Journal Search Engine
Search Advanced Search Adode Reader(link)
Download PDF Export Citaion korean bibliography PMC previewer
ISSN : 1226-7155(Print)
ISSN : 2287-6618(Online)
International Journal of Oral Biology Vol.45 No.1 pp.1-7
DOI : https://doi.org/10.11620/IJOB.2020.45.1.1

Malignant transformation of oral lichen planus and related genetic factors

Eurim C. Hwang1, Se-Young Choi1, Jeong Hee Kim2,3*
1Department of Oral Physiology, School of Dentistry, Seoul National University, Seoul 03080, Republic of Korea
2Department of Oral Biochemistry and Molecular Biology, College of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
3Department of KHU-KIST Converging Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
*Correspondence to: Jeong Hee Kim, E-mail: jhkimh@khu.ac.kr
February 17, 2020 March 18, 2020 March 20, 2020

Abstract


Oral lichen planus (OLP) is a chronic inflammatory disease observed in approximately 0.5–2.2% of the population, and it is recognized as a premalignant lesion that can progress into oral squamous cell carcinoma (OSCC). The rate of malignant transformation is approximately 1.09–2.3%, and the risk factors for malignant transformation are age, female, erosive type, and tongue site location. Malignant transformation of OLP is likely related to the low frequency of apoptotic phenomena. Therefore, apoptosis-related genetic factors, like p53, BCL-2, and BAX are reviewed. Increased p53 expression and altered expression of BCL-2 and BAX were observed in OLP patients, and the malignant transformation rate in these patients was relatively higher. The involvement of microRNA (miRNA) in the malignant transformation of OLP is also reviewed. Because autophagy is involved in cell survival and death through the regulation of various cellular processes, autophagy-related genetic factors may function as factors for malignant transformation. In OLP, decreased levels of ATG9B mRNA and a higher expression of IGF1 were observed, suggesting a reduction in cell death and autophagic response. Activated IGF1-PI3K/AKT/mTor cascade may play an important role in a signaling pathway related to the malignant transformation of OLP to OSCC. Recent research has shown that miRNAs, such as miR-199 and miR-122, activate the cascade, increasing the prosurvival and proproliferative signals.



초록


    © The Korean Academy of Oral Biology. All rights reserved.

    This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/bync/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

    Introduction

    Oral lichen planus (OLP) is commonly observed chronic inflammatory disease of the mucous membrane surface of the mouth [1-3]. This disease is observed in about 0.5–2.2% of the populations [3-8]. Incidence rate of OLP is related with older age, female and location of the lesion. Among the patients, incidence of 30–60 year-old female is the highest [3-9]. Clinically, OLP mainly manifests three different forms: reticular, atrophic and erosive. They are generally simplified into two categories: erosive (erosive lesion) and non-erosive (reticular and atrophic lesions) [3,10,11].

    The relationship of OLP and squamous cell carcinoma has attracted people’s attention and studies on this subject have been performed [3,12,13]. According to the World Health Organization (WHO), OLP is categorized into the group of potentially malignant disorders, while its most severe complication is the progression into oral squamous cell carcinoma (OSCC) [14,15]. Possibility of the malignant transformation of OLP has been hypothesized and researchers have been searching for the evidence for malignant transformation. The OLP was recognized as pre-malignant by WHO, but the rate of malignant transformation remains unknown. Moreover, the factors involved in the malignant transformation of OLP to OSCC is not much known. Therefore, in this brief review, the malignant transformation of OLP and related genetic factors, especially in relation with apoptosis and autophagy, are discussed.

    Malignant Transformation to Oral Squamous Cell Carcinoma

    For many years, the possibility of malignant transformation of OLP has been discussed and reported [12,13]. Since OLP is longstanding inflammation, patients with OLP are believed to be at higher risk for OSCC. In order to elucidate the relationship between OLP and OSCC, meta-analysis studies, systematic review, and population-based studies have been performed.

    A meta-analysis study was performed in order to investigate the possible risk factors for OLP malignant transformation to OSCC [16]. MEDLINE, Scopus and Web of Science were used as database. Results showed that approximately 1.1% of OLP patients develop OSCC. A higher incidence of malignant transformation was found among smokers, alcoholics, and hepatitis C virus (HCV)-infected patients compared to patients without these risk factors [3,17]. The involvement of HCV as a risk factor of OLP malignant transformation was also reported in other studies [3,17]. However, the authors reported that these associations should be further investigated.

    In order to determine the malignant transformation rate of OLP and its risk factors, data bases of PubMed, Scopus and Web of Science were systemically reviewed [13]. The results of the systematic review showed that 92 of 6,559 OLP patients developed OSCC, and thus the malignant transformation rate was found to be 1.37%. This study suggested that erosive type, female and tongue site should be considered as risk factors for malignant transformation of OLP. Similar risk factors for OLP malignant transformation such as age, female and erosive form were suggested by other study [18]. These studies also suggested that to diagnose OLP, strict clinical and histological criteria to diagnose OLP and sounder methodological observation studies should be performed.

    Another systematic approach was performed to evaluate the relationship between malignant transformation and OLP [12]. It was reported that among 7,806 patients with OLP, 85 developed SCC. The overall rate of transformation was 1.09 % for OLP. Average age of the patients was 60.8 years. A slight predominance of female patients among those who experienced malignant transformation was noticed. The most common subsite of malignant transformation was the tongue. The average time from diagnosis of OLP to transformation was 51.4 months.

    A population-based retrospective cohort study showed that among the 303 OLP patients 7 had OSCC at 20 years after OLP incidence with malignant transformation rate of 2.3% [19]. The median time for the development of OSCC in OLP patients was 2.9 years which was 14.7 years earlier than the reference group. Patients with OLP, particularly the erosive type, have an increased incidence of OSCC development.

    These studies showed that the malignant transformation rate of OLP to OSCC was approximately 1.09–2.3%. The high-risk group is older age, female, erosive type, and tongue site location. Patients with other inflammatory disease such as HCV can be categorized as a high-risk group. The OLP patients in these categories need to be monitored carefully with regular observation and continued follow-up should be done.

    Genetic Factors Related to Oral Lichen Planus

    1. Apoptosis related genetic factors

    Apoptosis, a form of programmed cell death, is a selective physiological process that plays an important role in the balance between cell replication and cell death [20]. A wide range of stimuli can be integrated to trigger the irreversible decision of cell death. The low frequency of apoptotic phenomena in epithelial cells of OLP suggests the possibility of it creating a favorable substrate for malignant transformation [21].

    Protein p53 is a transcription factor involved in DNA repair and destruction of defective cells through the induction of apoptosis and cell cycle arrest [22-24]. Increased expression of p53 in keratinocytes in OLP and the suprabasal layer localization of p53 in 20% of OLP cases was reported [25]. The association between suprabasal expression of p53 and increased risk of malignant transformation was suggested in other studies [26,27]. Protein p53 is confined to the basal cell layer in benign lesion and normal mucosa. Out of 35 patients with premalignant lesion, 7 patients showed p53 expression above the basal cell layer and 6 of these 7 developed carcinomas. In some cases, both p53 and MDM-2 was overexpressed in OLP compared to normal mucosa [27]. MDM-2 acts as an E3 ubiquitin ligase of p53 and functions as a major negative regulator of p53 [28].

    Immunohistochemical expression of p53 was compared between normal mucosa and OLP. The positive ratio of p53 in OLP patients was reported to be 69.9% [29] and 64% [30]. In some studies, even higher positive ratio of p53 was reported; 86.7% [31] and 90.9% [32], respectively. These studies, showing relatively high positive ratio of p53, claimed that mutant form of p53 was reacted. Thus revealing the presence of the mutation in the p53 protein and hence OLP has potential malignant transformation.

    A follow up study was done with OLP patients who exhibited increased p53 expression. After 96 months of follow-up, enhanced p53 expression was observed in patients progressed to OSCC [33]. This observation suggested the involvement of p53 in malignant transformation to OSCC. A recent study showed that the level of unstimulated whole salivary p53 in subjects with OLP and OSCC was higher (5.63 ± 1.08) compared to only OLP (0.94 ± 0.31) [34]. All of these studies suggest the involvement of p53 in malignant transformation of OLP to OSCC.

    Proteins of B-cell lymphoma-2 (BCL-2) control the intrinsic mitochondrial apoptosis pathway [35,36]. Among programmed cell death, intrinsic apoptosis is known as the most common type of cell death [37]. BCL-2 proteins with anti-apoptotic properties were discovered, which includes B-cell lymphomaextra- large (BCL-XL), myeloid cell leukemia‑1 (MCL-1), and BCL-2-like protein 2 (BCL-W) [36,38,39]. BCL-2 associated X protein (BAX) is a homologous binding partner of BCL-2. BAX has a pro-apoptotic property unlike other anti-apoptotic BCL-2 proteins. Among pro-apoptotic proteins, BAX, BCL-2 antagonist killer 1 (BAK), and BCL-2-related antagonist killer 1 (BOK) belong to the effector protein sub-class [23,36,39-41].

    The altered expression of BCL-2 and BAX, and increase in capase-3, an executor protease of apoptosis were observed in lichen planus biopsy samples [42]. In OLP cases, higher rate of positive expression for BCL-2 (97%) was observed. It was suggested that overexpression of BCL-2 is associated with OLP and appears to play a role in malignant transformation [43]. In addition, an increased expression of BCL-2 was found in the lymphocytic infiltrate which is characteristically encountered in OLP lesions. The expression of BAX was elevated in epithelial basement keratin in OLP group compared to that in the control group, which includes healthy subjects [44]. In other study, 92% of OLP patients had a positive expression of BCL-2 in inflammatory infiltrate [45].

    Immunohistochemical expression of BCL-2 was compared between normal mucosa and OLP. There was no positive expression in normal mucosa, but in OLP patient group, OLP positive rate was found to be 19.9% [29]. Similarly, BCL-2 positive rate of 16.7% in OLP was found in other study [46]. Strong expression of MCL-1 was observed in OLP and oral cancer cell lines. The authors suggested that MCL-1 could be a novel biomarker for the malignant potential of OLP [47]. Positive expression of BCL-2 was reported in 25 of 30 OSCC cases [48].

    These data suggest that BCL-2 plays an important role in the inhibition of apoptosis of lymphocytes, while BAX is involved in the induction of apoptosis of keratinocytes. Alterations in the expression of BCL-2/BAX may be involved in malignant transformation of OLP to OSCC.

    Micro RNA (miRNA) is small non-coding RNA molecule of 21–25 nucleotide that functions in RNA silencing and posttranscriptional regulation of gene expression. It usually functions by base-pairing with complementary sequences with mRNA molecules [49,50]. Recently, increasing evidence suggested that miRNAs are involved in the regulation of pathogenesis of OLP [51-53]. It was shown that miRNA-27b- 3p regulates keratinocyte apoptosis through cyclinD/BCL- 2 signaling [54]. Overexpression of miR-802 in OLP induces apoptosis of oral keratinocyte by targeting bcl-2 mRNA. However, vitamin D (VD)/VD receptor signaling could suppress the apoptosis by targeting miRNA-805 [55]. It was confirmed that the level of miR-26a and 26-b are decreased in OLP patients. In addition, mi-RNA 26a/b have protective action in OLP by inhibiting apoptosis of oral keratinocytes by targeting Protein Kinase C δ and cluster of the differentiation 38 (CD38) genes [56].

    2. Autophagy related genetic factors

    Autophagy is the major lysosomal intracellular degradation system that involves the delivery of cytoplasmic cargo to the lysosome. Autophagy is essential in survival, differentiation, development and homeostasis [57,58]. To promote cell survival or death, autophagy can be activated in response to various cellular and environmental stress conditions [59,60].

    In order to investigate the role of autophagy in T cell on OLP pathogenesis, mRNA expression of autophagy-associated gene in OLP patients as analyzed [61]. Insulin-like growth factor 1 (IGF1) gene expression in the blood of OLP patients was significantly higher than that in controls. The high expression was observed specially in female and middle-aged OLP patients compared to males and other age groups. IGF1 signaling could reduce cell death and possibly inhibit the autophagic response under the condition of nutritional stress [62,63]. Autophagy related 9 homolog B (ATG9B) mRNA level was significantly lower in OLP patients. Other genes such as hepatocyte growth factor-regulated tyrosine kinase substrate (HGS), estrogen receptor 1 (ESR1) and synuclein alpha (SNCA) showed no significant difference between experimental and control groups. These data suggest that dysregulation of T cell autophagy is involved in immune response of OLP.

    IGF1 signaling, initiated from activation of IGF1 receptor (IGF1R), plays critical role in regulation of cell growth and survival [64,65]. IGF1 signaling pathway has been identified to be one of the regulators of the autophagy [66]. Phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTor) cascade is an important intracellular signaling pathway in cell quiescence, proliferation, protein synthesis, cancer and autophagy. And it is activated after the phosphorylation of IGF1R. The expression of p-IGF1R was observed to be increased in OLP patients [67]. It is also reported that the expression of pAkt, p-mTor, ULK1 and LC3B are significantly increased in local T cells of OLP patients compared with that in controls [68]. The level of microtubule associated protein 1 light chain 3β (LC3B) was higher in non-erosive OLP than in erosive OLP and control group. Thus, LC3B may be used as a marker to monitor the disease severity [68]. Taken together, these results suggest that activated IGF1-PI3K/Akt/mTor-autophagy pathway may have a critical role in local T-cell mediated immunoregulatory mechanism of OLP.

    Recently, Wang et al. [69] used miRNA microarray and found that miR-122 and miR-199 demonstrated significant decrease in OLP patient group. By searching online data base, it was predicted that mTor was the target of miR-199 and Akt1 was the target of miR-122. These miRNAs may serve as potential therapeutic targets [69]. These results demonstrated that PI3K/Akt/mTor signaling pathway is activated in OLP patients. As a result, a cascade of pro-survival and pro-proliferative signals are triggered, a transduction that may affect the basal cells in OLP patients. Thus, the lifetime of these cells may be extended, which may cause dysplastic variations and may increase the risk of malignant transformation during OLP [69,70].

    Conflicts of Interest

    No potential conflict of interest relevant to this article was reported.

    Figure

    Table

    Reference

    1. Zhang L, Michelsen C, Cheng X, Zeng T, Priddy R, Rosin MP. Molecular analysis of oral lichen planus. A premalignant lesion? Am J Pathol 1997;151:323-7.
    2. Georgescu SR, Ene CD, Nicolae I, Mitran M, Musetescu A, Matei C, Rusu LC, Tampa M. Reflectometric analysis for identification of various pathological conditions associated with lichen planus. Rev Chim 2017;68:1103-8.
    3. Giannetti L, Dello Diago AM, Spinas E. Oral lichen planus. J Biol Regul Homeost Agents 2018;32:391-5.
    4. Farhi D, Dupin N. Pathophysiology, etiologic factors, and clinical management of oral lichen planus, part I: facts and controversies. Clin Dermatol 2010;28:100-8.
    5. Torrente-Castells E, Figueiredo R, Berini-Aytés L, Gay- Escoda C. Clinical features of oral lichen planus. A retrospective study of 65 cases. Med Oral Patol Oral Cir Bucal 2010;15:e685-90.
    6. Bombeccari GP, Guzzi G, Tettamanti M, Giannì AB, Baj A, Pallotti F, Spadari F. Oral lichen planus and malignant transformation: a longitudinal cohort study. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2011;112:328-34.
    7. Shen Z, Liu W, Zhu L, Feng J, Tang G, Zhou Z. A retrospective clinicopathological study on oral lichen planus and malignant transformation: analysis of 518 cases. Med Oral Patol Oral Cir Bucal 2012;17:e943-7.
    8. Bardellini E, Amadori F, Flocchini P, Bonadeo S, Majorana A. Clinicopathological features and malignant transformation of oral lichen planus: a 12-years retrospective study. Acta Odontol Scand 2013;71:834-40.
    9. Silverman S Jr, Gorsky M, Lozada-Nur F. A prospective follow-up study of 570 patients with oral lichen planus: persistence, remission, and malignant association. Oral Surg Oral Med Oral Pathol 1985;60:30-4.
    10. Zhou G, Zhang J, Ren XW, Hu JY, Du GF, Xu XY. Increased Eurim C. Hwang, et al. Malignant transformation of OLP and genetic factors B7-H1 expression on peripheral blood T cells in oral lichen planus correlated with disease severity. J Clin Immunol 2012;32:794-801.
    11. Lorenzini G, Viviano M, Chisci E, Chisci G, Picciotti M. A comparative immunohistochemical and immunophenotypical study on lymphocytes expression in patients affected by oral lichen planus. J Oral Pathol Med 2013;42:642-7.
    12. Fitzpatrick SG, Hirsch SA, Gordon SC. The malignant transformation of oral lichen planus and oral lichenoid lesions: a systematic review. J Am Dent Assoc 2014;145:45-56.
    13. Giuliani M, Troiano G, Cordaro M, Corsalini M, Gioco G, Lo Muzio L, Pignatelli P, Lajolo C. Rate of malignant transformation of oral lichen planus: a systematic review. Oral Dis 2019;25:693-709.
    14. Shen ZY, Liu W, Feng JQ, Zhou HW, Zhou ZT. Squamous cell carcinoma development in previously diagnosed oral lichen planus: de novo or transformation? Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2011;112:592-6.
    15. Peng Q, Zhang J, Ye X, Zhou G. Tumor-like microenvironment in oral lichen planus: evidence of malignant transformation? Expert Rev Clin Immunol 2017;13:635-43.
    16. Aghbari SMH, Abushouk AI, Attia A, Elmaraezy A, Menshawy A, Ahmed MS, Elsaadany BA, Ahmed EM. Malignant transformation of oral lichen planus and oral lichenoid lesions: a meta-analysis of 20095 patient data. Oral Oncol 2017;68:92-102.
    17. Abbate G, Foscolo AM, Gallotti M, Lancella A, Mingo F. Neoplastic transformation of oral lichen: case report and review of the literature. Acta Otorhinolaryngol Ital 2006;26:47-52.
    18. Marder MZ, Deesen KC. Transformation of oral lichen planus to squamous cell carcinoma: a literature review and report of case. J Am Dent Assoc 1982;105:55-60.
    19. Laniosz V, Torgerson RR, Ramos-Rodriguez AJ, Ma JE, Mara KC, Weaver AL, Bruce AJ. Incidence of squamous cell carcinoma in oral lichen planus: a 25-year population-based study. Int J Dermatol 2019;58:296-301.
    20. Meier P, Finch A, Evan G. Apoptosis in development. Nature 2000;407:796-801.
    21. Bascones-Ilundain C, González-Moles MA, Campo-Trapero J, Gil-Montoya JA, Esparza-Gómez GC, Cano-Sánchez J, Bascones-Martínez A. No differences in caspase-3 and Bax expression in atrophic-erosive vs. reticular oral lichen planus. J Eur Acad Dermatol Venereol 2008;22:204-12.
    22. Gotz C, Montenarh M. P53 and its implication in apoptosis (review). Int J Oncol 1995;6:1129-35.
    23. Chen J. The cell-cycle arrest and apoptotic functions of p53 in tumor initiation and progression. Cold Spring Harb Perspect Med 2016;6:a026104.
    24. Charni M, Aloni-Grinstein R, Molchadsky A, Rotter V. p53 on the crossroad between regeneration and cancer. Cell Death Differ 2017;24:8-14.
    25. Tanda N, Mori S, Saito K, Ikawa K, Sakamoto S. Expression of apoptotic signaling proteins in leukoplakia and oral lichen planus: quantitative and topographical studies. J Oral Pathol Med 2000;29:385-93.
    26. Cruz IB, Snijders PJ, Meijer CJ, Braakhuis BJ, Snow GB, Walboomers JM, van der Waal I. p53 expression above the basal cell layer in oral mucosa is an early event of malignant transformation and has predictive value for developing oral squamous cell carcinoma. J Pathol 1998;184:360-8.
    27. Oliveira Alves M, Balducci I, Rodarte Carvalho Y, Cabral L, Nunes F, Almeida J. Evaluation of the expression of p53, MDM2, and SUMO-1 in oral lichen planus. Oral Dis 2013;19:775-80.
    28. Wade M, Wong ET, Tang M, Stommel JM, Wahl GM. Hdmx modulates the outcome of p53 activation in human tumor cells. J Biol Chem 2006;281:33036-44.
    29. Shailaja G, Kumar JV, Baghirath PV, Kumar U, Ashalata G, Krishna AB. Estimation of malignant transformation rate in cases of oral epithelial dysplasia and lichen planus using immunohistochemical expression of Ki-67, p53, BCL-2, and BAX markers. Dent Res J (Isfahan) 2015;12:235-42.
    30. Taniguchi Y, Nagao T, Maeda H, Kameyama Y, Warnakulasuriya KA. Epithelial cell proliferation in oral lichen planus. Cell Prolif 2002;35 Suppl 1:103-9.
    31. Fakhrjou A, Toutounchi SJS. Morphologic evaluation of P53 apoptotic signaling responses and proliferative activity of Ki- 67 in oral lichen planus, oral squamous cell carcinoma and normal specimens. J Med Sci 2012;12:51-6.
    32. Acay RR, Felizzola CR, de Araújo N, de Sousa SO. Evaluation of proliferative potential in oral lichen planus and oral lichenoid lesions using immunohistochemical expression of p53 and Ki67. Oral Oncol 2006;42:475-80.
    33. Valente G, Pagano M, Carrozzo M, Carbone M, Bobba V, Palestro G, Gandolfo S. Sequential immunohistochemical p53 expression in biopsies of oral lichen planus undergoing malignant evolution. J Oral Pathol Med 2001;30:135-40.
    34. Agha-Hosseini F, Mirzaii-Dizgah I, Miri-Zarandi N. Unstimulated salivary p53 in patients with oral lichen planus and squamous cell carcinoma. Acta Med Iran 2015;53:439-43.
    35. Oltersdorf T, Elmore SW, Shoemaker AR, Armstrong RC, Augeri DJ, Belli BA, Bruncko M, Deckwerth TL, Dinges J, Hajduk PJ, Joseph MK, Kitada S, Korsmeyer SJ, Kunzer AR, Letai A, Li C, Mitten MJ, Nettesheim DG, Ng S, Nimmer PM, O’Connor JM, Oleksijew A, Petros AM, Reed JC, Shen W, Tahir SK, Thompson CB, Tomaselli KJ, Wang B, Wendt MD, Zhang H, Fesik SW, Rosenberg SH. An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature 2005;435:677-81.
    36. Edlich F. BCL-2 proteins and apoptosis: recent insights and unknowns. Biochem Biophys Res Commun 2018;500:26-34.
    37. Hotchkiss RS, Strasser A, McDunn JE, Swanson PE. Cell death. N Engl J Med 2009;361:1570-83.
    38. Chen HC, Kanai M, Inoue-Yamauchi A, Tu HC, Huang Y, Ren D, Kim H, Takeda S, Reyna DE, Chan PM, Ganesan YT, Liao CP, Gavathiotis E, Hsieh JJ, Cheng EH. An interconnected hierarchical model of cell death regulation by the BCL-2 family. Nat Cell Biol 2015;17:1270-81.
    39. Campbell KJ, Tait SWG. Targeting BCL-2 regulated apoptosis in cancer. Open Biol 2018;8:180002.
    40. Oltvai ZN, Milliman CL, Korsmeyer SJ. Bcl-2 heterodimerizes in vivo with a conserved homolog, Bax, that accelerates programmed cell death. Cell 1993;74:609-19.
    41. Westphal D, Kluck RM, Dewson G. Building blocks of the apoptotic pore: how Bax and Bak are activated and oligomerize during apoptosis. Cell Death Differ 2014;21:196-205.
    42. Abdel-Latif AM, Abuel-Ela HA, El-Shourbagy SH. Increased caspase-3 and altered expression of apoptosis-associated proteins, Bcl-2 and Bax in lichen planus. Clin Exp Dermatol 2009;34:390-5.
    43. Arreaza AJ, Rivera H, Correnti M. Expression of COX-2 and bcl-2 in oral lichen planus lesions and lichenoid reactions. Ecancermedicalscience 2014;8:411.
    44. Li XR, Guo J, Li XM. [The expression and changes of apoptosis protein Bcl-2 and Bax in oral lichen planus]. Shanghai Kou Qiang Yi Xue 2015;24:465-9. Chinese.
    45. Pigatti FM, Taveira LA, Soares CT. Immunohistochemical expression of Bcl-2 and Ki-67 in oral lichen planus and leukoplakia with different degrees of dysplasia. Int J Dermatol 2015;54:150-5.
    46. Sousa FA, Paradella TC, Carvalho YR, Rosa LE. Immunohistochemical expression of PCNA, p53, bax and bcl-2 in oral lichen planus and epithelial dysplasia. J Oral Sci 2009;51:117- 21.
    47. Shin JA, Seo JM, Oh S, Cho SD, Lee KE. Myeloid cell leukemia- 1 is a molecular indicator for malignant transformation of oral lichen planus. Oncol Lett 2016;11:1603-7.
    48. Sutariya RV, Manjunatha BS. Immunohistochemical study of p21 and Bcl-2 in leukoplakia, oral submucous fibrosis and oral squamous cell carcinoma. J Exp Ther Oncol 2016;11:285-92.
    49. Ambros V. The functions of animal microRNAs. Nature 2004;431:350-5.
    50. Lu TX, Rothenberg ME. MicroRNA. J Allergy Clin Immunol 2018;141:1202-7.
    51. Hwang HW, Mendell JT. MicroRNAs in cell proliferation, cell death, and tumorigenesis. Br J Cancer 2006;94:776-80.
    52. Gassling V, Hampe J, Açil Y, Braesen JH, Wiltfang J, Häsler R. Disease-associated miRNA-mRNA networks in oral lichen planus. PLoS One 2013;8:e63015.
    53. Ma H, Wu Y, Yang H, Liu J, Dan H, Zeng X, Zhou Y, Jiang L, Chen Q. MicroRNAs in oral lichen planus and potential miRNA- mRNA pathogenesis with essential cytokines: a review. Oral Surg Oral Med Oral Pathol Oral Radiol 2016;122:164-73.
    54. Chen J, Wang Y, Du G, Zhang W, Cao T, Shi L, Wang Y, Mi J, Tang G. Down-regulation of miRNA-27b-3p suppresses keratinocytes apoptosis in oral lichen planus. J Cell Mol Med 2019;23:4326-37.
    55. Zhao B, Xu N, Li R, Yu F, Zhang F, Yang F, Ge X, Li YC, Du J. Vitamin D/VDR signaling suppresses microRNA-802-induced apoptosis of keratinocytes in oral lichen planus. FASEB J 2019;33:1042-50.
    56. Du J, Gao R, Wang Y, Nguyen T, Yang F, Shi Y, Liu T, Liao W, Li R, Zhang F, Ge X, Zhao B. MicroRNA-26a/b have protective roles in oral lichen planus. Cell Death Dis 2020;11:15.
    57. Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell 2008;132:27-42.
    58. Mizushima N, Komatsu M. Autophagy: renovation of cells and tissues. Cell 2011;147:728-41.
    59. Baehrecke EH. Autophagy: dual roles in life and death? Nat Rev Mol Cell Biol 2005;6:505-10.
    60. Levine B, Mizushima N, Virgin HW. Autophagy in immunity and inflammation. Nature 2011;469:323-35.
    61. Tan YQ, Zhang J, Du GF, Lu R, Chen GY, Zhou G. Altered autophagy- associated genes expression in T cells of oral lichen planus correlated with clinical features. Mediators Inflamm 2016;2016:4867368.
    62. Gu Y, Wang C, Cohen A. Effect of IGF-1 on the balance between autophagy of dysfunctional mitochondria and apoptosis. FEBS Lett 2004;577:357-60.
    63. Troncoso R, Vicencio JM, Parra V, Nemchenko A, Kawashima Y, Del Campo A, Toro B, Battiprolu PK, Aranguiz P, Chiong M, Yakar S, Gillette TG, Hill JA, Abel ED, Leroith D, Lavandero S. Energy-preserving effects of IGF-1 antagonize starvationinduced cardiac autophagy. Cardiovasc Res 2012;93:320-9.
    64. Wang F, Tan YQ, Zhang J, Zhou G. Insulin-like growth factor 1 exhibits the pro-autophagic and anti-apoptotic activity on T cells of oral lichen planus. Int J Biol Macromol 2019;133:640-6.
    65. Smith TJ. Insulin-like growth factor-I regulation of immune function: a potential therapeutic target in autoimmune diseases? Pharmacol Rev 2010;62:199-236.
    66. Fu J, Hao L, Tian Y, Liu Y, Gu Y, Wu J. miR-199a-3p is involved in estrogen-mediated autophagy through the IGF-1/ mTOR pathway in osteocyte-like MLO-Y4 cells. J Cell Physiol 2018;233:2292-303.
    67. Ma RJ, Tan YQ, Zhou G. Aberrant IGF1-PI3K/AKT/MTOR signaling pathway regulates the local immunity of oral lichen planus. Immunobiology 2019;224:455-61.
    68. Zhang N, Zhang J, Tan YQ, Du GF, Lu R, Zhou G. Activated Akt/mTOR-autophagy in local T cells of oral lichen planus. Int Immunopharmacol 2017;48:84-90.
    69. Wang L, Wu W, Chen J, Li Y, Xu M, Cai Y. miR‑122 and miR‑199 synergistically promote autophagy in oral lichen planus by targeting the Akt/mTOR pathway. Int J Mol Med 2019;43:1373-81.
    70. Roopashree MR, Gondhalekar RV, Shashikanth MC, George J, Thippeswamy SH, Shukla A. Pathogenesis of oral lichen planus--a review. J Oral Pathol Med 2010;39:729-34.