HIF-1α Induces Genetic Instability by Transcriptionally Downregulating MutSα Expression
缺氧诱导因子(HIF-1α)与肿瘤病理的关系

缺氧诱导因子(HIF-1α)与肿瘤病理的关系
李锁严;其木格;刘晓辉;李蕾;李占勇
【期刊名称】《赤峰学院学报(自然科学版)》
【年(卷),期】2009(025)002
【摘要】缺氧是实体瘤生长中存在的普遍现象,它和肿瘤的发展、浸润和转移密切相关.缺氧诱导因子(hypoxia-inducible faftor-1,HIF-1)是缺氧条件下广泛存在于哺乳动物及人体的一种转录因子,HIF-1是由α亚基和β亚基组成的异源二聚体,HIF-1α代表HIF-1的活性,其表达与肿瘤增殖、侵袭和转移、肿瘤新生血管生成等特征密切相关.HIF-1α对临床的诊断和对癌症治疗手段和新药物的研发有着重要的应用价值.
【总页数】2页(P61-62)
【作者】李锁严;其木格;刘晓辉;李蕾;李占勇
【作者单位】赤峰学院,医学院,内蒙古赤峰024000;赤峰学院,医学院,内蒙古赤峰024000;赤峰学院,医学院,内蒙古赤峰024000;赤峰学院,医学院,内蒙古赤峰024000;赤峰学院,医学院,内蒙古赤峰024000
【正文语种】中文
【中图分类】R73
【相关文献】
1.缺氧诱导因子HIF-1α在食管癌中的表达和微血管密度MVD的关系 [J], 王永占
2.HIF-1α和VEGF与肿瘤病理的关系 [J], 刘诚;唐维平
3.缺氧诱导因子1α(HIF-1α)与脑肿瘤研究进展 [J], 丁新民;李旭光;程高鹏
4.缺氧诱导因子(HIF-1α)与肿瘤病理的关系 [J], 李锁严;其木格;刘晓辉;李蕾;李占勇;
5.缺氧诱导因子1α(HIF-1α)及其在脑肿瘤中表达 [J], 高利民
因版权原因,仅展示原文概要,查看原文内容请购买。
HIF-1α基因沉默对人脉络膜黑色素瘤细胞上皮—间质转化的影响的开题报告

HIF-1α基因沉默对人脉络膜黑色素瘤细胞上皮—间质转化的影响的开题报告背景:脉络膜黑色素瘤(Choroidal melanoma,CM)是一种特殊的黑色素瘤,其起源于视网膜和脉络膜的黑素细胞。
CM是一种潜在恶性肿瘤,可能会导致眼球和视觉的丧失,以及全身的转移,其预后较差。
因此,深入了解CM的发病机制和治疗方法尤为重要。
研究表明,上皮—间质转化(Epithelial-mesenchymal transition,EMT)在肿瘤的发生和发展中扮演着重要角色。
HIF-1α是EMT的一个关键调节因子,在肿瘤的侵袭和转移中起着重要作用。
HIF-1α的高表达可以促进EMT,并增强肿瘤细胞的浸润和转移能力。
因此,沉默HIF-1α基因可能有助于抑制CM的发生和发展。
研究目的:本研究旨在探究HIF-1α基因沉默对人脉络膜黑色素瘤细胞上皮—间质转化的影响,为CM的治疗提供新的理论和实践依据。
研究方法:1.实验材料:人脉络膜黑色素瘤细胞(如MM28细胞),siRNA、质粒、脂质体转染试剂、Western blot试剂盒等。
2.实验组分:对照组、转染siRNA的实验组、空白质粒转染的对照组。
3.实验步骤:①分别将MM28细胞转染siRNA、空白质粒和对照组,按组织培养条件培养48小时。
②分别通过Western blot检测HIF-1α、E-cadherin和N-cadherin 的表达程度。
4.数据分析:采用SPSS19.0统计软件对实验数据进行统计学分析,利用单因素方差分析进行组间比较,并进行人口统计学描述,探究HIF-1α基因沉默对人脉络膜黑色素瘤细胞上皮—间质转化的影响。
预期成果:本研究将揭示HIF-1α在EMT调节中的作用,为CM的治疗提供理论和实践依据,为深入研究CM的发生和发展机制提供新思路和研究角度。
HIF-1—低氧与固有免疫的联系者

HIF-1—低氧与固有免疫的联系者沙继斌【摘要】近年来研究发现在低氧与固有免疫这两个古老、保守的应激反应之间存在密切联系.尤其是低氧应答的核心元件低氧诱导因子l(hypoxia inducible factor 1,HIFl)与机体固有免疫的重要调节者核转录因子κB(nuclear transcription factor,NF-κB)之间存在相互调节,并可影响彼此下游产物的表达.就该方面研究进展做一综述,以期为运动性免疫抑制的机理研究提供新的依据.%Recent studies showed that there may be a hardwire or softwire link between these two ancient and conservative process,hypoxia and innate immunity.The relationship between the core hypoxia response element HIF1 (hypoxia inducible factor 1) and NF-κB (Nuclear Transcription facto r) was focused.They regulate each other' s function concomitant with downstream effects.It might provide a new insight for exercise-induced immunosuppression.【期刊名称】《生物学杂志》【年(卷),期】2013(030)003【总页数】4页(P77-80)【关键词】固有免疫;低氧诱导因子1;核转录因子κB【作者】沙继斌【作者单位】山东体育学院基础理论系,济南250102;西安交通大学生命科学与技术学院,西安710049【正文语种】中文【中图分类】R392.1人体是一个多种调节机制与调节网络并存的复杂生物体,研究已经证实运动应激越强烈,则细胞所生成的活性氧数目越多[1]。
HIF-1α在成人急性淋巴细胞白血病患者骨髓单个核细胞中的表达及意义

HIF-1α在成人急性淋巴细胞白血病患者骨髓单个核细胞中的表达及意义云清英;徐刚【期刊名称】《中国现代医学杂志》【年(卷),期】2017(027)021【摘要】Objective To investigate the expression and significance of hypoxia-inducible factor-1α (HIF-1α) in bone marrow mononuclear cells of adult patients with acute lymphoblastic leukemia (ALL).Methods Seventy-three adult patients with newly-diagnosed ALL were selected.In the same period,30 samples of bone marrow from non-leukemic patients were selected as the control group.Bone marrow mononuclear cells were harvested and isolated.The expression of HIF-1α gene in bone marrow mononuclear cell was detected by qRT-PCR.The effect of the expression of HIF-1α gene on the relapse-free survival time was analyzed by Kaplan-Meier.Results The relative expression level of HIF-1α m RNA in bone marrow mononuclear ceils of the adult patients with newly diagnosed ALL was (1.86 ± 0.18),which was significantly higher than that of the control group (1.17 ± 0.09),and the difference was statistically significant (t=25.020,P=0.000).The relative expression levels of HIF-1α mRNA in bone marrow mononuclear cells of the patients with B-ALL and T-ALL were higher than those of the control group (P < 0.05),while there was no significant difference between those with B-ALL and T-ALL(t=0.807,P=0.211).The relative expression levels of HIF-1α mRNA in bone marrow mnnonuclear cells of Philadelphia chromosome (Ph)-positive and Ph-negative patients were higher than those of the control group (P<0.05),while there was no significant difference between Ph-positive group and Ph-negative group (t=1.314,P=0.096).The relative expression levels of HIF-1α mRNA in bone marrow mononuclear cells of the B-ALL and T-ALL patients were unrelated with sex,age,leukocyte level or risk grade (P>0.05).According to the cutoff value of P25 of the relative expression level of HIF-1α mRNA,the patients were divided into the low expression group (n =18) and the high expression group (n =55).During the follow-up period,4 cases in the low expression group had recurrence,the recurrence rate was 22.2%;20 cases in the high expression group had recrudescence,the recurrence rate was 36.4%.Kaplan-Meier survival analysis showed that the relapse-free survival time in the low expression group was 11.2 months,which was longer than that of the high expression group (8.6 months),but the difference was not statistically significant(x2=1.771,P=0.183).Conclusions HIF-1α is highly expressed in bone marrow mononuclear cells of adult patients with newly diagnosed ALL.It may be associated with chemotherapy resistance and recurrence.%目的探讨低氧诱导因子-1α(HIF-1α)在成人急性淋巴细胞白血病(ALL)患者骨髓单个核细胞中的表达及意义.方法选取73例成人初诊ALL患者,同期留取非白血病骨髓30份作为对照组,采集并分离骨髓单个核细胞,利用实时荧光定量聚合酶链反应技术检测骨髓单个核细胞中HIF-1α基因表达,利用KaplanMeier生存分析分析HIF-1 α基因表达对患者无复发生存时间的影响.结果成人初诊ALL患者骨髓单个核细胞中HIF-1αmRNA相对表达量(1.86±0.18),高于对照组(1.17±0.09),差异有统计学意义(=25.020,P=0.000);B系-ALL(B-ALL)和T系-ALL(T-ALL)患者骨髓单个核细胞中HIF-1αmRNA相对表达量均高于对照组(P<0 05),而B-ALL和T-ALL患者之间差异无统计学意义(t=0.807,P=0.211);费城染色体(Ph)阳性和Ph阴性患者HIF-1 αmRNA相对表达量均高于对照组(P<0.05),而Ph阳性和Ph阴性患者之间差异无统计学意义(t=1.314,P=0.096);B-ALL和T-ALL成人初诊ALL患者骨髓单个核细胞中HIF-1 α信使核糖核酸(rnRNA)相对表达量均与性别、年龄、白细胞水平和危险度分级无关(P>0.05);以HIF-1 αmRNA相对表达量的P25为截点,将患者分为低表达组(n=18)和高表达组(n=55),低表达组随访中复发4例,复发率22.2%,高表达组随访中复发20例,复发率36.4%,Kaplan-Meier生存分析显示,低表达组无复发生存时间11.2个月,长于高表达组的8.6个月,但差异无统计学意义(X2=1.771,P=0.183).结论 HIF-1α在成人初诊ALL患者骨髓单个核细胞中呈高表达,可能与ALL患者化疗耐药及复发相关.【总页数】5页(P49-53)【作者】云清英;徐刚【作者单位】山东中医药大学第二附属医院检验科,山东济南250001;中国医科大学第四医院,辽宁沈阳110000【正文语种】中文【中图分类】R733.71【相关文献】1.β-catenin和MMP-7在成人急性淋巴细胞白血病中的表达及意义 [J], 王石松2.CDX2基因在成人急性淋巴细胞白血病中的表达及临床意义 [J], 申淑珍;白晓川;刘娟3.成人急性淋巴细胞白血病患者p73基因异常表达及临床意义 [J], 吴朝阳;金洁;季勇慧;钱震;杨小飞;盛晓静;张永宁;钱军4.宫颈鳞癌患者组织中HIF-1α和VEGF的表达及在放疗中的意义 [J], 潘秀花; 白力; 谢福川; 江晓聪; 蓝玉宏; 肖伟5.GLUT3在成人急性淋巴细胞白血病中的表达及意义 [J], 朱鸿燕;阳梅;何川因版权原因,仅展示原文概要,查看原文内容请购买。
《HIF-1α介导DNA甲基化调控S100B转录促进肝癌细胞生长和转移机制研究》范文

《HIF-1α介导DNA甲基化调控S100B转录促进肝癌细胞生长和转移机制研究》篇一一、引言肝癌是全球范围内高发的恶性肿瘤之一,其发病机制复杂且尚未完全明确。
近年来,越来越多的研究表明,缺氧诱导因子-1α(HIF-1α)在肝癌的发生、发展过程中起着关键作用。
本文旨在探讨HIF-1α如何通过介导DNA甲基化调控S100B转录,进而促进肝癌细胞生长和转移的机制。
二、HIF-1α与肝癌HIF-1α是一种在缺氧环境下表达的蛋白质,能够调节细胞对缺氧环境的适应。
在肝癌组织中,HIF-1α的表达往往升高,与肿瘤的恶性程度和患者的预后密切相关。
HIF-1α通过调控多种基因的表达,参与肝癌细胞的增殖、侵袭和转移等过程。
三、DNA甲基化与基因表达DNA甲基化是一种重要的表观遗传学修饰方式,能够影响基因的表达。
在肝癌中,DNA甲基化的变化与肿瘤的发生、发展密切相关。
甲基化程度的变化能够调控肿瘤相关基因的转录活性,进而影响肿瘤细胞的生物学行为。
四、HIF-1α介导的DNA甲基化调控S100B转录S100B是一种钙结合蛋白,在多种肿瘤中表达异常。
研究发现,HIF-1α能够通过介导DNA甲基化来调控S100B的转录。
具体来说,HIF-1α与DNA甲基转移酶相互作用,导致S100B基因启动子区域的甲基化程度增加,从而抑制S100B的转录。
这一过程可能进一步促进了肝癌细胞的生长和转移。
五、S100B与肝癌细胞生长和转移S100B在肝癌组织中的表达与肝癌细胞的生长和转移密切相关。
一方面,S100B能够促进肝癌细胞的增殖和侵袭;另一方面,S100B还能够影响肿瘤微环境,促进肿瘤血管生成和免疫逃逸。
因此,S100B的异常表达可能加速肝癌的进展。
六、HIF-1α调控S100B转录促进肝癌细胞生长和转移的机制HIF-1α通过介导DNA甲基化调控S100B的转录,进一步促进肝癌细胞生长和转移的机制如下:首先,HIF-1α在缺氧环境下表达增加,并与DNA甲基转移酶相互作用;其次,这种相互作用导致S100B基因启动子区域甲基化程度增加,从而抑制S100B 的转录;最后,S100B的表达降低进一步促进了肝癌细胞的生长和转移。
HIF-1与疾病相关性研究进展

Research Progress on the Relationship between HIF-1 and Diseases
XU Lei, YANG Guang-lu
HIF-1 具 有 两 个 亚 基,HIF-1α(120-kDa)和 HIF-1β(9194-kDa),均为 Per-Arnt-Sim(PAS)家系的碱性螺旋 - 环 - 螺旋 (bHLH)蛋白 [1]。HIF-1α 和 HIF-1β 蛋白含有异二聚体形成和反 式激活结构域(TAD)所需的 bHLH 和 PAS 基序,这是结合转录共 激活因子和刺激转录所必需的 [2]。HIF-1α 是缺氧信号的主要调 节因子,也是表达最普遍的。HIF-1α 和 HIF-1β 蛋白识别并结合 共有 DNA 序列,在缺氧反应基因的调节启动子区域内,称为缺氧 反应元件(HRE)。在正常氧浓度(常氧)存在下,HIF-1β 相对稳 定,而 HIF-1α 与 VHL 肿瘤抑制蛋白结合,导致 HIF-1α 的泛素化 和快速降解 。 [2-3] 然而在缺氧情况下,HIF-1α 会ห้องสมุดไป่ตู้积并与 HIF1β 结合,从而形成能够启动转录的活性 HIF 复合物 。 [2,4] 在细胞 核内,HIF-1 复合物与缺氧反应基因的启动子区域上的缺氧反应 元件(HRE)结合,募集其他转录因子,如 P-CREB 和 P-STAT3, 启动并诱导这些基因的转录,进而引发组织细胞的一系列耐氧适 应性反应,缺氧反应基因包括 VEGF,促红细胞生成素(EPO),葡 萄糖转运蛋白和糖酵解没,细胞存活因子,细胞表面受体,细胞外 基质蛋白和转录因子的基因 。 [2,3,5,6,7]
低氧诱导因子hif-1α 适应 能量代谢 文献解读

低氧诱导因子HIF-1α在人体内是一种重要的生物活性蛋白质,它在缺氧情况下对能量代谢起着至关重要的调控作用。
本文将围绕HIF-1α在能量代谢中的作用进行文献解读,以期全面了解该蛋白对人体功能的影响。
1. HIF-1α的基本介绍HIF-1α是一种由基因HIF1A编码的蛋白质,其编码基因位于人类染色体14q23.2-q24.1上,由该基因转录、翻译得到的蛋白质主要分布在细胞的细胞质内。
HIF-1α的主要功能是在细胞缺氧时,通过调节多种基因的表达,以适应低氧环境。
其中,其对能量代谢的调控作用备受研究者的关注。
2. HIF-1α与能量代谢研究表明,HIF-1α在细胞缺氧时能够促进糖酵解途径的进行,增加葡萄糖转化为丙酮酸和乳酸的速率,从而增加ATP的产生。
HIF-1α还可以抑制线粒体的功能,减少线粒体呼吸链的活性,从而减少氧化磷酸化的过程,进一步节约细胞内氧气的利用。
通过这些方式,HIF-1α能够在细胞缺氧时维持细胞内的能量供应,保障细胞正常的生理功能。
3. HIF-1α与疾病的关系近年来的研究发现,HIF-1α在多种疾病的发生发展中发挥着重要的作用。
在肿瘤的发生过程中,肿瘤组织由于生长速度快、造血不足等原因,常常处于低氧状态,HIF-1α的异常活化对肿瘤的代谢、侵袭和转移等过程起着重要的调节作用。
另外,在心脏缺血再灌注损伤、糖尿病等多种疾病中,HIF-1α的异常表达也与疾病的发生发展密切相关。
4. HIF-1α的研究进展目前,针对HIF-1α的研究已经取得了许多重要的进展。
通过基因工程技术,研究者可以对HIF-1α基因进行敲除或过表达,从而揭示了该基因在细胞能量代谢中的重要作用。
另外,一些研究还发现了HIF-1α的调控机制,比如HIF-1α的翻译后修饰、HIF-1α与其他蛋白的相互作用等,这为进一步揭示HIF-1α的功能机制打下了重要的基础。
5. 未来的研究方向虽然HIF-1α在能量代谢中的作用已经得到了一定程度的解析,但其在细胞生理和病理过程中的复杂调控机制仍有待进一步研究。
HIF-1在肿瘤细胞中的研究进展

HIF-1在肿瘤细胞中的研究进展【中图分类号】R418【文献标识码】B【文章编号】1005-0515(2011)02-0038-01肿瘤最主要的特征是肿瘤细胞的失控性生长,不断增加的细胞数及高代谢状态将导致绝大多数肿瘤耗氧量的增加,造成肿瘤内缺氧微环境的形成,且在实体瘤中表现更明显。
肿瘤细胞适应缺氧的策略,一是提高糖酵解的速率,二是形成多血管体系,在肿瘤发生发展过程中,肿瘤增殖速度超过血管生成速度就会造成局部组织缺氧,肿瘤组织内血管结构及功能的异常,肿瘤细胞血供减少以及快速增殖导致肿瘤细胞耗氧量增加。
肿瘤组织的缺氧在肿瘤病理过程中异常重要,缺氧与肿瘤细胞的生长、分化、浸润、转移等生物学行为都有关系,而且导致肿瘤细胞对放疗、化疗耐受抵抗及治疗的失败[1]。
诱导因子-1( Hypoxia-inducible factor-1,HIF-1)HIF-1是在缺氧条件下存在于哺乳动物和人体内的一种转录因子,是由Semeza和Wang于1992年在低氧的肝癌细胞株Hep3B细胞的核提取物中发现的一种蛋白特异性结合于红细胞生成素基因增强子的寡核苷酸序列。
缺氧诱导因子-1作为细胞平衡和缺氧诱导基因表达的中心调节因子,可调控一系列靶基因(如VEGF,GLUT1,GLUT3)的转录,在肿瘤的增殖、转移以及发生、发展中起着重要作用。
它不仅在肿瘤细胞及其转移细胞中过度表达,而且能诱导肿瘤组织中异常基因的表达,同时对肿瘤细胞的生长、肿瘤新生血管形成密切相关。
1HIF-1a功能1.1促进红细胞生成。
肿瘤乏氧条件下HIF-1表达增加,诱导促红细胞生成素(erythropoiesis,EPO)受体表达增加。
EPO在许多组织及细胞包括红细胞、肿瘤细胞等存在,是促红细胞生成的刺激因子。
HIF-1诱导EPO表达增加可促进红细胞生成,增加血液氧的运输,减轻肿瘤组织缺氧,增强肿瘤细胞的适应性[2]。
1.2促进肿瘤血管形成及能量代谢。
HIF-1的目的基因中包含有许多与葡萄糖代谢和糖酵解相关的酶,缺氧状态下,肿瘤细胞通过HIF-1上调这些酶的表达,使细胞适应缺氧状态。
- 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、"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、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)。
Molecular Cell,Vol.17,793–803,March18,2005,Copyright©2005by Elsevier Inc.DOI10.1016/j.molcel.2005.02.015HIF-1␣Induces Genetic Instabilityby Transcriptionally DownregulatingMutS␣ExpressionMinori Koshiji,1Kenneth K.-W.To,1pressor p53or the apoptosis inhibitor Bcl2substantiallyreduced cell death(Graeber et al.,1996).Hypoxia also Stefanie Hammer,1Kensuke Kumamoto,1induces gene amplifications(Rice et al.,1986)by driv-Adrian L.Harris,2Paul Modrich,3,4ing fusion and subsequent reintegration of double min-and L.Eric Huang1,*1Laboratory of Human Carcinogenesisutes into chromosomal fragile sites,generating homo-geneously stained regions(Coquelle et al.,1998). National Cancer InstituteMoreover,hypoxia enriches DNA mismatch repair-defi-National Institutes of Healthcient cells,thereby augmenting microsatellite instability Bethesda,Maryland20892(MIN)in human colon carcinomas(Kondo et al.,2001). 2Molecular Oncology LaboratoryGenetic instability is regarded as a hallmark of most Weatherall Institute for Molecular Medicinehuman cancers.The resulting genetic changes include John Radcliffe Hospitalthose at the nucleotide level(such as base substitu-Cancer Research UKtions and deletions or insertions of a few nucleotides) Oxford OX39DSand those at the chromosomal level involving chromo-United Kingdomsome loss or gain,translocations,and rearrangements 3Howard Hughes Medical Institute(Lengauer et al.,1998).Most human tumors are geneti-Duke University Medical Centercally heterogeneous,presumably resulting from a mu-Durham,North Carolina27710tator phenotype early during their evolution(Loeb et al., 4Department of Biochemistry2003).The molecular mechanisms underlying these ge-Duke Universitynetic alterations have been the subject of continued in-Durham,North Carolina27710vestigation.The human DNA mismatch repair(MMR)systemplays a crucial role in safeguarding genomic integrityby correcting DNA replication errors and by blocking Summaryrecombination events between divergent DNA se-quences(Kolodner,1996;Modrich and Lahue,1996). Hypoxia promotes genetic instability by undefinedMMR has also been implicated in the cellular response mechanisms.The transcription factor HIF-1␣is crucialto certain classes of DNA damage(Modrich and Lahue, for the cellular response to hypoxia and is frequently1996;Li,1999)and may have some function in nucleo-overexpressed in human cancers,resulting in thetide excision repair(Kolodner,1996).Mammalian MMR activation of genes essential for cell survival.Here,proteins include homologs of Escherichia coli MutS we demonstrate that HIF-1␣is responsible for genetic(MSH2,MSH3,and MSH6)and MutL(MLH1and PMS2). instability at the nucleotide level by inhibiting MSH2They form MutSα(MSH2-MSH6),MutSβ(MSH2-MSH3), and MSH6,thereby decreasing levels of the MSH2-and MutLα(MLH1-PMS2)heterodimers(Modrich and MSH6complex,MutS␣,which recognizes base mis-Lahue,1996;Buermeyer et al.,1999;Kolodner and Mar-matches.HIF-1␣displaces the transcriptional ac-sischky,1999).Whereas the MutSαheterodimer recog-tivator Myc from Sp1binding to repress MutS␣nizes both base-base and insertion and/or deletion expression in a p53-dependent manner;Sp1servesmismatches,MutSβspecificity is restricted to the latter as a molecular switch by recruiting HIF-1␣to the gene(Genschel et al.,1998).In addition,MutSαforms a ter-promoter under hypoxia.Furthermore,in human spo-nary complex with MutLαand presumably plays a role radic colon cancers,HIF-1␣overexpression is statis-in the initiation of mismatch repair(Blackwell et al., tically associated with the loss of MSH2expression,2001).Mutations in the MMR genes are associated with especially when p53is immunochemically undetecta-the development of both hereditary and sporadic can-ble.These findings indicate that the regulation ofcers,and germline mutations in MSH2or MLH1are the DNA repair is an integral part of the hypoxic re-most prevalent cause of hereditary nonpolyposis colo-sponse,providing molecular insights into the mecha-rectal cancers(Buermeyer et al.,1999).Hereditary nisms underlying hypoxia-induced genetic instability.MSH6defects have also been implicated in the devel-opment of late-onset familial colon tumors(Akiyama et Introduction al.,1997;Miyaki et al.,1997).Despite the functional importance of MMR in main-Hypoxia—oxygen deficiency in tissues—is a key factor taining genetic stability,little is known about the regula-of tumor microenvironment that has been tightly asso-tory mechanisms underlying MMR gene expression. ciated with tumor progression,metastasis,and resis-Hypermethylation of the MLH1promoter has been ob-tance to chemotherapy and radiation therapy(Young et served in colon cancer with MIN(Kane et al.,1997).p53 al.,1988;Brizel et al.,1996;Hockel et al.,1996).Hyp-binds to the MSH2promoter for transcriptional activa-oxic microenvironment is linked to DNA base substitu-tion(Scherer et al.,1996;Warnick et al.,2001).Myc has tions and deletions in tumors(Reynolds et al.,1996).been implicated by serial analysis of gene expression Severe hypoxia selects for tumor cells that have de-as a positive regulator of MSH2expression(Menssen fects in apoptosis;genetic alterations of the tumor sup-and Hermeking,2002).In addition,several functionalSp1sites exist in the MSH6promoter(Gazzoli and Ko-lodner,2003).However,how hypoxia induces MIN re-*Correspondence:huange@Molecular Cell794Figure1.Hypoxia Induces Genetic Instabilityby Specifically Inhibiting MutSαExpressionin a p53-Dependent Manner(A)HCT116cells were either subjected tohypoxia or infected with recombinant adeno-viruses expressing GFP or⌬ODD for48hr.The dinucleotide-repeat marker D17S250was used for MIN analysis.Arrows denoteinduced MIN.(B)HEC59cells were treated and analyzedas above.(C)In-frame(IF)and out-of-frame(OF)β-galactosidase reporters were used to ex-amine hypoxia-induced genetic instability incell types as indicated.The results were pre-sented as means+standard deviations fromthree independent experiments in duplicate.*p<0.05;**p<0.01;RBU,relativeβ-galacto-sidase activity;N,normoxia;and H,hypoxia.(D)HCT116cells were subjected to hypoxiafor16hr and analyzed for hypoxic repressionof MSH2and MSH6protein levels by West-ern blot analysis.HIF-1α,p53,andβ-actinwere included as controls.(E)Normal human small airway epithelialcells(SAEC)and fibroblasts(MRC5)weresubjected to hypoxia as above.The mRNAlevels of mismatch repair genes,as indi-cated,were determined with real-time PCR.PGK1served as a positive control for hyp-oxic induction.Representative results fromthree independent experiments in triplicatewere presented as means±standard errors.(F)HCT116(TP53+/+),its TP53−/−mutant,andHeLa cells were treated as above and exam-ined with real-time PCR.mains unclear.Recently,MLH1expression was shown markers(Boland et al.,1998).Figure1A shows that to be specifically inhibited by hypoxia,resulting in func-hypoxia reproducibly increased MIN in MLH1-deficient tional MMR deficiency(Mihaylova et al.,2003),even HCT116cells,as assayed with the dinucleotide-repeat though the proposed mechanism of hypoxia-induced marker D17S250.However,no such change was de-histone deacetylation apparently cannot account for tected in MSH2-deficient HEC59cells(Figure1B),sug-the specific downregulation of MLH1.To ascertain the gesting the requirement of MSH2for hypoxia-induced mechanisms of hypoxia-induced genetic instability,we e of the mononucleotide-repeat marker BAT26 examined the role of HIF-1α,a key regulator of hypoxia-yielded a similar result in HCT116cells(see Figure S1A responsive genes(Giaccia et al.,2003;Huang and in the Supplemental Data available with this article Bunn,2003;Pugh and Ratcliffe,2003;Semenza,2003;online).Poellinger and Johnson,2004),in DNA mismatch repair To provide additional evidence for the role of MSH2 and demonstrated that HIF-1αis responsible for hyp-in hypoxia-induced MIN,we employed reporter con-oxia-induced genetic instability by inhibiting MutSαstructs containing multiple copies of CA dinucleotide gene expression.repeats that precede theβ-galactosidase reporter geneeither in frame(IF)or out of frame(OF)(Parsons et al., Results1993).Hypoxia not only decreased the expression ofthe IF reporter by>40%,but also increased the OF re-porter activity by w3-fold in HCT116cells(Figure1C). Hypoxia Induces Genetic Instability by InhibitingAgain,no such changes were observed in HEC59cells. MutS␣ExpressionUnlike its parent cells,the MSH2-proficient HEC59+Ch2 To demonstrate hypoxic effects on genetic instability,we first examined microsatellite DNA with specific cells(Aebi et al.,1996)exhibited an w25%decrease ofHIF-1αRepresses MutSαExpression795Figure2.HIF-1αIs Necessary and Sufficient to Induce Genetic Instability by Downregulating MutSαExpression(A)HCT116cells were transfected with siRNAs targeting HIF1A or HIF2A and then subjected to hypoxic treatment.Luciferase siRNA(luc) was used as a negative control.MSH2,MSH6,and PGK1mRNA levels were determined with real-time PCR.Representative results are shown as in Figure1E.(B)Cells were treated as in(A)and assayed for specific protein levels,as indicated,by sequential probing of the same blot with the corre-sponding antibodies.(C)HCT116cells were infected with adenoviruses expressing⌬ODD(Ad-⌬ODD)or transcriptionally inactive⌬ODD(Ad-LCLL)for16hr.Cells without treatment(−)or infected with Ad-GFP served as controls.Transcripts of MSH2and MSH6,along with HIF1A,D ODD,hypoxia-inducible BHLHB2,and ACTB were determined with conventional PCR and quantified with densitometry.The numerical results in reference to ACTB levels are indicated.(D)HCT116cells were subjected to the same treatment as above,and protein levels of specified genes were sequentially probed with specific antibodies.Molecular weights are indicated on the left.(E)HCT116+Ch3cells were analyzed for MMR gene expression with real-time PCR after adenoviral infection as indicated.(F)Effects of Ad-⌬ODD on genetic instability were examined with IF and OF reporters in HCT116and HEC59cells.The results were presented as in Figure1C.**p<0.01.the IF reporter activity and an w30%increase in the MLH1remained essentially unchanged(Figure1E).In OF reporter activity in response to hypoxia.Similar re-contrast,the hypoxia-inducible gene PGK1exhibited a sults were obtained with MLH1-proficient HCT116cells4-fold increase.Similar results were obtained in the (HCT116+Ch3,Figure S1B).Furthermore,we tested thehuman MRC5fibroblasts(Figure1E),as well as in IF and OF reporters in MMR-proficient MCF-7and MMR-proficient MCF-7and HepG2cells(Figure S2B).As expected,hypoxia also decreased MSH2and MSH6 HepG2cells.Again,the hypoxic effects were also sta-tistically significant(Figure S2A).Taken together,these protein levels in HCT116cells(Figure1D).findings indicate the involvement of MSH2in hypoxia-However,in human cervical carcinoma HeLa cells, induced genetic instability.the expression of MLH1(Figure S3A),but not MSH2 To determine whether hypoxia alters MSH2expres-and MSH6(Figure1F),was slightly downregulated, sion,we investigated a panel of MMR genes in the nor-consistent with a previous report(Mihaylova et al., mal human diploid small airway epithelial cells.Results2003).In addition,no significant hypoxia-induced MIN from Taqman assays showed that hypoxia markedly re-was detected with the BAT26marker(Figure S3B).p53 pressed MSH2and MSH6expression by>50%,butis inactivated in HeLa cells(Scheffner et al.,1990),andMolecular Cell796Figure3.Requirement of Myc and p53forMutSαExpression(A)HCT116cells were transfected with MYC-specific siRNA and subsequently assayedfor MutSαexpression in normoxia and hyp-oxia with real-time PCR.Representative re-sults are shown as in Figure1E.(B)Effects of MYC siRNA were examined bysequential probing with respective antibod-ies in Western blotting.(C)TP53expression was knocked down inHCT116TP53+/+cells by specific siRNA orrestored in HCT116TP53−/−cells with ade-noviral infection.The mRNA levels of speci-fied genes were determined by real-timePCR.(D)MSH2and MSH6protein levels were alsoexamined in the cells treated as above byWestern blot analysis.p53levels were alsodetermined.the possible role of p53was therefore examined for the hypoxia.Figure2C shows that the infection of HCT116 hypoxic repression of MSH2and MSH6.In contrast to cells with recombinant adenoviruses expressing a sta-the result from wild-type HCT116,the TP53null cells ble HIF-1α(Ad-⌬ODD)(Koshiji et al.,2004)resulted in (Bunz et al.,1998)not only lost hypoxic repression but the downregulation of both MSH2and MSH6and the also exhibited a marked decrease in the basal expres-upregulation of BHLHB2,a known hypoxia-inducible sion of MMR genes(Figure1F).Thus,we conclude that gene(Wykoff et al.,2000).Interestingly,adenoviruses hypoxia promotes genetic instability by specifically in-expressing a stable yet transcriptionally inactive HIF-hibiting MutSαexpression in a p53-dependent manner.1α(Ad-LCLL)(Koshiji et al.,2004)also inhibited MutSαexpression but was unable to activate BHLHB2.Thisfinding suggested that HIF-1αtransactivation domains HIF-1␣,but Not HIF-2␣,Is Critical for Hypoxicare not required for the inhibitory effect.Consistently, Inhibition of Mismatch RepairAd-LLCL decreased protein levels of MutSαsubunits Hypoxia activates HIF-1αas well as HIF-2α/EPAS(Tian(Figure2D).Furthermore,in HCT116+Ch3cells,Ad-et al.,1997),another member of the HIF-αfamily.ToLCLL infection specifically downregulated MSH2and address whether HIF-αis responsible for hypoxic inhi-MSH6expression(Figure2E).These results argue that bition of MMR,we employed small interfering RNAHIF-1αtranscriptional activity is dispensable for inhibit-(siRNA)targeting either HIF1A or HIF2A.As shown ining MutSαexpression.Figure2A,HIF1A,but not HIF2A,siRNA specificallyTo test whether HIF-1αpromotes genetic instability, abrogated hypoxic repression of MSH2and MSH6.Re-we again resorted to the IF and OF reporters and analy-sults in Figure2B confirmed the loss of HIF-1αand HIF-sis of MIN.Ad-⌬ODD infection significantly altered the 2α(Figure S4A)and the unique role of HIF-1αin mediat-reporter activities in HCT116,but not in HEC59(Figure ing the hypoxic repression.2F).Similarly,Ad-⌬ODD infection gave rise to MIN in To substantiate the role of HIF-1αin MutSαexpres-HCT116cells,but not in HEC59(Figures1A,1B,and sion,we asked whether a stable HIF-1αin normoxiawould recapitulate the repression of MMR genes by S1A).Altogether,we conclude that HIF-1αis responsi-HIF-1αRepresses MutSαExpression797To substantiate the role of p53in hypoxic inhibitionof MutSα,we also employed TP53siRNA.Silencing ofTP53expression decreased MSH2and MSH6mRNAlevels by>30%in HCT116cells(Figure3C).Conversely,introduction of p53to TP53null HCT116cells raisedthe basal level similar to that in wild-type HCT116.Moreover,the p53effect on MutSαexpression wasconfirmed by Western blotting(Figure3D).In addition,forced expression of p53in TP53null HCT116cells in-creased the MSH2promoter activity(Figure S5).There-fore,both Myc and p53are required for basal expres-sion of MutSα.To establish the relevance of the above transcriptionfactors to the transcriptional regulation of MutSαgenes,we analyzed their binding to the MSH2andMSH6promoters in vivo with chromatin immunopre-cipitations(ChIP).The MSH2proximal promoter(−1200to+1)was divided into three regions(Figure4A),one ofwhich(−780to−392)showed binding of these tran-scription factors.This region was narrowed down fur-ther to−671to−518.Likewise,the MSH6promoter wasmapped to−644to−300.The ChIP results from theminimal regions revealed that although p53and Mycbound to the MSH2promoter in normoxic HCT116cells,hypoxic treatment markedly reduced the bindingbut induced HIF-1αbinding(Figure4B).Similar resultswere obtained with the MSH6promoter,reminiscent ofthe changes taking place in the CDKN1A promoter(Koshiji et al.,2004).However,unlike the CDKN1A prox-imal promoter,the MutSαgene promoters showed noMiz-1binding.Moreover,p53binding was greatly inhib-Figure4.Myc Displacement by Hypoxia from MSH2and MSH6Pro-ited by hypoxia in the MSH2and MSH6promoters butmoters Is p53Dependent was unaffected in the CDKN1A distal promoter(Koshiji (A)Schematic representation of the MSH2and MSH6proximal pro-et al.,2004).Furthermore,in hypoxic TP53null cells, moters.Double-arrowed lines indicate regions that were PCR-Myc binding remained unchanged in the MSH2and amplified in ChIP.Representative data of the shadowed regions are MSH6promoters but markedly diminished in theshown in(B).CDKN1A proximal promoter(Figure4B),suggesting the (B)Chromatin immunoprecipitations of normoxic and hypoxicdifferential requirement of p53for the displacement of HCT116cells were performed by using antibodies against indi-Myc from MutSαgenes and CDKN1A promoters.Alto-cated transcription factors.Both TP53+/+and TP53−/−cells wereused.Specific primers flanking regions of the MSH2,MSH6,and gether,we infer that hypoxic repression of MutSαCDKN1A proximal promoters were used to amplify the immuno-genes results from the gain of HIF-1αbinding,in con-precipitated genomic DNA.Sheared genomic DNA prior to immu-comitance with the loss of Myc and p53binding from noprecipitations served as input,and immunoglobulin(IgG)servedthe promoter.as a control.HIF-1␣Competes against Myc for Sp1Bindingble for hypoxia-induced genetic instability by downreg-Previously,we showed that HIF-1αforms a weak com-ulating MutSαgenes.plex with Myc,indicative of an indirect interaction(Ko-shiji et al.,2004).The above ChIP of the MutSαgene HIF-1␣Requires Myc and p53to Inhibit MutS␣promoters identified constitutive binding of Sp1,which Previously,we proposed that the N-terminal HIF-1αin-is known to interact with Myc(Gartel et al.,2001)andto engage in a hypoxic response(Discher et al.,1998). dependently regulates a distinct set of hypoxia-respon-sive genes via antagonizing Myc activity(Koshiji et al.,Figure5A shows that knockdown of SP1with siRNA 2004).MSH2is activated by Myc(Menssen and Her-(Figure S4B)abrogated MutSαexpression in both nor-meking,2002)and is repressed herein by a transcrip-moxia and hypoxia,suggesting an essential role for tionally inactive HIF-1α,characteristic of genes regu-Sp1in the transcriptional regulation of MutSαgenes. lated by the HIF-1α-Myc pathway.To confirm the In addition,SP1siRNA abolished hypoxic induction of involvement of Myc in MSH2and MSH6expression,wePGK1(Pfeifer et al.,1990).To establish the functional knocked down MYC expression with RNA interference.relevance of Sp1,we asked whether HIF-1αcompetes Consequently,MYC siRNA not only markedly reducedfor Sp1binding.Figure5B shows that anti-Sp1immu-MSH2and MSH6basal expression but also abrogated noprecipitation captured Myc only in normoxic extract the hypoxic repression(Figures3A and3B),confirmingand HIF-1αinstead in hypoxic extract.Moreover,knock-the essential role of Myc for maintaining MutSαex-down of HIF-1αlevels by specific siRNA restored Myc pression.binding in hypoxia,arguing for the pivotal role of Sp1Molecular Cell798Figure5.Sp1Is Essential for Recruiting HIF-1αonto the MSH2Promoter(A)HCT116cells were transfected with SP1siRNA,and subsequently the expression ofMutSαgenes was determined with real-timePCR.Representative results are shown as inFigure1E.(B)Normoxic and hypoxic HCT116cell ly-sates were immunoprecipitated with anti-Sp1antibody(α-Sp1IP),followed by Westernblotting with anti-Myc and anti-HIF-1αanti-bodies(bottom).Input levels of HIF-1αandMyc were determined by direct Western blot(top).HIF1A expression was also silenced byspecific siRNA.(C)In vitro-translated,[35S]methionine-labeled(with asterisks)Myc or HIF-1αwas coimmu-noprecipitated with anti-Sp1antibody againstendogenous Sp1.The addition of unlabeledHIF-1αabrogated Myc coprecipitation,whereas the addition of unlabeled Myc hadno effect on HIF-1αcoprecipitation.Input:10%of lysates without immunoprecipi-tations.(D)An MSH2luciferase reporter was co-transfected into HCT116cells with vectorsexpressing p53,Myc,or⌬ODD.Cotransfec-tion with pcDNA3(−)was used as a control.The reporter activity was presented as rela-tive luciferase activity(RLU)as means+standard deviations.An MSH2mutant re-porter(MSH2-Sp1)with a mutation at a puta-tive Sp1site was also examined.The datafrom three independent experiments in du-plicate were presented as means+standarddeviations.**p<0.01.(E)MRC5cell extract was assayed for Sp1DNA binding with[32P]-labeled oligonucleo-tides containing a consensus Sp1element(Sp1*),a putative Sp1site in the MSH2pro-moter(MSH2*),or a mutated Sp1site(MSH2m*).A100-fold excess of unlabeledoligonucleotides of the consensus Sp1(Sp1),wild-type MSH2(MSH2),or mutatedMSH2(MSH2m)was used for competition.An arrowhead denotes Sp1binding and anarrow denotes Sp3binding.(F)Normoxic and hypoxic MRC5extracts were subjected to supershift with the addition of antibodies against Sp1(α-Sp1),HIF-1α(α-HIF), and Sp3(α-Sp3).Minus symbol,no antibody added.in the HIF-1α-Myc pathway.Consistently,an in vitro re-bona fide Sp1binding to oligonucleotides derived from constitution assay demonstrated that HIF-1αdomi-the MSH2promoter(Figure S6A).Moreover,in MRC5 nated Sp1binding(Figure5C).Taken together,these cell extract,Sp1binding to the consensus Sp1oligonu-findings indicate that HIF-1αdisplaces Myc bindingcleotides or the wild-type MSH2oligonucleotides was from the target gene promoters via its interaction with competed for by either of the oligonucleotides in ex-Sp1.cess,but not by those with the same mutation used in Next,we constructed a luciferase reporter driven by the reporter assay above(Figure5E).Furthermore,we the MSH2minimal promoter region that not only boundutilized specific antibodies for supershift to ascertain Myc,Sp1,HIF-1α,and p53(Figure4),but also exhibited the nature of the Sp1complex in MRC5cell extract hypoxic repression(Figure S5).In accordance with the(Figure5F).The anti-HIF-1αantibody abolished the results aforementioned,expression of p53or Myc hypoxic Sp1binding,but not the normoxic one,indicat-markedly stimulated the MSH2promoter,whereas ex-ing that Sp1recruits HIF-1αto the MSH2promoter in pression of a stable HIF-1α,⌬ODD(Huang et al.,1998),hypoxia.Taken together,these findings corroborate thepivotal role of Sp1in mediating the HIF-1α-Myc significantly repressed the reporter activity(Figure5D).Remarkably,mutation of a putative Sp1binding ele-pathway.ment in the promoter abolished Myc stimulation as wellas⌬ODD inhibition.In contrast,the p53stimulatory ef-The N-Terminal HIF-1␣Is Responsiblefect was only slightly affected.To confirm that Sp1for Inducing Genetic Instabilitybinds to this site,we performed electrophoretic mobil-Previously,we mapped out the N-terminal HIF-1αity shift assays with recombinant Sp1and showed a(amino acids1–329)that weakly interacts with Myc;HIF-1αRepresses MutSαExpression799Hence,we argue that HIF-1αdownregulates MutSαex-pression via its N-terminal interaction with Sp1.An Inverse Association between HIF-1␣and MSH2in Human Colon Cancer Tissueswith Undetectable p53To demonstrate the role of HIF-1αin MSH2expressionin human cancers,we performed immunohistochemicalstaining on80specimens of nonhereditary colon can-cers(Table S1).The four most common types of stain-ing,as shown in Figure7A,fall into two groups,p53-undetectable(indicating p53wild-type or loss of p53expression,left two columns)and p53-detectable(indi-cating p53mutations,right two columns).The formergroup includes specimens stained positive for MSH2orHIF-1α.The latter group depicts those that stained HIF-1αpositive or both HIF-1αpositive and MSH2positive.In total,35%of the cases showed MSH2positive stain-ing and HIF-1αnegative staining,whereas27.5%showed the reverse.(Figure7B).Additionally,25%ofthe cases stained both positive,and12.5%were bothnegative.Statistical analyses showed a two-tailed pvalue of<0.05by Fisher’s exact test and an odds ratioof0.325,suggesting an inverse association betweenHIF-1αand MSH2expression in these specimens.Con-sistent with the requirement of wild-type p53for MSH2inhibition by hypoxia,stratified analysis of the p53-undetectable cases yielded a p value of<0.001with anodds ratio of0.076.By contrast,in the p53-detectablegroup,no statistical significance was observed.There-fore,these results are consistent with our hypothesisthat overexpressed HIF-1αsuppresses MSH2expres-sion in a p53-dependent manner.DiscussionFigure6.The N-Terminal HIF-1αInteracts with Sp1and InducesIn this study,we ascertained the mechanism underlying Genetic Instabilityhypoxia-promoted genetic instability by focusing on (A)HIF-1α1–329,HIF-1α1–167,and⌬ODD and its R27G mutantthe role of HIF-1αin MutSαexpression.HIF-1αhas were translated in vitro and subjected to anti-Sp1immunoprecipi-tation.Input:10%of lysates without immunoprecipitations.been demonstrated as a transcriptional activator that (B)HIF-1α1–329and the above mutants were tested for their effect is responsible for the upregulation of an expanding on wild-type MSH2promoter(MSH2)and the promoter with a mu-group of hypoxia-responsive genes(Harris,2002;Sem-tation at the Sp1binding site(MSH2-Sp1).The results were pre-enza,2003).However,to gain a comprehensive under-sented as in Figure5D.**p<0.01.standing of the hypoxic response,hypoxia-downregu-(C)HIF-1α1–329and its R27G mutant were examined together withlated genes merit an equal amount of attention(Koshiji other HIF-1αmutants as specified for their ability to induce geneticinstability with the IF and OF reporters.The results were presented and Huang,2004),even though until now whether and, as in Figure1C.if so,how HIF-1αplays such a role in gene suppressionwas unclear.We demonstrate that HIF-1αacts as atranscriptional repressor of MSH2and MSH6.Although whereas further C-terminal deletion(HIF-1α1–167)the underlying mechanism conforms to the HIF-1α-Myc eliminates the binding(Koshiji et al.,2004).Consistently,pathway identified in the hypoxic activation of CDKN1A Figure6A shows that Sp1captured HIF-1α1–329,but(Koshiji et al.,2004),the outcome is just the opposite. not HIF-1α1–167.In addition,Sp1interacted with Of particular interest is an absolute requirement of wild-⌬ODD and a R27G mutant unable to bind DNA(Koshijitype p53for MSH2and MSH6repression,but not for et al.,2004).To confirm that HIF-1α1–329is sufficient CDKN1A activation(Goda et al.,2003;Koshiji et al.,2004).Apart from the HIF-1αdisplacement of Myc from to downregulate MSH2expression,we tested effects ofHIF-1α1–329on the MSH2reporter.Results in Figure the MSH2and MSH6promoters,p53binding was also 6B show that HIF-1α1–329,but not HIF-1α1–167,sig-decreased by hypoxia,in sharp contrast to the unal-nificantly inhibited the MSH2promoter activity.As ex-tered binding in the CDKN1A distal promoter(Koshiji et pected,no such effect was seen when the Sp1bindingal.,2004).p53’s crucial role was further substantiated site was mutated.Furthermore,transfection with HIF-by the finding that in TP53−/−HCT116cells,hypoxia 1α1–329also reversed the IF and OF reporter activitiesshowed no effect on Myc binding to the MutSαgene (Figure6C).Similar results were obtained with a HIF-1αpromoters,again distinct from a decreased Myc bind-1–329(R27G)mutant,but not with HIF-1α1–167.ing in the CDK1NA promoter.Therefore,it would be。