Pub Date : 2018-07-01DOI: 10.1158/1538-7755.DISP17-B59
April E. Deveaux, Bi-Dar Wang, B. LaCroix, Brendon M. Patierno, Dadong Zhang, K. Owzar, N. Lee, D. George, J. Freedman, S. Patierno
Background: Age-adjusted incidence and mortality rates for prostate cancer (PCa) among African American (AA) men are significantly greater than among white men. Much of this disparity remains after controlling for behavioral, social, and neighborhood determinants of health, which is consistent with a biologic contribution. Evidence suggests that the genetic etiology and molecular progression of PCa are different in AAs versus whites. Our laboratory has identified novel alternatively spliced genes in AA versus white PCa biopsy tissue that track with population-based prostate tumor aggressiveness. This work addresses the urgent need to interrogate novel molecular mechanisms driving more aggressive PCa in AAs to aid in development of new biomarkers and therapeutic targets for improved prognosis and treatment of aggressive disease. Methods: We have analyzed RNA from human PCa biopsy tissue from 20 AA and 15 white patients using exon arrays. From these data, we prioritized targets for further study by identifying those associated with androgen signaling and PCa and those that have also been shown to be alternatively spliced in breast, lung, and liver cancer. To validate race-related alternative splicing of the insulin receptor (INSR) gene, we have used qPCR and targeted sequencing of RNA from population-specific PCa cell lines and patient-derived explants. In addition, we have knocked down the INSR splice variants in population-specific PCa cell lines using isoform-specific siRNAs and assessed resulting alterations in proliferation using a WST-1 proliferation assay. Current studies include assessing additional resulting alterations in expression of selected key targets in INSR signaling using Western blot analysis. In addition, studies are under way to treat population-specific PCa cell lines with INSR signaling inhibitors and assess resulting alterations in PCa cell biology. Results: Exon array analysis of RNA from AA and white PCa biopsy tissue revealed 934 differentially expressed exons in 861 corresponding genes. Thirty-nine targets have been prioritized for further study. We have validated race-related alternative splicing of INSR, involving differential exon inclusion/skipping of exon 11, in population-specific PCa cell lines and patient-derived explants, with the exclusion of exon 11 occurring more often in preclinical models of white PCa. Knockdown of INSR splice variants in population-specific PCa cell lines has shown that decreased expression of the isoform excluding exon 11 inhibits proliferation while decreased expression of the isoform including exon 11 has no effect on proliferation. Further studies to assess additional resulting alterations in expression of selected key targets in INSR signaling are under way. In addition, studies are under way to treat population-specific PCa cell lines with INSR signaling inhibitors and assess resulting alterations in PCa cell biology. Conclusions: These studies identify differential splicing of INSR
背景:非裔美国人(AA)男性前列腺癌(PCa)的年龄校正发病率和死亡率明显高于白人男性。在控制了健康的行为、社会和邻里决定因素之后,这种差异在很大程度上仍然存在,这与生物学的贡献是一致的。有证据表明,遗传病因学和分子进展的前列腺癌是不同的白人与白人。我们的实验室已经在AA和白色PCa活检组织中发现了新的选择性剪接基因,这些基因与基于人群的前列腺肿瘤侵袭性有关。这项工作解决了迫切需要询问在AAs中驱动更具侵袭性PCa的新分子机制,以帮助开发新的生物标志物和治疗靶点,以改善预后和治疗侵袭性疾病。方法:我们使用外显子阵列分析了20例AA和15例白人患者PCa活检组织的RNA。根据这些数据,我们确定了进一步研究的优先目标,确定了那些与雄激素信号和PCa相关的目标,以及那些在乳腺癌、肺癌和肝癌中被证明是选择性剪接的目标。为了验证与种族相关的胰岛素受体(INSR)基因的选择性剪接,我们使用了qPCR和来自人群特异性PCa细胞系和患者来源的外植体的RNA靶向测序。此外,我们使用同种异型特异性sirna敲除了种群特异性PCa细胞系中的INSR剪接变体,并使用WST-1增殖试验评估了由此产生的增殖变化。目前的研究包括使用Western blot分析评估INSR信号传导中选定关键靶点表达的额外变化。此外,用INSR信号抑制剂治疗人群特异性PCa细胞系的研究正在进行中,并评估PCa细胞生物学的变化。结果:对AA和白色PCa活检组织的RNA进行外显子阵列分析,发现861个相应基因中有934个差异外显子表达。已确定39个目标优先进行进一步研究。我们已经在人群特异性PCa细胞系和患者来源的外植体中验证了与种族相关的INSR选择性剪接,包括外显子11的差异包涵/跳跃,而外显子11的排除在白人PCa临床前模型中更常见。在群体特异性PCa细胞系中敲低INSR剪接变异体表明,不包括外显子11的异构体表达减少可抑制增殖,而包括外显子11的异构体表达减少对增殖没有影响。目前正在进行进一步的研究,以评估INSR信号传导中选定关键靶点表达的额外变化。此外,用INSR信号抑制剂治疗人群特异性PCa细胞系的研究正在进行中,并评估PCa细胞生物学的变化。结论:这些研究确定了AA和白色PCa中INSR的不同剪接以及排除外显子11的INSR异构体是前列腺肿瘤侵袭性的促进因素。进一步探究前列腺癌差异背后的机制及其对靶向治疗的影响,有可能减少AA男性的前列腺癌差异,并改善由该机制驱动的侵袭性疾病的所有种族男性的预后。引文格式:April Deveaux, Bi-Dar Wang, Bonnie Lacroix, Brendon Patierno,张大东,Kouros Owzar, Norman Lee, Daniel George, Jennifer Freedman, Steven Patierno。种族相关的胰岛素受体剪接差异:前列腺癌差异的新靶点[摘要]。见:第十届AACR会议论文集:种族/少数民族和医疗服务不足人群的癌症健康差异科学;2017年9月25-28日;亚特兰大,乔治亚州。费城(PA): AACR;癌症流行病学杂志,2018;27(7增刊):摘要nr B59。
{"title":"Abstract B59: Race-related differential splicing of the insulin receptor: A novel target underlying prostate cancer disparities","authors":"April E. Deveaux, Bi-Dar Wang, B. LaCroix, Brendon M. Patierno, Dadong Zhang, K. Owzar, N. Lee, D. George, J. Freedman, S. Patierno","doi":"10.1158/1538-7755.DISP17-B59","DOIUrl":"https://doi.org/10.1158/1538-7755.DISP17-B59","url":null,"abstract":"Background: Age-adjusted incidence and mortality rates for prostate cancer (PCa) among African American (AA) men are significantly greater than among white men. Much of this disparity remains after controlling for behavioral, social, and neighborhood determinants of health, which is consistent with a biologic contribution. Evidence suggests that the genetic etiology and molecular progression of PCa are different in AAs versus whites. Our laboratory has identified novel alternatively spliced genes in AA versus white PCa biopsy tissue that track with population-based prostate tumor aggressiveness. This work addresses the urgent need to interrogate novel molecular mechanisms driving more aggressive PCa in AAs to aid in development of new biomarkers and therapeutic targets for improved prognosis and treatment of aggressive disease. Methods: We have analyzed RNA from human PCa biopsy tissue from 20 AA and 15 white patients using exon arrays. From these data, we prioritized targets for further study by identifying those associated with androgen signaling and PCa and those that have also been shown to be alternatively spliced in breast, lung, and liver cancer. To validate race-related alternative splicing of the insulin receptor (INSR) gene, we have used qPCR and targeted sequencing of RNA from population-specific PCa cell lines and patient-derived explants. In addition, we have knocked down the INSR splice variants in population-specific PCa cell lines using isoform-specific siRNAs and assessed resulting alterations in proliferation using a WST-1 proliferation assay. Current studies include assessing additional resulting alterations in expression of selected key targets in INSR signaling using Western blot analysis. In addition, studies are under way to treat population-specific PCa cell lines with INSR signaling inhibitors and assess resulting alterations in PCa cell biology. Results: Exon array analysis of RNA from AA and white PCa biopsy tissue revealed 934 differentially expressed exons in 861 corresponding genes. Thirty-nine targets have been prioritized for further study. We have validated race-related alternative splicing of INSR, involving differential exon inclusion/skipping of exon 11, in population-specific PCa cell lines and patient-derived explants, with the exclusion of exon 11 occurring more often in preclinical models of white PCa. Knockdown of INSR splice variants in population-specific PCa cell lines has shown that decreased expression of the isoform excluding exon 11 inhibits proliferation while decreased expression of the isoform including exon 11 has no effect on proliferation. Further studies to assess additional resulting alterations in expression of selected key targets in INSR signaling are under way. In addition, studies are under way to treat population-specific PCa cell lines with INSR signaling inhibitors and assess resulting alterations in PCa cell biology. Conclusions: These studies identify differential splicing of INSR ","PeriodicalId":146931,"journal":{"name":"Cell, Molecular, and Tumor Biology","volume":"16 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2018-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"129496078","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2018-07-01DOI: 10.1158/1538-7755.DISP17-B63
Aristeidis G. Telonis, I. Rigoutsos
Triple-negative breast cancer (TNBC) is a subtype of breast cancer that is characterized by marked differences between Black/African-American (B/Aa) and White (Wh) women. Despite the great progress that has been achieved towards understanding the causes of race disparities in TNBC, there have been no system-level investigations of the problem from the standpoint of molecular oncology. We used the transcriptomic data from The Cancer Genome Atlas (TCGA) repository to study the normal breast and TNBC samples from Wh and B/Aa TNBC individuals that are contained in it. Specifically, our analysis integrated transcriptomic data from messenger RNAs (mRNAs), microRNAs (miRNAs), miRNA isoforms (isomiRs), and tRNA-derived fragments (tRFs). IsomiRs and tRFs are constitutive regulatory molecules that target genes and pathways through RNA interference (RNAi). In addition to RNAi, isomiRs and tRFs also have other modes of action that are not yet understood. IsomiRs and tRFs represent recent and very important advances in our understanding of post-transcriptional regulation in the cell. What makes them particularly relevant for this study is our having shown that the “footprint” of isomiRs and tRFs that are active in a cell depends strongly on a person9s gender, race, and population origin, as well as on tissue type, tissue state, and disease type/subtype. We analyzed and compared TCGA samples from normal breast and TNBC and did so separately for B/Aa and Wh donors. As expected, we found many mRNAs to be differentially expressed. More importantly, when we compared B/Aa and Wh TNBC patients we found many mRNAs to be differentially wired. By this we mean that, even though the two groups of patients have many regulators and effectors in common, there are very extensive differences in the manner regulators interact with effectors in B/Aa and in Wh TNBC patients, respectively. Our accumulated evidence suggests strongly that this differential wiring is an important driver of the phenotypic differences that we observe between B/Aa and Wh TNBC patients. Our studies show that there is a multitude of regulatory molecules that are involved in this rewiring and the racial disparities in TNBC. Among isomiRs, we identified numerous isomiRs that arise from the mir-200c and mir-21 miRNA precursors, and from the oncogenic miR-17/92 and miR-183/96/182 clusters. Among tRFs, we identified many tRFs that arise from nuclear (e.g., Glycine, Leucine) and mitochondrial tRNAs (e.g., Valine, Proline). We showcase this race-specific rewiring with the help of MAPK and Wnt signaling, two metastasis-related pathways. The data-driven approaches that we employed in the analysis enabled us to carry out comprehensive studies of the transcriptomes of two states (normal and cancer) and two races (B/Aa and Wh). Our methodology allowed us to dissect the complex regulatory events that are mediated by isomiRs and tRFs. It also helped us unravel the significant roles that isomiRs and tRFs have in reshap
三阴性乳腺癌(TNBC)是乳腺癌的一种亚型,其特征在黑人/非裔美国人(B/Aa)和白人(Wh)妇女之间存在显著差异。尽管在了解TNBC中种族差异的原因方面已经取得了很大的进展,但还没有从分子肿瘤学的角度对这个问题进行系统级的调查。我们利用来自the Cancer Genome Atlas (TCGA)知识库的转录组学数据,研究了其中包含的Wh和B/Aa TNBC个体的正常乳腺和TNBC样本。具体来说,我们的分析整合了信使rna (mrna)、微rna (miRNAs)、miRNA异构体(isomir)和trna衍生片段(trf)的转录组学数据。异构体和trf是通过RNA干扰(RNAi)靶向基因和途径的组成性调控分子。除了RNAi外,isomir和trf还有其他尚未了解的作用模式。isomir和trf代表了我们对细胞转录后调控的理解的最新和非常重要的进展。使它们与本研究特别相关的是,我们已经表明,在细胞中活跃的异构体和trf的“足迹”在很大程度上取决于人的性别、种族和人口来源,以及组织类型、组织状态和疾病类型/亚型。我们分析和比较了正常乳腺和TNBC的TCGA样本,并分别对B/Aa和Wh供体进行了分析和比较。正如预期的那样,我们发现许多mrna是差异表达的。更重要的是,当我们比较B/Aa和Wh TNBC患者时,我们发现许多mrna的连接方式不同。我们的意思是,尽管两组患者有许多共同的调节剂和效应器,但在B/Aa和Wh TNBC患者中,调节剂与效应器相互作用的方式存在非常广泛的差异。我们积累的证据强烈表明,这种不同的线路是我们在B/Aa和Wh TNBC患者之间观察到的表型差异的重要驱动因素。我们的研究表明,在TNBC中,有大量的调控分子参与了这种重新布线和种族差异。在异构体中,我们发现了许多来自mir-200c和mir-21 miRNA前体以及致癌miR-17/92和miR-183/96/182簇的异构体。在trf中,我们发现许多trf来自核(如甘氨酸、亮氨酸)和线粒体trna(如缬氨酸、脯氨酸)。我们在MAPK和Wnt信号(两种与转移相关的途径)的帮助下展示了这种种族特异性的重新布线。我们在分析中采用的数据驱动方法使我们能够对两种状态(正常和癌症)和两个种族(B/Aa和Wh)的转录组进行全面研究。我们的方法使我们能够剖析由异构体和trf介导的复杂调控事件。它还帮助我们揭示了异构体和trf在两个种族中重塑mRNA表达的差异中的重要作用。总的来说,我们的研究结果表明,系统生物学水平的途径可能在TNBC和其他癌症的差异研究中证明是富有成效的。引文格式:Aristeidis G. Telonis, Isidore Rigoutsos。种族和三阴性乳腺癌:非编码RNA研究进展和系统生物学水平分析揭示了关键的分子差异[摘要]。见:第十届AACR会议论文集:种族/少数民族和医疗服务不足人群的癌症健康差异科学;2017年9月25-28日;亚特兰大,乔治亚州。费城(PA): AACR;癌症流行病学杂志,2018;27(7增刊):摘要nr B63。
{"title":"Abstract B63: Race and triple-negative breast cancer: Advances in noncoding RNA research together with a systems-biology-level analysis uncover key molecular differences","authors":"Aristeidis G. Telonis, I. Rigoutsos","doi":"10.1158/1538-7755.DISP17-B63","DOIUrl":"https://doi.org/10.1158/1538-7755.DISP17-B63","url":null,"abstract":"Triple-negative breast cancer (TNBC) is a subtype of breast cancer that is characterized by marked differences between Black/African-American (B/Aa) and White (Wh) women. Despite the great progress that has been achieved towards understanding the causes of race disparities in TNBC, there have been no system-level investigations of the problem from the standpoint of molecular oncology. We used the transcriptomic data from The Cancer Genome Atlas (TCGA) repository to study the normal breast and TNBC samples from Wh and B/Aa TNBC individuals that are contained in it. Specifically, our analysis integrated transcriptomic data from messenger RNAs (mRNAs), microRNAs (miRNAs), miRNA isoforms (isomiRs), and tRNA-derived fragments (tRFs). IsomiRs and tRFs are constitutive regulatory molecules that target genes and pathways through RNA interference (RNAi). In addition to RNAi, isomiRs and tRFs also have other modes of action that are not yet understood. IsomiRs and tRFs represent recent and very important advances in our understanding of post-transcriptional regulation in the cell. What makes them particularly relevant for this study is our having shown that the “footprint” of isomiRs and tRFs that are active in a cell depends strongly on a person9s gender, race, and population origin, as well as on tissue type, tissue state, and disease type/subtype. We analyzed and compared TCGA samples from normal breast and TNBC and did so separately for B/Aa and Wh donors. As expected, we found many mRNAs to be differentially expressed. More importantly, when we compared B/Aa and Wh TNBC patients we found many mRNAs to be differentially wired. By this we mean that, even though the two groups of patients have many regulators and effectors in common, there are very extensive differences in the manner regulators interact with effectors in B/Aa and in Wh TNBC patients, respectively. Our accumulated evidence suggests strongly that this differential wiring is an important driver of the phenotypic differences that we observe between B/Aa and Wh TNBC patients. Our studies show that there is a multitude of regulatory molecules that are involved in this rewiring and the racial disparities in TNBC. Among isomiRs, we identified numerous isomiRs that arise from the mir-200c and mir-21 miRNA precursors, and from the oncogenic miR-17/92 and miR-183/96/182 clusters. Among tRFs, we identified many tRFs that arise from nuclear (e.g., Glycine, Leucine) and mitochondrial tRNAs (e.g., Valine, Proline). We showcase this race-specific rewiring with the help of MAPK and Wnt signaling, two metastasis-related pathways. The data-driven approaches that we employed in the analysis enabled us to carry out comprehensive studies of the transcriptomes of two states (normal and cancer) and two races (B/Aa and Wh). Our methodology allowed us to dissect the complex regulatory events that are mediated by isomiRs and tRFs. It also helped us unravel the significant roles that isomiRs and tRFs have in reshap","PeriodicalId":146931,"journal":{"name":"Cell, Molecular, and Tumor Biology","volume":"282 ","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2018-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"113983031","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2018-07-01DOI: 10.1158/1538-7755.DISP17-C46
S. Nomura, C. Dash, L. Rosenberg, J. Palmer, L. Adams-Campbell
Objective: The objective of this project was to evaluate associations of fruit and vegetable intake, according to cigarette smoking history, with lung cancer incidence among U.S. Black women. Methods: The Black Women9s Health Study is a prospective cohort study (analytic cohort=46,889) among Black women between the ages 21-69 at baseline (1995). Fruit and vegetable intake (collected in 1995 and 2001only) and smoking history were ascertained via questionnaires at baseline and during follow-up (every other year). Combined fruit and vegetable ( 1 servings/day), and cruciferous vegetable ( 1 servings/day) intakes were evaluated. Cigarette smoking measures that were evaluated include: 1) current smoking status (never, former, current 2 tests were utilized to assess interactions between smoking history and fruit and vegetable intake. Results: More than half the women reported no history of cigarette smoking (66.4%), while 18.4% were former smokers and 15.1% were current smokers ( Conclusion: Low fruit and vegetable intake was widespread in this population of U.S. Black women, but results do not support an association between fruit and vegetable intake and lung cancer incidence, regardless of cigarette smoking history. Citation Format: Sarah JO Nomura, Chiranjeev Dash, Lynn Rosenberg, Julie Palmer, Lucile L. Adams-Campbell. Fruit and vegetable intake and lung cancer incidence among Black women according to cigarette smoking status [abstract]. In: Proceedings of the Tenth AACR Conference on the Science of Cancer Health Disparities in Racial/Ethnic Minorities and the Medically Underserved; 2017 Sep 25-28; Atlanta, GA. Philadelphia (PA): AACR; Cancer Epidemiol Biomarkers Prev 2018;27(7 Suppl):Abstract nr C46.
目的:该项目的目的是评估美国黑人妇女中,根据吸烟史,水果和蔬菜摄入量与肺癌发病率的关系。方法:黑人妇女健康研究是一项前瞻性队列研究(分析队列=46,889),研究对象为基线年龄在21-69岁之间的黑人妇女(1995)。在基线和随访期间(每隔一年)通过问卷调查确定水果和蔬菜摄入量(仅在1995年和2001年收集)和吸烟史。评估了水果和蔬菜(1份/天)和十字花科蔬菜(1份/天)的摄入量。评估的吸烟措施包括:1)目前吸烟状况(从不,以前,目前2项测试用于评估吸烟史与水果和蔬菜摄入量之间的相互作用。结果:超过一半的女性报告没有吸烟史(66.4%),而18.4%是前吸烟者,15.1%是当前吸烟者(结论:低水果和蔬菜摄入量在美国黑人女性人群中普遍存在,但结果不支持水果和蔬菜摄入量与肺癌发病率之间的关联,无论吸烟史如何。引用格式:Sarah JO Nomura, Chiranjeev Dash, Lynn Rosenberg, Julie Palmer, Lucile L. Adams-Campbell。吸烟状况下黑人女性果蔬摄入量与肺癌发病率的关系[摘要]。见:第十届AACR会议论文集:种族/少数民族和医疗服务不足人群的癌症健康差异科学;2017年9月25-28日;亚特兰大,乔治亚州。费城(PA): AACR;癌症流行病学杂志,2018;27(7增刊):摘要nr C46。
{"title":"Abstract C46: Fruit and vegetable intake and lung cancer incidence among Black women according to cigarette smoking status","authors":"S. Nomura, C. Dash, L. Rosenberg, J. Palmer, L. Adams-Campbell","doi":"10.1158/1538-7755.DISP17-C46","DOIUrl":"https://doi.org/10.1158/1538-7755.DISP17-C46","url":null,"abstract":"Objective: The objective of this project was to evaluate associations of fruit and vegetable intake, according to cigarette smoking history, with lung cancer incidence among U.S. Black women. Methods: The Black Women9s Health Study is a prospective cohort study (analytic cohort=46,889) among Black women between the ages 21-69 at baseline (1995). Fruit and vegetable intake (collected in 1995 and 2001only) and smoking history were ascertained via questionnaires at baseline and during follow-up (every other year). Combined fruit and vegetable ( 1 servings/day), and cruciferous vegetable ( 1 servings/day) intakes were evaluated. Cigarette smoking measures that were evaluated include: 1) current smoking status (never, former, current 2 tests were utilized to assess interactions between smoking history and fruit and vegetable intake. Results: More than half the women reported no history of cigarette smoking (66.4%), while 18.4% were former smokers and 15.1% were current smokers ( Conclusion: Low fruit and vegetable intake was widespread in this population of U.S. Black women, but results do not support an association between fruit and vegetable intake and lung cancer incidence, regardless of cigarette smoking history. Citation Format: Sarah JO Nomura, Chiranjeev Dash, Lynn Rosenberg, Julie Palmer, Lucile L. Adams-Campbell. Fruit and vegetable intake and lung cancer incidence among Black women according to cigarette smoking status [abstract]. In: Proceedings of the Tenth AACR Conference on the Science of Cancer Health Disparities in Racial/Ethnic Minorities and the Medically Underserved; 2017 Sep 25-28; Atlanta, GA. Philadelphia (PA): AACR; Cancer Epidemiol Biomarkers Prev 2018;27(7 Suppl):Abstract nr C46.","PeriodicalId":146931,"journal":{"name":"Cell, Molecular, and Tumor Biology","volume":"97 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2018-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"115664953","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2018-07-01DOI: 10.1158/1538-7755.DISP17-B56
Adaugo Q. Ohandjo, C. Dill, C. Dongquan, J. Lillard
The prevalence of prostate cancer (PCa) has been a worldwide burden. Cancer of the prostate is the most prevalent form of cancer in men and the second leading cause of death due to cancer in men in the United States and Europe. There is a major health disparity associated with PCa; the number of fatalities associated with this disease is more significant in African-American (AA) men than in European American (EA) men. Disease progression occurs despite hormone ablation therapy, which leads to a condition called castration-resistant prostate cancer (CRPC). Further complicating the matter, CRPC is a disease that affects a variety of patients differently, which can be problematic for physicians to provide standardized treatments with similar outcomes. AAs with CRPC can experience significantly lower rates of overall survival, faster rates of tumor progression, and poor responses to chemotherapy compared to EAs with this disease. We previously showed that CXCL13:CXCR5 axis plays a role in PCa pathobiology and is also associated with PCa aggressiveness; CXCR5 signaling mediates PCa cell growth, migration, invasion, and docetaxel resistance. To further understand the implication of CXCR5-CXCL13 in CRPC, we characterize the reason behind the health disparities seen across groups of PCa patients by evaluating the difference in regulatory molecules (miRNAs) on CXCL13-CXCR5 signaling. We found the expression profile of CXCL13 and CXCR5 RNA to be associated with miRNAs (miR-140-3p.1, miR-24-3p, miR-200c, miR-221-3p/22-3p, miR-192-5p/215-5p, miR-214-5p). Using RNA-seq datasets from the Gene Expression Omnibus (GEO) and the Sequence Read Archive (SRA), CXCL13 and CXCR5 RNA as well as associated miRNAs (miR-140-3p.1, miR-24-3p, miR-221-3p/22-3p, miR-192-5p/215-5p, and miR-214-5p) were defined by etiologic disparities among PCa patients; this correlation is different among AA men compared to EA men. Understanding PCa and specifically CRPC disparities requires a better understanding of the race-specific differences in prostate tumor biology, molecules leading to prostate inflammation, and the mechanisms associated with CRPC therapeutic resistance and its aggressive phenotype. Citation Format: Adaugo Queen Ohandjo, Courtney Dill, Chen Dongquan, James W. Lillard, Jr.. Regulation of CXCR5-CXCL13 signaling in prostate cancer etiologic disparities [abstract]. In: Proceedings of the Tenth AACR Conference on the Science of Cancer Health Disparities in Racial/Ethnic Minorities and the Medically Underserved; 2017 Sep 25-28; Atlanta, GA. Philadelphia (PA): AACR; Cancer Epidemiol Biomarkers Prev 2018;27(7 Suppl):Abstract nr B56.
前列腺癌(PCa)的患病率一直是一个世界性的负担。前列腺癌是男性中最常见的癌症,也是美国和欧洲男性因癌症死亡的第二大原因。与前列腺癌相关的主要健康差异;与此疾病相关的死亡人数在非洲裔美国人(AA)中比在欧洲裔美国人(EA)中更显著。尽管激素消融治疗,疾病仍会进展,导致一种称为去势抵抗性前列腺癌(CRPC)的疾病。更复杂的是,CRPC是一种影响不同患者的疾病,这对于医生提供具有相似结果的标准化治疗可能是一个问题。与患有这种疾病的ea相比,患有CRPC的AAs的总生存率明显较低,肿瘤进展速度更快,对化疗的反应较差。我们之前的研究表明,CXCL13:CXCR5轴在前列腺癌的病理生物学中发挥作用,也与前列腺癌的侵袭性有关;CXCR5信号通路介导PCa细胞生长、迁移、侵袭和多西他赛耐药。为了进一步了解CXCR5-CXCL13在CRPC中的意义,我们通过评估CXCL13-CXCR5信号调控分子(mirna)的差异,描述了不同PCa患者健康差异背后的原因。我们发现CXCL13和CXCR5 RNA的表达谱与mirna (miR-140-3p)相关。1、miR-24-3p、miR-200c、miR-221-3p/22-3p、miR-192-5p/215-5p、miR-214-5p)。使用来自基因表达综合(GEO)和序列读取档案(SRA)的RNA-seq数据集,CXCL13和CXCR5 RNA以及相关的miRNAs (miR-140-3p)。1、通过前列腺癌患者的病因差异来定义miR-24-3p、miR-221-3p/22-3p、miR-192-5p/215-5p和miR-214-5p;与EA男性相比,AA男性的这种相关性有所不同。了解前列腺癌和特异性CRPC差异,需要更好地了解前列腺肿瘤生物学、导致前列腺炎症的分子以及CRPC治疗耐药及其侵袭性表型的相关机制。引文格式:Adaugo Queen Ohandjo, Courtney Dill,陈东泉,James W. Lillard, Jr..CXCR5-CXCL13信号在前列腺癌病因差异中的调控[摘要]。见:第十届AACR会议论文集:种族/少数民族和医疗服务不足人群的癌症健康差异科学;2017年9月25-28日;亚特兰大,乔治亚州。费城(PA): AACR;癌症流行病学杂志,2018;27(7增刊):摘要nr B56。
{"title":"Abstract B56: Regulation of CXCR5-CXCL13 signaling in prostate cancer etiologic disparities","authors":"Adaugo Q. Ohandjo, C. Dill, C. Dongquan, J. Lillard","doi":"10.1158/1538-7755.DISP17-B56","DOIUrl":"https://doi.org/10.1158/1538-7755.DISP17-B56","url":null,"abstract":"The prevalence of prostate cancer (PCa) has been a worldwide burden. Cancer of the prostate is the most prevalent form of cancer in men and the second leading cause of death due to cancer in men in the United States and Europe. There is a major health disparity associated with PCa; the number of fatalities associated with this disease is more significant in African-American (AA) men than in European American (EA) men. Disease progression occurs despite hormone ablation therapy, which leads to a condition called castration-resistant prostate cancer (CRPC). Further complicating the matter, CRPC is a disease that affects a variety of patients differently, which can be problematic for physicians to provide standardized treatments with similar outcomes. AAs with CRPC can experience significantly lower rates of overall survival, faster rates of tumor progression, and poor responses to chemotherapy compared to EAs with this disease. We previously showed that CXCL13:CXCR5 axis plays a role in PCa pathobiology and is also associated with PCa aggressiveness; CXCR5 signaling mediates PCa cell growth, migration, invasion, and docetaxel resistance. To further understand the implication of CXCR5-CXCL13 in CRPC, we characterize the reason behind the health disparities seen across groups of PCa patients by evaluating the difference in regulatory molecules (miRNAs) on CXCL13-CXCR5 signaling. We found the expression profile of CXCL13 and CXCR5 RNA to be associated with miRNAs (miR-140-3p.1, miR-24-3p, miR-200c, miR-221-3p/22-3p, miR-192-5p/215-5p, miR-214-5p). Using RNA-seq datasets from the Gene Expression Omnibus (GEO) and the Sequence Read Archive (SRA), CXCL13 and CXCR5 RNA as well as associated miRNAs (miR-140-3p.1, miR-24-3p, miR-221-3p/22-3p, miR-192-5p/215-5p, and miR-214-5p) were defined by etiologic disparities among PCa patients; this correlation is different among AA men compared to EA men. Understanding PCa and specifically CRPC disparities requires a better understanding of the race-specific differences in prostate tumor biology, molecules leading to prostate inflammation, and the mechanisms associated with CRPC therapeutic resistance and its aggressive phenotype. Citation Format: Adaugo Queen Ohandjo, Courtney Dill, Chen Dongquan, James W. Lillard, Jr.. Regulation of CXCR5-CXCL13 signaling in prostate cancer etiologic disparities [abstract]. In: Proceedings of the Tenth AACR Conference on the Science of Cancer Health Disparities in Racial/Ethnic Minorities and the Medically Underserved; 2017 Sep 25-28; Atlanta, GA. Philadelphia (PA): AACR; Cancer Epidemiol Biomarkers Prev 2018;27(7 Suppl):Abstract nr B56.","PeriodicalId":146931,"journal":{"name":"Cell, Molecular, and Tumor Biology","volume":"101 2 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2018-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"122974629","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2018-07-01DOI: 10.1158/1538-7755.disp17-pr04
B. LaCroix, Brendon M. Patierno, B. Sullenger, D. George, S. Patierno, J. Freedman
{"title":"Abstract PR04: Development of a novel therapeutic splice-switching oligonucleotide targeting race-related androgen receptor signaling and aggressive prostate cancer","authors":"B. LaCroix, Brendon M. Patierno, B. Sullenger, D. George, S. Patierno, J. Freedman","doi":"10.1158/1538-7755.disp17-pr04","DOIUrl":"https://doi.org/10.1158/1538-7755.disp17-pr04","url":null,"abstract":"","PeriodicalId":146931,"journal":{"name":"Cell, Molecular, and Tumor Biology","volume":"179 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2018-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"123840641","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2018-07-01DOI: 10.1158/1538-7755.DISP17-B58
E. Girsch, B. Bao, C. Mitrea, W. Sakr, G. Dyson, I. Powell, Aliccia Bollig-Fischer
Background: African American men (AAM) have a 60% higher risk of being diagnosed with prostate cancer and a 2 to 3 times greater risk of dying from the disease compared to European American men (EAM). We previously reported evidence of molecular underpinnings for these disparities from high-throughput gene expression analysis of prostate cancer (PCa) specimens. The data showed that proinflammatory cytokine signaling factors, including IL6 and TGFB1, are significantly over-represented in PCa specimens from AAM. The limitation of that study, however, is that it did not investigate the potential molecular diversity for high-grade PCa, and it did not consider adjacent noncancer tissue. Methods: We conducted a pilot RNA-sequencing study of high-grade PCa, GS greater or equal to 7(4+3), and matched non-cancer adjacent prostate tissue specimens from AAM and EAM. We also studied the molecular biology of PCa cell lines derived from AAM and EAM tumors. Results: According to our genomic analysis, IL6 was once again relevant to our research, this time upregulated in PCa specimens from EAM, but also higher in the stromal compartment in AAM. Also, we noted that whether or not a PCa cell line expresses IL6 appears to be linked to TP53 mutation status. Moreover, cell lines derived from AAM (MDA-PCa-2b and RC77T), which are TP53 wild-type, did not express IL6 and showed an increased stem cell-like phenotype in response to IL6 treatment that was dose dependent. PCa cell lines that are TP53 mutant and expressed IL6 did not respond to exogenous IL6 treatment. We uncovered that in responsive PCa cell lines, IL6 treatment induced mRNA and protein expression of the epigenetic reader methyl CpG binding domain protein 2 (MBD2), more specifically the alternative mRNA splicing variant MBD2_v2. Further investigation validated that this short isoform promotes self-renewal and expansion of PCa stem cell-like cells. Conclusion: The outcomes suggest that there may be a dual nature for IL6 signaling in PCa that remains to be understood, yet contributes to the molecular diversity of high-grade prostate cancer and race disparities. Citation Format: Emily Girsch, Bin Bao, Cristina Mitrea, Wael A. Sakr, Gregory Dyson, Isaac Powell, Aliccia Bollig-Fischer. The role of epigenetic reader and alternative mRNA splicing variant MBD2_v2 in IL6 signaling in prostate cancer [abstract]. In: Proceedings of the Tenth AACR Conference on the Science of Cancer Health Disparities in Racial/Ethnic Minorities and the Medically Underserved; 2017 Sep 25-28; Atlanta, GA. Philadelphia (PA): AACR; Cancer Epidemiol Biomarkers Prev 2018;27(7 Suppl):Abstract nr B58.
背景:非洲裔美国男性(AAM)被诊断患有前列腺癌的风险比欧洲裔美国男性(EAM)高60%,死于该疾病的风险是欧洲裔美国男性(EAM)的2至3倍。我们之前报道了前列腺癌(PCa)标本高通量基因表达分析中这些差异的分子基础证据。数据显示,促炎细胞因子信号因子,包括IL6和TGFB1,在AAM的PCa标本中显着过度代表。然而,该研究的局限性在于,它没有研究高级别PCa的潜在分子多样性,也没有考虑邻近的非癌组织。方法:我们对高级别前列腺癌、GS≥7(4+3)以及AAM和EAM非癌旁前列腺组织标本进行了一项先导rna测序研究。我们还研究了来自AAM和EAM肿瘤的PCa细胞系的分子生物学。结果:根据我们的基因组分析,IL6再次与我们的研究相关,这次在EAM的PCa标本中上调,但在AAM的基质室中也较高。此外,我们注意到PCa细胞系是否表达IL6似乎与TP53突变状态有关。此外,来自AAM的细胞系(MDA-PCa-2b和RC77T)是TP53野生型,它们不表达IL6,并且在IL6的剂量依赖性作用下表现出增加的干细胞样表型。TP53突变和表达IL6的PCa细胞系对外源性IL6治疗没有反应。我们发现,在响应性PCa细胞系中,IL6处理诱导表观遗传解读器甲基CpG结合域蛋白2 (MBD2)的mRNA和蛋白表达,更具体地说是替代mRNA剪接变体MBD2_v2。进一步的研究证实,这种短同种异构体促进了PCa干细胞样细胞的自我更新和扩增。结论:这些结果表明,前列腺癌中il - 6信号可能具有双重性质,这仍有待了解,但有助于高级别前列腺癌的分子多样性和种族差异。引文格式:Emily Girsch, Bin Bao, Cristina Mitrea, Wael A. Sakr, Gregory Dyson, Isaac Powell, alicia bolligi - fischer。表观遗传解读子和备选mRNA剪接变体MBD2_v2在前列腺癌中IL6信号传导中的作用[摘要]。见:第十届AACR会议论文集:种族/少数民族和医疗服务不足人群的癌症健康差异科学;2017年9月25-28日;亚特兰大,乔治亚州。费城(PA): AACR;癌症流行病学杂志,2018;27(7增刊):摘要nr B58。
{"title":"Abstract B58: The role of epigenetic reader and alternative mRNA splicing variant MBD2_v2 in IL6 signaling in prostate cancer","authors":"E. Girsch, B. Bao, C. Mitrea, W. Sakr, G. Dyson, I. Powell, Aliccia Bollig-Fischer","doi":"10.1158/1538-7755.DISP17-B58","DOIUrl":"https://doi.org/10.1158/1538-7755.DISP17-B58","url":null,"abstract":"Background: African American men (AAM) have a 60% higher risk of being diagnosed with prostate cancer and a 2 to 3 times greater risk of dying from the disease compared to European American men (EAM). We previously reported evidence of molecular underpinnings for these disparities from high-throughput gene expression analysis of prostate cancer (PCa) specimens. The data showed that proinflammatory cytokine signaling factors, including IL6 and TGFB1, are significantly over-represented in PCa specimens from AAM. The limitation of that study, however, is that it did not investigate the potential molecular diversity for high-grade PCa, and it did not consider adjacent noncancer tissue. Methods: We conducted a pilot RNA-sequencing study of high-grade PCa, GS greater or equal to 7(4+3), and matched non-cancer adjacent prostate tissue specimens from AAM and EAM. We also studied the molecular biology of PCa cell lines derived from AAM and EAM tumors. Results: According to our genomic analysis, IL6 was once again relevant to our research, this time upregulated in PCa specimens from EAM, but also higher in the stromal compartment in AAM. Also, we noted that whether or not a PCa cell line expresses IL6 appears to be linked to TP53 mutation status. Moreover, cell lines derived from AAM (MDA-PCa-2b and RC77T), which are TP53 wild-type, did not express IL6 and showed an increased stem cell-like phenotype in response to IL6 treatment that was dose dependent. PCa cell lines that are TP53 mutant and expressed IL6 did not respond to exogenous IL6 treatment. We uncovered that in responsive PCa cell lines, IL6 treatment induced mRNA and protein expression of the epigenetic reader methyl CpG binding domain protein 2 (MBD2), more specifically the alternative mRNA splicing variant MBD2_v2. Further investigation validated that this short isoform promotes self-renewal and expansion of PCa stem cell-like cells. Conclusion: The outcomes suggest that there may be a dual nature for IL6 signaling in PCa that remains to be understood, yet contributes to the molecular diversity of high-grade prostate cancer and race disparities. Citation Format: Emily Girsch, Bin Bao, Cristina Mitrea, Wael A. Sakr, Gregory Dyson, Isaac Powell, Aliccia Bollig-Fischer. The role of epigenetic reader and alternative mRNA splicing variant MBD2_v2 in IL6 signaling in prostate cancer [abstract]. In: Proceedings of the Tenth AACR Conference on the Science of Cancer Health Disparities in Racial/Ethnic Minorities and the Medically Underserved; 2017 Sep 25-28; Atlanta, GA. Philadelphia (PA): AACR; Cancer Epidemiol Biomarkers Prev 2018;27(7 Suppl):Abstract nr B58.","PeriodicalId":146931,"journal":{"name":"Cell, Molecular, and Tumor Biology","volume":"46 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2018-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"126642220","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2018-07-01DOI: 10.1158/1538-7755.DISP17-B75
Eleni Stampouloglou, N. Kingston, Chih-Sheng Yang, Liye Zhang, S. Monti, X. Varelas
The aberrant activity of the transcriptional regulators TAZ and YAP (TAZ/YAP) drives many oncogenic traits, including the resistance to cell death, sustaining cell growth, and control of cell fate. Aggressive breast cancers in particular exhibit high TAZ/YAP levels and activity, and thus understanding the roles of these factors may offer therapeutic opportunity for these poorly treatable diseases. Here, we report that TAZ/YAP play an important role in driving breast cancer dependency to exogenous glutamine, an abundant amino acid that has emerged as central for tumor growth. We observed that breast cancer cells with high levels of TAZ/YAP are more sensitive to glutamine deprivation, and that knockout of TAZ/YAP in these cells alleviated defects resulting from glutamine. Rescue experiments with glutamine-derived metabolites suggested an essential role for glutamate and α-ketoglutarate (AKG) in TAZ/YAP-driven cell growth. Analysis of enzymes mediating the conversion of glutamate to α-ketoglutarate (AKG) showed that TAZ/YAP induce the expression of distinct transaminases, and that TAZ/YAP activity positively correlates with transaminase expression in breast cancer patients. Notably, the transaminase inhibitor aminooxyacetate (AOA) repressed cell growth in a TAZ/YAP-dependent manner, identifying transamination as a potential vulnerable metabolic requirement for TAZ/YAP-driven breast cancer. Considering the link between breast cancer risk and obesity, and the connection between obesity-driven metabolic changes to glutamine metabolism, our study offers a potential direction for future therapeutic targeting of aggressive breast cancers in underserved populations, where obesity is becoming a prevalent factor. Citation Format: Eleni Stampouloglou, Nathan Kingston, Chih-Sheng Yang, Liye Zhang, Stefano Monti, Xaralabos Varelas. Nutritional profiling reveals glutamine-utilizing transaminases as a metabolic vulnerability of TAZ/YAP-driven breast cancer cells [abstract]. In: Proceedings of the Tenth AACR Conference on the Science of Cancer Health Disparities in Racial/Ethnic Minorities and the Medically Underserved; 2017 Sep 25-28; Atlanta, GA. Philadelphia (PA): AACR; Cancer Epidemiol Biomarkers Prev 2018;27(7 Suppl):Abstract nr B75.
{"title":"Abstract B75: Nutritional profiling reveals glutamine-utilizing transaminases as a metabolic vulnerability of TAZ/YAP-driven breast cancer cells","authors":"Eleni Stampouloglou, N. Kingston, Chih-Sheng Yang, Liye Zhang, S. Monti, X. Varelas","doi":"10.1158/1538-7755.DISP17-B75","DOIUrl":"https://doi.org/10.1158/1538-7755.DISP17-B75","url":null,"abstract":"The aberrant activity of the transcriptional regulators TAZ and YAP (TAZ/YAP) drives many oncogenic traits, including the resistance to cell death, sustaining cell growth, and control of cell fate. Aggressive breast cancers in particular exhibit high TAZ/YAP levels and activity, and thus understanding the roles of these factors may offer therapeutic opportunity for these poorly treatable diseases. Here, we report that TAZ/YAP play an important role in driving breast cancer dependency to exogenous glutamine, an abundant amino acid that has emerged as central for tumor growth. We observed that breast cancer cells with high levels of TAZ/YAP are more sensitive to glutamine deprivation, and that knockout of TAZ/YAP in these cells alleviated defects resulting from glutamine. Rescue experiments with glutamine-derived metabolites suggested an essential role for glutamate and α-ketoglutarate (AKG) in TAZ/YAP-driven cell growth. Analysis of enzymes mediating the conversion of glutamate to α-ketoglutarate (AKG) showed that TAZ/YAP induce the expression of distinct transaminases, and that TAZ/YAP activity positively correlates with transaminase expression in breast cancer patients. Notably, the transaminase inhibitor aminooxyacetate (AOA) repressed cell growth in a TAZ/YAP-dependent manner, identifying transamination as a potential vulnerable metabolic requirement for TAZ/YAP-driven breast cancer. Considering the link between breast cancer risk and obesity, and the connection between obesity-driven metabolic changes to glutamine metabolism, our study offers a potential direction for future therapeutic targeting of aggressive breast cancers in underserved populations, where obesity is becoming a prevalent factor. Citation Format: Eleni Stampouloglou, Nathan Kingston, Chih-Sheng Yang, Liye Zhang, Stefano Monti, Xaralabos Varelas. Nutritional profiling reveals glutamine-utilizing transaminases as a metabolic vulnerability of TAZ/YAP-driven breast cancer cells [abstract]. In: Proceedings of the Tenth AACR Conference on the Science of Cancer Health Disparities in Racial/Ethnic Minorities and the Medically Underserved; 2017 Sep 25-28; Atlanta, GA. Philadelphia (PA): AACR; Cancer Epidemiol Biomarkers Prev 2018;27(7 Suppl):Abstract nr B75.","PeriodicalId":146931,"journal":{"name":"Cell, Molecular, and Tumor Biology","volume":"1 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2018-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"124397941","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2018-07-01DOI: 10.1158/1538-7755.DISP17-B73
Anthony Williams, Larischa de Wet, Marc Gillard, Steven Kregel, Tzintzuni I. Garcia, R. Szmulewitz, D. V. Griend
Background: Although organ-confined, moderately graded prostate cancer has encouraging cure rates, progression to castration-resistant prostate cancer (CRPC) from advanced, high-grade lesions represents the lethal form of this malignancy, for which current therapeutic modalities are only palliative; patients with CRPC will die from their disease. Importantly, African-American (AA) men exhibit both a higher incidence of prostate cancer diagnoses and are at higher risk of developing CRPC as compared to other racial groups. Currently, the mechanisms driving prostate cancer disparities in AA men are poorly understood, and new biomarkers and novel drugs to address this disparity are in high demand to improve personalized medicine and clinical management of prostate cancer in AA men. Work by our group and others has identified SOX2 (sex determining region Y-box 2), an essential transcription factor for maintaining the survival and pluripotency of undifferentiated embryonic stem cells (ESCs), to be associated with aggressive prostate cancer and that the constitutive overexpression of SOX2 in a hormone-sensitive, SOX2-negative prostate cancer cell line is sufficient to generate a castration-resistant phenotype both in vitro and in vivo. Further, our data demonstrate canonical SOX2 transcriptional cofactors OCT4 and NANOG are frequently not expressed in prostate cancer, implying that SOX2 has unique, non-stem cell gene targets and binding partners within malignant prostate malignancies. Methods and Results: To elucidate such SOX2 gene targets in CRPC, we performed SOX2 chromatin immunoprecipitation and sequencing (ChIP-Seq) in prostate cancer cells, revealing SOX2 binding of several interesting and functionally important gene target promoters, including FOXP1, an AR target gene and transcription factor known to play a role in disparate aggressiveness of prostate cancer in African-American men. Based on these findings, we hypothesize that upon AR ablation in prostate cancer treatment, FOXP1, a tumor suppressor demonstrated to be associated with disparate prostate cancer aggressiveness in AA men, is repressed by SOX2 to drive CRPC in AA men. Work to test this hypothesis is currently under way, and includes (1) assessment of differential FOXP1 gene expression using MDA-PCa-2a, MDA-PCa-2b, and EA0066-hT, prostate cancer cell lines derived from AA men, and CWRR1, CWR22Rv1, and LNCaP, prostate cancer cell lines derived from CA men overexpressing SOX2 in the context of AR ablation; and (2) FOXP1 ChIP-Seq paired with RNA-Seq in tumor tissues prospectively collected from AA and CA men with prostate cancer undergoing radical prostatectomy. Conclusion: The work proposed herein represents an in-depth exploration of basic SOX2 biology in the context of CRPC, is highly innovative and translational, and has transformative potential to improve clinical patient management and eradicate disparities in CRPC. Note: This abstract was not presented at the conference. Citation
背景:虽然器官受限、中度分级的前列腺癌具有令人鼓舞的治愈率,但从晚期、高级别病变进展为去势抵抗性前列腺癌(CRPC)是这种恶性肿瘤的致命形式,目前的治疗方式仅是姑息性的;CRPC患者会死于疾病。重要的是,与其他种族相比,非裔美国人(AA)男性既表现出更高的前列腺癌诊断发病率,也有更高的CRPC发病风险。目前,AA男性前列腺癌差异的机制尚不清楚,迫切需要新的生物标志物和新药来解决这一差异,以改善AA男性前列腺癌的个性化治疗和临床管理。我们的研究小组和其他研究人员发现,SOX2(性别决定区Y-box 2)是维持未分化胚胎干细胞(ESCs)存活和多能性的重要转录因子,与侵袭性前列腺癌有关,SOX2在激素敏感、SOX2阴性的前列腺癌细胞系中组成性过表达足以在体外和体内产生抗去势表型。此外,我们的数据表明,典型的SOX2转录辅因子OCT4和NANOG在前列腺癌中经常不表达,这意味着SOX2在恶性前列腺恶性肿瘤中具有独特的非干细胞基因靶点和结合伙伴。方法和结果:为了阐明SOX2基因在CRPC中的靶点,我们在前列腺癌细胞中进行了SOX2染色质免疫沉淀和测序(ChIP-Seq),揭示了SOX2结合几个有趣且功能重要的基因靶启动子,包括FOXP1,一个已知在非裔美国男性前列腺癌不同侵袭性中起作用的AR靶基因和转录因子。基于这些发现,我们假设在前列腺癌治疗的AR消融中,FOXP1(一种被证明与AA男性不同前列腺癌侵袭性相关的肿瘤抑制因子)被SOX2抑制,从而驱动AA男性的CRPC。验证这一假设的工作目前正在进行中,包括(1)在AR消融背景下,使用MDA-PCa-2a、MDA-PCa-2b和EA0066-hT(来自AA男性的前列腺癌细胞系)和CWRR1、CWR22Rv1和LNCaP(来自过表达SOX2的CA男性的前列腺癌细胞系)评估FOXP1基因的差异表达;(2)在行根治性前列腺切除术的AA和CA前列腺癌患者肿瘤组织中FOXP1 ChIP-Seq与RNA-Seq配对。结论:本文提出的工作代表了在CRPC背景下对SOX2基础生物学的深入探索,具有高度的创新性和转化性,并具有改善临床患者管理和消除CRPC差异的变革潜力。注:本摘要未在会议上发表。引文格式:Anthony Williams, Larischa de Wet, Marc Gillard, Steve Kregel, Tzintzuni Garcia, Russell Szmulewitz, Don Vander Griend。SOX2调控FOXP1作为驱动非裔美国男性前列腺癌不同侵袭性的机制[摘要]。见:第十届AACR会议论文集:种族/少数民族和医疗服务不足人群的癌症健康差异科学;2017年9月25-28日;亚特兰大,乔治亚州。费城(PA): AACR;癌症流行病学杂志,2018;27(7增刊):摘要nr B73。
{"title":"Abstract B73: SOX2 regulation of FOXP1 as a mechanism driving disparate aggressiveness of prostate cancer in African-American men","authors":"Anthony Williams, Larischa de Wet, Marc Gillard, Steven Kregel, Tzintzuni I. Garcia, R. Szmulewitz, D. V. Griend","doi":"10.1158/1538-7755.DISP17-B73","DOIUrl":"https://doi.org/10.1158/1538-7755.DISP17-B73","url":null,"abstract":"Background: Although organ-confined, moderately graded prostate cancer has encouraging cure rates, progression to castration-resistant prostate cancer (CRPC) from advanced, high-grade lesions represents the lethal form of this malignancy, for which current therapeutic modalities are only palliative; patients with CRPC will die from their disease. Importantly, African-American (AA) men exhibit both a higher incidence of prostate cancer diagnoses and are at higher risk of developing CRPC as compared to other racial groups. Currently, the mechanisms driving prostate cancer disparities in AA men are poorly understood, and new biomarkers and novel drugs to address this disparity are in high demand to improve personalized medicine and clinical management of prostate cancer in AA men. Work by our group and others has identified SOX2 (sex determining region Y-box 2), an essential transcription factor for maintaining the survival and pluripotency of undifferentiated embryonic stem cells (ESCs), to be associated with aggressive prostate cancer and that the constitutive overexpression of SOX2 in a hormone-sensitive, SOX2-negative prostate cancer cell line is sufficient to generate a castration-resistant phenotype both in vitro and in vivo. Further, our data demonstrate canonical SOX2 transcriptional cofactors OCT4 and NANOG are frequently not expressed in prostate cancer, implying that SOX2 has unique, non-stem cell gene targets and binding partners within malignant prostate malignancies. Methods and Results: To elucidate such SOX2 gene targets in CRPC, we performed SOX2 chromatin immunoprecipitation and sequencing (ChIP-Seq) in prostate cancer cells, revealing SOX2 binding of several interesting and functionally important gene target promoters, including FOXP1, an AR target gene and transcription factor known to play a role in disparate aggressiveness of prostate cancer in African-American men. Based on these findings, we hypothesize that upon AR ablation in prostate cancer treatment, FOXP1, a tumor suppressor demonstrated to be associated with disparate prostate cancer aggressiveness in AA men, is repressed by SOX2 to drive CRPC in AA men. Work to test this hypothesis is currently under way, and includes (1) assessment of differential FOXP1 gene expression using MDA-PCa-2a, MDA-PCa-2b, and EA0066-hT, prostate cancer cell lines derived from AA men, and CWRR1, CWR22Rv1, and LNCaP, prostate cancer cell lines derived from CA men overexpressing SOX2 in the context of AR ablation; and (2) FOXP1 ChIP-Seq paired with RNA-Seq in tumor tissues prospectively collected from AA and CA men with prostate cancer undergoing radical prostatectomy. Conclusion: The work proposed herein represents an in-depth exploration of basic SOX2 biology in the context of CRPC, is highly innovative and translational, and has transformative potential to improve clinical patient management and eradicate disparities in CRPC. Note: This abstract was not presented at the conference. Citation ","PeriodicalId":146931,"journal":{"name":"Cell, Molecular, and Tumor Biology","volume":"51 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2018-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"131515805","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2018-07-01DOI: 10.1158/1538-7755.DISP17-B68
M. Nava, Nwamaka A. Amobi, Nathan R Zemke, A. Berk, R. Farias-Eisner, J. Vadgama, Yanyuan Wu
Introduction: The purpose of the study is to examine the crosstalk between HER2/EGFR and Wnt signaling in HER2-positive (HER2+) breast cancer cells. Human epidermal growth factor receptors (HER) constitute a family of four transmembrane receptors (HER1-HER4). Ligand binding to HER1 (EGFR), HER3, and HER4 results in heterodimerization with other HER family members, including HER2. HER2+ breast cancer is characterized by an amplification of the HER2 gene, resulting in an increase in HER2 protein presence on the surface of cells, magnification of downstream intracellular signaling, and enhanced responsiveness to ligand stimulation (e.g., EGF). Approximately 20-30% of breast cancers have HER2 amplifications. The Wnt pathway is highly conserved in mammals and overexpression of some Wnt family members results in cancer. Wnts are ligands that bind to Frizzled/LRP receptors to initiate downstream signaling that results in the stabilization and nuclear translocation of β-catenin. Once in the nucleus, β-catenin associates with activators such as TCF/LEF, SMADs, ATF2, and KLF4 to promote the transcription of many target genes. EGFR and Wnt crosstalk has been observed in various cell types following EGF treatment. As an example, EGF treatment of human epidermoid carcinoma cells results in β-catenin nuclear translocation and activation of TCF/LEF dependent reporters. Mitogen-Activated Protein Kinases (MAPKs), which are activated following EGF stimulation, have been demonstrated to inhibit GSK3β, a negative regulator of β-catenin. However, no genome-wide analysis has been conducted to determine what genes are regulated by β-catenin following EGF treatment in HER2+ breast cancer cells. We sought to determine the modulation of gene expression following the stimulation of HER2+ breast cancer cells with EGF and to investigate the mechanisms that underlie the changes observed in gene expression. Our studies have revealed exciting and novel findings that elucidate the effects of EGFR signaling on the epigenetic landscape. Specifically, we have identified putative β-catenin targets that become activated following EGFR stimulation. We hypothesize that EGFR signaling promotes the activation of specific β-catenin genes in order to alter cellular identity. Methods: RNA-seq and ChIP-seq for H3K18ac and H3K27ac was conducted following an EGF treatment time course in SKBR3 cells. The levels of several proteins of interest were determined by Western blot analysis. The cellular localization of proteins of interest was examined using biochemically fractionated lysates followed by Western blot analysis. Results: RNA-seq analysis following an EGF treatment time course revealed that approximately 2,200 genes are either upregulated or downregulated compared to untreated cells. Moreover, the expression profiles clearly demonstrated waves of transcription. Next, we determined the status of H3K18ac and H3K27ac using ChIP-seq following an EGF time course. We found that H3K18ac and H3K
简介:本研究的目的是研究HER2阳性(HER2+)乳腺癌细胞中HER2/EGFR和Wnt信号之间的串扰。人表皮生长因子受体(HER)由四个跨膜受体(HER1-HER4)组成。与HER1 (EGFR)、HER3和HER4结合的配体导致与其他HER家族成员(包括HER2)异源二聚化。HER2+乳腺癌的特征是HER2基因扩增,导致细胞表面HER2蛋白存在增加,下游细胞内信号传导放大,对配体刺激(如EGF)的反应性增强。大约20-30%的乳腺癌有HER2扩增。Wnt通路在哺乳动物中高度保守,一些Wnt家族成员的过度表达会导致癌症。wnt是与卷曲/LRP受体结合的配体,启动下游信号传导,导致β-连环蛋白的稳定和核易位。一旦进入细胞核,β-catenin与TCF/LEF、SMADs、ATF2和KLF4等激活因子结合,促进许多靶基因的转录。EGFR和Wnt串扰在EGF处理后的各种细胞类型中被观察到。例如,EGF对人表皮样癌细胞的处理导致β-catenin核易位和TCF/LEF依赖报告基因的激活。有丝分裂原活化蛋白激酶(MAPKs)在EGF刺激后被激活,已被证明可以抑制β-连环蛋白的负调节因子GSK3β。然而,目前还没有进行全基因组分析来确定在HER2+乳腺癌细胞中EGF治疗后β-catenin调节哪些基因。我们试图确定EGF刺激HER2+乳腺癌细胞后基因表达的调节,并研究观察到的基因表达变化的机制。我们的研究揭示了令人兴奋和新颖的发现,阐明了EGFR信号在表观遗传景观中的作用。具体来说,我们已经确定了在EGFR刺激后被激活的β-catenin靶点。我们假设EGFR信号可以促进特定β-连环蛋白基因的激活,从而改变细胞身份。方法:在SKBR3细胞中进行EGF处理后,对H3K18ac和H3K27ac进行RNA-seq和ChIP-seq检测。通过Western blot分析确定几种感兴趣的蛋白的水平。感兴趣的蛋白质的细胞定位使用生化分离裂解物进行检测,然后进行Western blot分析。结果:EGF处理时间过程后的RNA-seq分析显示,与未处理的细胞相比,约有2200个基因上调或下调。此外,表达谱清楚地显示了转录波。接下来,我们使用ChIP-seq根据EGF时间过程确定H3K18ac和H3K27ac的状态。我们发现,与未处理的细胞相比,egf处理后1小时内H3K18ac和H3K27ac在全球范围内增加。我们对所有激活基因的-1000bp到+200bp的转录因子结合位点进行了基序发现搜索,并确定每一波转录都有一些独特的假定调节因子。正如预期的那样,egf处理后1h和2h激活的基因含有c-Jun和JunD结合位点。令人惊讶的是,TCF3、TCF5和LEF1基序在一些基因中富集,在EGF处理后的6h、16h和24h达到表达高峰。最后,我们对细胞区室进行生化分离,检测到EGF处理后染色质相关β-连环蛋白的增加,表明EGFR和Wnt信号组分之间存在串扰。我们计划在EGF治疗后确定β-catenin的全基因组定位。结论:我们的数据表明,EGFR和Wnt信号组分之间的串扰可能调节β-catenin靶基因,导致HER2+细胞对治疗产生耐药性。引文格式:Miguel Nava, Nwamaka Amobi, Nathan Zemke, Arnold Berk, Robin Farias-Eisner, Jay Vadgama, Yanyuan Wu。EGFR信号介导的转录调节和可能与Wnt信号组分的串扰[摘要]。见:第十届AACR会议论文集:种族/少数民族和医疗服务不足人群的癌症健康差异科学;2017年9月25-28日;亚特兰大,乔治亚州。费城(PA): AACR;癌症流行病学杂志,2018;27(7增刊):摘要nr B68。
{"title":"Abstract B68: EGFR signaling mediated modulation of transcription and probable crosstalk with components of Wnt signaling","authors":"M. Nava, Nwamaka A. Amobi, Nathan R Zemke, A. Berk, R. Farias-Eisner, J. Vadgama, Yanyuan Wu","doi":"10.1158/1538-7755.DISP17-B68","DOIUrl":"https://doi.org/10.1158/1538-7755.DISP17-B68","url":null,"abstract":"Introduction: The purpose of the study is to examine the crosstalk between HER2/EGFR and Wnt signaling in HER2-positive (HER2+) breast cancer cells. Human epidermal growth factor receptors (HER) constitute a family of four transmembrane receptors (HER1-HER4). Ligand binding to HER1 (EGFR), HER3, and HER4 results in heterodimerization with other HER family members, including HER2. HER2+ breast cancer is characterized by an amplification of the HER2 gene, resulting in an increase in HER2 protein presence on the surface of cells, magnification of downstream intracellular signaling, and enhanced responsiveness to ligand stimulation (e.g., EGF). Approximately 20-30% of breast cancers have HER2 amplifications. The Wnt pathway is highly conserved in mammals and overexpression of some Wnt family members results in cancer. Wnts are ligands that bind to Frizzled/LRP receptors to initiate downstream signaling that results in the stabilization and nuclear translocation of β-catenin. Once in the nucleus, β-catenin associates with activators such as TCF/LEF, SMADs, ATF2, and KLF4 to promote the transcription of many target genes. EGFR and Wnt crosstalk has been observed in various cell types following EGF treatment. As an example, EGF treatment of human epidermoid carcinoma cells results in β-catenin nuclear translocation and activation of TCF/LEF dependent reporters. Mitogen-Activated Protein Kinases (MAPKs), which are activated following EGF stimulation, have been demonstrated to inhibit GSK3β, a negative regulator of β-catenin. However, no genome-wide analysis has been conducted to determine what genes are regulated by β-catenin following EGF treatment in HER2+ breast cancer cells. We sought to determine the modulation of gene expression following the stimulation of HER2+ breast cancer cells with EGF and to investigate the mechanisms that underlie the changes observed in gene expression. Our studies have revealed exciting and novel findings that elucidate the effects of EGFR signaling on the epigenetic landscape. Specifically, we have identified putative β-catenin targets that become activated following EGFR stimulation. We hypothesize that EGFR signaling promotes the activation of specific β-catenin genes in order to alter cellular identity. Methods: RNA-seq and ChIP-seq for H3K18ac and H3K27ac was conducted following an EGF treatment time course in SKBR3 cells. The levels of several proteins of interest were determined by Western blot analysis. The cellular localization of proteins of interest was examined using biochemically fractionated lysates followed by Western blot analysis. Results: RNA-seq analysis following an EGF treatment time course revealed that approximately 2,200 genes are either upregulated or downregulated compared to untreated cells. Moreover, the expression profiles clearly demonstrated waves of transcription. Next, we determined the status of H3K18ac and H3K27ac using ChIP-seq following an EGF time course. We found that H3K18ac and H3K","PeriodicalId":146931,"journal":{"name":"Cell, Molecular, and Tumor Biology","volume":"1 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2018-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"123131160","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2018-07-01DOI: 10.1158/1538-7755.DISP17-B72
S. Ramakrishnan, Xuan Peng, Qianya Qi, Q. Hu, G. Azabdaftari, E. Pop, J. Mohler, K. Attwood, Li Yan, Jianmin Wang, A. Woloszynska-Read
Background: African-American (AA) men are more often diagnosed with early-onset aggressive prostate cancer than European American (EA) men. Prostate cancer health disparities between AA and EA men are attributed to socioeconomic as well as biologic differences existing between the two groups. The purpose of this study is to investigate how genetic and epigenetic alterations, specifically DNA methylation, contribute to early-onset aggressive prostate cancer in AA men. Methods: We performed 450K methylation array and RNA-sequencing on radical prostatectomy specimens from AA (n=32) and EA (n=5) men treated at Roswell Park Cancer Institute (RPCI). To increase analytical power, we included AA (n=22) and EA (n=172) patients from The Cancer Genome Atlas (TCGA) database. We found no significant difference in the average age between EA and AA men in the RPCI cohort, but the TCGA cohort consisted of AA men who had significantly lower average and median age compared to EA men (57 and 56 vs 61 and 62; p=0.019). We assessed baseline androgen receptor (AR) protein levels in matched benign and malignant AA (n=95) and EA (n=93) radical prostatectomy tissues from RPCI using immunohistochemistry. Gleason score 4+3 were classified as low and high aggressive tumors, respectively. Results: Unsupervised hierarchical clustering of DNA methylation levels in prostate cancers revealed 9 clusters. We focused on the 2 largest clusters, Cluster 1 (n=133) and Cluster 3 (n=73). Cluster 1 consisted predominantly of low aggressive disease (p=0.00002) with lower serum prostate specific antigen (PSA) values (8.97 vs 13.35 ng/ml) compared to Cluster 3. Following this trend, AA patients, but not EA patients, in Cluster 1 had better overall (57 vs 50 months, p=0.48) and disease-free time (47 vs 22 months, p=0.01) as compared to Cluster 3. ERG (ETS-related gene) fusion-positive prostate cancer is believed to be more aggressive than fusion-negative prostate cancer due to increased ERG gene transcription. We did not observe a difference in fusion status between the two DNA methylation clusters. However, overall DNA methylation was higher in fusion-negative samples as compared to fusion-positive samples in the TCGA dataset. ERG is known to bind to and reduce PSA, a direct downstream AR target. In the TCGA dataset, AA fusion-negative patients had higher average and median preoperative PSA values (18.38, 20.15 ng/ml) as compared with AA fusion-positive (7.51, 6.70 ng/ml), EA fusion-positive (9.43, 7.00 ng/ml), and EA fusion-negative patients (11.47, 7.50 ng/ml). There was no significant difference in the AR transcript and AR protein levels between fusion-positive and -negative AA and EA men. AR protein expression in RPCI cohort tissue microarrays showed that adjacent nontumor tissues from AA men had higher percent AR positive nuclei (p Conclusions: Analysis of the RPCI and TCGA cohorts indicated that DNA methylation separated low and high aggressive prostate cancer in AA men. Further analy
背景:非裔美国人(AA)男性比欧洲裔美国人(EA)男性更常被诊断为早发性侵袭性前列腺癌。AA和EA男性之间的前列腺癌健康差异归因于两组之间存在的社会经济和生物学差异。本研究的目的是探讨遗传和表观遗传改变,特别是DNA甲基化,如何促进AA男性早发性侵袭性前列腺癌。方法:我们对Roswell Park癌症研究所(RPCI)治疗的AA (n=32)和EA (n=5)男性根治性前列腺切除术标本进行450K甲基化阵列和rna测序。为了提高分析能力,我们从癌症基因组图谱(TCGA)数据库中纳入了AA (n=22)和EA (n=172)患者。我们发现在RPCI队列中EA和AA男性的平均年龄没有显著差异,但TCGA队列中AA男性的平均和中位年龄明显低于EA男性(57岁和56岁vs 61岁和62岁;p = 0.019)。我们使用免疫组化技术评估了RPCI根治性前列腺切除术后的良性和恶性AA (n=95)和EA (n=93)配对组织的基线雄激素受体(AR)蛋白水平。Gleason评分4+3分为低侵袭性肿瘤和高侵袭性肿瘤。结果:前列腺癌DNA甲基化水平的无监督分层聚类显示了9个簇。我们专注于2个最大的集群,集群1 (n=133)和集群3 (n=73)。聚类1主要由低侵袭性疾病组成(p=0.00002),与聚类3相比,血清前列腺特异性抗原(PSA)值较低(8.97 vs 13.35 ng/ml)。根据这一趋势,第1组AA患者比第3组EA患者总体上(57 vs 50个月,p=0.48)和无病时间(47 vs 22个月,p=0.01)更好。ERG (ets相关基因)融合阳性前列腺癌被认为比融合阴性前列腺癌更具侵袭性,因为ERG基因转录增加。我们没有观察到两个DNA甲基化簇之间融合状态的差异。然而,与TCGA数据集中的融合阳性样本相比,融合阴性样本的总体DNA甲基化更高。已知ERG结合并降低PSA,这是AR的直接下游靶标。在TCGA数据集中,AA融合阴性患者的平均和中位术前PSA值(18.38,20.15 ng/ml)高于AA融合阳性患者(7.51,6.70 ng/ml), EA融合阳性患者(9.43,7.00 ng/ml)和EA融合阴性患者(11.47,7.50 ng/ml)。融合阳性和阴性AA和EA男性的AR转录物和AR蛋白水平无显著差异。RPCI队列组织芯片中AR蛋白表达显示,AA男性的邻近非肿瘤组织中AR阳性核的比例更高(p)。结论:RPCI和TCGA队列分析表明,DNA甲基化区分了AA男性的低侵袭性和高侵袭性前列腺癌。进一步的分析将包括更多的患者,并确定集群1和集群3之间特定CpG位点的差异。我们的研究结果表明,DNA甲基化改变在ERG融合阴性疾病患者中可能很重要,因为这些前列腺癌表现出更高水平的总体DNA甲基化。最后,我们观察到邻近非肿瘤组织中较高的AR表达表明,与EA男性相比,AA男性中存在潜在的遗传变异,导致早发性高侵袭性前列腺癌。引用格式:Swathi Ramakrishnan, Xuan Peng, Qianya Qi, Qiang Hu, Gissou Azabdaftari, Elena Pop, James Mohler, christopher Attwood, Li Yan, Jianmin Wang, Anna Woloszynska-Read。DNA甲基化和基因改变与非裔美国男性侵袭性前列腺癌有关。见:第十届AACR会议论文集:种族/少数民族和医疗服务不足人群的癌症健康差异科学;2017年9月25-28日;亚特兰大,乔治亚州。费城(PA): AACR;癌症流行病学杂志,2018;27(7增刊):摘要nr B72。
{"title":"Abstract B72: DNA methylation and genetic alterations contribute to aggressive prostate cancer in African American men","authors":"S. Ramakrishnan, Xuan Peng, Qianya Qi, Q. Hu, G. Azabdaftari, E. Pop, J. Mohler, K. Attwood, Li Yan, Jianmin Wang, A. Woloszynska-Read","doi":"10.1158/1538-7755.DISP17-B72","DOIUrl":"https://doi.org/10.1158/1538-7755.DISP17-B72","url":null,"abstract":"Background: African-American (AA) men are more often diagnosed with early-onset aggressive prostate cancer than European American (EA) men. Prostate cancer health disparities between AA and EA men are attributed to socioeconomic as well as biologic differences existing between the two groups. The purpose of this study is to investigate how genetic and epigenetic alterations, specifically DNA methylation, contribute to early-onset aggressive prostate cancer in AA men. Methods: We performed 450K methylation array and RNA-sequencing on radical prostatectomy specimens from AA (n=32) and EA (n=5) men treated at Roswell Park Cancer Institute (RPCI). To increase analytical power, we included AA (n=22) and EA (n=172) patients from The Cancer Genome Atlas (TCGA) database. We found no significant difference in the average age between EA and AA men in the RPCI cohort, but the TCGA cohort consisted of AA men who had significantly lower average and median age compared to EA men (57 and 56 vs 61 and 62; p=0.019). We assessed baseline androgen receptor (AR) protein levels in matched benign and malignant AA (n=95) and EA (n=93) radical prostatectomy tissues from RPCI using immunohistochemistry. Gleason score 4+3 were classified as low and high aggressive tumors, respectively. Results: Unsupervised hierarchical clustering of DNA methylation levels in prostate cancers revealed 9 clusters. We focused on the 2 largest clusters, Cluster 1 (n=133) and Cluster 3 (n=73). Cluster 1 consisted predominantly of low aggressive disease (p=0.00002) with lower serum prostate specific antigen (PSA) values (8.97 vs 13.35 ng/ml) compared to Cluster 3. Following this trend, AA patients, but not EA patients, in Cluster 1 had better overall (57 vs 50 months, p=0.48) and disease-free time (47 vs 22 months, p=0.01) as compared to Cluster 3. ERG (ETS-related gene) fusion-positive prostate cancer is believed to be more aggressive than fusion-negative prostate cancer due to increased ERG gene transcription. We did not observe a difference in fusion status between the two DNA methylation clusters. However, overall DNA methylation was higher in fusion-negative samples as compared to fusion-positive samples in the TCGA dataset. ERG is known to bind to and reduce PSA, a direct downstream AR target. In the TCGA dataset, AA fusion-negative patients had higher average and median preoperative PSA values (18.38, 20.15 ng/ml) as compared with AA fusion-positive (7.51, 6.70 ng/ml), EA fusion-positive (9.43, 7.00 ng/ml), and EA fusion-negative patients (11.47, 7.50 ng/ml). There was no significant difference in the AR transcript and AR protein levels between fusion-positive and -negative AA and EA men. AR protein expression in RPCI cohort tissue microarrays showed that adjacent nontumor tissues from AA men had higher percent AR positive nuclei (p Conclusions: Analysis of the RPCI and TCGA cohorts indicated that DNA methylation separated low and high aggressive prostate cancer in AA men. Further analy","PeriodicalId":146931,"journal":{"name":"Cell, Molecular, and Tumor Biology","volume":"221 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2018-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"114431388","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}