首页> 外文学位 >DNA Repair Capacity as a Marker of Breast Cancer Susceptibility.
【24h】

DNA Repair Capacity as a Marker of Breast Cancer Susceptibility.

机译:DNA修复能力是乳腺癌易感性的标志。

获取原文
获取原文并翻译 | 示例

摘要

Introduction: The wide-ranging prognostic implications of a breast cancer diagnosis highlight the need to better enable women to make informed decisions regarding screening and treatment options. As several cancer susceptibility syndromes have been linked to germline mutations resulting in defective DNA repair, including the predisposition to breast cancer due to BRCA1 and BRCA2 mutations, more subtle defects in DNA repair capacity may contribute to the components driving differential susceptibility within the general population. Hence, understanding the role of DNA repair capacity in breast cancer onset may aid in the development of a more comprehensive risk profile, thereby furthering the effort to target relevant populations for early screening. In the studies undertaken for this dissertation, we employed various methodologies capturing endpoints across different repair pathways detectable in blood to both further elucidate the etiologic basis of breast cancer development and leverage the information into the potential development of a screening biomarker. Methods: For the phenotypic assessment of nucleotide excision repair (NER) capacity, we developed an ELISA-based method to determine benzo(a)pyrene diolepoxide (BPDE)-DNA adduct capacity in lymphoblastoid cell lines. Gene expression levels were assessed with pre-designed Taqman kits in RNA-derived cDNAs from mononuclear cells using a real-time PCR-based platform. Methylation analysis was conducted with in-house designed assays on bisulfite-converted DNA from mononuclear cells using a pyrosequencing platform. Finally, single nucleotide polymorphisms (SNP) genotyping was assessed in DNA derived from white blood cells with pre-designed Taqman SNP genotyping assays using a real-time PCR-based platform. All studies were conducted in sister-sets enrolled in the New York site within the Breast Cancer Family Registry and all statistical analysis was conducted using the R Foundation for Statistical Computing (2011). Results: We did not detect an association between the ELISA-based phenotypic assessment of NER capacity in the lymphoblastoid cells lines of the sister-sets (n=246, 114 sister-sets) and breast cancer risk (OR = 1.0, 95%CI=0.95, 1.04). Furthermore, we did not observe a correlation with previously determined NER capacity in the same population using an immunohistochemical-based method (r= -0.01, p=0.86). In our gene expression study (n=569, 218 sister-sets), women in the lowest tertile of ATM expression had a heightened risk of breast cancer compared to women in the highest tertile of expression, adjusted for age at blood draw and smoking status (OR=2.12, 95%CI=1.09, 4.12). This association was largely restricted to women with an extended family history of breast cancer (pinteraction = 0.06). Additionally, women in the lowest tertile of MSH2 expression also had a heightened risk of breast cancer compared to women in the highest tertile of expression, adjusted for age at blood draw and smoking status (OR=2.75, 95%CI=1.31, 5.79). The association observed between reductions in ATM expression level and breast cancer risk was lost upon incorporating previously determined end-joining capacity of EcoRI-generated sticky end substrates (OR=1.28, 95%CI=0.15, 11.2) and HincII-generated blunt end substrates (OR=1.55, 95%CI=0.15, 15.5) into the model, suggesting that the impact on risk due to reductions in ATM expression maybe partially driven by the reduction in double strand break repair capacity. In our study investigating breast cancer risk due to the impact of epigenetic modulation on DNA repair gene activity (n=569, 218 sister-sets), no association with risk was observed due to differential promoter methylation levels of BRCA1 (OR=1.09, 95%CI=0.98, 1.20), MLH1 (OR=1.19, 95%CI=0.91, 1.55) or MSH2 (OR=0.89, 95%CI=0.48, 1.64). Furthermore, no correlation between BRCA1 and expression (r=-0.05, p=0.39) or MSH2 methylation and expression (r=-0.04, p=0.39) was observed. Finally, our mismatch repair genotyping study (n=714, 313 sister-sets) indicated an association between the variant MutY_rs3219489 (OR=2.23, 95%CI=1.10, 4.52) and breast cancer risk, as well as a borderline association with risk due to the variant MSH2_rs2303428 (OR=1.71, 95%CI=0.99, 2.95). Furthermore, a protective effect was observed due to the variant MLH3_rs175080, restricted to women without an extended family history of breast cancer (pinteraction = 0.03). Conclusion: These studies suggest that the deregulation of targets spanning various DNA repair pathways contribute to the risk of familial breast cancer.
机译:简介:乳腺癌诊断的广泛预后意义突显了需要使妇女更好地做出有关筛查和治疗选择的明智决定的必要性。由于几种癌症易感性综合症与种系突变相关联,导致DNA修复缺陷,包括由于BRCA1和BRCA2突变而导致的乳腺癌易感性,DNA修复能力中更细微的缺陷可能会导致推动普通人群中不同易感性的成分。因此,了解DNA修复能力在乳腺癌发病中的作用可能有助于开发更全面的风险概况,从而进一步致力于针对相关人群进行早期筛查。在针对本论文的研究中,我们采用了多种方法来捕获血液中可检测到的不同修复途径的终点,从而进一步阐明乳腺癌发展的病因基础,并利用信息将其应用于筛选生物标志物的潜在发展。方法:为了对核苷酸切除修复(NER)能力进行表型评估,我们开发了一种基于ELISA的方法来测定淋巴母细胞样细胞系中苯并(a)py二醇环氧(BPDE)-DNA加合物的能力。使用基于实时PCR的平台,使用预先设计的Taqman试剂盒评估来自单核细胞的RNA衍生cDNA中的基因表达水平。使用焦磷酸测序平台,通过内部设计的分析方法对单核细胞中亚硫酸氢盐转化的DNA进行了甲基化分析。最后,使用基于实时PCR的平台,通过预先设计的Taqman SNP基因分型测定法,评估了白细胞DNA中的单核苷酸多态性(SNP)基因型。所有研究均在乳腺癌家族登记处纽约站点的姐妹研究中进行,所有统计分析均使用R Foundation for Statistical Computing(2011)进行。结果:我们未发现姐妹组(n = 246、114个姐妹组)的淋巴母细胞系中基于ELISA的NER能力表型评估与乳腺癌风险(OR = 1.0、95%CI)之间存在关联= 0.95,1.04)。此外,我们未发现使用基于免疫组织化学的方法与同一人群中先前确定的NER能力相关(r = -0.01,p = 0.86)。在我们的基因表达研究中(n = 569,共218个姐妹组),ATM表达最低三分位数的女性与最高表达三分位数的女性相比,经抽血和吸烟状况的年龄调整后患乳腺癌的风险更高(OR = 2.12,95%CI = 1.09,4.12)。这种关联在很大程度上仅限于有乳腺癌家族史的女性(互动= 0.06)。此外,与最高三分位数表达水平的女性相比,经MSH2表达最低三分位数表达的女性,经抽血年龄和吸烟状况调整后,患乳腺癌的风险也较高(OR = 2.75,95%CI = 1.31,5.79) 。通过结合先前确定的EcoRI产生的粘性末端底物(OR = 1.28,95%CI = 0.15,11.2)和HincII产生的钝端底物的末端结合能力,丧失了ATM表达水平降低与乳腺癌风险之间的联系(OR = 1.55,95%CI = 0.15,15.5)纳入模型,表明ATM表达降低对风险的影响可能部分由双链断裂修复能力的降低所驱动。在我们的研究中,由于表观遗传调控对DNA修复基因活性的影响(n = 569,218个姐妹集),导致乳腺癌的风险,未观察到与BRCA1启动子甲基化水平差异相关的风险(OR = 1.09,95) %CI = 0.98,1.20),MLH1(OR = 1.19,95%CI = 0.91,1.55)或MSH2(OR = 0.89,95%CI = 0.48,1.64)。此外,未观察到BRCA1与表达(r = -0.05,p = 0.39)或MSH2甲基化与表达(r = -0.04,p = 0.39)之间的相关性。最后,我们的错配修复基因分型研究(n = 714,313个姐妹集)表明,变异MutY_rs3219489(OR = 2.23,95%CI = 1.10,4.52)与乳腺癌风险之间存在关联,并且与风险之间存在临界关联由于版本为MSH2_rs2303428(OR = 1.71,95%CI = 0.99,2.95)。此外,由于变体MLH3_rs175080,还观察到了保护作用,该变体仅限于没有大范围乳腺癌家族史的女性(互动= 0.03)。结论:这些研究表明,跨越多种DNA修复途径的靶标的失控导致家族性乳腺癌的风险。

著录项

  • 作者

    Kappil, Maya A.;

  • 作者单位

    Columbia University.;

  • 授予单位 Columbia University.;
  • 学科 Health Sciences Public Health.;Health Sciences Epidemiology.;Biology Molecular.
  • 学位 Dr.P.H.
  • 年度 2014
  • 页码 147 p.
  • 总页数 147
  • 原文格式 PDF
  • 正文语种 eng
  • 中图分类
  • 关键词

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号