Identification of 67 histone marks and histone lysine crotonylation

合集下载

WHO_TRS_996_annex09翻译

WHO_TRS_996_annex09翻译

Annex9组织机构进行体内生物等效性研究指南背景2014年的一场非正式讨论会上,在世界卫生组织(WHO)药学准备工作规范专家委员会的第49次会议,讨论产生关于可能修正组织机构进行体内生物等效性研究指南(WHO技术报告系列,No. 937, Annex9, 2006)。

WHO药学准备工作规范专家委员会同意,鉴于新的事态发展,将会准备一条修正法案。

新的指南不仅考虑多来源指南的修订,并会考虑创造良好数据管理下的新的指南。

法案也会考虑自从2006年评价和稽查BE试验领域的经验。

在稽查者们重复发现相同问题的领域,新的指南将会提供阐述,增补的细节也已加入生物分析。

指南也会更加注重项目安全性和数据完整性。

在第一版工作文件的基础上,第二版结合了很多的意见和反馈,有来自于公众、WHO资格预审团队(PQT)的意见,也有来自于2015年举行的关于数据管理、生物等效性、GMP、药品稽查的讨论会。

WHO/PQT建立于2001年,是为了保障采购的药用产品满足WHO关于质量、安全性、有效性的规范和标准(http://www.who.int/prequal/)。

特别的是,要求报送的产品档案所有必要的内容经评估都是可接受的,成品药(FPP)以及API的生产地点满足WHO的GMP要求。

由于报送WHO/PQT的产品通常是多来源的(仿制的),一般通过在比如合同研究组织CRO(也叫做临床研究机构)进行的BE试验来证明治疗等价性。

对于资格预审的产品至关重要的是,除了上述的要求,申办方BE试验使用的CRO公司要满足WHO的药物临床试验质量管理规范GCP,考虑优良实验室规范GLP和质量控制(QC)305实验室管理规范来保证数据的完整性和可追溯性。

除此之外,如果存在当地的法律法规,CRO应该得到各自国家药品局的认可。

如果国家规定需要,BE试验应该获得国家监督管理局的授权。

因此报送资格预审的产品涉及到BE试验中执行和分析的,需要保证满足WHO相关的规范和标准,以便为WHO的稽查做准备。

应用地球化学元素丰度数据手册-原版

应用地球化学元素丰度数据手册-原版

应用地球化学元素丰度数据手册迟清华鄢明才编著地质出版社·北京·1内容提要本书汇编了国内外不同研究者提出的火成岩、沉积岩、变质岩、土壤、水系沉积物、泛滥平原沉积物、浅海沉积物和大陆地壳的化学组成与元素丰度,同时列出了勘查地球化学和环境地球化学研究中常用的中国主要地球化学标准物质的标准值,所提供内容均为地球化学工作者所必须了解的各种重要地质介质的地球化学基础数据。

本书供从事地球化学、岩石学、勘查地球化学、生态环境与农业地球化学、地质样品分析测试、矿产勘查、基础地质等领域的研究者阅读,也可供地球科学其它领域的研究者使用。

图书在版编目(CIP)数据应用地球化学元素丰度数据手册/迟清华,鄢明才编著. -北京:地质出版社,2007.12ISBN 978-7-116-05536-0Ⅰ. 应… Ⅱ. ①迟…②鄢…Ⅲ. 地球化学丰度-化学元素-数据-手册Ⅳ. P595-62中国版本图书馆CIP数据核字(2007)第185917号责任编辑:王永奉陈军中责任校对:李玫出版发行:地质出版社社址邮编:北京市海淀区学院路31号,100083电话:(010)82324508(邮购部)网址:电子邮箱:zbs@传真:(010)82310759印刷:北京地大彩印厂开本:889mm×1194mm 1/16印张:10.25字数:260千字印数:1-3000册版次:2007年12月北京第1版•第1次印刷定价:28.00元书号:ISBN 978-7-116-05536-0(如对本书有建议或意见,敬请致电本社;如本社有印装问题,本社负责调换)2关于应用地球化学元素丰度数据手册(代序)地球化学元素丰度数据,即地壳五个圈内多种元素在各种介质、各种尺度内含量的统计数据。

它是应用地球化学研究解决资源与环境问题上重要的资料。

将这些数据资料汇编在一起将使研究人员节省不少查找文献的劳动与时间。

这本小册子就是按照这样的想法编汇的。

五氯苯酚检测标准技术分析与展望

五氯苯酚检测标准技术分析与展望

检测认证五氯苯酚检测标准技术分析与展望■ 宋玉峰(山东省产品质量检验研究院)摘 要:五氯苯酚因其防霉杀菌作用广泛应用于多个领域,在保护消费品品质的同时存在潜在健康风险。

科学有效地检测识别这类物质是了解与防范其安全风险的基础。

本文梳理了国内外五氯苯酚痕量迁移物现行检测标准,概述并比较其迁移物提取的前处理实验流程和检测技术方法,通过前处理流程及检测技术的梳理解析,分析比较过程的复杂性和操控性、试剂的环保性,探讨实验技术的优化及改进。

本文以期为五氯苯酚检测过程中绿色环保的改进、提高检测过程的可操控性等提供参考。

关键词:五氯苯酚,有害物质迁移,前处理技术,健康风险DOI编码:10.3969/j.issn.1002-5944.2023.15.036Prospect and Technological Analysis of DetectionStandard for trace pentachlorophenol MigrationSONG Yu-feng(Shandong Institute for Product Quality Inspection)Abstract:Pentachlorophenol is extensively applied in many fields due to its anti-bacterial and anti-fungal effects, which facilitates the quality of products and has health risks. Effective identification of these substances is the basis of understanding and preventing safety risks. The paper sorts out current detection standards to trace phentachlorophenol migration at home and abroad. The paper presents and compares the pretreatment experimental processes and technological testing methods, analyzes the complexity, controllability, and environmental effects of reagents by sorting out pretreatment processes and testing methods, and discusses the improvement of experimental technologies. The paper provides reference for improving the greenness and controllability of pentachlorophenol detection process.Keywords: pentachlorophenol, migration of harmful substances, pretreatment technology, health risk0 引 言五氯苯酚及其盐和酯类物质防霉杀菌效果好,价格优势明显,使用方便,作除草剂、防腐剂和防霉剂广泛用于多个领域。

marked manuscript

marked manuscript

Quality evaluation of Flos Lonicerae through a simultaneous determination of seven saponins by HPLC with ELSDXing-Yun Chai1, Song-Lin Li2, Ping Li1*1Key Laboratory of Modern Chinese Medicines and Department of Pharmacognosy, China Pharmaceutical University, Nanjing, 210009, People’s Republic of China2Institute of Nanjing Military Command for Drug Control, Nanjing, 210002, People’s Republic of China*Corresponding author: Ping LiKey Laboratory of Modern Chinese Medicines and Department of Pharmacognosy, China Pharmaceutical University, Nanjing 210009, People’s Republic of China.E-mail address: lipingli@Tel.: +86-25-8324-2299; 8539-1244; 135********Fax: +86-25-8532-2747AbstractA new HPLC coupled with evaporative light scattering detection (ELSD) method has been developed for the simultaneous quantitative determination of seven major saponins, namely macranthoidinB (1), macranthoidin A (2), dipsacoside B (3), hederagenin-28-O-β-D-glucopyranosyl(6→1)-O-β-D- glucopyranosyl ester (4), macranthoside B (5), macranthoside A (6), and hederagenin-3-O-α-L-arabinopyranosyl(2→1)-O-α-L-rhamnopyranoside (7)in Flos Lonicerae, a commonly used traditional Chinese medicine (TCM) herb.Simultaneous separation of these seven saponins was achieved on a C18 analytical column with a mixed mobile phase consisting of acetonitrile(A)-water(B)(29:71 v/v) acidified with 0.5% acetic acid. The elution was operated from keeping 29%A for 10min, then gradually to 54%B from 10 to 25 min on linear gradient, and then keep isocratic elution with 54%B from 25 to 30min.The drift tube temperature of ELSD was set at 106℃, and with the nitrogen flow-rate of 2.6 l/min. All calibration curves showed good linear regression (r2 0.9922) within test ranges. This method showed good reproducibility for the quantification of these seven saponins in Flos Lonicerae with intra- and inter-day variations of less than 3.0% and 6.0% respectively. The validated method was successfully applied to quantify seven saponins in five sources of Flos Lonicerae, which provides a new basis of overall assessment on quality of Flos Lonicerae.Keywords: HPLC-ELSD; Flos Lonicerae; Saponins; Quantification1. IntroductionFlos Lonicerae (Jinyinhua in Chinese), the dried buds of several species of the genus Lonicera (Caprifoliaceae), is a commonly used traditional Chinese medicine (TCM) herb. It has been used for centuries in TCM practice for the treatment of sores, carbuncles, furuncles, swelling and affections caused by exopathogenic wind-heat or epidemic febrile diseases at the early stage [1]. Though four species of Lonicera are documented as the sources of Flos Lonicerae in China Pharmacopeia (2000 edition), i.e. L. japonica, L. hypoglauca,L. daystyla and L. confusa, other species such as L. similes and L. macranthoides have also been used on the same purpose in some local areas in China [2]. So it is an important issue to comprehensively evaluate the different sources of Flos Lonicerae, so as to ensure the clinical efficacy of this Chinese herbal drug.Chemical and pharmacological investigations on Flos Lonicerae resulted in discovering several kinds of bioactive components, i.e. chlorogenic acid and its analogues, flavonoids, iridoid glucosides and triterpenoid saponins [3]. Previously, chlorogenic acid has been used as the chemical marker for the quality evaluation of Flos Lonicerae,owing to its antipyretic and antibiotic property as well as its high content in the herb. But this compound is not a characteristic component of Flos Lonicerae, as it has also been used as the chemical marker for other Chinese herbal drugs such as Flos Chrysanthemi and so on[4-5]. Moreover, chlorogenic acid alone could not be responsible for the overall pharmacological activities of Flos Lonicerae[6].On the other hand, many studies revealed that triterpenoidal saponins of Flos Lonicerae possess protection effects on hepatic injury caused by Acetaminophen, Cd, and CCl4, and conspicuous depressant effects on swelling of ear croton oil [7-11]. Therefore, saponins should also be considered as one of the markers for quality control of Flos Lonicerae. Consequently, determinations of all types of components such as chlorogenic acid, flavonoids, iridoid glucosides and triterpenoidal saponins in Flos Lonicerae could be a better strategy for the comprehensive quality evaluation of Flos Lonicerae.Recently an HPLC-ELSD method has been established in our laboratory for qualitative and quantitative determination of iridoid glucosides in Flos Lonicerae [12]. But no method was reported for the determination of triterpenoidal saponins in Flos Lonicera. As a series studies on the comprehensive evaluation of Flos Lonicera, we report here, for the first time, the development of an HPLC-ELSD method for simultaneous determination of seven triterpenoidal saponins in the Chinese herbal drug Flos Lonicerae, i.e.macranthoidin B (1), macranthoidin A (2), dipsacoside B (3), hederagenin-28-O-β-D-glucopyranosyl(6→1)-O-β-D- glucopyranosyl ester (4), macranthoside B (5), macranthoside A (6), and hederagenin-3-O-α-L-arabinopyranosyl(2→1)-O-α-L-rhamnopyranoside (7) (Fig. 1).2. Experimental2.1. Samples, chemicals and reagentsFive samples of Lonicera species,L. japonica from Mi county, HeNan province (LJ1999-07), L. hypoglauca from Jiujang county, JiangXi province (LH2001-06), L. similes from Fei county, ShanDong province (LS2001-07), L. confuse from Xupu county, HuNan province (LC2001-07), and L. macranthoides from Longhu county, HuNan province (LM2000-06) respectively, were collected in China. All samples were authenticated by Dr. Ping Li, professor of department of Pharmacognosy, China Pharmaceutical University, Nanjing, China. The voucher specimens were deposited in the department of Pharmacognosy, China Pharmaceutical University, Nanjing, China. Seven saponin reference compounds: macranthoidin B (1), macranthoidin A (2), dipsacoside B (3), hederagenin-28-O-β-D-glucopyranosyl(6→1)-O-β-D- glucopyranosyl ester (4), macranthoside B (5), macranthoside A (6), and hederagenin-3-O-α-L-arabinopyranosyl(2→1)-O-α-L-rhamnopyranoside (7) were isolated previously from the dried buds of L. confusa by repeated silica gel, sephadex LH-20 and Rp-18 silica gel column chromatography, their structures were elucidated by comparison of their spectral data (UV, IR, MS, 1H- NMR and 13C-NMR) with references [13-15]. The purity of these saponins were determined to be more than 98% by normalization of the peak areas detected by HPLC with ELSD, and showed very stable in methanol solution.HPLC-grade acetonitrile from Merck (Darmstadt, Germany), the deionized water from Robust (Guangzhou, China), were purchased. The other solvents, purchased from Nanjing Chemical Factory (Nanjing, China) were of analytical grade.2.2. Apparatus and chromatographic conditionsAglient1100 series HPLC apparatus was used. Chromatography was carried out on an Aglient Zorbax SB-C18 column(250 4.6mm, 5.0µm)at a column temperature of 25℃.A Rheodyne 7125i sampling valve (Cotati, USA) equipped with a sample loop of 20µl was used for sample injection. The analog signal from Alltech ELSD 2000 (Alltech, Deerfield, IL, USA)was transmitted to a HP Chemstation for processing through an Agilent 35900E (Agilent Technologies, USA).The optimum resolution was obtained by using a linear gradient elution. The mobile phase was composed of acetonitrile(A) and water(B) which acidified with 0.5% acetic acid. The elution was operated from keeping 29%A for 10min, then gradually to 54%B from 10 to 25 min in linear gradient, and back to the isocratic elution of 54%B from 25 to 30 min.The drift tube temperature for ELSD was set at 106℃and the nitrogen flow-rate was of 2.6 l/min. The chromatographic peaks were identified by comparing their retention time with that of each reference compound tried under the same chromatographic conditions with a series of mobile phases. In addition, spiking samples with the reference compounds further confirmed the identities of the peaks.2.3. Calibration curvesMethanol stock solutions containing seven analytes were prepared and diluted to appropriate concentration for the construction of calibration curves. Six concentrationof the seven analytes’ solution were injected in triplicate, and then the calibration curves were constructed by plotting the peak areas versus the concentration of each analyte. The results were demonstrated in Table1.2.4. Limits of detection and quantificationMethanol stock solution containing seven reference compounds were diluted to a series of appropriate concentrations with methanol, and an aliquot of the diluted solutions were injected into HPLC for analysis.The limits of detection (LOD) and quantification (LOQ) under the present chromatographic conditions were determined at a signal-to-noise ratio (S/N) of 3 and 10, respectively. LOD and LOQ for each compound were shown in Table1.2.5. Precision and accuracyIntra- and inter-day variations were chosen to determine the precision of the developed assay. Approximate 2.0g of the pulverized samples of L. macranthoides were weighted, extracted and analyzed as described in 2.6 Sample preparation section. For intra-day variability test, the samples were analyzed in triplicate for three times within one day, while for inter-day variability test, the samples were examined in triplicate for consecutive three days. Variations were expressed by the relative standard deviations. The results were given in Table 2.Recovery test was used to evaluate the accuracy of this method. Accurate amounts of seven saponins were added to approximate 1.0g of L. macranthoides,and then extracted and analyzed as described in 2.6 Sample preparation section. The average recoveries were counted by the formula: recovery (%) = (amount found –original amount)/ amount spiked ×100%, and RSD (%) = (SD/mean) ×100%. The results were given in Table 3.2.6. Sample preparationSamples of Flos Lonicerae were dried at 50℃until constant weight. Approximate 2.0g of the pulverized samples, accurately weighed, was extracted with 60% ethanol in a flask for 4h. The ethanol was evaporated to dryness with a rotary evaporator. Residue was dissolved in water, followed by defatting with 60ml of petroleum ether for 2 times, and then the water solution was evaporated, residue was dissolved with methanol into a 25ml flask. One ml of the methanol solution was drawn and transferred to a 5ml flask, diluted to the mark with methanol. The resultant solution was at last filtrated through a 0.45µm syringe filter (Type Millex-HA, Millipore, USA) and 20µl of the filtrate was injected to HPLC system. The contents of the analytes were determined from the corresponding calibration curves.3. Results and discussionsThe temperature of drift tube and the gas flow-rate are two most important adjustable parameters for ELSD, they play a prominent role to an analyte response. In ourprevious work [12], the temperature of drift tube was optimized at 90°C for the determination of iridoids. As the polarity of saponins are higher than that of iridoids, more water was used in the mobile phase for the separation of saponins, therefore the temperature for saponins determination was optimized systematically from 95°C to 110°C, the flow-rate from 2.2 to 3.0 l/min. Dipsacoside B was selected as the testing saponin for optimizing ELSD conditions, as it was contained in all samples. Eventually, the drift tube temperature of 106℃and a gas flow of 2.6 l/min were optimized to detect the analytes. And these two exact experimental parameters should be strictly controlled in the analytical procedure [16].All calibration curves showed good linear regression (r2 0.9922) within test ranges. Validation studies of this method proved that this assay has good reproducibility. As shown in Table 2, the overall intra- and inter-day variations are less than 6% for all seven analytes. As demonstrated in Table 3, the developed analytical method has good accuracy with the overall recovery of high than 96% for the analytes concerned. The limit of detection (S/N=3) and the limit of quantification (S/N=10) are less than 0.26μg and 0.88μg respectively (Table1), indicating that this HPLC-ELSD method is precise, accurate and se nsitive enough for the quantitative evaluation of major non- chromaphoric saponins in Flos Lonicerae.It has been reported that there are two major types of saponins in Flos Lonicerae, i.e. saponins with hederagenin as aglycone and saponins with oleanolic acid as the aglycone [17]. But hederagenin type saponins of the herb were reported to have distinct activities of liver protection and anti-inflammatory [7-11]. So we adoptedseven hederagenin type saponins as representative markers to establish a quality control method.The newly established HPLC-ELSD method was applied to analyze seven analytes in five plant sources of Flos Lonicerae, i.e. L. japonica,L. hypoglauca,L. confusa,L. similes and L. macranthoides(Table 4). It was found that there were remarkable differences of seven saponins contents between different plant sources of Flos Lonicerae. All seven saponins analyzed could be detected in L. confusa and L. hypoglauca, while only dipsacoside B was detected in L. japonica. Among all seven saponins interested, only dipsacoside B was found in all five plant species of Flos Lonicerae analyzed, and this compound was determined as the major saponin with content of 53.7 mg/g in L. hypoglauca. On the other hand, macranthoidin B was found to be the major saponin with the content higher than 41.0mg/g in L. macranthoides,L. confusa, and L. similis, while the contents of other analytes were much lower.In our previous study [12], overall HPLC profiles of iridoid glucosides was used to qualitatively and quantitatively distinguish different origins of Flos Lonicerae. As shown in Fig.2, the chromatogram profiles of L. confusa, L. japonica and L. similes seem to be similar, resulting in the difficulty of clarifying the origins of Flos Lonicerae solely by HPLC profiles of saponins, in addition to the clear difference of the HPLC profiles of saponins from L. macranthoides and L. hypoglauca.Therefore, in addition to the conventional morphological and histological identification methods, the contents and the HPLC profiles of saponins and iridoids could also be used as accessory chemical evidence toclarify the botanical origin and comprehensive quality evaluation of Flos Lonicerae.4. ConclusionsThis is the first report on validation of an analytical method for qualification and quantification of saponins in Flos Lonicerae. This newly established HPLC-ELSD method can be used to simultaneously quantify seven saponins, i.e. macranthoidin B, macranthoidin A, dipsacoside B, hederagenin-28-O-β-D-glucopyranosyl(6→1)-O-β-D- glucopyranosyl ester, macranthoside B, macranthoside A, and hederagenin-3-O-α-L-arabinopyranosyl(2→1)-O-α-L-rhamnopyranoside in Flos Lonicerae. Together with the HPLC profiles of iridoids, the HPLC-ELSD profiles of saponins could also be used as an accessory chemical evidence to clarify the botanical origin and comprehensive quality evaluation of Flos Lonicerae.AcknowledgementsThis project is financially supported by Fund for Distinguished Chinese Young Scholars of the National Science Foundation of China (30325046) and the National High Tech Program(2003AA2Z2010).[1]Ministry of Public Health of the People’s Republic of China, Pharmacopoeia ofthe People’s Republic of China, V ol.1, 2000, p. 177.[2]W. Shi, R.B. Shi, Y.R. Lu, Chin. Pharm. J., 34(1999) 724.[3]J.B. Xing, P. Li, D.L. Wen, Chin. Med. Mater., 26(2001) 457.[4]Y.Q. Zhang, L.C. Xu, L.P. Wang, J. Chin. Med. Mater., 21(1996) 204.[5] D. Zhang, Z.W. Li, Y. Jiang, J. Pharm. Anal., 16(1996) 83.[6]T.Z. Wang, Y.M. Li, Huaxiyaoxue Zazhi, 15(2000) 292.[7]J.ZH. Shi, G.T. Liu. Acta Pharm. Sin., 30(1995) 311.[8]Y. P. Liu, J. Liu, X.SH. Jia, et al. Acta Pharmacol. Sin., 13 (1992) 209.[9]Y. P. Liu, J. Liu, X.SH. Jia, et al. Acta Pharmacol. Sin., 13 (1992) 213.[10]J.ZH. Shi, L. Wan, X.F. Chen.ZhongYao YaoLi Yu LinChuang, 6 (1990) 33.[11]J. Liu, L. Xia, X.F. Chen. Acta Pharmacol. Sin., 9 (1988) 395[12]H.J. Li, P. Li, W.C. Ye, J. Chromatogr. A 1008(2003) 167-72.[13]Q. Mao, D. Cao, X.SH. Jia. Acta Pharm. Sin., 28(1993) 273.[14]H. Kizu, S. Hirabayashi, M. Suzuki, et al. Chem. Pharm. Bull., 33(1985) 3473.[15]S. Saito, S. Sumita, N. Tamura, et al. Chem Pharm Bull., 38(1990) 411.[16]Alltech ELSD 2000 Operating Manual, Alltech, 2001, p. 16. In Chinese.[17]J.B. Xing, P. Li, Chin. Med. Mater., 22(1999) 366.Fig. 1 Chemical structures of seven saponins from Lonicera confusa macranthoidin B (1), macranthoidin A (2), dipsacoside B (3), hederagenin-28-O-β-D-glucopyranosyl(6→1)-O-β-D- glucopyranosyl ester (4), macranthoside B (5), macranthoside A (6), and hederagenin-3-O-α-L-arabinopyranosyl(2→1)-O-α-L-rhamnopyranoside (7)Fig. 2Representative HPLC chromatograms of mixed standards and methanol extracts of Flos Lonicerae.Column: Agilent Zorbax SB-C18 column(250 4.6mm, 5.0µm), temperature of 25℃; Detector: ELSD, drift tube temperature 106℃, nitrogen flow-rate 2.6 l/min.A: Mixed standards, B: L. confusa, C: L. japonica, D: L. macranthoides, E: L. hypoglauca, F: L. similes.Table 1 Calibration curves for seven saponinsAnalytes Calibration curve ar2Test range(μg)LOD(μg)LOQ(μg)1 y=6711.9x-377.6 0.9940 0.56–22.01 0.26 0.882 y=7812.6x-411.9 0.9922 0.54–21.63 0.26 0.843 y=6798.5x-299.0 0.9958 0.46–18.42 0.22 0.724 y=12805x-487.9 0.9961 0.38–15.66 0.10 0.345 y=4143.8x-88.62 0.9989 0.42–16.82 0.18 0.246 y=3946.8x-94.4 0.9977 0.40–16.02 0.16 0.207 y=4287.8x-95.2 0.9982 0.42–16.46 0.12 0.22a y: Peak area; x: concentration (mg/ml)Table 2 Reproducibility of the assayAnalyteIntra-day variability Inter-day variability Content (mg/g) Mean RSD (%) Content (mg/g) Mean RSD (%)1 46.1646.2846.2246.22 0.1346.2245.3647.4226.33 2.232 5.385.385.165.31 2.405.285.345.045.22 3.043 4.374.304.184.28 2.244.284.464.024.255.204 nd1)-- -- nd -- --5 1.761.801.821.79 1.701.801.681.841.77 4.706 1.281.241.221.252.451.241.341.201.26 5.727 tr2)-- -- tr -- -- 1): not detected; 2): trace. RSD (%) = (SD/Mean) ×100%Table 3 Recovery of the seven analytesAnalyteOriginal(mg) Spiked(mg)Found(mg)Recovery(%)Mean(%)RSD(%)1 23.0823.1423.1119.7122.8628.1042.7346.1351.0199.7100.699.399.8 0.722.692.672.582.082.913.164.735.515.7698.197.6100.698.8 1.632.172.152.091.732.182.623.884.404.6598.8103.297.799.9 2.94nd1)1.011.050.980.981.101.0297.0104.8104.1102.0 4.250.880.900.910.700.871.081.561.752.0197.197.7101.898.9 2.660.640.620.610.450.610.751.081.211.3397.796.796.096.8 0.97tr2)1.021.101.081.031.111.07100.9102.799.1100.9 1.81): not detected; 2): trace.a Recovery (%) = (Amount found –Original amount)/ Amount spiked ×100%, RSD (%) = (SD/Mean) ×100%Table 4 Contents of seven saponins in Lonicera spp.Content (mg/g)1 2 3 4 5 6 7 L. confusa45.65±0.32 5.13±0.08 4.45±0.11tr1) 2.04±0.04tr 1.81±0.03 L. japonica nd2)nd 3.44±0.09nd nd nd nd L. macranthoides46.22±0.06 5.31±0.13 4.28±0.10 tr 1.79±0.03 1.25±0.03 tr L. hypoglauca11.17±0.07 nq3)53.78±1.18nd 1.72±0.02 2.23±0.06 2.52±0.04 L. similes41.22±0.25 4.57±0.07 3.79±0.09nd 1.75±0.02tr nd 1): trace; 2): not detected.. 3) not quantified owing to the suspicious purity of the peak.。

Cancer Epigenetics From Mechanism to Therapy

Cancer Epigenetics From Mechanism to Therapy

Leading EdgeReviewCancer Epigenetics:From Mechanism to TherapyMark A.Dawson1,2and Tony Kouzarides1,*1Gurdon Institute and Department of Pathology,University of Cambridge,Tennis Court Road,Cambridge CB21QN,UK2Department of Haematology,Cambridge Institute for Medical Research and Addenbrooke’s Hospital,University of Cambridge,Hills Road, Cambridge CB20XY,UK*Correspondence:t.kouzarides@/10.1016/j.cell.2012.06.013The epigenetic regulation of DNA-templated processes has been intensely studied over the last15 years.DNA methylation,histone modification,nucleosome remodeling,and RNA-mediated target-ing regulate many biological processes that are fundamental to the genesis of cancer.Here,we present the basic principles behind these epigenetic pathways and highlight the evidence suggest-ing that their misregulation can culminate in cancer.This information,along with the promising clin-ical and preclinical results seen with epigenetic drugs against chromatin regulators,signifies that it is time to embrace the central role of epigenetics in cancer.Chromatin is the macromolecular complex of DNA and histone proteins,which provides the scaffold for the packaging of our entire genome.It contains the heritable material of eukaryotic cells.The basic functional unit of chromatin is the nucleosome. It contains147base pairs of DNA,which is wrapped around a histone octamer,with two each of histones H2A,H2B,H3, and H4.In general and simple terms,chromatin can be subdi-vided into two major regions:(1)heterochromatin,which is highly condensed,late to replicate,and primarily contains inac-tive genes;and(2)euchromatin,which is relatively open and contains most of the active genes.Efforts to study the coordi-nated regulation of the nucleosome have demonstrated that all of its components are subject to covalent modification,which fundamentally alters the organization and function of these basic tenants of chromatin(Allis et al.,2007).The term‘‘epigenetics’’was originally coined by Conrad Wad-dington to describe heritable changes in a cellular phenotype that were independent of alterations in the DNA sequence. Despite decades of debate and research,a consensus definition of epigenetics remains both contentious and ambiguous(Berger et al.,2009).Epigenetics is most commonly used to describe chromatin-based events that regulate DNA-templated pro-cesses,and this will be the definition we use in this review. Modifications to DNA and histones are dynamically laid down and removed by chromatin-modifying enzymes in a highly regulated manner.There are now at least four different DNA modifications(Baylin and Jones,2011;Wu and Zhang,2011) and16classes of histone modifications(Kouzarides,2007;Tan et al.,2011).These are described in Table1.These modifications can alter chromatin structure by altering noncovalent interac-tions within and between nucleosomes.They also serve as docking sites for specialized proteins with unique domains that specifically recognize these modifications.These chromatin readers recruit additional chromatin modifiers and remodeling enzymes,which serve as the effectors of the modification.The information conveyed by epigenetic modifications plays a critical role in the regulation of all DNA-based processes, such as transcription,DNA repair,and replication.Conse-quently,abnormal expression patterns or genomic alterations in chromatin regulators can have profound results and can lead to the induction and maintenance of various cancers.In this Review,we highlight recent advances in our understanding of these epigenetic pathways and discuss their role in oncogen-esis.We provide a comprehensive list of all the recurrent cancer mutations described thus far in epigenetic pathways regulating modifications of DNA(Figure2),histones(Figures3,4,and5), and chromatin remodeling(Figure6).Where relevant,we will also emphasize existing and emerging drug therapies aimed at targeting epigenetic regulators(Figure1).Characterizing the EpigenomeOur appreciation of epigenetic complexity and plasticity has dramatically increased over the last few years following the development of several global proteomic and genomic technol-ogies.The coupling of next-generation sequencing(NGS)plat-forms with established chromatin techniques such as chromatin immunoprecipitation(ChIP-Seq)has presented us with a previ-ously unparalleled view of the epigenome(Park,2009).These technologies have provided comprehensive maps of nucleo-some positioning(Segal and Widom,2009),chromatin confor-mation(de Wit and de Laat,2012),transcription factor binding sites(Farnham,2009),and the localization of histone(Rando and Chang,2009)and DNA(Laird,2010)modifications.In addi-tion,NGS has revealed surprising facts about the mammalian transcriptome.We now have a greater appreciation of the fact that most of our genome is transcribed and that noncoding RNA may play a fundamental role in epigenetic regulation(Ama-ral et al.,2008).Most of the complexity surrounding the epigenome comes from the modification pathways that have been identified.12Cell150,July6,2012ª2012Elsevier Inc.Recent improvements in the sensitivity and accuracy of mass spectrometry (MS)instruments have driven many of these discoveries (Stunnenberg and Vermeulen,2011).Moreover,although MS is inherently not quantitative,recent advances in labeling methodologies,such as stable isotope labeling by amino acids in cell culture (SILAC),isobaric tags for relative and absolute quantification (iTRAQ),and isotope-coded affinity tag (ICAT),have allowed a greater ability to provide quantitative measurements (Stunnenberg and Vermeulen,2011).These quantitative methods have generated ‘‘protein recruit-ment maps’’for histone and DNA modifications,which contain proteins that recognize chromatin modifications (Bartke et al.,2010;Vermeulen et al.,2010).Many of these chromatin readers have more than one reading motif,so it is important to under-stand how they recognize several modifications either simulta-neously or sequentially.The concept of multivalent engagement by chromatin-binding modules has recently been explored by using either modified histone peptides (Vermeulen et al.,2010)or in-vitro-assembled and -modified nucleosomes (Bartkeet al.,2010;Ruthenburg et al.,2011).The latter approach in particular has uncovered some of the rules governing the recruit-ment of protein complexes to methylated DNA and modified histones in a nucleosomal context.The next step in our under-standing will require a high-resolution in vivo genomic approach to detail the dynamic events on any given nucleosome during the course of gene expression.Epigenetics and the Cancer ConnectionThe earliest indications of an epigenetic link to cancer were derived from gene expression and DNA methylation studies.These studies are too numerous to comprehensively detail in this review;however,the reader is referred to an excellent review detailing the history of cancer epigenetics (Feinberg and Tycko,2004).Although many of these initial studies were purely correl-ative,they did highlight a potential connection between epige-netic pathways and cancer.These early observations have been significantly strengthened by recent results from the Inter-national Cancer Genome Consortium (ICGC).Whole-genomeTable 1.Chromatin Modifications,Readers,and Their Function Chromatin Modification NomenclatureChromatin-Reader MotifAttributed Functionand Cit,citrulline.Reader domains:MBD,methyl-CpG-binding domain;PHD,plant homeodomain;MBT,malignant brain tumor domain;PWWP,proline-tryptophan-tryptophan-proline domain;BRCT,BRCA1C terminus domain;UIM,ubiquitin interaction motif;IUIM,inverted ubiquitin interaction motif;SIM,sumo interaction motif;and PBZ,poly ADP-ribose binding zinc finger.aThese are established binding modules for the posttranslational modification;however,binding to modified histones has not been firmly established.Cell 150,July 6,2012ª2012Elsevier Inc.13sequencing in a vast array of cancers has provided a catalog of recurrent somatic mutations in numerous epigenetic regulators (Forbes et al.,2011;Stratton et al.,2009).A central tenet in analyzing these cancer genomes is the identification of ‘‘driver’’mutations (causally implicated in the process of oncogenesis).A key feature of driver mutations is that they are recurrently found in a variety of cancers,and/or they are often present at a high prevalence in a specific tumor type.We will mostly concentrate our discussions on suspected or proven driver mutations in epigenetic regulators.For instance,malignancies such as follicular lymphoma contain recurrent mutations of the histone methyltransferase MLL2in close to 90%of cases (Morin et al.,2011).Similarly,UTX ,a histone demethylase,is mutated in up to 12histologi-cally distinct cancers (van Haaften et al.,2009).Compilation of the epigenetic regulators mutated in cancer highlights histone acetylation and methylation as the most widely affected epige-netic pathways (Figures 3and 4).These and other pathways that are affected to a lesser extent will be described in the following sections.Deep sequencing technologies aimed at mapping chromatin modifications have also begun to shed some light on the origins of epigenetic abnormalities in cancer.Cross-referencing of DNA methylation profiles in human cancers with ChIP-Seq data for histone modifications and the binding of chromatinregulators have raised intriguing correlations between cancer-associated DNA hypermethylation and genes marked with ‘‘bivalent’’histone modifications in multipotent cells (Easwaran et al.,2012;Ohm et al.,2007).These bivalent genes are marked by active (H3K4me3)and repressive (H3K27me3)histone modi-fications (Bernstein et al.,2006)and appear to identify transcrip-tionally poised genes that are integral to development and lineage commitment.Interestingly,many of these genes are targeted for DNA methylation in cancer.Equally intriguing are recent comparisons between malignant and normal tissues from the same individuals.These data demonstrate broad domains within the malignant cells that contain significant alter-ations in DNA methylation.These regions appear to correlate with late-replicating regions of the genome associated with the nuclear lamina (Berman et al.,2012).Although there remains little mechanistic insight into how and why these regions of the genome are vulnerable to epigenetic alterations in cancer,these studies highlight the means by which global sequencing plat-forms have started to uncover avenues for further investigation.Genetic lesions in chromatin modifiers and global alterations in the epigenetic landscape not only imply a causative role for these proteins in cancer but also provide potential targets for therapeutic intervention.A number of small-molecule inhibitors have already been developed against chromatin regulators (Figure 1).These are at various stages of development,andthreeFigure 1.Epigenetic Inhibitors as Cancer TherapiesThis schematic depicts the process for epigenetic drug development and the current status of various epigenetic therapies.Candidate small molecules are first tested in vitro in malignant cell lines for specificity and phenotypic response.These may,in the first instance,assess the inhibition of proliferation,induction of apoptosis,or cell-cycle arrest.These phenotypic assays are often coupled to genomic and proteomic methods to identify potential molecular mechanisms for the observed response.Inhibitors that demonstrate potential in vitro are then tested in vivo in animal models of cancer to ascertain whether they may provide therapeutic benefit in terms of survival.Animal studies also provide valuable information regarding the toxicity and pharmacokinetic properties of the drug.Based on these preclinical studies,candidate molecules may be taken forward into the clinical setting.When new drugs prove beneficial in well-conducted clinical trials,they are approved for routine clinical use by regulatory authorities such as the FDA.KAT,histone lysine acetyltransferase;KMT,histone lysine methyltransferase;RMT,histone arginine methyltransferase;and PARP,poly ADP ribose polymerase.14Cell 150,July 6,2012ª2012Elsevier Inc.of these(targeting DNMTs,HDACs,and JAK2)have already been granted approval by the US Food and Drug Administra-tion(FDA).This success may suggest that the interest in epige-netic pathways as targets for drug discovery had been high over the past decade.However,the reality is that thefield of drug discovery had been somewhat held back due to concerns over the pleiotropic effects of both the drugs and their targets. Indeed,some of the approved drugs(against HDACs)have little enzyme specificity,and their mechanism of action remains contentious(Minucci and Pelicci,2006).The belief and investment in epigenetic cancer therapies may now gain momentum and reach a new level of support following the recent preclinical success of inhibitors against BRD4,an acetyl-lysine chromatin-binding protein(Dawson et al.,2011; Delmore et al.,2011;Filippakopoulos et al.,2010;Mertz et al., 2011;Zuber et al.,2011).The molecular mechanisms governing these impressive preclinical results have also been largely uncovered and are discussed below.This process is pivotal for the successful progression of these inhibitors into the clinic. These results,along with the growing list of genetic lesions in epigenetic regulators,highlight the fact that we have now entered an era of epigenetic cancer therapies.Epigenetic Pathways Connected to CancerDNA MethylationThe methylation of the5-carbon on cytosine residues(5mC)in CpG dinucleotides was thefirst described covalent modifica-tion of DNA and is perhaps the most extensively characterized modification of chromatin.DNA methylation is primarily noted within centromeres,telomeres,inactive X-chromosomes,and repeat sequences(Baylin and Jones,2011;Robertson,2005). Although global hypomethylation is commonly observed in malignant cells,the best-studied epigenetic alterations in cancerare the methylation changes that occur within CpG islands, which are present in 70%of all mammalian promoters.CpG island methylation plays an important role in transcriptional regu-lation,and it is commonly altered during malignant transforma-tion(Baylin and Jones,2011;Robertson,2005).NGS platforms have now provided genome-wide maps of CpG methylation. These have confirmed that between5%–10%of normally unme-thylated CpG promoter islands become abnormally methylated in various cancer genomes.They also demonstrate that CpG hypermethylation of promoters not only affects the expression of protein coding genes but also the expression of various noncoding RNAs,some of which have a role in malignant trans-formation(Baylin and Jones,2011).Importantly,these genome-wide DNA methylome studies have also uncovered intriguing alterations in DNA methylation within gene bodies and at CpG‘‘shores,’’which are conserved sequences upstream and downstream of CpG islands.The functional relevance of these regional alterations in methylation are yet to be fully deciphered, but it is interesting to note that they have challenged the general dogma that DNA methylation invariably equates with transcriptional silencing.In fact,these studies have established that many actively transcribed genes have high levels of DNA methylation within the gene body,suggesting that the context and spatial distribution of DNA methylation is vital in transcrip-tional regulation(Baylin and Jones,2011).Three active DNA methyltransferases(DNMTs)have been identified in higher eukaryotes.DNMT1is a maintenance methyl-transferase that recognizes hemimethylated DNA generated during DNA replication and then methylates newly synthesized CpG dinucleotides,whose partners on the parental strand are already methylated(Li et al.,1992).Conversely,DNMT3a and DNMT3b,although also capable of methylating hemimethylated DNA,function primarily as de novo methyltransferases to estab-lish DNA methylation during embryogenesis(Okano et al.,1999). DNA methylation provides a platform for several methyl-binding proteins.These include MBD1,MBD2,MBD3,and MeCP2. These in turn function to recruit histone-modifying enzymes to coordinate the chromatin-templated processes(Klose and Bird,2006).Although mutations in DNA methyltransferases and MBD proteins have long been known to contribute to developmental abnormalities(Robertson,2005),we have only recently become aware of somatic mutations of these key genes in human malig-nancies(Figure2).Recent sequencing of cancer genomes has identified recurrent mutations in DNMT3A in up to25%of patients with acute myeloid leukemia(AML)(Ley et al.,2010). Importantly,these mutations are invariably heterozygous and are predicted to disrupt the catalytic activity of the enzyme. Moreover,their presence appears to impact prognosis(Patel et al.,2012).However,at present,the mechanisms bywhich Figure2.Cancer Mutations Affecting Epigenetic Regulators of DNA MethylationThe5-carbon of cytosine nucleotides are methylated(5mC)by a family of DNMTs.One of these,DNMT3A,is mutated in AML,myeloproliferative diseases(MPD),and myelodysplastic syndromes(MDS).In addition to its catalytic activity,DNMT3A has a chromatin-reader motif,the PWWP domain, which may aid in localizing this enzyme to chromatin.Somatically acquired mutations in cancer may also affect this domain.The TET family of DNA hydroxylases metabolizes5mC into several oxidative intermediates,including 5-hydroxymethylcytosine(5hmC),5-formylcytosine(5fC),and5-carbox-ylcytosine(5caC).These intermediates are likely involved in the process of active DNA demethylation.Two of the three TET family members are mutated in cancers,including AML,MPD,MDS,and CMML.Mutation types are as follows:M,missense;F,frameshift;N,nonsense;S,splice site mutation;and T,translocation.Cell150,July6,2012ª2012Elsevier Inc.15these mutations contribute to the development and/or mainte-nance of AML remains elusive.Understanding the cellular consequences of normal and aber-rant DNA methylation remains a key area of interest,especially because hypomethylating agents are one of the few epigenetic therapies that have gained FDA approval for routine clinical use(Figure1).Although hypomethylating agents such as azaci-tidine and decitabine have shown mixed results in various solid malignancies,they have found a therapeutic niche in the myelo-dysplastic syndromes(MDS).Until recently,this group of disor-ders was largely refractory to therapeutic intervention,and MDS was primarily managed with supportive care.However,several large studies have now shown that treatment with azacitidine, even in poor prognosis patients,improves their quality of life and extends survival time.Indeed,azacitidine is thefirst therapy to have demonstrated a survival benefit for patients with MDS (Fenaux et al.,2009).The molecular mechanisms governing the impressive responses seen in MDS are largely unknown. However,recent evidence would suggest that low doses of these agents hold the key to therapeutic benefit(Tsai et al., 2012).It is also emerging that the combinatorial use of DNMT and HDAC inhibitors may offer superior therapeutic outcomes (Gore,2011).DNA Hydroxy-Methylation and Its Oxidation Derivatives Historically,DNA methylation was generally considered to be a relatively stable chromatin modification.However,early studies assessing the global distribution of this modification during embryogenesis had clearly identified an active global loss of DNA methylation in the early zygote,especially in the male pronucleus.More recently,high-resolution genome-wide mapping of this modification in pluripotent and differentiated cells has also confirmed the dynamic nature of DNA methylation, evidently signifying the existence of an enzymatic activity within mammalian cells that either erases or alters this chromatin modification(Baylin and Jones,2011).In2009,two seminal manuscripts describing the presence of5-hydroxymethylcyto-sine(5hmC)offered thefirst insights into the metabolism of 5mC(Kriaucionis and Heintz,2009;Tahiliani et al.,2009).The ten-eleven translocation(TET1–3)family of proteins have now been demonstrated to be the mammalian DNA hydroxy-lases responsible for catalytically converting5mC to5hmC. Indeed,iterative oxidation of5hmC by the TET family results in further oxidation derivatives,including5-formylcytosine(5fC) and5-carboxylcytosine(5caC).Although the biological signifi-cance of the5mC oxidation derivatives is yet to be established, several lines of evidence highlight their importance in transcrip-tional regulation:(1)they are likely to be an essential intermediate in the process of both active and passive DNA demethylation,(2) they preclude or enhance the binding of several MBD proteins and,as such,will have local and global effects by altering the recruitment of chromatin regulators,and(3)genome-wide mapping of5hmC has identified a distinctive distribution of this modification at both active and repressed genes,including its presence within gene bodies and at the promoters of bivalently marked,transcriptionally poised genes(Wu and Zhang,2011). Notably,5hmC was also mapped to several intergenic cis-regu-latory elements that are either functional enhancers or insulator elements.Consistent with the notion that5hmC is likely to have a role in both transcriptional activation and silencing, the TET proteins have also been shown to have activating and repressive functions(Wu and Zhang,2011).Genome-wide mapping of TET1has demonstrated it to have a strong prefer-ence for CpG-rich DNA and,consistent with its catalytic function, it also been localized to regions enriched for5mC and5hmC. The TET family of proteins derive their name from the initial description of a recurrent chromosomal translocation, t(10;11)(q22;q23),which juxtaposes the MLL gene with TET1in a subset of patients with AML(Lorsbach et al.,2003).Notably, concurrent to the initial description of the catalytic activity for the TET family of DNA hydroxylases,several reports emerged describing recurrent mutations in TET2in numerous hematolog-ical malignancies(Cimmino et al.,2011;Delhommeau et al., 2009;Langemeijer et al.,2009)(Figure2).Interestingly,TET2-deficient mice develop a chronic myelomonocytic leukemia (CMML)phenotype,which is in keeping with the high prevalence of TET2mutations in patients with this disease(Moran-Crusio et al.,2011;Quivoron et al.,2011).The clinical implications of TET2mutations have largely been inconclusive;however,in some subsets of AML patients,TET2mutations appear to confer a poor prognosis(Patel et al.,2012).Early insights into the process of TET2-mediated oncogenesis have revealed that the patient-associated mutations are largely loss-of-function muta-tions that consequently result in decreased5hmC levels and a reciprocal increase in5mC levels within the malignant cells that harbor them.Moreover,mutations in TET2also appear to confer enhanced self-renewal properties to the malignant clones (Cimmino et al.,2011).Histone ModificationsIn1964,Vincent Allfrey prophetically surmised that histone modifications might have a functional influence on the regulation of transcription(Allfrey et al.,1964).Nearly half a century later, thefield is still grappling with the task of unraveling the mecha-nisms underlying his enlightened statement.In this time,we have learned that these modifications have a major influence, not just on transcription,but in all DNA-templated processes (Kouzarides,2007).The major cellular processes attributed to each of these modifications are summarized in Table1.The great diversity in histone modifications introduces a remarkable complexity that is slowly beginning to be ing transcription as an example,we have learned that multiple coexisting histone modifications are associated with activation,and some are associated with repression. However,these modification patterns are not static entities but a dynamically changing and complex landscape that evolves in a cell context-dependent fashion.Moreover,active and repres-sive modifications are not always mutually exclusive,as evi-denced by‘‘bivalent domains.’’The combinatorial influence that one or more histone modifications have on the deposition, interpretation,or erasure of other histone modifications has been broadly termed‘‘histone crosstalk,’’and recent evidence would suggest that crosstalk is widespread and is of great bio-logical significance(Lee et al.,2010).It should be noted that the cellular enzymes that modify histones may also have nonhistone targets and,as such,it has been difficult to divorce the cellular consequences of individual histone modifications from the broader targets of many of these16Cell150,July6,2012ª2012Elsevier Inc.enzymes.In addition to their catalytic function,many chromatin modifiers also possess‘‘reader’’domains allowing them to bind to specific regions of the genome and respond to information conveyed by upstream signaling cascades.This is important, as it provides two avenues for therapeutically targeting these epigenetic regulators.The residues that line the binding pocket of reader domains can dictate a particular preference for specific modification states,whereas residues outside the binding pocket contribute to determining the histone sequence specificity.This combination allows similar reader domains to dock at different modified residues or at the same amino acid displaying different modification states.For example,some methyl-lysine readers engage most efficiently with di/tri-methyl-ated lysine(Kme2/3),whereas others prefer mono-or unmethy-lated lysines.Alternatively,when the same lysines are now acet-ylated,they bind to proteins containing bromodomains(Taverna et al.,2007).The main modification binding pockets contained within chromatin-associated proteins is summarized in Table1. Many of the proteins that modify or bind these histone modifi-cations are misregulated in cancer,and in the ensuing sections, we will discuss the most extensively studied histone modifica-tions in relation to oncogenesis and novel therapeutics. Histone Acetylation.The Nε-acetylation of lysine residues is a major histone modification involved in transcription,chromatin structure,and DNA repair.Acetylation neutralizes lysine’s posi-tive charge and may consequently weaken the electrostatic interaction between histones and negatively charged DNA.For this reason,histone acetylation is often associated with a more ‘‘open’’chromatin conformation.Consistent with this,ChIP-Seq analyses have confirmed the distribution of histone acetyla-tion at promoters and enhancers and,in some cases,throughout the transcribed region of active genes(Heintzman et al.,2007; Wang et al.,2008).Importantly,lysine acetylation also serves as the nidus for the binding of various proteins with bromodo-mains and tandem plant homeodomain(PHD)fingers,which recognize this modification(Taverna et al.,2007).Acetylation is highly dynamic and is regulated by the competing activities of two enzymatic families,the histone lysine acetyltransferases(KATs)and the histone deacetylases (HDACs).There are two major classes of KATs:(1)type-B,which are predominantly cytoplasmic and modify free histones,and(2) type-A,which are primarily nuclear and can be broadly classifiedinto the GNAT,MYST,and CBP/p300families.KATs were thefirst enzymes shown to modify histones.The importance of thesefindings to cancer was immediately apparent,as one of these enzymes,CBP,was identified by its ability to bind the transforming portion of the viral oncoprotein E1A(Bannister and Kouzarides,1996).It is now clear that many,if not most,of the KATs have been implicated in neoplastic transformation,and a number of viral oncoproteins are known to associate with them.There are numerous examples of recur-rent chromosomal translocations(e.g.,MLL-CBP[Wang et al., 2005]and MOZ-TIF2[Huntly et al.,2004])or coding mutations (e.g.,p300/CBP[Iyer et al.,2004;Pasqualucci et al.,2011]) involving various KATs in a broad range of solid and hematolog-ical malignancies(Figure3).Furthermore,altered expression levels of several of the KATs have also been noted in a range of cancers(Avvakumov and Coˆte´,2007;Iyer et al.,2004).In some cases,such as the leukemia-associated fusion gene MOZ-TIF2,we know a great deal about the cellular conse-quences of this translocation involving a MYST family member. MOZ-TIF2is sufficient to recapitulate an aggressive leukemia in murine models;it can confer stem cell properties and reacti-vate a self-renewal program when introduced into committed hematopoietic progenitors,and much of this oncogenic potential is dependent on its inherent and recruited KAT activity as well as its ability to bind to nucleosomes(Deguchi et al.,2003;Huntly et al.,2004).Despite these insights,the great conundrum with regards to unraveling the molecular mechanisms by which histone acetyl-transferases contribute to malignant transformation has been dissecting the contribution of altered patterns in acetylation on histone and nonhistone proteins.Although it is clear that global histone acetylation patterns are perturbed in cancers(Fraga Figure 3.Cancer Mutations Affecting Epigenetic Regulators Involved in Histone AcetylationThese tables provide somatic cancer-associated mutations identified in histone acetyltransferases and proteins that contain bromodomains(which recognize and bind acetylated histones).Several histone acetyltransferases possess chromatin-reader motifs and,thus,mutations in the proteins may alter both their catalytic activities as well as the ability of these proteins to scaffold multiprotein complexes to chromatin.Interestingly,sequencing of cancer genomes to date has not identified any recurrent somatic mutations in histone deacetylase enzymes.Abbreviations for the cancers are as follows: AML,acute myeloid leukemia;ALL,acute lymphoid leukemia;B-NHL,B-cell non-Hodgkin’s lymphoma;DLBCL,diffuse large B-cell lymphoma;and TCC, transitional cell carcinoma of the urinary bladder.Mutation types are as follows:M,missense;F,frameshift;N,nonsense;S,splice site mutation;T, translocation;and D,deletion.Cell150,July6,2012ª2012Elsevier Inc.17。

循证医学PICO

循证医学PICO

[结果]疗效评价按实体肿瘤的疗效评价标准(Response EvaluationCriteria In Solid Tumors RECISTl.1)标准进行。手术组总有效率(CR+PR): 96.8%。非手术组总有效率:93.0%。1、2、3年生存率为93.2%、85.1%、 63.5%。手术组与非手术组1、2、3年生存率分别为93.54%、87.09%、 77.41%,93.02%、83.72%、55.82%,手术组内观察:pI期、pIIa期、 pIIb期1、2、3年生存率分别为:100%、90.90%、90.90%,100%、 88.88%、88.88%,81.81%、63.63%、54.54%。手术组与非手术组1、2 年生存率无明显差异,3年生存率手术组优于非手术组(P<0.05)。手术 组内分析:pI期~pIIa期生存率无差异,IIb期生存率明显降低。差异有统计 学意义(P<0.05)。[结论]在早期小细胞肺癌患者治疗方案的选择中:单 纯化、放疗相比,手术治疗联合化疗或联合化疗+放疗的治疗手段,能明显 延长小细胞肺癌患者生存时间。肿瘤TN分期对早期小细胞肺癌的预后均有 明显影响。
证据3
来源数据库:PubMed Journal DOI:10.3390/ijms160511439 关键词:CC chemokine ligand 2 (CCL2); SCLC; blood–brain barrier (BBB); brain metastasis; transendothelial migration; visfatin; 摘要:Small-cell lung cancer (SCLC) is characterized as an aggressive tumor with brain metastasis. Although preventing SCLC metastasis to the brain is immensely important for survival, the molecular mechanisms of SCLC cells penetrating the blood-brain barrier (BBB) are largely unknown. Herein, we present evidence that elevated levels of visfatin in the serum of SCLC patients were associated with brain metastasis, and visfain was increased in NCI-H446 cells, a SCLC cell line, during interacting with human brain microvascular endothelial cells (HBMEC). Using in vitro BBB model, we found that visfatin could promote NCI-H446 cells migration across HBMEC monolayer, while the effect was inhibited by knockdown of visfatin. Furthermore, our findings indicated that CC chemokine ligand 2 (CCL2) was involved in visfatin-mediated NCI-H446 cells transendothelial migtation. Results also showed that the upregulation of CCL2 in the co-culture system was reversed by blockade of visfatin. In particular, visfatin-induced CCL2 was attenuated by specific inhibitor of PI3K/Akt signaling in NCI-H446 cells. Taken together, we demonstrated that visfatin was a prospective target for SCLC metastasis to brain, and understanding the molecular mediators would lead to effective strategies for inhibition of SCLC brain metastasis.

妊娠期糖代谢异常的母儿遗传问题

妊娠期糖代谢异常的母儿遗传问题

妊娠期糖代谢异常的母儿遗传问题祝彩霞;王子莲【期刊名称】《妇产与遗传(电子版)》【年(卷),期】2015(005)002【总页数】5页(P13-17)【关键词】妊娠期糖代谢异常;母婴遗传;糖尿病,妊娠;糖尿病合并妊娠【作者】祝彩霞;王子莲【作者单位】510080 广州, 中山大学附属第一医院妇产科;510080 广州, 中山大学附属第一医院妇产科【正文语种】中文【中图分类】R714.256随着经济发展和人们生活水平的提高,妊娠期糖代谢异常的发病率逐年升高[1]。

妊娠期糖代谢异常包括妊娠期糖尿病(gestational diabetes mellitus,GDM)和孕前糖尿病(pregestational diabetesmellitus,PGDM)。

其中,妊娠前患有1型或2型糖尿病称为孕前糖尿病,其对母儿产生严重不良影响。

妊娠期糖尿病在不同的种族和发展中国家发病率有所不同,国际糖尿病联合会(International Diabetes Federation)统计分析全球大约有16%的孕妇出现血糖升高,新标准下我国妊娠合并糖尿病的患病率高达17.5%。

妊娠期糖代谢异常不仅促进各种妊娠期并发症的发生,还与胚胎发育、子代生长发育及远期并发症相关[2]。

研究表明,围产期监护能改善妊娠期糖代谢异常孕妇的妊娠结局,其中严格的血糖控制、规律的产前检查和早期诊治并发症有助于提高活产率、降低母儿不良并发症的发生[3-6]。

糖尿病的遗传学病因至今仍然不明,目前发现至少40个等位基因与2型糖尿病的发病相关[7],与1型糖尿病相关的易感基因则有60多个[8]。

但基因遗传仅能解释5%~10%的糖尿病的发病,其他因素还包括表观遗传、营养条件、环境等因素的作用。

本文对妊娠期糖尿病对母儿遗传代谢的影响综述如下。

一、糖代谢异常对胚胎发育的影响研究发现,妊娠期糖代谢异常影响胚胎发育,从而导致胎儿宫内生长受限、母胎代谢异常、胚胎发育异常等。

第六届全国生物信息学与系统生物学学术大会

第六届全国生物信息学与系统生物学学术大会

第六届全国生物信息学与系统生物学学术大会
暨国际生物信息学前沿研讨会
会议时间:2014年10月6日— 9日
会议地点:南京市中山东路307号钟山宾馆(江苏省会议中心)
会议日程概要
会议期间联系:侯越,谢建明,孙啸
会议日程
3
四、主题报告、专题报告(三)10月7日下午,309会议室
五、10月8日上午, 三楼大会堂
六、主题报告、专题报告(四)10月8日下午,307会议室
5
八、主题报告、专题报告(六)10月8日下午,309会议室
九、会议墙报交流10月8日晚上,20:00开始,金陵厅
十、青年沙龙10月8日晚上,20:00开始,307会议室
欢迎青年科研工作者参加。

7
十一、10月9日上午, 三楼大会堂。

  1. 1、下载文档前请自行甄别文档内容的完整性,平台不提供额外的编辑、内容补充、找答案等附加服务。
  2. 2、"仅部分预览"的文档,不可在线预览部分如存在完整性等问题,可反馈申请退款(可完整预览的文档不适用该条件!)。
  3. 3、如文档侵犯您的权益,请联系客服反馈,我们会尽快为您处理(人工客服工作时间:9:00-18:30)。

Resource Identification of67Histone Marksand Histone Lysine Crotonylationas a New Type of Histone ModificationMinjia Tan,1,6Hao Luo,1,6Sangkyu Lee,1,6Fulai Jin,2Jeong Soo Yang,1Emilie Montellier,3Thierry Buchou,3Zhongyi Cheng,1Sophie Rousseaux,3Nisha Rajagopal,2Zhike Lu,1Zhen Ye,2Qin Zhu,4Joanna Wysocka,5Yang Ye,4 Saadi Khochbin,3Bing Ren,2and Yingming Zhao1,*1Ben May Department of Cancer Research,The University of Chicago,Chicago,IL60637,USA2Ludwig Institute for Cancer Research and Department of Cellular and Molecular Medicine,University of California San Diego School of Medicine,9500Gilman Drive,La Jolla,CA92093,USA3INSERM,U823;Universite´Joseph Fourier-Grenoble1;Institut Albert Bonniot,Faculte´de Me´decine,Domaine de la Merci,38706La Tronche Cedex,France4Shanghai Institute of Materia Medica,Chinese Academy of Sciences,555Zu Chong Zhi Road,Shanghai201203,P.R.China5Department of Chemical and Systems Biology,Stanford University School of Medicine,Stanford,CA94305,USA6These authors contributed equally to this work*Correspondence:yingming.zhao@DOI10.1016/j.cell.2011.08.008SUMMARYWe report the identification of67previously unde-scribed histone modifications,increasing the current number of known histone marks by about70%. We further investigated one of the marks,lysine cro-tonylation(Kcr),confirming that it represents an evolutionarily-conserved histone posttranslational modification.The unique structure and genomic localization of histone Kcr suggest that it is mecha-nistically and functionally different from histone lysine acetylation(Kac).Specifically,in both human somatic and mouse male germ cell genomes,histone Kcr marks either active promoters or potential en-hancers.In male germinal cells immediately fol-lowing meiosis,Kcr is enriched on sex chromosomes and specifically marks testis-specific genes,in-cluding a significant proportion of X-linked genes that escape sex chromosome inactivation in haploid cells.These results therefore dramatically extend the repertoire of histone PTM sites and designate Kcr as a specific mark of active sex chromosome-linked genes in postmeiotic male germ cells. INTRODUCTIONMounting evidence suggests that histone PTMs play a crucial role in diverse biological processes,such as cell differentiation and organismal development,and that aberrant modification of histones contributes to diseases such as cancer(Berdasco and Esteller,2010;Fullgrabe et al.,2011).At least eleven types of PTMs have been reported at over60different amino acid resi-dues on histones,including histone methylation,acetylation,propionylation,butyrylation,formylation,phosphorylation,ubiq-uitylation,sumoylation,citrullination,proline isomerization,and ADP ribosylation(Martin and Zhang,2007;Ruthenburg et al., 2007).Histone PTMs are thought to contribute to the regulation of chromatin-templated processes via two major mechanisms (Kouzarides,2007;Ruthenburg et al.,2007).First,histone PTMs can directly modulate the packaging of chromatin either by altering the net charge of histone molecules or by altering inter-nucleosomal interactions,thereby regulating chromatin structure and the access of DNA-binding proteins such as tran-scription factors.Second,histone PTMs regulate chromatin structure and function by recruiting PTM-specific binding pro-teins,which recognize modified histones via specialized struc-tural folds such as bromo-,chromo-and PHD domains(Wysocka et al.,2005,2006;Zeng and Zhou,2002).Alternatively,histone PTMs can also function by inhibiting the interaction of specific binders with chromatin.PTM-induced changes in protein interac-tions between chromatin and its binding partners are in turn translated into biological outcomes(Margueron et al.,2005). While the majority of known histone PTMs are located within the N-terminal tail domain of core histones,PTMs of crucial importance for histone-DNA and histone-histone interactions have also been found in the globular domain of core histones (Cosgrove et al.,2004;Garcia et al.,2007c;Mersfelder and Par-thun,2006).Novel PTM sites occurring outside of the N-terminal tails continue to be discovered,generally with the aid of sequence and modification-specific antibodies or by unbiased mass spec-trometry(MS)methods(Chu et al.,2006;Garcia et al.,2007b; Johnson et al.,2004;Wisniewski et al.,2007).The recent discovery of O-GlcNAc modification(Sakabe et al.,2010) suggests that additional histone PTMs may yet be discovered. Here,we used an integrated,mass spectrometry-based proteomics approach,which takes advantage of in vitro propio-nylation,efficient peptide separation using isoelectric focusing1016Cell146,1016–1028,September16,2011ª2011Elsevier Inc.(OFFGEL),and the high sensitivity of the LTQ Orbitrap Velos mass spectrometer to carry out a comprehensive analysis of histone PTMs.With this approach,we achieved high sequence coverage of peptide mapping in core and linker histones, ranging from87%–100%,which in turn resulted in the identifi-cation of67new PTM sites.These histone marks expand the total number of known histone PTMs by about70%.Inter-estingly,our results show that histones are intensely modified at various residues not only in the N-terminal tail,but also within globular domains.Among the modifications,we identi-fied tyrosine hydroxylation(Yoh)and lysine crotonylation(Kcr) as two novel histone mark types.Finally,we demonstrated that histone Kcr is a robust indicator of active promoters and could be an important signal in the control of male germ cell differentiation.RESULTSExperimental DesignHistone proteins are characterized by a high ratio of both lysine and arginine residues(Garcia et al.,2007a;Zee et al.,2010).As a result,tryptic digestions of histones tend to yield peptides that are relatively small and hydrophilic,which are difficult for subsequent detection by MS due to poor retention by the C18 RP-HPLC column.This problem can be addressed by chemical derivatization(e.g.,lysine propionylation)of amine groups in the protein(N-terminal amines,and free and monomethylated lysine ε-amino groups)before or after tryptic digestion(Garcia et al., 2007a).Similarly,lysine propionylation of core histones,before or after tryptic digestion,can generate complementary peptide sequences that boost the sequence coverage of peptide mapping by MS.Additionally,IEF separation of the tryptic digest into12fractions will further reduce peptide complexity and improve dynamic range.Using this rationale,we designed an integrated approach for the systematic analysis of histone PTMs which maximizes both sequence coverage and sensitivity,leading to the identifi-cation of many novel PTM sites.In this method,MS analysis was carried out in histone proteolytic peptides that were gener-ated by four parallel methods(Figure1A):(1)Tryptic digestion of core histones without an in vitro chemical derivatization reac-tion;(2)tryptic peptides that were in vitro propionylated after tryptic digestion;(3)tryptic peptides that were generated by tryptic digestion of in vitro propionylated histone proteins;and (4)tryptic peptides that were generated by tryptic in-gel diges-tion of the individual histone proteins.We used PTMap,an algorithm capable of identifying all possible PTMs of a protein(Chen et al.,2009),to analyze all acquired MS/MS data and identify histone peptides with or without a PTM.As anticipated,sequence coverage by MS mapping was significantly improved after in vitro propionylation, either before or after tryptic digestion(Figure1B).Among the four methods,Method III(in vitro propionylation before tryptic diges-tion of histones)achieved the highest sequence coverage of histones H1.2(100%),H2A(90.7%),and H2B(94.4%).Method IV gave the best coverage for histones H3(87.3%)and H4 (82.3%).In aggregate,we achieved sequence coverage of 100%of H1.2,90.7%of H2A,100%of H2B,91%of H3,and87.3%of H4.To our knowledge,this represents the highest reported sequence coverage for peptide mapping in histones.Using this approach,we identified130unique PTM sites, which not only confirmed63previously known histone PTMs, but also revealed67novel ones,including28Kcr sites,18lysine monomethylation(Kme)sites,1lysine dimethylation(Kme2)site, 4lysine formylation(Kfo)sites,2lysine acetylation(Kac)sites,8 arginine monomethylation(Rme)sites,and6tyrosine hydroxyl-ation(Yoh)sites(Figure1C).A summary of the non-Kcr modification sites and Kcr sites identified in this study is shown in Figures1D and1E,respec-tively.All the MS/MS spectra for the identified histone PTM peptides were carefully verified as previously reported(Chen et al.,2005).We confirmed the identification of10novel non-Kcr PTM sites by high-resolution MS/MS(Figure S1available online).Identification and validation of these non-Kcr PTMs are included in Extended Experimental Procedures. Characterization of the Novel Histone PTM sitesA core histone protein typically consists of an unstructured N-terminus,a globular core including a central histone-fold domain,and a conformationally mobile C-terminal tail(Garcia et al.,2007c;Mersfelder and Parthun,2006).The central histone-fold domain consists of three a helices and two loops that are known to be involved in histone pair-pair and histone-DNA interaction sites(McGhee and Felsenfeld,1980).The majority of known histone PTM sites were previously identified in the N-terminal regions of histones.In this study,39novel non-Kcr PTM sites were identified.Inter-estingly,among these sites,only four sites were mapped to the N-terminal domains,while25were mapped to central histone-fold domains,and another ten were mapped to the C-terminal domains(Figure1D).Six PTM sites(including three monomethy-lated residues at H2BR79,H3R63,and H4K77,1acetylated residue at H3K122,and2hydroxylated sites at H2BY83and H4Y88)are located at the histone-fold domains(Figure S1M). H3R63and H4K77participate in DNA interactions(Arents and Moudrianakis,1993;Luger et al.,1997;Mersfelder and Parthun, 2006),while H2BR79,H2BY83,and H4Y88participate in the H2B-H4interaction.While the amino acid residues located on the outer nucleosome surface do not contact DNA,they are known to regulate chromatin structure(Mersfelder and Parthun, 2006).Five PTM sites were mapped at the outer nucleosome surface,among which we found3novel PTM sites,including monomethyllysine and formyllysine at H2BK116,and dimethylly-sine at H4K59(Figure S1M).Given the important roles performed by these residues in nucleosome structure and DNA binding,it is highly likely that these PTM sites will have significant impacts on transcriptional and epigenetic regulation.Identification and Validation of Kcr Residues in Histones A PTM will induce a structural change in the substrate residue and therefore a change of its molecular weight.Interestingly, on28lysine residues of core histone peptides,our analysis identified a mass shift of+68Da that does not match the shift associated with any known PTM(Figure1E).This result sug-gested the possible presence of a previously unreported histone mark.Cell146,1016–1028,September16,2011ª2011Elsevier Inc.1017B(%)CPTMsLinkerhistoneCore histonesTotalH1.2 H2A H2B H3 H4# ofidentifiedsites3112313323130Protein sequenceValidationSequencecoverage# of novelsites171019101167Kme 736-218Kme2 ----11Kfo 1-21-4Kac 1--1-2Rme -21238Yoh 112-26alignmentHPLC-MS/MSKcr 7486328SGRGKQGGKAR GNYSER HLQLAIR KKTESHHKS…KASGP…K…K…KK…Y…K…K…K…KLNK…K…K…K…K…K…KKK62268151819956841709213183memeacac12376663187me54me me me mefofomemeacfofoohH1.2memefo fo fomemeDac me me meoh fo memeSGRGKQGGKAR...GNYSER…HLQLAIR…KKTESHHK…...K…KKGSKKAVTKAQK…K..Y..K..K..R…YNK…R..K…KAVTK…515120ac ac2023me57851081169983379711H2Bmefoacmeme me me memefofoohfo1216acac acme3446fo fo oh591254288391118H2A119me meme meme2me32ememme fomefofomeacX: Novel sitesX: Known sitesSGRGK…K..K...KRHRK…K..R…YEETR…K..R…KRK…YALK…ac58121620ac ac ac me19779513me5567518853H4me me me meme2fofo oh oxmefofo79acfome4914182736566379ac ac82122132acacme2ARTKQTARK…K…K…KAARK…K..K..R..K...KDIQLR…H3me meacac me2acE Kcr: lysine crotonylation Kac: lysine acetylationH2A Human: NH2-SGRG K QGG K ARA K A K TR………LLR K GNY………AVLLP KK TESHH K AKGK-COOHKcr KcrKcr Kcr52191181163513195Human:Mouse:Kcr KcrH1.2 Human: NH2-SET…PR K ASGP…ALK K AL…GL K SLVS K GTLVQT K G……KA K K…ATVT K KVA………KK-COOH33167Kcr89r cKr cK96KcrHuman:Mouse:63Kcr84r cKr cK158Kcr Kcrr cKr cKr cKy y y yKac Kac Kac Kac KacH2B Human: NH2-PEPA K SAPAP KK GS KK AVT K AQ K K………RSR K ESYSI………TK YTSSK-COOH51215342024Kac Kac Kac Kac Kacr cKr cK111623KcrKcr Kcr Kcr KcrHuman:Mouse:Kcr KcrKcrKcr Kcr Kcr KcrKcrKcrH3 Human: NH2-ART K QTAR K STGG K APR K QLAT K AAR K S……RYQ K ST…RIRGERA-COOHKcrKcr KcrKcrKcrHuman:Mouse:KcrKcrKcr KcrKcrKcr KcrH4 Human: NH2-SGRG K GG K GLG K GGA K RHR K VLRDNIQG………TLYGFGG-COOHKac Kac Kac KacKcrKac58121620Kcr KcrHuman:Mouse:Kcr r c Kr cK23Kac Kac Kac Kac2718149Kac654KacKacFigure1.Experimental Strategy and Results for Identified Histone PTM Sites(A)Schematic diagram of the experimental design for comprehensive mapping of PTM sites in linker and core histones from HeLa cells.Histone extracts were in-solution trypticly digested without chemical propionylation(Method I),chemically propionylated after in-solution tryptic digestion(Method II),chemically propionylated before in-solution tryptic digestion(Method III),and in-gel digested after SDS-PAGE gel separation.Samples from Methods I and II were further subjected to IEF fractionation to generate12fractions.(B)Peptide sequence coverage of linker and core histones in each of the four methods is shown.(C)A table summarizing all the PTM sites identified by this study.Abbreviations:me,monomethylation;me2,dimethylation;me3,trimethylation;fo,formylation; ac,acetylation;oh,hydroxylation;and cr,crotonylation.(D)A diagram showing sites of histone PTMs other than Kcr identified in this study.Amino acid residue number is indicated below its sequence.Gray and blank boxes indicate N-terminal and globular core domains,respectively.1018Cell146,1016–1028,September16,2011ª2011Elsevier Inc.To determine the nature of this modification,we selected one of these peptides,PEPAK +68SAPAPK (modified at H2BK5),for further analysis.After manual inspection of the high-resolu-tion MS data (precursor ion mass at m/z 580.8181),we deter-mined the accurate mass shift of this modification was +68.0230Da.By setting the mass tolerance to ±0.01Da ($9ppm,which is within the mass accuracy of our mass spec-trometer),and specifying a maximum of 2nitrogen atoms,we were able to deduce the possible element compositions of the modification group as either C 4H 4O or H 6NO 3.The former,C 4H 5O (mass shift plus one proton),is the only reasonable molecular formula of this modification.There were 4possible structures consistent with the element composition:Kcr (Figures 2A and 2B),vinylacetyllysine (3-butenoyllysine),methacryllysine,and cyclopropanecarboxyllysine (Figure S2A).As crotonyl-CoA is an important and abundant intermediate (Figure 2C)in meta-bolic pathways of butyryl-CoA and acetyl-CoA,we focused on Kcr as the PTM candidate most likely to cause the mass shift.To test if the identified mass shift of +68.0230Da was caused by Kcr,we synthesized the Kcr peptide,PEPAK cr SAPAPK,and compared its MS/MS spectrum with that of the in vivo-derived peptide.The in vivo modified peptide bearing a lysine residue(E)Illustrations of histone Kcr sites in human HeLa cells and mouse MEF cells.All Kcr sites are shown in red and underlined.Previously reported Kac sites are shown in blue.See also Figure S1and Supplemental Information 1,Supplemental Information 2,Supplemental Information 3,and Supplemental Information 4.A+ X-CoAX = acetyl, crotonylCrotonyllysineLysineBAcetyllysineen i s y l l y t e c A e n i s y l l y n o t o r C CMultiple stepsFigure 2.Short-Chain Lysine Acylations(A)An illustration of the enzymatic reactions for lysine acetylation by lysine acetyltransferases (KATs)using acetyl-CoA as a cofactor,and a hypothesized mechanism for Kcr using crotonyl-CoA as a cofactor.(B)Ball-and-stick models of a crotonyl group and an acetyl group.The three-dimensional arrangement of four carbons and one oxygen of the crotonyl group are rigid and located in the same plane (left).The two olefinic carbons of the crotonyl group are shown in yellow.In contrast,the tetrahedral CH 3in the acetyl group (right)can be rotated such that it is structurally very different from the crotonyl group.(C)Crotonyl-CoA metabolism pathways.Crotonyl-CoA was generated from butyryl-CoA or glutaryl-CoA,and oxidized to acetyl-CoA through multiple steps.Cell 146,1016–1028,September 16,2011ª2011Elsevier Inc.1019with a mass shift of +68.0230Da,the synthetic Kcr peptide with the same peptide sequence (PEPAK cr SAPAPK),and the mixture of the two peptides exhibited almost identical parent massesand high-resolution MS/MS spectra (Figures 3A–3C).In addition,the mixture of the in vivo and synthetic peptides coeluted in HPLC/MS analysis (Figure 3D).These results indicated thatABm/zC408061.82In Vivo408061.12Synthetic4080Retention Time (min)60.08MixtureDFigure 3.Identification and Verification of a Kcr Peptide,PEPAK cr SAPAPKKcr indicates a crotonyllysine residue.(A–C)High-resolution MS/MS spectrum of a tryptic peptide,PEPAKSAPAPK,with a mass of +68.0230Da at its Lys5residue identified from in vivo histone H2B (A),its synthetic Kcr counterpart (B),and a peptide mixture of the in vivo-derived tryptic peptide and its synthetic counterpart (C),each showing the same MS/MS fragmentation patterns and the same precursor ion mass.Inset shows their precursor ion masses.(D)Extracted ion chromatograms (XICs)of the in vivo-derived PEPAK +68.0230SAPAPK peptide,the synthetic Kcr counterpart,and their mixture by nano-HPLC/MS/MS analysis using a reversed-phase HPLC column,showing the coelution of the two peptides.1020Cell 146,1016–1028,September 16,2011ª2011Elsevier Inc.the identified mass shift of +68.0230Da was very likely caused by Kcr.To further confirm Kcr in histones,we generated a pan anti-body against Kcr.This pan anti-Kcr antibody specifically recog-nized a peptide library bearing Kcr,but not four other peptide libraries in which the fixed lysine residue was unmodified,acet-ylated,propionylated,or butyrylated (Figure 4A);the specificity of pan anti-Kac antibody was confirmed likewise (Figure S2B).The specificity of the pan anti-Kcr antibody was further demon-strated by western blotting with three different bovine serum albumin (BSA)derivatives,where peptide lysines were chemi-cally modified by a crotonyl,vinylacetyl,or methacryl group,respectively.The result showed that pan anti-Kcr antibody recognized only the lysine crotonylated BSA,but not the unmod-ified,lysine vinylacetylated or lysine methacrylated BSA (Fig-ure S2C).This pan anti-Kcr antibody was subsequently used for western blotting and immunostaining of Kcr signal.The antibody detected a Kcr signal among all core histone proteins:H2A,H2B,H3,H4,and linker histone H1.In each pro-tein,the signal could be efficiently competed away by a peptide library bearing a Kcr,but not peptide libraries bearing an unmod-ified lysine (Figure 4B),metharcryllysine (Figure S2D),acetylly-sine,propionyllysine,or butyryllysine (Figure S2E).The strong Kcr signals detected in histones indicated that this modificationKKacKprKbuKcrPeptideA H1CompetitionK KcrWB: anti-KcrCB1 ng 5 ng25 ngCore HistonesH1Core HistonesBlue stainingCrotonate (mM) 0 50 100WB: anti-Kcr Blue stainingDc K K rEm/zCompetition Blue staining WB: anti-KcrBlue stainingFigure 4.Detection of Kcr in Histones by Western Blotting.(A)Specificity of pan anti-Kcr antibody demonstrated by dot-spot assay using five peptide libraries with indicated amount (ng).Each peptide library contains 13residues CXXXXXKXXXXXX,where X is a mixture of 19amino acids (excluding cysteine),C is cysteine,and the 7th residue is a fixed lysine residue:unmodified lysine (K),Kac,propionyllysine (Kpr),butyryllysine (Kbu),and Kcr.(B)Detection of Kcr in histones.Western blotting was carried out using the histones from HeLa cells with competition of a peptide library bearing a fixed unmodified lysine (K)or Kcr.(C)Dynamics of histone Kcr in response to crotonate.The histone proteins extracted from human prostate cancer cell line Du145incubated with 0,50,or 100mM crotonate for 24hr,were western blotted with anti-Kcr pan antibody.(D)MS/MS spectrum of PEPA K D4-cr SAPAPK identified from D 4-crotonate-labeled sample.The mixture of D 4-,D 3-and D 2-crotonyl groups was used for the identification of D 4-crotonyl peptide.(E)Kcr signals in core histones of S.cerevisiae ,C.elegans ,D.melanogaster (S2),M.musculus (MEF),as well as H.sapiens (HeLa)cells by western blotting analysis with competition.See also Figure S2.Cell 146,1016–1028,September 16,2011ª2011Elsevier Inc.1021is present in nuclei and is associated with chromosomes. Indeed,immunostaining using the pan antibody showed that Kcr mainly existed in nuclei(Figure S2F;see also the analysis below of spermatogenic cells).Isotopic labeling is an established method to confirm in vivo protein modifications that was previously used for the study of histone Kac(Allis et al.,1985).We rationalized that D4-crotonate can be converted into crotonyl-CoA in vivo,which then functions as a lysine crotonylation donor.Consistent with this hypothesis, the histone Kcr signal from HeLa cells increased dramatically after cells were cultured with crotonate(Figure4C).After tryptic digestion of histones from D4-crotonate labeled HeLa cells,we carried out peptide immunoprecipitation using the anti-Kcr antibody.The enriched Kcr peptides were then subjected to HPLC/MS/MS analysis and protein sequence alignment,which confirmed Kcr on histone H2BK5by D4-crotonate labeling(Fig-ure4D).(Note:The D4-crotonic acid was mixed with D3-and D2-crotonic acid(Figure S2G).This characteristic isotopic distri-bution was used for the identification of D4-crotonyl peptide.) Histone Kcr Sites in Human CellsTo identify histone Kcr sites,we used a Mascot sequence align-ment to analyze MS/MS data derived from HeLa histones,using Kcr(+68.02621Da)as a variable modification.These analyses led to the identification of28Kcr sites in human histones(Fig-ure1E).In addition,19of these28identified sites were confirmed by in vivo D4-crotonate labeling experiments.The Kcr peptides and their corresponding MS/MS spectra are included in Supple-mental Information1and Supplemental Information2.In summary,we have usedfive independent methods–MS/ MS and HPLC coelution of synthetic peptides,D4-crotonate labeling,western blotting,and immunostaining–to show that histone Kcr exists in cells,and identified28lysines on various histones that are subject to this PTM.Histone Kcr Is an Evolutionarily Conserved Histone PTM To test if lysine crotonylation is present in histones from other eukaryotic cells,we isolated histones from yeast S.cerevisiae, C.elegans,Drosophila S2cells,mouse embryonicfibroblast (MEF)cells,as well as human HeLa cells.Kcr signals were de-tected among core histones from allfive species by western blotting(Figure4E).Taking advantage of affinity enrichment using the pan anti-Kcr antibody and HPLC/MS/MS,we identified 24Kcr sites on mouse MEF cells(Figure1E).All of the annotated MS/MS spectra for Kcr mouse histone peptides are included in Supplemental Information3.Taken together,our results revealed that Kcr is an evolutionarily conserved histone mark appearing in eukaryotic cells from a wide range of species. Histone Kcr and Kac Are Mechanisticallyand Functionally DifferentIn order to examine whether histone Kcr is derived from promis-cuous histone acetyltransferase(HAT)activity,we overex-pressed two HATs,CBP and p300,in293T cells and tested for histone crotonylation levels by western blotting.We found that overexpression of CBP or p300led to the enhancement of histone Kac but did not significantly change the levels of histone Kcr(Figure S2H).Using afluorometric assay,we also demonstrated that histone lysine deacetylases(HDACs)1,2,3,and6exhibit potent lysine deacetylation activities,but have very weak or no effect on lysine decrotonylation.For example,lysine decrotonylation activity was not detected with HDAC6exposure,though the enzyme’s deacetylation activity was very strong in this assay(Figure S2I). These results suggest that enzymes required for the addition and removal of Kcr on histone proteins may be different from those for histone Kac regulation.Genome-wide Mapping of Histone Kcr in Human CellsIn order to explore the in vivo function of histone Kcr,we per-formed ChIP-seq analysis with the pan anti-Kcr antibody to determine the genomic distribution of histones with this modifi-cation in the human fetal lungfibroblast IMR90cell line.As a control,we compared the Kcr distribution with previously ob-tained ChIP-seq results for H3K4me3(marking gene promoters) and H3K4me1(marking enhancers)distribution(Heintzman et al.,2007).Strikingly,we found an abundance(totaling84,435peaks)of this histone modification in the human genome.Histone Kcr was largely associated with active chromatin,including both the transcription starting site(TSS)and regions previously predicted to be enhancers(Hawkins et al.,2010)(Figures5A and5B).The majority(68%)of histone Kcr peaks was associated with either promoter or predicted enhancer regions(Figure5B). At promoters,histone Kcr showed the strongest enrichment flanking TSS,in contrast to the observation that H3K4me3was more enriched downstream of TSS(Figure5C).At predicted enhancers,we observed a strong enrichment of Kcr in agree-ment with the H3K4me1pattern at these sites(Figure5D). Furthermore,we also observed a strong correlation between gene expression and Kcr level at promoters(Figure5E).Taken together,these data strongly support a role for histone Kcr in gene regulation,especially at promoters and enhancers.In addition,we also compared Kcr sites with histone lysine acetylation sites revealed by ChIP-seq assay using a pan anti-Kac antibody.The result showed that histone crotonylation generally occupies similar locations as acetylation in IMR90cells as there is a significant overlap between Kac and Kcr peaks(Fig-ure S3A).These results suggested that in resting somatic cells, open chromatins are simultaneously marked by both histone Kac and Kcr.Given thefinding that these two marks are cata-lyzed by different sets of enzymes,we assumed that the regula-tion of Kcr would likely have different spatial and temporal dynamics from Kac.To test this prediction,we explored the function of Kcr in the highly dynamic spermatogenesis process. Histone Kcr Marks Testis-Specific Genes Activatedin Postmeiotic CellsWe next investigated the genomic distribution of histone Kcr in spermatogenic cells,where a very specific gene expression pro-gram directs key steps of differentiation(Boussouar et al.,2008; Gaucher et al.,2010).Wefirst examined the global dynamics of Kcr during mouse spermatogenesis using immunohistochem-istry.We observed an intense labeling of Kcr during elongating steps9–11in spermatids(Figure6A),which coincides with a1022Cell146,1016–1028,September16,2011ª2011Elsevier Inc.。

相关文档
最新文档