区变应性鼻炎常见变应原调查分析[J]. 山东大学耳鼻喉眼学报, 2006, 20(6): 520 -521 .
[4] 李海燕,赵秋良,韩晓攀,于丽 . 阻塞性睡眠呼吸暂停综合征患者自由基损伤与高血压的关系[J]. 山东大学耳鼻喉眼学报, 2006, 20(6): 527 -529 .
[5] 宋西成,张华,张庆泉 . 血循环DNA的检测与头颈部肿瘤的诊断[J]. 山东大学耳鼻喉眼学报, 2006, 20(5): 385 -387 .
[6] 詹善强,孔凡俐,操文娟,汤杰,沈菊,詹文波 . 电刀切除扁桃体236例[J]. 山东大学耳鼻喉眼学报, 2006, 20(6): 559 -560 .
[7] 张 虎,艾 琴 . 甲状腺癌再手术15例[J]. 山东大学耳鼻喉眼学报, 2007, 21(2): 166 -167 .
[8] 胡 明,李招娜,李云杰,陶祥臣 . 硬性透气性角膜接触镜对圆锥角膜的塑形作用[J]. 山东大学耳鼻喉眼学报, 2007, 21(2): 171 -173 .
[9] 娄 锋 . 喉上神经阻滞预防喉显微手术心血管反应[J]. 山东大学耳鼻喉眼学报, 2008, 22(2): 173 -175 .
[10] 陈伟雄 王跃建 曾勇 何发尧 张剑利 郑立岗 汤苏成. 胸骨上小切口无注气内镜甲状腺手术的临床体会[J]. 山东大学耳鼻喉眼学报, 2009, 23(4): 24 -26 .