HDACi联合ATRA重启宫颈癌细胞RARβ2表达及其抗肿瘤机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
宫颈癌最常见的女性生殖道恶性肿瘤,发病率和病死率居生殖道恶性肿瘤首位。每年全世界大约有53万新发病例,约50%患者死于该疾病,其中的80%来自发展中国家。早期宫颈癌一般推荐采用手术治疗,对于晚期转移和复发的病例,则失去了手术机会,常规采用化疗和放疗。然而,由于耐药的发生,患者身体耐受状况的恶化,放、化疗效果十分有限。因此,深入探讨宫颈癌发病机制,寻找更为安全有效、靶向性强的宫颈癌防治办法意义重大。
     靶向抑制组蛋白脱乙酰化酶(histone deacetylase, HDAC)是一种目前非常有前景的肿瘤治疗手段。组蛋白的乙酰化修饰是一种重要的基因表达调控模式,由HDAC和组蛋白乙酰化转移酶(histone acetyl-transferase, HAT)共同调节。研究证实,脱乙酰化作用过度,导致一系列肿瘤抑制基因下调/沉默,在肿瘤的发生过程起到关键作用;在宫颈癌组织中,HDAC活性异常增高;宫颈脱落细胞学发现组蛋白H3乙酰化水平与宫颈上皮内瘤变(cervival intraepithelial neoplasia, CIN)程度相关。因此,组蛋白脱乙酰化酶抑制剂(histone deacetylase inhibitor, HDACi)是一种潜在的宫颈癌靶向治疗药物。
     全反式维甲酸(all-trans retinoic acid, ATRA)是一种上皮细胞生长和分化的调节剂,可以诱导发生转化的细胞重新进入分化程序,而被用于肿瘤的防治。ATRA与受体(retinoic acid receptor, RAR)结合,尤其是RARβ2,才能发挥抗肿瘤作用。RARβ2作为一种重要的肿瘤抑制基因,由于启动子区域的甲基化和/或去乙酰化修饰而在包括宫颈癌在内很多肿瘤中的表达被沉默。有研究报道,HDACi可以上调多种肿瘤RARβ2的表达。因此,HDACi联合ATRA能否重启宫颈细胞RARβ2的表达,并发挥协同抗肿瘤作用值得深入研究。
     本课题将分析临床宫颈癌组织中组蛋白乙酰化水平与肿瘤抑制基因(Tumor suppressor genes, TSGs)表达及相关病理参数的关系;深入探讨HDACi (VPA和SAHA)联合ATRA对宫颈癌细胞RARβ2的表达的影响,从表观遗传修饰角度来阐明调控机制;通过体外实验,研究HDACi联合ATRA的抗宫颈癌作用,并深入探讨分子机制;建立人宫颈癌移植瘤模型,进一步评价疗效和验证分子机制。
     主要研究结果如下:
     1宫颈癌组织中组蛋白乙酰化水平和RARβ2及其下游基因的表达显著降低,甚至缺失:选取高、中、低分化的宫颈鳞癌组织65例,免疫组化检测AcH3、RARβ2、 E-cadherin和β-catenin的表达并评分。结果显示,随着宫颈癌组织分化程度的降低,AcH3、RARβ2、E-cadherin和p-catenin的表达逐渐降低、甚至缺失;组蛋白H3的乙酰化水平与RARβ2、E-cadherin和β-catenin的表达呈显著的正相关性(r=0.560, r=0.731, and r=0.733, P均<0.01)。表明,宫颈癌的进展过程中存在表观遗传修饰的异常,后者可能参与调控肿瘤抑制基因RARβ2. E-cadherin和β-catenin的表达。
     2HDACi联合ATRA显著提高宫颈癌细胞株AcH3水平,重启RARβ2表达:HDACi联合ATRA处理宫颈癌细胞株HeLa和SiHa48h后,基因表达谱芯片和Q-PCR结果证实RARβ2表达重启,转录水平显著上调,而RARα和RARγ则为组成性表达;WB显示组蛋白乙酰化水平显著提高,RARβ2表达量显著上升。
     3AcH3与RARβ2启动子区域RARE结合,启动RARβ2表达:以往研究证实,组蛋白H3第9位赖氨酸乙酰化与基因转录活化相关。我们采用H3K9ac抗体进行ChIP实验,结果显示HDACi联合ATRA通过提高H3K9ac水平,后者再与RARβ2启动子区RARE结合,从而重启RARβ2表达。
     4HDACi联合ATRA显著抑制宫颈癌细胞增殖
     1) HDACi联合ATRA抑制宫颈癌细胞增殖能力: MTT实验显示,HDACi与ATRA联合有效抑制宫颈癌细胞增殖,细胞增殖抑制率显著高于VPA和ATRA单一应用(P<0.01),甚至优于1mgL顺铂对宫颈癌细胞的抑制作用(P<0.01);ATRA对pIRES2-RARβ2-EGFP稳转宫颈癌细胞株的抑制作用与联合用药效果无显著差异(P>0.05),表明HDACi重启宫颈癌细胞RARβ2表达,提供了与ATRA协同抗肿瘤作用的分子基础。药物处理后,Ki67的表达明显减少,细胞集落形成能力被显著抑制,联合用药效果优于任一药物单用(P<0.01),再次验证了HDACi和ATRA的协同/相加抑制宫颈癌细胞增殖的作用。
     2) HDACi联合ATRA引起宫颈癌细胞G1期阻滞:HDACi和ATRA处理宫颈癌细胞株48h后检测细胞周期,结果显示联合用药使细胞株发生G1期阻滞;WB结果表明,HDACi和ATRA显著上调P21CIP1和P53的表达,而p-Stat3水平显著降低,Cyclin D1水平仅轻微下调。
     3) HDACi联合ATRA诱导宫颈癌细胞分化:细胞进入分化程序前需脱离细胞周期,WB和免疫组化结果证实,HDACi联合ATRA显著提高上皮细胞分化标志物Filaggrin、Loricrin、和Involucrin的表达;FACS和WB结果显示,药物处理后未引起细胞凋亡的显著增加。
     4) HDACi联合ATRA通过E-cadherin/PI3K/Akt通路诱导细胞分化:WB显示药物联合处理上调E-cadherin/β-catenin和p-Akt水平;分别采用LE-135拮抗RARβ2、siRNA沉默E-cadherin、LY-294002抑制PI3K活性、和siRNA沉默Akt1/2表达,可以阻断诱导分化作用。表明,HDACi联合ATRA重启RARβ2表达,通过E-cadherin-PI3K/Akt通路诱导宫颈癌细胞分化。
     5HDACi联合ATRA抑制裸鼠皮下移植瘤的生长:分别将5×106HeLa和SiHa细胞裸鼠背部皮下注射,建立移植瘤模型。治疗4周后,联合用药显著抑制肿瘤生长,肿瘤体积显著小于VPA组和ATRA组(P<0.01),与顺铂治疗组无显著差异(P>0.05)。与体外实验结果一致,WB显示药物联合处理显著提高组蛋白乙酰化水平,重启宫颈癌细胞RARβ2并上调P21CIP1和P53的表达,而Stat3磷酸化水平显著降低。免疫组化显示,联合药物处理抑制Ki67表达,增强上皮细胞终末分化标志Filaggrin、Loricrin、和Involucrin的表达。
     6建立人宫颈癌组织裸鼠皮下移植瘤模型,验证HDACi联合ATRA对宫颈癌的治疗作用
     1) HDACi联合ATRA抑制人宫颈癌组织移植瘤的生长:收集人低分化宫颈鳞癌组织,经过裸鼠皮下2次移植建立移植瘤模型。药物治疗4周后,联合用药显著抑制肿瘤生长(P<0.01),对照组、VPA组、ATRA组和联合用药组肿瘤平均体积分别316.65±94.58mm3,119.83±7.74mm3, and208.64±76.61mm3,66.84±19.10mm3;肿瘤抑制率(percentage of tumor-growth inhibition,%TGI)分别为59.39%,37.58%,和73.33%。
     2) HDACi联合ATRA诱导移植瘤分化及细胞凋亡:药物治疗4周后,剥离肿瘤组织,HE染色可见联合治疗组出现细胞角化和少量角化珠形成,未见核分裂相,而对照组仍维持来源宫颈癌组织的低分化状态,核分裂相多见。免疫组化显示,HDACi联合ATRA抑制Ki67表达,提高AcH3、RARβ2、及Loricrin、和Involucrin的表达;TUNEL检测显示,VPA治疗后凋亡细胞增加,联合治疗组凋亡细胞增加最为显著(P<0.01)。
     3) HDACi联合ATRA通过表观遗传调控机制重启RARβ2而发挥抗肿瘤作用:药物治疗4周后,免疫组化和WB结果表明组蛋白H3乙酰化水平显著提高;ChIP结果显示,H3K9ac可以与RARβ2启动子核心区域RARE结合,从而启动RARβ2的表达;与体外实验结果一致,Q-PCR和WB结果显示重启RARβ2表达后,其下游E-cadherin和上皮细胞分化标志物Involucrin和Loricrin的表达显著上调;同时P21CIP1、P53和活化的Caspase3表达上调,Stat3磷酸化水平显著降低,Bcl2的表达出现下调。
     综上所述,HDACi单独和联合ATRA可以通过表观遗传途径重启宫颈癌细胞RARβ2的表达,继而提供了两者协同/相加抗肿瘤作用的分子基础;HDACi联合ATRA使宫颈癌细胞脱离细胞周期,通过E-cadherin/PI3K/Akt通路诱导细胞分化是其抗肿瘤作用的主要机制,细胞凋亡也发挥了一定作用。本研究不仅进一步完善了宫颈癌的发病机制,而且阐明了HDACi联合ATRA对宫颈癌诱导分化治疗的效果及信号途径,为临床宫颈癌的防治提供了新的策略,但是治疗效果还需要通过临床试验进一步验证和评价。
Cervical cancer is one of the most frequent diseases in women and a considerable cause of morbidity and mortality among them. It is the second more frequent type of cancer in women worldwide, preceded only by breast and colorectal cancer; each year approximately500,000women worldwide are diagnosed with invasive cervical cancer and more than half of them die of this disease. Eighty percent of these deaths occur in developing countries. Surgery is the recommended treatment for early-stage cervical cancer, but in advanced stages, namely recurrent and metastatic cases, surgery has very limited value. Additionally, the tumors at advanced stages are commonly resistant to chemotherapy and these patients also frequently have a poor performance status, which limits use of aggressive chemotherapy or radiation. Thus the majority of patients die as a result of uncontrolled disease. Therefore, new therapeutic approaches with fewer adverse effects are needed for cervical cancer.
     One targeted class of compounds of particular interest is the histone deacetylase (HDAC) inhibitors. Histone acetylation is a critical regulatory mechanism for gene expression. The aberrant deacetylation of critical genes involved in differentiation, apoptosis or cell cycle arrest has been reported to contribute to the pathogenesis of malignancy. In cervical cancer, it has been reported that HDACs are overexpressed, and the presence of phosphorylated and acetylated forms of histone H3in cytologic smears has demonstrated a marked association of histone H3modifications with the progression of the disease from cervical intraepithelial neoplasia (CIN) I to CIN II and CIN III. HDAC inhibitors are therefore attractive target compounds for cervical cancer treatment.
     All-trans retinoic acid (ATRA) is a potent regulator of cellular growth and differentiation, including normal epithelial cell differentiation; it can function as a chemopreventive agent and is an effective inhibitor of chemical and viral carcinogenesis. The antineoplastic pathways induced by ATRA are regulated predominantly by RARβ (particularly the β2isoform). However, the expression of RARβ2is frequently decreased or lost in many cancer types, including cervical carcinomas. Recent studies have shown that the loss of RARβ2is attributed to the silencing of its promoter region due to histone hypermethylation and deacetylation. The use of HDAC inhibitors, alone and in combination with ATRA, has been shown to restore the expression of silenced RARβ2, thus HDAC inhibitors enhance the growth inhibitory and proapoptotic actions of ATRA in many types of cancer cells. Therefore, whether the promising combination of VPA and ATRA results in the reexpression of silenced RARβ2, and consequently has additive effects on growth suppression in cervical cancer cells, merits further investigation.
     In this study, we investigated histone H3acetylation and tumor suppressor genes (TSGs) expression in cervical cancer and its association with clinicopathological parameters. The efficacy of combined treatment with HDACi and ATRA was examed in restoring RARβ2expression and anti-neoplastic activity in cervical cancer. Furthermore, we evaluated the therapeutic potential of the HDACi combined with ATRA in treating a tumour xenograft model derived from human cervical carcinoma.
     1The expression of AcH3, RARβ2and its downstream genes was downregulated or even silenced. In the65cancerous tissues, the immunoreactivity intensity of these four parameters was strong in well-differentiated tumours and reduced in moderately differentiated carcinoma, with weak or negative labelling in poorly differentiated carcinomas. There was a statistically significant correlation between H3acetylation and the expression of RARβ2, E-cadherin, and β-catenin (r=0.560, r=0.731, and r=0.733,respectively, P<0.01). These indicate that transcriptional silencing by histone deacetylation is one of the well-established mechanisms of tumor suppressor genes inactivation.
     2The combination of HDACi and ATRA restores RARβ2expression via increasing the level of acetylated histone H3. In our experiments, an immunoblot analysis showed that HDACi, either alone or combined with ATRA, strongly induced the hyperacetylation of histone H3. Intriguely, the expression of RARJ32was significantly induced, with50-to90-fold upregulation in combination-treated cells but not detected in untreated control cells. We did not detect any differences in the expression of RARa and RARy between the different treatments, but both were constitutively expressed in both cell lines after treatment.
     3HDACi in combination with ATRA restore RARβ2expression through RARβ2-RARE. The hyperacetylation of lysine9on histone H3(H3K9ac) is usually associated with actively transcribed genes. To further understand the mechanism for histone acetylation-induced re-expression of RARβ2, we performed a ChIP assay using an anti-H3K9ac antibody to determine the binding of acetylated histone H3to the RAR(32promoter region. This combined treatment was more effective at inducing histone acetylation at the RARβ2-RARE region than any single drug. These results indicate that the RARE in the RARβ2promoter is functional and that histone acetylation at the RARβ2-RARE region is closely correlated with RAR(32re-expression in cervical cancer cells.
     4Combined treatment with HDACi and ATRA inhibits the growth of cervical cancer cells.
     1) Combined treatment with HDACi and ATRA enhances growth inhibition in cervical cancer cells. The growth inhibition rate of the combined treatment was significantly higher than that of each drug used alone(P<0.01). The effect of the combination drug treatment was even superior to that of the cytotoxic drug cisplatin (1mg/L) administered alone (P<0.01). Furthermore, the combination of HDACi and ATRA exerted additive effects on growth inhibition. The inhibition rate of the cell lines transfected with pIRES2-RARβ2-EGFP treated with ATRA only was similar as that treated with the combination. Consistent with the decreased cell proliferation, the expression of Ki67, a marker for cell proliferation, and colony formation were significantly decreased in cervical cancer cells treated with VPA and ATRA after48h of incubation.
     2) Combined treatment with HDACi and ATRA causes G1phase arrest. To assess the specific impact of the combination of HDACi and ATRA on cell proliferation, a flow cytometric analysis of cellular DNA content was performed. An evident G1arrest was observed in cervical cancer cell lines treated with HDACi and ATRA for48h. To determine the causes of the G1phase arrest, we examined the expression of cell cycle regulators (Fig.3B). We observed that VPA combined with ATRA induced an evident up-regulation of P21and P53but a significantly decreased level of p-Stat3expression. However, the expression of cyclin D1was only moderately changed.
     3) HDACi potentiates ATRA-induced differentiation in cervical cancer cells. Immunoblotting further confirmed that the combinatorial treatment definitely led to cell differentiation but not apoptosis in the cervical cancer cell lines. Consistent with Annexin V labeling, HDACi alone or in combination with ATRA at the designated concentration induced no major changes in the expression of Bcl-2or Caspase-3and its cleavage products. However, HDACi potentiated ATRA-induced differentiation even at a pharmacological dose of ATRA (1μmol/L) as revealed by the expression of the differentiation markers Filaggrin, Loricrin, and Involucrin.
     4) E-cadherin engagement and the activation of the PI3K/Akt pathway determine the differentiation of cervical cancer cells. The above results suggested that when exposed to a combination of HDACi and ATRA, cervical cancer cells exit the cell cycle and undergo differentiation. E-cadherin and phosphorylated Akt was strongly induced by the combination of drugs and continued to increase with up to48h of exposure, which indicates that PI3K/Akt signaling plays a role in the control of cervical cancer cell differentiation. Blocking the PI3K/Akt pathway by either LE135(selective antagonist of RAR02), the PI3K pharmacological inhibitor LY294002, or specific siRNA for E-cadherin and Aktl/2, caused a significantly decreased expression of the terminal differentiation markers Filaggrin, Loricrin, and Involucrin.
     5Combined treatment with HDACi and ATRA inhibits the progression of tumor xenografts. Consistently, the combination of VPA and ATRA in vivo caused a statistically significant reduction in tumor volume compared with the single-drug treatments (P<0.01). Importantly, the average tumor volume in the mice that received VPA and ATRA was not significantly different from that of the mice treated with cisplatin alone (P>0.05), suggesting the valid chemotherapeutic potential of this drug combination in cervical cancer. Further, we also found that VPA combined with ATRA induced histone H3hyperacetylation, restored RARβ2expression, and up-regulated P53and P21, whereas this treatment down-regulated p-Stat3in vivo. A pathological examination showed a significant repression of Ki67and the greatest level of induction of the differentiation signature in the combination-treated tumors.
     6To evaluate the therapeutic effect of HDACi combined with ATRA on the tumour xenografts derived from human donors.
     1) Combined treatment of HDACi and ATRA inhibits the progression of tumour xenografts. To determine the effect of VPA and ATRA on tumor xenograft growth, animals received vehicle as a control, VPA (300mg/kg/d i.p.), ATRA (15mg/kg/d i.g.), or the combination treatment for28days as the tumors were established. After28days, the mean tumor volume for the combination treatment group was66.84±19.10mm3, while the tumor volumes for mice treated with vehicle, VPA, and ATRA were316.65±94.58mm3,119.83±7.74mm3, and208.64±76.61mm3, respectively. These data indicate that the xenografts were suppressed by59.39%,37.58%, and73.33%(percentage of tumor-growth inhibition,%TGI) for the VPA, ATRA, and combined treatments, respectively.
     2) Combined treatment of HDACi and ATRA induces cell differentiation and part apoptosis in the tumour xenografts. Importantly, the histological appearance showed that VPA and the combination treatment improved the extent of histological differentiation in the tumor xenografts. Cancer cells from xenografts treated with VPA alone or in combination with ATRA have abundant eosinophilic keratinized cytoplasm, while mitotic figures are absent or noted only occasionally. However, cancer cells in the tissues from both the control and ATRA-treated groups were poorly differentiated. Mitotic figures were common (2-4per high-power field in the control group, but≤2per high-power field in ATRA-treated group), and the cytoplasm was eosinophilic to amphophilic, while keratinization was minimal or absent, similar to the original carcinoma. These observations were further verified by immunohistochemical staining for involucrin and loricrin (markers of terminal epithelium differentiation) in tissues from tumor xenografts. Treatment with VPA in combination with ATRA led to a significant decrease in Ki67-positive cells. In addition to inducing differentiation, VPA also promotes apoptosis. To explore this effect, we performed TUNEL staining to evaluate apoptosis in tumor xenografts. There was a significant increase in apoptotic cells in the combination-treated tumors.
     3) HDACi and ATRA restore RARβ2expression via epigenetic modification and sequentially inhibit tumor growth. VPA treatment significantly increased the level of acetylated H3compared to the control and the ATRA group. When combined with ATRA, VPA exhibited an evident additive effect in histone epigenetic modification. The ChIP assay showed that VPA combined with ATRA caused a significant increase in the level of histone acetylation at the RARβ2-promoter region. Therefore, the combination treatment reactivated RARβ2expression via epigenetic modification, sequentially enhancing the expression of target genes, including E-cadherin, Involucrin, and Loricrin. Furthermore, a concomitant increase in the levels of P21CIP1, P53, and activated caspase3as well as a decrease in the levels of p-Stat3and Bcl2were noted.
     In conclusion, HDACi in combination with ATRA has additive effects on the inhibition of cell proliferation and on retarding rumor growth in cervical cancer by restoring RARβ2expression via epigenetic modulation and sequentially promoting differentiation through the PI3K/Akt pathway. Based on these results, a therapeutic regimen combining HDACi and ATRA could be useful in the treatment of cervical cancer, but clinical trials are needed to further evaluate the potential of this combination therapy.
引文
1. Soerjomataram I, Lortet-Tieulent J, Parkin DM, Ferlay J, Mathers C, Forman D, Bray F:Global burden of cancer in 2008:a systematic analysis of disability-adjusted life-years in 12 world regions. Lancet 2012, 380:1840-1850.
    2. Tomljenovic L, Wilyman J, Vanamee E, Bark T, Shaw CA:HPV vaccines and cancer prevention, science versus activism. Infectious agents and cancer 2013,8:6.
    3. Fothergill T, McMillan NA:Papillomavirus virus-like particles activate the PI3-kinase pathway via alpha-6 beta-4 integrin upon binding. Virology 2006,352:319-328.
    4. Shaikh F, Sanehi P, Rawal R:Molecular screening of compounds to the predicted Protein-Protein Interaction site of Rbl-E7 with p53-E6 in HPV. Bioinformation 2012,8:607-612.
    5. Maher DM, Bell MC, O'Donnell EA, Gupta BK, Jaggi M, Chauhan SC: Curcumin suppresses human papillomavirus oncoproteins, restores p53, Rb, and PTPN13 proteins and inhibits benzo[a]pyrene-induced upregulation of HPV E7. Molecular carcinogenesis 2011,50:47-57.
    6. Mighty KK, Laimins LA:The role of human papillomaviruses in oncogenesis. Recent results in cancer research Fortschritte der Krebsforschung Progres dans les recherches sur le cancer 2014,193:135-148.
    7. De-Castro Arce J, Gockel-Krzikalla E, Rosl F:Retinoic acid receptor beta silences human papillomavirus-18 oncogene expression by induction of de novo methylation and heterochromatinization of the viral control region. The Journal of biological chemistry 2007,282:28520-28529.
    8. Sun LL, Cao DY, Yang JX, Li H, Zhou XR, Song ZQ, Cheng XM, Chen J, Shen K:Population-based case-control study on DAPK1, RAR-beta2 and MGMT methylation in liquid-based cytology. Archives of gynecology and obstetrics 2012,285:1433-1439.
    9. Zannoni GF, Prisco MG, Vellone VG, De Stefano I, Scambia G, Gallo D: Changes in the expression of oestrogen receptors and E-cadherin as molecular markers of progression from normal epithelium to invasive cancer in elderly patients with vulvar squamous cell carcinoma. Histopathology 2011,58:265-275.
    10. Sun Y, Tokar EJ, Waalkes MP:Overabundance of putative cancer stem cells in human skin keratinocyte cells malignantly transformed by arsenic. Toxicological sciences:an official journal of the Society of Toxicology 2012, 125:20-29.
    11. Mighty KK, Laimins LA:p63 is necessary for the activation of human papillomavirus late viral functions upon epithelial differentiation. Journal of virology 2011,85:8863-8869.
    12. William WN, Jr.:Oral premalignant lesions:any progress with systemic therapies? Current opinion in oncology 2012,24:205-210.
    13. Weinstock MA, Bingham SF, Digiovanna JJ, Rizzo AE, Marcolivio K, Hall R, Eilers D, Naylor M, Kirsner R, Kalivas J, et al:Tretinoin and the prevention of keratinocyte carcinoma (Basal and squamous cell carcinoma of the skin):a veterans affairs randomized chemoprevention trial. The Journal of investigative dermatology 2012,132:1583-1590.
    14. Ocadiz-Delgado R, Castaneda-Saucedo E, Indra AK, Hernandez-Pando R, Flores-Guizar P, Cruz-Colin JL, Recillas-Targa F, Perez-Ishiwara G, Covarrubias L, Gariglio P:RXRalpha deletion and E6E7 oncogene expression are sufficient to induce cervical malignant lesions in vivo. Cancer letters 2012,317:226-236.
    15. Abu J, Batuwangala M, Herbert K, Symonds P:Retinoic acid and retinoid receptors:potential chemopreventive and therapeutic role in cervical cancer. The lancet oncology 2005,6:712-720.
    16. Rana B, Veal GJ, Pearson AD, Redfern CP:Retinoid X receptors and retinoid response in neuroblastoma cells. Journal of cellular biochemistry 2002,86:67-78.
    17. Wu S, Zhang D, Zhang ZP, Soprano DR, Soprano KJ:Critical role of both retinoid nuclear receptors and retinoid-X-receptors in mediating growth inhibition of ovarian cancer cells by all-trans retinoic acid. Oncogene 1998, 17:2839-2849.
    18. Kanki K, Akechi Y, Ueda C, Tsuchiya H, Shimizu H, Ishijima N, Toriguchi K, Hatano E, Endo K, Hirooka Y, Shiota G:Biological and clinical implications of retinoic acid-responsive genes in human hepatocellular carcinoma cells. Journal of hepatology 2013.
    19. Lefebvre B, Brand C, Flajollet S, Lefebvre P:Down-regulation of the tumor suppressor gene retinoic acid receptor beta2 through the phosphoinositide 3-kinase/Akt signaling pathway. Molecular endocrinology 2006, 20:2109-2121.
    20. Song S, Guan B, Men T, Hoque A, Lotan R, Xu XC:Antitumor effect of retinoic acid receptor-beta2 associated with suppression of cyclooxygenase-2. Cancer prevention research 2009,2:274-280.
    21. Faria TN, Mendelsohn C, Chambon P, Gudas LJ:The targeted disruption of both alleles of RARbeta(2) in F9 cells results in the loss of retinoic acid-associated growth arrest. The Journal of biological chemistry 1999, 274:26783-26788.
    22. Song X, Ye D, Liu B, Cui J, Zhao X, Yi L, Liang J, Song J, Zhang Z, Zhao Q: Combination of all-trans retinoic acid and a human papillomavirus therapeutic vaccine suppresses the number and function of immature myeloid cells and enhances antitumor immunity. Cancer science 2009, 100:334-340.
    23. Feng D, Cao Z, Li C, Zhang L, Zhou Y, Ma J, Liu R, Zhou H, Zhao W, Wei H, Ling B:Combination of valproic acid and ATRA restores RARbeta2 expression and induces differentiation in cervical cancer through the PI3K/Akt pathway. Current molecular medicine 2012,12:342-354.
    24. Russo D, Durante C, Bulotta S, Puppin C, Puxeddu E, Filetti S, Damante G: Targeting histone deacetylase in thyroid cancer. Expert opinion on therapeutic targets 2013,17:179-193.
    25. Lee H, Seo SY, Tiwari I, Jang KL:Epstein-Barr Virus latent membrane protein 1 overcomes all-trans retinoic acid-induced apoptosis by inhibiting retinoic acid receptor-beta(2) expression. Biochemical and biophysical research communications 2012,423:313-318.
    26. Tao MH, Mason JB, Marian C, McCann SE, Platek ME, Millen A, Ambrosone C, Edge SB, Krishnan SS, Trevisan M, et al:Promoter methylation of E-cadherin, p16, and RAR-beta(2) genes in breast tumors and dietary intake of nutrients important in one-carbon metabolism. Nutrition and cancer 2011,63:1143-1150.
    27. Ksiaa F, Ziadi S, Amara K, Korbi S, Trimeche M:Biological significance of promoter hypermethylation of tumor-related genes in patients with gastric carcinoma. Clinica chimica acta; international journal of clinical chemistry 2009,404:128-133.
    28. Sun J, Xu X, Liu J, Liu H, Fu L, Gu L:Epigenetic regulation of retinoic acid receptor beta2 gene in the initiation of breast cancer. Medical oncology 2011,28:1311-1318.
    29. Jung JK, Park SH, Jang KL:Hepatitis B virus X protein overcomes the growth-inhibitory potential of retinoic acid by downregulating retinoic acid receptor-beta2 expression via DNA methylation. The Journal of general virology 2010,91:493-500.
    30. Cras A, Darsin-Bettinger D, Balitrand N, Cassinat B, Soulie A, Toubert ME, Delva L, Chomienne C:Epigenetic patterns of the retinoic acid receptor beta2 promoter in retinoic acid-resistant thyroid cancer cells. Oncogene 2007,26:4018-4024.
    31. Samartzis N, Imesch P, Dedes KJ, Samartzis EP, Fedier A, Fink D, Caduff R, Fehr MK:Expression pattern of class I histone deacetylases in vulvar intraepithelial neoplasia and vulvar cancer:a tissue microarray study. BMC cancer 2011,11:463.
    32. Schroeder FA, Lewis MC, Fass DM, Wagner FF, Zhang YL, Hennig KM, Gale J, Zhao WN, Reis S, Barker DD, et al:A Selective HDAC 1/2 Inhibitor Modulates Chromatin and Gene Expression in Brain and Alters Mouse Behavior in Two Mood-Related Tests. PloS one 2013,8:e71323.
    33. Bandres E, Agirre X, Bitarte N, Ramirez N, Zarate R, Roman-Gomez J, Prosper F, Garcia-Foncillas J:Epigenetic regulation of microRNA expression in colorectal cancer. International journal of cancer Journal international du cancer 2009,125:2737-2743.
    34. Chavez-Blanco A, Segura-Pacheco B, Perez-Cardenas E, Taja-Chayeb L, Cetina L, Candelaria M, Cantu D, Gonzalez-Fierro A, Garcia-Lopez P, Zambrano P, et al:Histone acetylation and histone deacetylase activity of magnesium valproate in tumor and peripheral blood of patients with cervical cancer. A phase I study. Molecular cancer 2005,4:22.
    35. Wentzensen N, Sherman ME, Schiffman M, Wang SS:Utility of methylation markers in cervical cancer early detection:appraisal of the state-of-the-science. Gynecologic oncology 2009,112:293-299.
    36. Jung S, Jeong D, Kim J, Yi L, Koo K, Lee J, Kim CJ, Kim CH, An S, Yang Y, et al:Epigenetic regulation of the potential tumor suppressor gene, hLHX6.1, in human cervical cancer. International journal of oncology 2011, 38:859-869.
    37. Zhang Z, Joh K, Yatsuki H, Zhao W, Soejima H, Higashimoto K, Noguchi M, Yokoyama M, Iwasaka T, Mukai T:Retinoic acid receptor beta2 is epigenetically silenced either by DNA methylation or repressive histone modifications at the promoter in cervical cancer cells. Cancer letters 2007, 247:318-327.
    38. Dhillon VS, Young AR, Husain SA, Aslam M:Promoter hypermethylation of MGMT, CDH1, RAR-beta and SYK tumour suppressor genes in granulosa cell tumours (GCTs) of ovarian origin. British journal of cancer 2004,90:874-881.
    39. Arapshian A, Kuppumbatti YS, Mira-y-Lopez R:Methylation of conserved CpG sites neighboring the beta retinoic acid response element may mediate retinoic acid receptor beta gene silencing in MCF-7 breast cancer cells. Oncogene 2000,19:4066-4070.
    40. Carafa V, Miceli M, Altucci L, Nebbioso A:Histone deacetylase inhibitors: a patent review (2009-2011). Expert opinion on therapeutic patents 2013, 23:1-17.
    41. Kim HJ, Bae SC:Histone deacetylase inhibitors:molecular mechanisms of action and clinical trials as anti-cancer drugs. American journal of translational research 2011,3:166-179.
    42. Boyle GM, Martyn AC, Parsons PG:Histone deacetylase inhibitors and malignant melanoma. Pigment cell research/sponsored by the European Society for Pigment Cell Research and the International Pigment Cell Society 2005,18:160-166.
    43. Yeo W, Chung HC, Chan SL, Wang LZ, Lim R, Picus J, Boyer M, Mo FK, Koh J, Rha SY, et al:Epigenetic therapy using belinostat for patients with unresectable hepatocellular carcinoma:a multicenter phase Ⅰ/Ⅱ study with biomarker and pharmacokinetic analysis of tumors from patients in the Mayo Phase Ⅱ Consortium and the Cancer Therapeutics Research Group. Journal of clinical oncology:official journal of the American Society of Clinical Oncology 2012,30:3361-3367.
    44. Juergens RA, Wrangle J, Vendetti FP, Murphy SC, Zhao M, Coleman B, Sebree R, Rodgers K, Hooker CM, Franco N, et al:Combination epigenetic therapy has efficacy in patients with refractory advanced non-small cell lung cancer. Cancer discovery 2011,1:598-607.
    45. Venugopal B, Baird R, Kristeleit RS, Plummer R, Cowan R, Stewart A, Fourneau N, Hellemans P, Elsayed Y, McClue S, et al:A Phase I Study of Quisinostat (JNJ-26481585), an Oral Hydroxamate Histone Deacetylase Inhibitor with Evidence of Target Modulation and Antitumor Activity, in Patients with Advanced Solid Tumors. Clinical cancer research:an official journal of the American Association for Cancer Research 2013, 19:4262-4272.
    46. Finazzi G, Vannucchi AM, Martinelli V, Ruggeri M, Nobile F, Specchia G, Pogliani EM, Olimpieri OM, Fioritoni G, Musolino C, et al:A phase II study of Givinostat in combination with hydroxycarbamide in patients with polycythaemia vera unresponsive to hydroxycarbamide monotherapy. British journal of haematology 2013,161:688-694.
    47. Qiu T, Zhou L, Zhu W, Wang T, Wang J, Shu Y, Liu P:Effects of treatment with histone deacetylase inhibitors in solid tumors:a review based on 30 clinical trials. Future oncology 2013,9:255-269.
    48. Sandor V, Bakke S, Robey RW, Kang MH, Blagosklonny MV, Bender J, Brooks R, Piekarz RL, Tucker E, Figg WD, et al:Phase I trial of the histone deacetylase inhibitor, depsipeptide (FR901228, NSC 630176), in patients with refractory neoplasms. Clinical cancer research:an official journal of the American Association for Cancer Research 2002,8:718-728.
    49. Campas-Moya C:Romidepsin for the treatment of cutaneous T-cell lymphoma. Drugs of today 2009,45:787-795.
    50. Muscal JA, Thompson PA, Horton TM, Ingle AM, Ahern CH, McGovern RM, Reid JM, Ames MM, Espinoza-Delgado I, Weigel BJ, Blaney SM:A phase I trial of vorinostat and bortezomib in children with refractory or recurrent solid tumors:a Children's Oncology Group phase I consortium study (ADVL0916). Pediatric blood & cancer 2013,60:390-395.
    51. Giaccone G, Raj an A, Berman A, Kelly RJ, Szabo E, Lopez-Chavez A, Trepel J, Lee MJ, Cao L, Espinoza-Delgado I, et al:Phase II study of belinostat in patients with recurrent or refractory advanced thymic epithelial tumors. Journal of clinical oncology:official journal of the American Society of Clinical Oncology 2011,29:2052-2059.
    52. Duvic M, Vu J:Update on the treatment of cutaneous T-cell lymphoma (CTCL):Focus on vorinostat. Biologics:targets & therapy 2007, 1:377-392.
    53. Hummel TR, Wagner L, Ahern C, Fouladi M, Reid JM, McGovern RM, Ames MM, Gilbertson RJ, Horton T, Ingle AM, et al:A pediatric phase 1 trial of vorinostat and temozolomide in relapsed or refractory primary brain or spinal cord tumors:a Children's Oncology Group phase 1 consortium study. Pediatric blood & cancer 2013,60:1452-1457.
    54. Sawangsang P, Sae-Teng C, Suprasert P, Srisomboon J, Khunamornpong S, Kietpeerakool C:Clinical significance of atypical glandular cells on Pap smears:experience from a region with a high incidence of cervical cancer. The journal of obstetrics and gynaecology research 2011,37:496-500.
    55. Liu LK, Jiang XY, Zhou XX, Wang DM, Song XL, Jiang HB:Upregulation of vimentin and aberrant expression of E-cadherin/beta-catenin complex in oral squamous cell carcinomas:correlation with the clinicopathological features and patient outcome. Modern pathology:an official journal of the United States and Canadian Academy of Pathology, Inc 2010,23:213-224.
    56. Chui X, Egami H, Yamashita J, Kurizaki T, Ohmachi H, Yamamoto S, Ogawa M:Immunohistochemical expression of the c-kit proto-oncogene product in human malignant and non-malignant breast tissues. British journal of cancer 1996,73:1233-1236.
    57. Yang Q, Tian Y, Liu S, Zeine R, Chlenski A, Salwen HR, Henkin J, Cohn SL: Thrombospondin-1 peptide ABT-510 combined with valproic acid is an effective antiangiogenesis strategy in neuroblastoma. Cancer research 2007, 67:1716-1724.
    58. Xu WS, Parmigiani RB, Marks PA:Histone deacetylase inhibitors: molecular mechanisms of action. Oncogene 2007,26:5541-5552.
    59. Mai A, Massa S, Rotili D, Cerbara I, Valente S, Pezzi R, Simeoni S, Ragno R: Histone deacetylation in epigenetics:an attractive target for anticancer therapy. Medicinal research reviews 2005,25:261-309.
    60. Sarshad A, Sadeghifar F, Louvet E, Mori R, Bohm S, Al-Muzzaini B, Vintermist A, Fomproix N, Ostlund AK, Percipalle P:Nuclear myosin lc facilitates the chromatin modifications required to activate rRNA gene transcription and cell cycle progression. PLoS genetics 2013,9:e1003397.
    61. Fandy TE, Gore SD:Epigenetic targets in human neoplasms. Epigenomics 2010,2:221-232.
    62. Valdes-Mora F, Song JZ, Statham AL, Strbenac D, Robinson MD, Nair SS, Patterson KI, Tremethick DJ, Stirzaker C, Clark SJ:Acetylation of H2A.Z is a key epigenetic modification associated with gene deregulation and epigenetic remodeling in cancer. Genome research 2012,22:307-321.
    63. Wilting RH, Dannenberg JH:Epigenetic mechanisms in tumorigenesis, tumor cell heterogeneity and drug resistance. Drug resistance updates reviews and commentaries in antimicrobial and anticancer chemotherapy 2012,15:21-38.
    64. Milon BC, Cheng H, Tselebrovsky MV, Lavrov SA, Nenasheva VV, Mikhaleva EA, Shevelyov YY, Nurminsky DI:Role of histone deacetylases in gene regulation at nuclear lamina. PloS one 2012,7:e49692.
    65. Giordano A, Egleston BL, Hajage D, Bland J, Hortobagyi GN, Reuben JM, Pierga JY, Cristofanilli M, Bidard FC:Establishment and validation of circulating tumor cell-based prognostic nomograms in first-line metastatic breast cancer patients. Clinical cancer research:an official journal of the American Association for Cancer Research 2013, 19:1596-1602.
    66. Ou JN, Torrisani J, Unterberger A, Provencal N, Shikimi K, Karimi M, Ekstrom TJ, Szyf M:Histone deacetylase inhibitor Trichostatin A induces global and gene-specific DNA demethylation in human cancer cell lines. Biochemical pharmacology 2007,73:1297-1307.
    67. Fan J, Eastham L, Varney ME, Hall A, Adkins NL, Chetel L, Sollars VE, Georgel P, Niles RM:Silencing and re-expression of retinoic acid receptor beta2 in human melanoma. Pigment cell & melanoma research 2010, 23:419-429.
    68. Dutta A, Sen T, Banerji A, Das S, Chatterjee A:Studies on Multifunctional Effect of All-Trans Retinoic Acid (ATRA) on Matrix Metalloproteinase-2 (MMP-2) and Its Regulatory Molecules in Human Breast Cancer Cells (MCF-7). Journal of oncology 2009,2009:627840.
    69. Li P, Pashmforoush M, Sucov HM:Mesodermal retinoic acid signaling regulates endothelial cell coalescence in caudal pharyngeal arch artery vasculogenesis. Developmental biology 2012,361:116-124.
    70. Suman S, Kurisetty V, Das TP, Vadodkar A, Ramos G, Lakshmanaswamy R, Damodaran C:Activation of AKT signaling promotes epithelial-mesenchymal transition and tumor growth in colorectal cancer cells. Molecular carcinogenesis 2013.
    71. Hirano T, Satow R, Kato A, Tamura M, Murayama Y, Saya H, Kojima H, Nagano T, Okabe T, Fukami K:Identification of Novel Small Compounds that Restore E-cadherin Expression and Inhibit Tumor Cell Motility and Invasiveness. Biochemical pharmacology 2013.
    72. Rajak H, Singh A, Raghuwanshi K, Kumar R, Dewangan PK, Veerasamy R, Sharma PC, Dixit A, Mishra P:A Structural Insight into Hydroxamic Acid Based Histone Deacetylase Inhibitors for the Presence of Anticancer Activity. Current medicinal chemistry 2013.
    73. Tang J, Yan H, Zhuang S:Histone deacetylases as targets for treatment of multiple diseases. Clinical science 2013,124:651-662.
    74. Hagelkruys A, Sawicka A, Rennmayr M, Seiser C:The biology of HDAC in cancer:the nuclear and epigenetic components. Handbook of experimental pharmacology 2011,206:13-37.
    75. New M, Olzscha H, La Thangue NB:HDAC inhibitor-based therapies:can we interpret the code? Molecular oncology 2012,6:637-656.
    76. Lin Z, Bazzaro M, Wang MC, Chan KC, Peng S, Roden RB:Combination of proteasome and HDAC inhibitors for uterine cervical cancer treatment. Clinical cancer research:an official journal of the American Association for Cancer Research 2009,15:570-577.
    77. Tan S, de Vries EG, van der Zee AG, de Jong S:Anticancer drugs aimed at E6 and E7 activity in HPV-positive cervical cancer. Current cancer drug targets 2012,12:170-184.
    78. Mora-Garcia Mde L, Duenas-Gonzalez A, Hernandez-Montes J, De la Cruz-Hernandez E, Perez-Cardenas E, Weiss-Steider B, Santiago-Osorio E, Ortiz-Navarrete VF, Rosales VH, Cantu D, et al:Up-regulation of HLA class-Ⅰ antigen expression and antigen-specific CTL response in cervical cancer cells by the demethylating agent hydralazine and the histone deacetylase inhibitor valproic acid. Journal of translational medicine 2006, 4:55.
    79. Takai N, Kira N, Ishii T, Nishida M, Nasu K, Narahara H:Novel chemotherapy using histone deacetylase inhibitors in cervical cancer. Asian Pacific journal of cancer prevention:APJCP 2011,12:575-580.
    80. Miao F, Natarajan R:Mapping global histone methylation patterns in the coding regions of human genes. Molecular and cellular biology 2005, 25:4650-4661.
    81. Xu XC, Mitchell MF, Silva E, Jetten A, Lotan R:Decreased expression of retinoic acid receptors, transforming growth factor beta, involucrin, and cornifin in cervical intraepithelial neoplasia. Clinical cancer research:an official journal of the American Association for Cancer Research 1999, 5:1503-1508.
    82. Myga-Nowak M, Pacholska-Bogalska J, Kwasniewski W, Kwasniewska A, Gozdzicka-Jozefiak A:Proliferation of cells and expression of RARs, RXRs and HPV viral E6 and E7 proteins in cervical cancer cell lines after treatment with ATRA. Annals of agricultural and environmental medicine: AAEM 2011,18:145-150.
    83. Khan MA, Jenkins GR, Tolleson WH, Creek KE, Pirisi L:Retinoic acid inhibition of human papillomavirus type 16-mediated transformation of human keratinocytes. Cancer research 1993,53:905-909.
    84. Meyskens FL, Jr., Surwit E, Moon TE, Childers JM, Davis JR, Dorr RT, Johnson CS, Alberts DS:Enhancement of regression of cervical intraepithelial neoplasia Ⅱ (moderate dysplasia) with topically applied all-trans-retinoic acid:a randomized trial. Journal of the National Cancer Institute 1994,86:539-543.
    85. Mongan NP, Gudas LJ:Valproic acid, in combination with all-trans retinoic acid and 5-aza-2'-deoxycytidine, restores expression of silenced RARbeta2 in breast cancer cells. Molecular cancer therapeutics 2005, 4:477-486.
    86. Tinkle CL, Lechler T, Pasolli HA, Fuchs E:Conditional targeting of E-cadherin in skin:insights into hyperproliferative and degenerative responses. Proceedings of the National Academy of Sciences of the United States of America 2004,101:552-557.
    87. Calautti E, Li J, Saoncella S, Brissette JL, Goetinck PF:Phosphoinositide 3-kinase signaling to Akt promotes keratinocyte differentiation versus death. The Journal of biological chemistry 2005,280:32856-32865.
    88. Lamb LE, Knudsen BS, Miranti CK:E-cadherin-mediated survival of androgen-receptor-expressing secretory prostate epithelial cells derived from a stratified in vitro differentiation model. Journal of cell science 2010, 123:266-276.
    89. Heliez C, Baricault L, Barboule N, Valette A:Paclitaxel increases p21 synthesis and accumulation of its AKT-phosphorylated form in the cytoplasm of cancer cells. Oncogene 2003,22:3260-3268.
    90. Holt SV, Logie A, Odedra R, Heier A, Heaton SP, Alferez D, Davies BR, Wilkinson RW, Smith PD:The MEK1/2 inhibitor, selumetinib (AZD6244; ARRY-142886), enhances anti-tumour efficacy when combined with conventional chemotherapeutic agents in human tumour xenograft models. British journal of cancer 2012,106:858-866.
    91. Tokita H, Tanaka N, Sekimoto K, Ueno T, Okamoto K, Fujimura S: Experimental model for combination chemotherapy with metronidazole using human uterine cervical carcinomas transplanted into nude mice. Cancer research 1980,40:4287-4294.
    92. Touma SE, Goldberg JS, Moench P, Guo X, Tickoo SK, Gudas LJ, Nanus DM: Retinoic acid and the histone deacetylase inhibitor trichostatin a inhibit the proliferation of human renal cell carcinoma in a xenograft tumor model. Clinical cancer research:an official journal of the American Association for Cancer Research 2005,11:3558-3566.
    93. Kumar P, Garg R, Bolden G, Pandey KN:Interactive roles of Ets-1, Spl, and acetylated histones in the retinoic acid-dependent activation of guanylyl cyclase/atrial natriuretic peptide receptor-A gene transcription. The Journal of biological chemistry 2010,285:37521-37530.
    94. Schug TT, Berry DC, Shaw NS, Travis SN, Noy N:Opposing effects of retinoic acid on cell growth result from alternate activation of two different nuclear receptors. Cell 2007,129:723-733.
    95. Hua S, Kittler R, White KP:Genomic antagonism between retinoic acid and estrogen signaling in breast cancer. Cell 2009,137:1259-1271.
    96. Tatebe H, Shimizu M, Shirakami Y, Sakai H, Yasuda Y, Tsurumi H, Moriwaki H:Acyclic retinoid synergises with valproic acid to inhibit growth in human hepatocellular carcinoma cells. Cancer letters 2009,285:210-217.
    1. Soerjomataram I, Lortet-Tieulent J, Parkin DM, Ferlay J, Mathers C, Forman D, Bray F:Global burden of cancer in 2008:a systematic analysis of disability-adjusted life-years in 12 world regions. Lancet 2012, 380:1840-1850.
    2. Tomljenovic L, Wilyman J, Vanamee E, Bark T, Shaw CA:HPV vaccines and cancer prevention, science versus activism. Infectious agents and cancer 2013,8:6.
    3. Fothergill T, McMillan NA:Papillomavirus virus-like particles activate the P13-kinase pathway via alpha-6 beta-4 integrin upon binding. Virology 2006,352:319-328.
    4. Shaikh F, Sanehi P, Rawal R:Molecular screening of compounds to the predicted Protein-Protein Interaction site of Rbl-E7 with p53-E6 in HPV. Bioinformation 2012,8:607-612.
    5. Maher DM, Bell MC, O'Donnell EA, Gupta BK, Jaggi M, Chauhan SC: Curcumin suppresses human papillomavirus oncoproteins, restores p53, Rb, and PTPN13 proteins and inhibits benzo[a]pyrene-induced upregulation of HPV E7. Molecular carcinogenesis 2011,50:47-57.
    6. Mighty KK, Laimins LA:The role of human papillomaviruses in oncogenesis. Recent results in cancer research Fortschritte der Krebsforschung Progres dans les recherches sur le cancer 2014,193:135-148.
    7. De-Castro Arce J, Gockel-Krzikalla E, Rosl F:Retinoic acid receptor beta silences human papillomavirus-18 oncogene expression by induction of de novo methylation and heterochromatinization of the viral control region. The Journal of biological chemistry 2007,282:28520-28529.
    8. Sun LL, Cao DY, Yang JX, Li H, Zhou XR, Song ZQ, Cheng XM, Chen J, Shen K:Population-based case-control study on DAPK1, RAR-beta2 and MGMT methylation in liquid-based cytology. Archives of gynecology and obstetrics 2012,285:1433-1439.
    9. Zannoni GF, Prisco MG, Vellone VG, De Stefano I, Scambia G, Gallo D: Changes in the expression of oestrogen receptors and E-cadherin as molecular markers of progression from normal epithelium to invasive cancer in elderly patients with vulvar squamous cell carcinoma. Histopathology 2011,58:265-275.
    10. Klopp AH, Eifel PJ:Chemoradiotherapy for cervical cancer in 2010. Current oncology reports 2011,13:77-85.
    11. Abu J, Batuwangala M, Herbert K, Symonds P:Retinoic acid and retinoid receptors:potential chemopreventive and therapeutic role in cervical cancer. The lancet oncology 2005,6:712-720.
    12. Ghiaur G, Yegnasubramanian S, Perkins B, Gucwa JL, Gerber JM, Jones RJ: Regulation of human hematopoietic stem cell self-renewal by the microenvironment's control of retinoic acid signaling. Proceedings of the National Academy of Sciences of the United States of America 2013.
    13. Lallemand-Breitenbach V, de The H:Retinoic acid plus arsenic trioxide, the ultimate panacea for acute promyelocytic leukemia? Blood 2013, 122:2008-2010.
    14. Kanki K, Akechi Y, Ueda C, Tsuchiya H, Shimizu H, Ishijima N, Toriguchi K, Hatano E, Endo K, Hirooka Y, Shiota G:Biological and clinical implications of retinoic acid-responsive genes in human hepatocellular carcinoma cells. Journal of hepatology 2013.
    15. McNamara S, Wang H, Hanna N, Miller WH, Jr.:Topoisomerase IIbeta negatively modulates retinoic acid receptor alpha function:a novel mechanism of retinoic acid resistance. Molecular and cellular biology 2008, 28:2066-2077.
    16. Petty WJ, Li N, Biddle A, Bounds R, Nitkin C, Ma Y, Dragnev KH, Freemantle SJ, Dmitrovsky E:A novel retinoic acid receptor beta isoform and retinoid resistance in lung carcinogenesis. Journal of the National Cancer Institute 2005,97:1645-1651.
    17. Suh YA, Lee HY, Virmani A, Wong J, Mann KK, Miller WH, Jr., Gazdar A, Kurie JM:Loss of retinoic acid receptor beta gene expression is linked to aberrant histone H3 acetylation in lung cancer cell lines. Cancer research 2002,62:3945-3949.
    18. Swift ME, Wallden B, Wayner EA, Swisshelm K:Truncated RAR beta isoform enhances proliferation and retinoid resistance. Journal of cellular physiology 2006,209:718-725.
    19. Feng D, Cao Z, Li C, Zhang L, Zhou Y, Ma J, Liu R, Zhou H, Zhao W, Wei H, Ling B:Combination of valproic acid and ATRA restores RARbeta2 expression and induces differentiation in cervical cancer through the PI3K/Akt pathway. Current molecular medicine 2012,12:342-354.
    20. Vucetic Z, Zhang Z, Zhao J, Wang F, Soprano KJ, Soprano DR:Acinus-S' represses retinoic acid receptor (RAR)-regulated gene expression through interaction with the B domains of RARs. Molecular and cellular biology 2008,28:2549-2558.
    21. Chen LI, Sommer KM, Swisshelm K:Downstream codons in the retinoic acid receptor beta -2 and beta -4 mRNAs initiate translation of a protein isoform that disrupts retinoid-activated transcription. The Journal of biological chemistry 2002,277:35411-35421.
    22. Faria TN, Mendelsohn C, Chambon P, Gudas LJ:The targeted disruption of both alleles of RARbeta(2) in F9 cells results in the loss of retinoic acid-associated growth arrest. The Journal of biological chemistry 1999, 274:26783-26788.
    23. Russo D, Durante C, Bulotta S, Puppin C, Puxeddu E, Filetti S, Damante G: Targeting histone deacetylase in thyroid cancer. Expert opinion on therapeutic targets 2013,17:179-193.
    24. Lee H, Seo SY, Tiwari I, Jang KL:Epstein-Barr Virus latent membrane protein 1 overcomes all-trans retinoic acid-induced apoptosis by inhibiting retinoic acid receptor-beta(2) expression. Biochemical and biophysical research communications 2012,423:313-318.
    25. Tao MH, Mason JB, Marian C, McCann SE, Platek ME, Millen A, Ambrosone C, Edge SB, Krishnan SS, Trevisan M, et al:Promoter methylation of E-cadherin, p16, and RAR-beta(2) genes in breast tumors and dietary intake of nutrients important in one-carbon metabolism. Nutrition and cancer 2011,63:1143-1150.
    26. Ksiaa F, Ziadi S, Amara K, Korbi S, Trimeche M:Biological significance of promoter hypermethylation of tumor-related genes in patients with gastric carcinoma. Clinica chimica acta; international journal of clinical chemistry 2009,404:128-133.
    27. Choi CH, Lee KM, Choi JJ, Kim TJ, Kim WY, Lee JW, Lee SJ, Lee JH, Bae DS, Kim BG:Hypermethylation and loss of heterozygosity of tumor suppressor genes on chromosome 3p in cervical cancer. Cancer letters 2007, 255:26-33.
    28. Wentzensen N, Sherman ME, Schiffman M, Wang SS:Utility of methylation markers in cervical cancer early detection:appraisal of the state-of-the-science. Gynecologic oncology 2009,112:293-299.
    29. Lee H, Woo YJ, Kim SS, Kim SH, Park BJ, Choi D, Jang KL:Hepatitis C virus Core protein overcomes all-trans retinoic acid-induced cell growth arrest by inhibiting retinoic acid receptor-beta2 expression via DNA methylation. Cancer letters 2013,335:372-379.
    30. Ye F, Xu XC:Benzo[a]pyrene diol epoxide suppresses retinoic acid receptor-beta2 expression by recruiting DNA (cytosine-5-)-methyltransferase 3A. Molecular cancer 2010,9:93.
    31. Lee S, Lee DK, Dou Y, Lee J, Lee B, Kwak E, Kong YY, Lee SK, Roeder RG, Lee JW:Coactivator as a target gene specificity determinant for histone H3 lysine 4 methyltransferases. Proceedings of the National Academy of Sciences of the United States of America 2006,103:15392-15397.
    32. Moison C, Senamaud-Beaufort C, Fourriere L, Champion C, Ceccaldi A, Lacomme S, Daunay A, Tost J, Arimondo PB, Guieysse-Peugeot AL:DNA methylation associated with polycomb repression in retinoic acid receptor beta silencing. FASEB journal:official publication of the Federation of American Societies for Experimental Biology 2013,27:1468-1478.
    33. Gudas LJ:Retinoids induce stem cell differentiation via epigenetic changes. Seminars in cell & developmental biology 2013.
    34. Jin B, Yao B, Li JL, Fields CR, Delmas AL, Liu C, Robertson KD:DNMT1 and DNMT3B modulate distinct polycomb-mediated histone modifications in colon cancer. Cancer research 2009,69:7412-7421.
    35. Fandy TE, Gore SD:Epigenetic targets in human neoplasms. Epigenomics 2010,2:221-232.
    36. Wang D, Xia X, Weiss RE, Refetoff S, Yen PM:Distinct and histone-specific modifications mediate positive versus negative transcriptional regulation of TSHalpha promoter. PloS one 2010,5:e9853.
    37. Schwartz YB, Kahn TG, Stenberg P, Ohno K, Bourgon R, Pirrotta V: Alternative epigenetic chromatin states of polycomb target genes. PLoS genetics 2010,6:e1000805.
    38. You JS, Kang JK, Lee EK, Lee JC, Lee SH, Jeon YJ, Koh DH, Ahn SH, Seo DW, Lee HY, et al:Histone deacetylase inhibitor apicidin downregulates DNA methyltransferase 1 expression and induces repressive histone modifications via recruitment of corepressor complex to promoter region in human cervix cancer cells. Oncogene 2008,27:1376-1386.
    39. Lin Z, Bazzaro M, Wang MC, Chan KC, Peng S, Roden RB:Combination of proteasome and HDAC inhibitors for uterine cervical cancer treatment. Clinical cancer research:an official journal of the American Association for Cancer Research 2009,15:570-577.
    40. Chavez-Blanco A, Segura-Pacheco B, Perez-Cardenas E, Taja-Chayeb L, Cetina L, Candelaria M, Cantu D, Gonzalez-Fierro A, Garcia-Lopez P, Zambrano P, et al:Histone acetylation and histone deacetylase activity of magnesium valproate in tumor and peripheral blood of patients with cervical cancer. A phase I study. Molecular cancer 2005,4:22.
    41. Gaillard E, Bruck N, Brelivet Y, Bour G, Lalevee S, Bauer A, Poch O, Moras D, Rochette-Egly C:Phosphorylation by PKA potentiates retinoic acid receptor alpha activity by means of increasing interaction with and phosphorylation by cyclin H/cdk7. Proceedings of the National Academy of Sciences of the United States of America 2006,103:9548-9553.
    42. Makkonen KM, Malinen M, Ropponen A, Vaisanen S, Carlberg C:Cell cycle regulatory effects of retinoic Acid and forskolin are mediated by the cyclin C gene. Journal of molecular biology 2009,393:261-271.
    43. Srinivas H, Xia D, Moore NL, Uray IP, Kim H, Ma L, Weigel NL, Brown PH, Kurie JM:Akt phosphorylates and suppresses the transactivation of retinoic acid receptor alpha. The Biochemical journal 2006,395:653-662.
    44. Lefebvre B, Brand C, Flajollet S, Lefebvre P:Down-regulation of the tumor suppressor gene retinoic acid receptor beta2 through the phosphoinositide 3-kinase/Akt signaling pathway. Molecular endocrinology 2006, 20:2109-2121.
    45. Srinivas H, Juroske DM, Kalyankrishna S, Cody DD, Price RE, Xu XC, Narayanan R, Weigel NL, Kurie JM:c-Jun N-terminal kinase contributes to aberrant retinoid signaling in lung cancer cells by phosphorylating and inducing proteasomal degradation of retinoic acid receptor alpha. Molecular and cellular biology 2005,25:1054-1069.
    46. Dragnev KH, Freemantle SJ, Spinella MJ, Dmitrovsky E:Cyclin proteolysis as a retinoid cancer prevention mechanism. Annals of the New York Academy of Sciences 2001,952:13-22.
    47. Tanaka T, Rodriguez de la Concepcion ML, De Luca LM:Involvement of all-trans-retinoic acid in the breakdown of retinoic acid receptors alpha and gamma through proteasomes in MCF-7 human breast cancer cells. Biochemical pharmacology 2001,61:1347-1355.
    48. Gianni M, Tarrade A, Nigro EA, Garattini E, Rochette-Egly C:The AF-1 and AF-2 domains of RAR gamma 2 and RXR alpha cooperate for triggering the transactivation and the degradation of RAR gamma 2/RXR alpha heterodimers. The Journal of biological chemistry 2003,278:34458-34466.
    49. Gianni M, Bauer A, Garattini E, Chambon P, Rochette-Egly C: Phosphorylation by p38MAPK and recruitment of SUG-1 are required for RA-induced RAR gamma degradation and transactivation. The EMBO journal 2002,21:3760-3769.
    50. Pathak S, Bhatla N, Singh N:Cervical cancer pathogenesis is associated with one-carbon metabolism. Molecular and cellular biochemistry 2012, 369:1-7.
    51. Moriwaki H, Shimizu M, Okuno M, Nishiwaki-Matsushima R: Chemoprevention of liver carcinogenesis with retinoids:Basic and clinical aspects. Hepatology research:the official journal of the Japan Society of Hepatology 2007,37 Suppl 2:S299-302.
    52. Ryu S, Stein JP, Chung CT, Lee YJ, Kim JH:Enhanced apoptosis and radiosensitization by combined 13-cis-retinoic acid and interferon-alpha2a; role of RAR-beta gene. International journal of radiation oncology, biology, physics 2001,51:785-790.
    53. Choo CK, Rorke EA, Eckert RL:Retinoid regulation of cell differentiation in a series of human papillomavirus type 16-immortalized human cervical epithelial cell lines. Carcinogenesis 1995,16:375-381.
    54. Jha AK, Nikbakht M, Parashar G, Shrivastava A, Capalash N, Kaur J: Reversal of hypermethylation and reactivation of the RARbeta2 gene by natural compounds in cervical cancer cell lines. Folia biologica 2010, 56:195-200.
    55. De la Cruz-Hernandez E, Perez-Plasencia C, Perez-Cardenas E, Gonzalez-Fierro A, Trejo-Becerril C, Chavez-Blanco A, Taja-Chayeb L, Vidal S, Gutierrez O, Dominguez GI, et al:Transcriptional changes induced by epigenetic therapy with hydralazine and magnesium valproate in cervical carcinoma. Oncology reports 2011,25:399-407.
    56. Sah JF, Eckert RL, Chandraratna RA, Rorke EA:Retinoids suppress epidermal growth factor-associated cell proliferation by inhibiting epidermal growth factor receptor-dependent ERK1/2 activation. The Journal of biological chemistry 2002,277:9728-9735.
    57. Ding Z, Green AG, Yang X, Chernenko G, Tang SC, Pater A:Retinoic acid inhibits telomerase activity and downregulates expression but does not affect splicing of hTERT:correlation with cell growth rate inhibition in an in vitro cervical carcinogenesis/multidrug-resistance model. Experimental cell research 2002,272:185-191.
    58. Guo JM, Xiao BX, Kang GZ, Liu DH, Chen H, Zhang S, Zhang XN: Suppression of telomerase activity and arrest at G1 phase in human cervical cancer HeLa cells by all-trans retinoic acid. International journal of gynecological cancer:official journal of the International Gynecological Cancer Society 2006,16:341-346.
    59. Lim JS, Park SH, Jang KL:All-trans retinoic acid induces cellular senescence by up-regulating levels of p16 and p21 via promoter hypomethylation. Biochemical and biophysical research communications 2011,412:500-505.
    60. Suzukawa K, Colburn NH:AP-1 transrepressing retinoic acid does not deplete coactivators or AP-1 monomers but may target specific Jun or Fos containing dimers. Oncogene 2002,21:2181-2190.
    61. Balasubramanian S, Chandraratna RA, Eckert RL:A novel retinoid-related molecule inhibits pancreatic cancer cell proliferation by a retinoid receptor independent mechanism via suppression of cell cycle regulatory protein function and induction of caspase-associated apoptosis. Oncogene 2005,24:4257-4270.
    62. Guan B, Li H, Yang Z, Hoque A, Xu Ⅹ:Inhibition of farnesoid X receptor controls esophageal cancer cell growth in vitro and in nude mouse xenografts. Cancer 2013,119:1321-1329.
    63. Woo YJ, Jang KL:All-trans retinoic acid activates E-cadherin expression via promoter hypomethylation in the human colon carcinoma HCT116 cells. Biochemical and biophysical research communications 2012,425:944-949.