3 Feng-1998-The tumor suppressor
靶向突变p53的小分子药物研究进展
靶向突变p53的小分子药物研究进展王玉玲;苏永南;暴亚锋;杨志宽;牟汉川;张继虹【期刊名称】《中国药理学通报》【年(卷),期】2018(034)003【摘要】Tumor suppressor p53 protein can regulate the tran-scription of target genes, to control cell apoptosis, aging and other life activities,but mutant p53 is prone to losing antitumor function, thus promoting tumor development. At present, p53 protein has become one of the hot targets for the treatment of cancer. This article mainly introduces the structure and mechanism of small molecular compounds with restoring activity of mutant p53 as the target.%肿瘤抑制因子p53蛋白可以调节靶基因转录,控制细胞凋亡、衰老等生命活动,但其容易发生突变,失去抑癌功能,促进肿瘤发生发展.目前p53蛋白已成为治疗肿瘤的热门靶点之一,该文主要介绍以突变p53为靶点恢复构象活性的小分子化合物结构及作用机制.【总页数】4页(P321-324)【作者】王玉玲;苏永南;暴亚锋;杨志宽;牟汉川;张继虹【作者单位】昆明理工大学医学院衰老与肿瘤分子遗传学实验室,云南昆明650500;昆明理工大学医学院衰老与肿瘤分子遗传学实验室,云南昆明 650500;昆明理工大学医学院衰老与肿瘤分子遗传学实验室,云南昆明 650500;昆明理工大学医学院衰老与肿瘤分子遗传学实验室,云南昆明 650500;昆明理工大学医学院衰老与肿瘤分子遗传学实验室,云南昆明 650500;昆明理工大学医学院衰老与肿瘤分子遗传学实验室,云南昆明 650500【正文语种】中文【中图分类】R-05;R341;R394.2;R730.5;R977.6;R979.1【相关文献】1.癌症中p53失活的分子机制和靶向p53的抗癌药物研究进展 [J], 周建林2.小分子靶向药物在皮肤病领域的研究进展 [J], 熊绮颖;叶瑞贤;张锡宝3.靶向肿瘤因子c-MYC基因启动区G4-DNA的小分子药物设计及核磁共振研究进展 [J], 胡晓东;蓝文贤;王春喜;曹春阳4.靶向PSMA放射性小分子药物研究进展 [J], 穆博帅;徐洋;刘志博5.靶向突变型p53的抗肿瘤药物研究进展 [J], 丁笠;张新跃因版权原因,仅展示原文概要,查看原文内容请购买。
在卵巢癌小鼠中,Trp53或Pik3ca的突变使I型卵巢癌向II型卵巢癌发展的过程中,转变为一种更有侵袭性的表型。
TUMORIGENESIS AND NEOPLASTIC PROGRESSIONType I to Type II Ovarian Carcinoma ProgressionMutant Trp53or Pik3ca Confers a More Aggressive Tumor Phenotype in a Mouse Model of Ovarian CancerRong Wu,*Suzanne J.Baker,y Tom C.Hu,*Kyle M.Norman,*Eric R.Fearon,*zx and Kathleen R.Cho*zxFrom the Departments of Pathology*and Internal Medicine,z and the Comprehensive Cancer Center,x University of Michigan Medical School,Ann Arbor, Michigan;and the Department of Developmental Neurobiology,y St.Jude Children’s Research Hospital,Memphis,TennesseeAccepted for publication December10,2012.Address correspondence to Kathleen R.Cho,M.D., Department of Pathology, University of Michigan Medical School,1506A.Alfred Taub-man BSRB,109Zina Pitcher, Ann Arbor,MI48109-2200.E-mail:kathcho@.A dualistic pathway model of ovarian carcinoma(OvCA)pathogenesis has been proposed:type I OvCAs are low grade,genetically stable,and relatively more indolent than type II OvCAs,most of which are high-grade serous carcinomas.Endometrioid OvCA(EOC)is a prototypical type I tumor,often harboring mutations that affect the Wnt and phosphatidylinositol3-kinase/AKT/mammalian target of rapamycin signaling pathways.Molecular and histopathologic analyses indicate type I and II OvCAs share over-lapping features,and a subset of EOCs may undergo type I/type II progression accompanied by acquisition of somatic TP53or PIK3CA mutations.We used a murine model of EOC initiated by conditional inactivation of the Apc and Pten tumor suppressor genes to investigate mutant Trp53or Pik3ca alleles as key drivers of type I/type II OvCA progression.In the mouse EOC model,the presence of somatic Trp53 or Pik3ca mutations resulted in shortened survival and more widespread metastasis.Activation of mutant Pik3ca alone had no demonstrable effect on the ovarian surface epithelium but resulted in papillary hyperplasia when coupled with Pten inactivation.Ourfindings indicate that the adverse prognosis associated with TP53and PIK3CA mutations in human cancers can be functionally replicated in mouse models of type I/type II OvCA progression.Moreover,the models should represent a robust platform for assessment of the contributions of Trp53or Pik3ca defects in the response of EOCs to conventional and targeted drugs.(Am J Pathol2013,182:1391e1399;/10.1016/j.ajpath.2012.12.031)Surgical pathologists continue to use traditional morphology-based schemes for classifying ovarian carcinomas(OvCAs). The schemes are based largely on the degree of resemblance of the OvCAs to non-neoplastic epithelia in the female genital tract.However,mounting clinicopathologic and molecular data have led Kurman and Shih1to propose a“dualistic”model of OvCA pathogenesis in which OvCAs are divided into two main categories,type I and type II.1e3 Type I OvCAs are suggested to be low-grade,relatively indolent and genetically stable tumors that typically arise from recognizable precursor lesions such as endometriosis or so-called borderline(low malignant potential)tumors.Type I OvCAs frequently harbor somatic mutations(eg,KRAS, BRAF,CTNNB1,PTEN)that dysregulate specific cell signaling pathway factors or certain chromatin remodeling complexes(eg,ARID1A).Type I OvCAs include most endometrioid,clear cell,and mucinous carcinomas and low-grade serous carcinomas.In contrast,type II OvCAs are proposed to be high-grade,biologically aggressive tumors from their outset,with a propensity for metastasis from small-volume primary lesions.Most type II OvCAs are high-grade serous carcinomas,virtually all of which harbor mutant TP53 alleles.4These tumors have a high level of chromosomal instability.5Although varied gene defects besides TP53 mutation have been identified in type II tumors,with the exception of frequent genetic/epigenetic inactivation of BRCA1/2and NF1,RB1,and CDK12mutations,each of the somatic gene defects is found in a small fraction of tumors.6e8Notably,recent data suggest that many(and perhaps most)high-grade serous carcinomas arise from Supported by National Cancer Institute grants RO1CA94172(K.R.C.) and RO1CA135554(S.J.B.)and Department of Defense grant W81XWH-10-2-0013(K.R.C.and E.R.F.).Copyrightª2013American Society for Investigative Pathology. Published by Elsevier Inc.All rights reserved./10.1016/The American Journal of Pathology,Vol.182,No.4,April2013epithelium in the fallopian tube,rather than the ovarian surface epithelium(OSE).9e11The identification of precursor lesions with p53protein overexpression and clonal TP53 mutations in the fallopian tube epithelium(including“p53 signature lesions”and tubal intraepithelial carcinomas) suggests TP53mutation is an early event in the pathogenesis of most type II OvCAs.12The dualistic pathway model represents an advance in conceptualizing OvCA pathogenesis,but the model is likely an oversimplified view of a complex group of cancers.For example,there are uncertainties about classification of clear cell carcinomas as type I versus type II,because their molecular features are more in keeping with type I tumors, but cytologic grade and clinical behavior are often more like type II tumors.3As another example,there is significant overlap in the morphologic and molecular features of high-grade serous and high-grade endometrioid OvCAs(hereafter referred to as EOCs)such that some pathologists now default the majority of gland-forming or near-solid cyto-logically high-grade OvCAs to the serous category and consider“true”high-grade EOCs to be rare or nonexistent.13 In this scenario,all low-grade EOCs would be classified as type I,and the category of high-grade EOCs would be eliminated.In a prior analysis of a substantial number of primary human EOCs,we found that mutations predicted to activate the canonical Wnt and phosphatidylinositol3-kinase(PI3K)/ AKT/mammalian target of rapamycin(mTOR)signaling pathways frequently co-occur in low-grade EOC,a prototyp-ical type I OvCA.14Activation of Wnt signaling typically occurs as a consequence of oncogenic CTNNB1mutations,or rarely bi-allelic APC inactivation,whereas PI3K/AKT/ mTOR signaling is usually activated because of PTEN inac-tivation and/or oncogenic PIK3CA mutations.Notably,we identified4of21tumors with gene expression and/or muta-tional profiles like type I EOCs,but the tumors had also acquired TP53mutations,suggesting type I/type II progression may occur in a sizeable subset(nearly20%)of EOCs.14Similarly,we identified four EOCs with mutations in both PTEN and PIK3CA,rather than a single mutation dys-regulating PI3K/AKT/mTOR signaling.Importantly,TP53 mutation has been associated with adverse outcome in women with endometrioid carcinomas of the endometrium and ovary,15e18and PIK3CA mutation has been linked to poor outcome in patients with several types of cancer,including carcinomas of the endometrium,breast,and colon.19e22The effect of PIK3CA mutations on the prognosis of patients with OvCA is unclear,because some studies have shown an adverse effect on outcome,whereas others showed no or even a favorable effect.23e25The notion that type II tumors can progress from type I tumors is not unique to EOCs.Indeed, type I/type II progression in serous carcinomas has also been described and is often associated with acquisition of TP53mutation.26e29We previously described a murine model for type I human EOC in which concurrent dysregulation of Wnt and PI3K/AKT/mTOR signaling is achieved through condi-tional inactivation of the Apc and Pten tumor suppressor genes in the OSE.14In this model,ovarian bursal injection of recombinant adenovirus expressing Cre recombinase (AdCre)in Apcflox/flox;Ptenfloxf/flox mice results in the devel-opment of EOC-like tumors with complete penetrance.The purpose of the present study was to determine the effect of mutant Trp53and Pik3ca on the ApcÀ/À;PtenÀ/Àmurine EOC tumor phenotype as models of type I/type II progression.Materials and MethodsStrains of Transgenic MiceIn Apcflox/flox;Ptenflox/flox mice,conditional inactivation of the Apc and Pten tumor suppressor genes in the mouse OSE (MOSE)results in ovarian tumor development with100% penetrance.14Trp53LSL-R172H/þ(01XM2),Trp53R270H/þ(01XM1),and Trp53flox/flox(01XC2)mice were purchased from the National Cancer Institute(Bethesda,MD)mouse repository and cross-bred with Apcflox/flox;Ptenflox/flox mice to generate the respective triple transgenic strains.Pik3ca LSL-E545K/þmice were developed in which a conditional mutant(LSL-E545K) allele is knocked into one of the endogenous Pik3ca loci.30 Cre-mediated deletion of a lox-STOP-lox cassette inserted upstream of thefirst coding exon activates expression of Pik3ca E545K from the endogenous locus.The mice were crossed with Apcflox/flox;Ptenflox/flox mice to generate triple transgenic Apcflox/flox;Ptenfloxflox;Pik3ca LSL-E545K/þmice.All strains were maintained on a mixed C57BL/6;FVB;129background. Induction of Murine Ovarian TumorsFor tumor induction,5Â107plaque-forming units of replication-incompetent AdCre(purchased from the University of Michigan’s Vector Core)with0.1%Evans blue(Sigma-Aldrich,Indianoplis,IN)were injected into the right ovarian bursal cavities of6-to10-week-old female mice as previously described.14,31In each mouse,the left ovarian bursa was not injected and served as control. Mouse Histopathology and ImmunohistochemistryAll tumor-bearing mice were euthanized per the Committee on Use and Care of Animals guidelines at the University of Michigan for end-stage illness and humane endpoints and then examined at necropsy.The genital tract and other major organs were collected from each mouse,fixed in10%(v/v) buffered formalin,and embedded in paraffin.H&E-stained tissue sections were evaluated by a board-certified surgical pathologist with expertise in gynecologic cancer diagnosis (K.R.C).Immunohistochemical staining was performed with standard methods;antigen e antibody complexes were detected with the avidin-biotin peroxidase method with the use of3,30-diaminobenzidine as the chromogenic substrate.Wu et al-The American Journal of PathologyAntibodies used in this study include rat anti-cytokeratin8 (CK8,#TROMA1;Developmental Studies Hybridoma Bank, University of Iowa);rabbit anti-p53(Vector Laboratories Inc., Burlingame,CA),rabbit anti e phospho-AKT(Ser473;#4060; Cell Signaling Technology,Inc.,Danvers,MA),rabbit anti-PTEN(clone138G6,#9559;Cell Signaling Technology, Inc.),mouse anti e b-catenin(Transduction Laboratories, Lexington,KY),goat anti e E-cadherin(R&D Systems,Minneapolis,MN),rabbit anti e WT-1(sc-192;Santa Cruz Biotechnology,Inc.,Dallas,TX),and rabbit anti-PAX8 (NBP1-74734;Novus Biologicals,Littleton,CO).Genotyping and Recombination Analysis of Mouse TumorsGenomic DNA was isolated from tail snips,ovarian tumors,or other organs,and PCR was performed with primers that allowed distinction between endogenous,genetically modified,and recombined alleles(primer sequences are available on request). Representative data are shown in Supplemental Figure S1. Statistical AnalysisGraphs were constructed and statistical functions were analyzed with GraphPad Prism version5(GraphPad Soft-ware Inc.,La Jolla,CA).Kaplan-Meier survival curves were compared separately for each experimental pair by log-rank (Mantel-Cox)tests;P<0.05was considered statistically significant.Metastasis between groups was compared with a c2contingency table test for independence.ResultsMore Aggressive Phenotype of Murine ApcÀ/À;PtenÀ/ÀOvarian Tumors with the Addition of Somatic Missense Trp53MutationsSomatic missense substitutions in the TP53gene which result in the R175H and R273H alleles are among the most frequently identified p53mutations in human cancers;both the R175H and R273H TP53alleles have been studied for their loss-of-function effects on the transcription activity of p53,as well as for potential gain-of-function effects.In OvCAs,R273H,R248W, and R175H are the most common missense TP53mutations (Catalogue of Somatic Mutations in Cancer;http://www.sanger. /perl/genetics/CGP/cosmic?action Z gene&ln Z TP53). As a means of modeling type I to type II progression of EOCs, we introduced the murine equivalents of the R273H(R270H) and R175H(R172H)mutations into Apcflox/flox;Ptenfloxflox mice. Specifically,Apcflox/flox;Ptenfloxflox mice were crossbred with mice that carried a conditional mutant Trp53LSL-R172H allele32or constitutive mutant Trp53R270H allele33to yield Apcflox/flox; Ptenfloxflox;Trp53LSL-R172H/þand Apcflox/flox;Ptenfloxflox;Figure1The phenotype of murine ApcÀ/À;PtenÀ/Àovarian tumors ismore aggressive with the addition of missense Trp53mutations.Kaplan-Meiersurvival curves of Apcflox/flox;Ptenfloxflox;Trp53LSL-R172H/þmice(n Z11)andApcflox/flox;Ptenfloxflox littermate controls(n Z9)(A)and Apcflox/flox;Ptenfloxflox;Trp53R270H/þmice(n Z11)and Apcflox/flox;Ptenfloxflox littermatecontrols(n Z10)(B)after ovarian bursal AdCre injection.H&E-stainedsections of ApcÀ/À;PtenÀ/À;Trp53R172H/þovarian tumors showing glandularepithelial differentiation admixed(C)with more poorly differentiated spindle-cell areas(D).Representative metastases on the surface of the liver(E)andkidney(F;aggregate of metastatic tumor cells indicated by an arrow)areshown.Immunohistochemical staining of p53in representative ovarian tumorsarising in Apcflox/flox;Ptenfloxflox;Trp53LSL-R172H/þ(G)and Apcflox/flox;Ptenfloxflox;Trp53R270H/þ(H)mice.Scale bars:100m m(C e H).Models of Ovarian Cancer Progression The American Journal of 1393Trp53R270H/þmice.Ovarian tumors were induced by injection of AdCre into the right ovarian bursa,and tumor-bearing mice were monitored for several weeks thereafter.Mice with ApcÀ/À; PtenÀ/Àovarian tumors also expressing R172H or R270H mutant p53had significantly shorter survival than littermate mice with ApcÀ/À;PtenÀ/Àtumors that had wild-type Trp53 alleles(P Z0.0001and P Z0.0227,respectively)(Figure1,A and B).Similar to the ApcÀ/À;PtenÀ/Àovarian tumors we previously described,tumors expressing mutant p53showed areas with glandular,overtly epithelial differentiation admixed with more poorly differentiated spindle-cell areas(Figure1,C and D).Metastatic carcinoma,represented by small tumor implants on the surface of abdominal organs,including liver and kidney(Figure1,E and F),was identified more frequently in mice expressing R172H mutant p53than Apcflox/flox;Ptenfloxflox; Trp53þ/þtumor-bearing littermates(P Z0.025).Microscopic lung metastases were observed only in mice whose tumors expressed mutant p53,albeit in a small fraction.Immunohis-tochemical staining showed nuclear accumulation of p53in the tumor cells expressing either the R172H or R270H mutant (Figure1,G and H);in some cases p53overexpression was focal and in others more diffuse.Data collected from ovarian tumor-bearing Apcflox/flox;Ptenfloxflox;Trp53LSL-R172H/þand Apcflox/flox;Ptenfloxflox;Trp53R270H/þmice are summarized in Table1.Mice Bearing ApcÀ/À;PtenÀ/À;Trp53À/ÀTumors Have the Shortest Survival and Frequently Develop Bulky Metastatic DiseaseBecause null and missense Trp53mutations have been shown to have different effects on the tumor phenotype in several mouse model systems34and roughly10%of TP53 mutations in human EOCs result in loss of p53protein expression(ie,nonsense,insertion,deletion;http://www. /search/s),we also wished to test the effects of Trp53deletion in the ApcÀ/À;PtenÀ/Àmodel of EOC. Notably,in humans,distant metastases are nearly eightfold more common in patients with OvCAs that carry TP53null mutations compared with those with missense mutations.35Apcflox/flox;Ptenfloxflox mice were crossed with mice harbor-ing conditional knockout(Trp53flox)alleles,allowing Cre-mediated deletion of Trp53exons2through10.36Ovarian tumors were induced in Apcflox/flox;Ptenfloxflox;Trp53flox/flox as well as Apcflox/flox;Ptenfloxflox;Trp53flox/þand Apcflox/flox; Ptenfloxflox;Trp53þ/þlittermates with bursal AdCre injection. Mice with ApcÀ/À;PtenÀ/À;Trp53À/Àovarian tumors had significantly reduced survival than mice with ApcÀ/À; PtenÀ/Àtumors in which one or both Trp53alleles were intact(P Z0.0006and P Z0.0004,respectively).No significant difference in survival was observed between mice whose tumors had Cre-mediated deletion of one Trp53 allele compared with mice with two intact alleles(P Z 0.3482)(Figure2A and Table1).At necropsy,all of the tumor-bearing Apcflox/flox;Ptenfloxflox;Trp53flox/flox mice had grossly visible abdominal metastases(Figure2B).Repre-sentative photomicrographs of tumors arising in Apcflox/flox; Ptenfloxflox;Trp53flox/flox and Apcflox/flox;Ptenfloxflox;Trp53flox/þmice are shown in Figure2,C and D.The distribution of tumor growth outside of the ovary was similar to that seen in patients with OvCA stage IV disease,because some mice showed deep extension of tumor into the parenchyma of liver(Figure2E)and/or kidney,and one mouse had metastasis to the lung(Figure2F).Metastatic carcinoma was observed significantly less frequently in tumor-bearing Apcflox/flox;Ptenfloxflox;Trp53þ/þlittermates(P Z0.016). ApcÀ/À;PtenÀ/À;p53À/Àovarian tumors and their metastases were poorly differentiated,but the epithelial components showed strong expression of E-cadherin(Figure2G)and cytokeratin8(not shown).As expected,the tumor cells showed complete absence of p53expression(Figure2H). EOC Development in Mice Requires Combined Defects in Canonical Wnt and PI3K/AKT/mTOR Signaling,Even in the Presence of Mutant p53Studies of human high-grade serous OvCAs and precursor lesions in the fallopian tube suggest that TP53mutations occur early and may be required for their development.In contrast,only a subset(z20%)of type I EOCs withTable1Effects of Missense and Null Trp53Mutations on the ApcÀ/À;PtenÀ/ÀTumor PhenotypeTrp53genotype Tumor size(mm3)Metastasis(n/N)P*Lungmetastasis(n/N)Mediansurvival(days)P yLSL-R172H/þ4752.7Æ1767.88/111/11570.0250.0001þ/þ4632.8Æ2178.62/90/976R270H/þ4331.2Æ1383.34/111/11710.7570.0227þ/þ4838.2Æ1868.13/100/1075flox/flox3068.7Æ877.09/91/957flox/flox vsflox/þ,0.0006flox/þ3137.6Æ1536.96/80.0160/864flox/flox vsþ/þ,0.0004þ/þ2422.8Æ1351.72/60/669.5flox/þvsþ/þ,0.3482*Determined by c2test.y Determined by log-rank test for median survival.flox,flox(del ex2-10).Wu et al-The American Journal of Pathologycanonical Wnt and PI3K/AKT/mTOR signaling pathway defects also have TP53mutations,suggesting that mutant p53is not required for EOC development but may be asso-ciated with type I to type II progression.In previous studies,we showed that EOCs fail to develop unless both Wnt and PI3K/AKT/mTOR signaling pathways are concomitantly dysregulated.Speci fically,no tumors formed after ovarian bursal AdCre injection in any of 61Apc flox/flox or 63Pten flox/flox mice.14Here,we wished to test whether Trp53point mutations (R172H or R270H)could cooperate with dysregulation of either canonical Wnt or PI3K/AKT/mTOR signaling in initi-ating EOCs in our mouse model.Ovarian bursal AdCre injec-tion was performed in Apc flox/flox ;Pten flox /þ;Trp53LSL-R172H /þ(n Z 2)and Apc flox /þ;Pten flox/flox ;Trp53R270H /þ(n Z 2)mice,and animals were monitored for up to 9months.Although only a small number of mice with each genotype were tested,none of the mice developed ovarian tumor,further supporting the conclusion that dysregulation of both Wnt and PI3K/AKT/mTOR signaling is required for murine EOC development,even in the presence of mutant p53.Mice with Apc À/À;Pten À/À;Pik3ca E545K /þEOCs Have Shortened Survival Compared with Mice with EOCs Lacking Mutant Pik3caIn our previous analysis of 21human EOCs with type I OvCA gene expression and mutational pro files,7had mutations predicted to dysregulate PI3K/AKT/mTOR signaling,including 3EOCs that had inactivating PTEN mutations in addition to activating PIK3CA mutations in exons 9or 20.14,37Because frequent mutations in PIK3CA exons 1to 7have been reported in endometrial adenocar-cinomas,38we subsequently evaluated these exons in our set of EOCs.One additional tumor (which also had mutant PTEN )was found to harbor two mutations in PIK3CA exon 1(R88Q and K111N;Cho laboratory).Both of these have previously been reported as gain-of-function muta-tions.39e 41To determine effects of mutant Pik3ca on our murine EOC tumor phenotype,Apc flox/flox ;Pten flox flox mice were crossed with mice in which a conditional mutant (LSL-E545K)allele is knocked into one of the endogenous Pik3ca loci.Ovarian bursal AdCre injection was used to induce tumors in Apc flox/flox ;Pten flox flox ;Pik3ca LSL-E545K /þand Apc flox/flox ;Pten flox flox ;Pik3ca þ/þlittermates.Mice with Apc À/À;Pten À/À;Pik3ca E545K /þtumors (n Z 11)had signi ficantly shorter survival (P Z 0.0003)and more frequent ascites and metastasis than littermate controls (n Z 11),with Apc À/À;Pten À/Àtumors lacking mutant Pik3ca (Figure 3A and Table 2).The addition of mutant Pik3ca had no appreciable effect on tumor morphology (Figure 3,B and C).As observed by Liang et al 42and Kinross et al,43activation of mutant Pik3ca alone (n Z 4)was insuf ficient to initiate tumors in the MOSE.However,in contrast to these other studies,we did not observe signi ficant epithelial hyperplasia in mice expressing only mutant Pik3ca in the MOSE (Figure 4A).Ovarian bursal injection of AdCre in Pten flox/flox ;Pik3ca LSL-E545K /þmice resulted in the development of nonepithelial hamartoma-like tumor masses (to be reported separately)in 9of 11mice similar to those reported in the soft tissue of humans with PTEN hamartoma tumor syndromes.44In addition,all 11Pten flox/flox ;Pik3ca LSL-E545K /þmice displayed micropapillary proliferation of the MOSE that resembled low-grade (type I)serous carci-noma between 9and 30weeks after AdCre administration (Figure 4,B and C).The hyperplastic epithelium showed strong expression of CK8,WT1,and PAX8,consistent with a Mulle-rian epithelial rather than mesothelial proliferation (Figure 4,D,E,and F).In addition,as expected,the hyperplastic epithelial cells were negative for Pten (Figure 4G)but showed elevated expression of p-AKT (Figure 4H).The changes inourFigure 2Mice bearing Apc À/À;Pten À/À;p53À/Àtumors have shortestsurvival and develop bulky metastatic disease.A :Kaplan-Meier survival curves of Apc flox/flox ;Pten flox flox ;Trp53flox/flox (n Z 9),Apc flox/flox ;Pten flox flox ;Trp53flox /þ(n Z 8),and Apc flox/flox ;Pten flox flox ;Trp53þ/þ(n Z 6)mice after ovarian bursal injection of AdCre.B :Image of representative tumor-bearing Apc flox/flox ;Pten flox flox ;Trp53flox/flox mouse showing grossly visible ovarian tumor (white star )and abdominal metastasis (black star ).Representative photomicro-graphs of H&E-stained sections showing primary ovarian tumor from Apc flox/flox ;Pten flox flox ;Trp53flox/flox mouse (C ),primary ovarian tumor from Apc flox/flox ;Pten flox flox ;Trp53flox /þmouse (D ),parenchymal liver metastasis (E ),and lung metastasis (F )in tumor-bearing Apc flox/flox ;Pten flox flox ;Trp53flox/flox mice.Immunohistochemical stains showing strong expression of E-cadherin (G )and absence of p53expression (H )in sections from a representative Apc À/À;Pten À/À;p53À/Àovarian tumor.Scale bars:100m m (C e H ).Models of Ovarian Cancer ProgressionThe American Journal of Pathology- 1395Pten flox/flox ;Pik3ca LSL-E545K /þmice are similar to the hyper-proliferative surface epithelium observed by Kinross et al 43after expression of Pik3ca H1047R in the MOSE.Mice with several other combinations of mutant Apc,Pten ,and Pik3ca alleles were also tested for their ability to form ovarian tumors after bursal AdCre injection (Table 2).In previous work,we showed that none of the 20Apc flox/flox ;Pten flox /þmice and only 1of the 20Apc flox/þ;Pten flox/flox mice developed OvCA after AdCre injection.14In this study,we found that AdCre induced OvCAs in two of five Apc flox /þ;Pten flox flox ;Pik3ca LSL-E545K /þmice,whereas the other three mice displayed hamartoma-like masses and micro-papillary proliferation of the MOSE similar to that observed inPten flox flox ;Pik3ca LSL-E545K /þmice.The carcinomas were not accompanied by hamartomatous lesions,presumably because the carcinomas progressed rapidly and may have overgrown early hamartomatous lesions.Similarly,four of six Apc flox flox ;Pten flox /þ;Pik3ca E545K /þmice developed OvCAs after AdCre injection.None of the Pten flox /þ;Pik3ca LSL-E545K (n Z 3)or Trp53R270/þ;Pik3ca LSL-E545K (n Z 4)mice developed ovarian surface epithelial alterations or tumors after AdCre injection within the 40-week surveillance period.Hence,tumor formation is most ef ficient when both alleles of Apc and Pten are inactivated,but addition of mutant Pik3ca increases the frequency of tumor formation when one copy of either Apc or Pten is intact.DiscussionUnderstanding differences in the biology and genetics of type I versus type II OvCAs is considered critical for identifying new therapeutic strategies that will improve outcome for patients with OvCA.Type I OvCAs are char-acterized as low-grade,slow-growing tumors that are more resistant to conventional chemotherapy but more likely to be responsive to hormonal therapy than their high-grade (type II)counterparts.45The type I tumors have a more favorable prognosis,largely because many are diagnosed when the tumors are con fined to the ovary and curable with surgical removal.It is important to emphasize that women with advanced-stage type I tumors have a poor prognosis.In a recent study of >600consecutive OvCAs,no signi ficant difference in progression-free or overall survival was observed between women with advanced-stage type I versus type II tumors.46Because many women with type I tumors will die of their disease,understanding the molecular events underlying type I tumor progression and metastasis and how type I and type II tumors differ are important goals.The dualistic model originally argued for the existence of two distinct pathogenetic pathways for OvCA development,but molecular and histopathologic analyses suggest that the two pathways are not mutually exclusive and that a sizeable subset of endometrioid (and smaller subset of serous)carcinomas may undergo type I /type II progression,perhaps associated with acquisition of TP53and/or PIK3CA mutations.The murine model systems described herein provide support for the adverse prognostic effects of TP53and PIK3CA mutations observed in human endometrial,ovarian,and other types of carcinomas.The murine EOCs that develop in our model system show morphologic features similar to their human counterparts,including distinct gland formation and occasional foci of squamous differentiation.Unlike typical primary human EOCs,the murine tumors also show areas of less-differentiated cells with spindle-cell morphology,possibly representing epithelial-mesenchymal transition.14It is dif ficult to compare survival of tumor-bearing mice with that of patients with EOC,in part because women with EOC are usually treated with surgical resection (with or without chemotherapy),andthisFigure 3Mice with Apc À/À;Pten À/À;Pik3ca E545K EOCs have shortenedsurvival.A :Kaplan-Meier survival curve of tumor-bearing Apc flox/flox ;Pten flox flox ;Pik3ca E545K /þ(n Z 11)and Apc flox/flox ;Pten flox flox (n Z 11)mice.Photomicrographs of representative primary OvCAs from Apc flox/flox ;Pten flox flox (B )and Apc flox/flox ;Pten flox flox ;Pik3ca E545K /þ(C )mice.Scale bars:100m m (B and C ).Wu et al-The American Journal of Pathology。
构建肺腺癌铁死亡预后模型
构建肺腺癌铁死亡预后模型摘要:目的:利用TCGA数据库构建肺腺癌铁死亡预后模型,旨在为肺腺癌患者预后提供理论依据。
方法:从TCGA数据库中下载肺腺癌及正常肺组织的基因表达谱及相关临床数据,筛选出与铁死亡相关的预后基因,运用LASSO回归模型构建铁死亡预后模型。
结果:单因素COX回归分析筛选出20个铁死亡相关基因,通过LASSO回归分析,最终筛选出13个铁死亡相关预后基因。
根据危险评分将患者分为高低风险组,与低风险组相比高风险组的总体生存率明显降低。
ROC曲线验证了模型的预测能力。
结论:ANGPTL7, DDIT4, SLC1A4, GDF15, SLC2A1,RRM2, ALOXE3, PHKG2, LINC00472, LINC00336, ALOX15,SLC7A11, 和GLS2的风险预测模型能够有效的对肺腺癌患者的预后进行预测。
关键词:肺腺癌铁死亡预后模型Construct a prognostic model of ferroptosis-related genes in lung adenocarcinomaMeng Cui, Jia Yang, Dan Liao, Lilong Cheng(Department of Medical laboratory, People's Hospital of Leshan, Leshan, Sichuan 614000)Abstract: Objective: A prognostic model of ferroptosis in lung adenocarcinoma was established by using TCGA database to provide theoretical basis for the prognosis of lung adenocarcinoma patients. Methods: the gene expression profiles and related clinical data of lung adenocarcinoma and normal lung tissues were downloaded from TCGA database, and the prognostic genes related to ferroptosis were screened. The prognosis model of ferroptosis-related gene was constructed by lasso regression model. Results: 20 ferroptosis-related genes were screened by univariate Cox regression analysis, and13ferroptosis-related prognostic genes were finally screened by lasso regression analysis. Patients were pided into high-risk and low-risk groups according to the risk score. Compared with the low-risk group, the overall survival rate of the high-risk group was significantly lower. ROC curve verifies the predictionability of the model. Conclusion: the risk prediction models of ANGPTL7, DDIT4,SLC1A4, GDF15, SLC2A1, RRM2, ALOXE3, PHKG2, LINC00472, LINC00336, ALOX15, SLC7A11 and GLS2 can effectively predict the prognosis of patients with lung adenocarcinoma.Key words: lung adenocarcinoma, ferroptosis , prognostic model肺癌是人类常见的恶性肿瘤,其发病率和死亡率已经位于癌症发病率和死亡率的首位,2020年全球新增肺癌患者220万,180万患者死于肺癌[1]。
四组细胞瘤块切片进行免疫组化分析...
图i克隆的鉴定A:RT--PCR鉴定结果B:westernblot鉴定结果I:阳性对照办2:MDA--MB--435/Ikbl(L),3:MDA一船一435几KBl(H)。
4:MDA一蛐--4352,5:MDA一船--435/vec图2各组细胞transwell细胞侵袭实验数据为4株肿瘤细胞3次独立实验的均数±标准差。
误差线代表标准误。
2图3各细胞MMP一2、删P一9、VEGF、BFGFmRNA的表达隋况A:RT—PCR检测结果l、2、3,4分别代表为MDA-MB-435细胞,MDA一蛐一435/vec细胞-叭—姗一435/LI(B1(H)细胞、D为MDA-MB-435/LKBl(L)细胞3图5MMP一2,删P一9活性的测定A:凝胶酶谱MMP-2,姗P一9检测结果1、2、3.4分别代表为MDA-惦一435细胞、MI)A-MB一435/vec细胞MI)A—MB-435/LKBl(H)细胞,D为~f1)A-14B一435/LKBl(L)细胞图4各细胞MMP一2、姗P一9、VEGF、BFGF蛋白的表达情况A:westernblot检测结果l、2,3,4分别代表为MDA-MB-435细胞、l¥)A-MB-435/vec细胞盼A一邺一435几I(Blm)细胞、D为如A-船一435/LKBl(L)细胞图6人乳癌裸鼠原位移植瘤情况MDA·MB-435/LKBl(L)MDA·MB·435/LKBl(H)MDA—MB一435NECMDA-MB-435图7各组鼠肺转移灶比较PuIm。
narymelastaslstum。
阽(HE’100)图8各移植瘤MMP一2、MMP--9、VEGF、BFGF蛋白的表达情况A:westernblot检测结果1、2、3代表MDA-MB-435细胞、4,5,6代表d4DA-MB-435/vec细胞7,8,9代表MDA-MB-435/LKBl(L)细胞、10,11.12代表hⅢA—m一435/u(Bl(H)细胞图9各组瘤组织微血管密度变化图10LKBl基因与微血管密度变化的关系LKB1基因与人乳腺癌细胞生长和侵袭的相关性研究作者:庄志刚学位授予单位:复旦大学1.Westerman AM.Entius MM.de Baar E Peutz-Jeghers syndrome:78-year follow-up of the original family 19992.Tiainen M.Ylikorkala A.Makela TP Growth arrest by the LKB1 tumor suppressor:induction ofp21(WAFl/CIP1) 2002(13)3.Shen Z.Wen n F The tumor suppressor gene LKB1 is assoclate with prognosis in human breast carcinoma 2002(07)4.Sapkota GP.Boudeau J.Deak M Phosphorylation of the protein kinase mutated in Peutz-Jeghers cancer syndrome,LKB1/STK11,at Ser431 by p90(RSK)and cAMP-dependent protein kinase,but not its farnesylation at Cys(433),is essential for LKB1 to suppress cell vrowth 2001(22)5.8apkota GP.Boudeau J.Deak M Identification and characterization of four novel phosphorylationsites(Ser31,Ser325,Thr336 and Thr366)on LKB1/STK11,the protein kinase mutated in Peutz-Jeghers cancer syndrome 2002(02)6.Forster LF.Defres S.Goudie DR An investigation of the Peutz-Jeghers gene(LKB1)in sporadic breast and colon cancers 20007.Chen J Lindblom Germline mutation screening of the STK11/LKB1 gene in familial breast cancer with LOH on 19p 20008.Kleiner DE.Stetler-Stevenson WG Matrix metalloproteinases and metastasis 19999.McCawley LJ.Matrisian LM Matrix metalloproteinases:they are not just for matrix anymore! 200110.Hojilla CV.Mohammed FF.Khokha R Matrix metalloproteinases and theirtissue inhibitors direct cell fate during cancer development 2003(10)11.Stamenkovic I Matrix metalloproteinases in tumor invasion andmetastasis 2000(06)12.John A.Tuszynski G The role of matrix metalloproteinases in tumor angiogenesis and tumor metastasis 2001(01)13.Freije JM.Balbin M.Pendas AM Matrix metalloproteinases and tumor progression 200314.Jones JL.Shaw JA.Pringle JH Primary breast myoepithelial cells exert an invasion-suppressor effect on breast cancer celis via paracrine down-regulation of MMP expression in fibroblasts and tumour cells 2003(04)15.Karuman P.Gozani O.Odze RD The Peutz-Jegher gene product LKB1 is a mediator of p53-dependent cell death[外文期刊] 200116.Wang JL.Sun Y.Wu S Gamma-irradiation induces matrix metalloproteinase Ⅱ expression in a p53-dependent manner 2000(04)17.Folkmen J Tumor angiogenesis:Therapeutic implication 197121.Tsutsui S.Kume M.Era S Prognostic value of microvessel density in invasive ductal carcinoma of the breast 2003(04)22.Weidner N.Semple J P.Welch W R.Folkman J Tumor angiogenesis and metastasis-correlat ion in invasive breast carcinoma 199123.Guidi AJ.Schnitt SJ.Fischer L Vascular permeability factor(vascular endothelial growthfactor)expresslon and anglogenesis in patients with ductal carcinoma in situ of the breast 199724.Ramanathan M.Giladi A.Leibovich SJ Regularion of vascularendothelial growth factor gene expression in murine macrophages by nitric oxide and hypoxia 2003(06)25.Ylikorkala A.Rossi DJ.Korsisaari N Vascular abnormalities and deregulation of VEGF in LKB1-deficient mice[外文期刊] 200126.Westerman AM.Entius MM.de Baar E Peutz-Jeghers syndrome:78-year follow-up of the original family 199927.Tiainen M.Ylikorkala A.Makela TP Growth arrest by the LKB1 tumor suppressor:induction ofp21(WAF1/CIP1) 2002(13)28.Jimenez AI.Fernandez P.Dominguez O Growth and molecular profile of lung cancer cells expressing ectopic LKB1:down-regulation of the phosphatidylinositol 3-phosphate kinase/PTEN pathway 2003(06)29.Ylikorkala A.Rossi DJ.Korsisaari N Vascular abnormalities and deregulation of VEGF in Lkb1-deficient mice[外文期刊] 200130.Miyoshi H.Nakau M.Ishikawa TO Gastrointestinal hamartomatous polyposis in lkb1 heterozygous knockout mice 2002(08)31.Rossi DJ.Ylikorkala A.Korsisaari N Induction of cyclooxygenase-2 in a mouse model of Peutz-Jeghers polyposis 2002(19)32.Bardeesy N.Sinha M.Hezel AF Loss of the Lkb1 tumor suppressor provokes intestinal polyposis but resistance to transformation 2002(6903)33.Nakau M.Miyoshi H.Seldin MF Hepatocelhlar carcinoma caused by loss of heterozygosity in lkb1 gene knockout mice 2002(16)34.Watts JL.Morton DG.Bestman j The C.elegans par-4 gene encodes a putative serine-threonine kinase required for establishing embryonic asymmetry35.Martin SG.St Johnston D A role for Drosophila LKB1 in anterior-posterior axis formation and epithelial polarity[外文期刊] 2003(6921)36.Baas AF.Boufeau J.Sapkota GP Activation of the tumour suppressor kinase LKB1 by the STE20-like pseudokinase STRAD 2003(12)37.Sapkota GP.Boudeau J.Deak M Phosphorylation of the protein kinase mutated in Peutz-Jeghers cancer syndrome,LKB1/STK11,at Ser431 by p90(RSK)and cAMP-dependent protein kinase,but not its farnesylation at Cys(433),is essential for LKB1 to suppress cell vrowth 2001(22)38.Sapkota GP.Boudeau J.Deak M Identification and characterization of four novel phosphorylation39.Smith DP.Rayter SI.Niederlander C LIP1,a cytoplasmic proteinfunctionally linked to the Peutz-Jeghers syndrome kinase LKB1 2001(25)40.Marignani PA.Kanai F.Carpenter CL LKB1 associates with Brg1 and is necessary for Brg1-induced growth arrest 2001(35)41.Karuman P.Gozani O.Odze RD The Peutz-Jegher gene product LKB1 is a mediator of p53-dependent cell death[外文期刊] 2001(06)42.Bignell GR.Barfoot R.Seal S Low frequency of somatic mutations in the LKB1/Peutz-Jeghers syndrome gene in sporadic breast cancer 1998(07)43.Forster LF.Defres S.Goudie DR An investigation of the Peutz-Jeghers gene(LKB1)in sporadic breast and colon cancers 2000(10)44.Chen J Lindblom Germline mutation screening of the STK11/LKB1 gene in familial breast cancer with LOH on 19p 2000(05)45.Shen Z.Wen n F The tumor suppressor gene Lkb1 is associate with prognosis in human breast carcinoma 2002(07)1.费菲原肌球蛋白-4在人类乳腺癌高低转移细胞株中的差异性表达及临床意义[学位论文]20052.沈赞抑癌基因LKB1在乳腺癌中的作用研究[学位论文]20023.王振军.严仲瑜.毕郭龙国人黑斑息肉病LKB1基因胚系突变的检测[期刊论文]-中华外科杂志2000,38 (2)4.董慧明上皮钙黏蛋白对人炎性乳腺癌细胞系生物学特性的影响[学位论文]20055.丁锦华微浸润在导管原位癌中的临床意义及MMP-2、Tenascin-C在导管原位癌中的表达[学位论文]20056.刘刚乳腺癌血管、淋巴管生成与转移预后的研究[学位论文]20037.张杰乳腺癌前哨淋巴结活检及骨髓播散肿瘤细胞的检测[学位论文]20058.王劲松Rab27A对人乳腺癌细胞生物学特性的影响及其机制的研究[学位论文]20079.宋科瑛耐药乳腺癌细胞MDR-MCF-7侵袭力增强机制探讨[学位论文]200310.李鹤成ERα和Her-2受体在人乳腺癌细胞株作用通路的实验研究及基质金属蛋白酶在淋巴结阴性乳腺癌的预后意义[学位论文]2005本文链接:/Thesis_Y952108.aspx。
分子肿瘤学3癌基因与抑癌基因
• 病毒癌基因常会出现碱基取代或碱基缺失
• 二者的同源序列有一定程度的差异,功能 上也有差异
2、细胞癌基因的特点:
1、广泛存在于生物界中; 2、基因序列高度保守; 3、它的作用通过其产物蛋白质来体现; 4、被激活后,可形成癌性的细胞转化基因。
3、原癌基因甲基化程度降低而激活,属于外基 因机制(epigenetic)
• DNA分子甲基化有稳定双螺旋结构,阻抑转录 的作用。如结肠腺癌和小细胞肺癌中,c-ras 基因比邻近正常组织中甲基化明显降低,导致 原癌基因激活
4、基因扩增(gene amplification)
原癌基因以某种不适当的方式被复制,拷贝增 多,过度表达。采用细胞内微注射法证实,正 常p21ras在高浓度时具有转化活性。
(6)核内转录因子:如c-myc ,l-myc等。 编码产物为反式作用因子,们于核内, 可与某些特定的DNA结合,影响复制、 转录,从而影响细胞的增殖、分化和凋 亡
(三)原癌基因活化的机制
1、逆转录病毒激活原癌基因 插入激活或插入致突变 转导激活
2、人类原癌基因的激活 点突变 基因易位(重排) 基因扩增 外基因机制
(1)Initiating Stage: 细胞受癌 性启动因子作用,DNA发生改变,成 为癌前细胞,但表型正常。各种干细 胞及处于分裂增殖中的细胞对启动因 子更敏感
(2)Promoting Stage: 在启动因子 和促癌因子的协同作用下,细胞出现 恶性。
• 启动因子:低剂量,一次接触,本身有 致癌性
激酶活性。
bcr
abl 9
22
t(9;22)(q34;q11)
9 bcr/abl
tumor suppressor genes
Tumor Suppressor GenesThe activation of cellular oncogenes represents only one of two distinct types of genetic alterations involved in tumor development; the other is inactivation of tumor suppressor genes. Oncogenes drive abnormal cell proliferation as a consequence of genetic alterations that either increase gene expression or lead to uncontrolled activity of the oncogene-encoded proteins. Tumor suppressor genes represent the opposite side of cell growth control, normally acting to inhibit cell proliferation and tumor development. In many tumors, these genes are lost or inactivated, thereby removing negative regulators of cell proliferation and contributing to the abnormal proliferation of tumor cells.Go to:Identification of Tumor Suppressor GenesThe first insight into the activity of tumor suppressor genes came from somatic cell hybridization experiments, initiated by Henry Harris and his colleagues in 1969. The fusion of normal cells with tumor cells yielded hybrid cells containing chromosomes from both parents (Figure 15.32). In most cases, such hybrid cells were not capable of forming tumors in animals. Therefore, it appeared that genes derived from the normal cell parent acted to inhibit (or suppress) tumor development. Definition of these genes at the molecular level came, however, from a different approach—the analysis of rare inherited forms of human cancer.Figure 15.32Suppression of tumorigenicity by cell fusion. Fusion of tumor cells with normal cells yields hybrids that contain chromosomes from both parents. Such hybrids are usually nontumorigenic.The first tumor suppressor gene was identified by studies of retinoblastoma, a rare childhood eye tumor. Provided that the disease is detected early, retinoblastoma can be successfully treated, and many patients survive to have families. Consequently, it was recognized that some cases of retinoblastoma are inherited. In these cases, approximately 50% of the children of an affected parent develop retinoblastoma,consistent with Mendelian transmission of a single dominant gene that confers susceptibility to tumor development (Figure 15.33).Figure 15.33Inheritance of retinoblastoma. Susceptibility to retinoblastoma is transmitted to approximately 50% of offspring. Affected and normal indivi- duals are indicated by purple and green symbols, respectively.Although susceptibility to retinoblastoma is transmitted as a dominant trait, inheritance of the susceptibility gene is not sufficient to transform a normal retinal cell into a tumor cell. All retinal cells in a patient inherit the susceptibility gene, but only a small fraction of these cells give rise to tumors. Thus, tumor development requires additional events beyond inheritance of tumor susceptibility. In 1971, Alfred Knudson proposed that the development of retinoblastoma requires two mutations, which are now known to correspond to the loss of both of the functional copies of the tumor susceptibility gene (the Rb tumor suppressor gene) that would be present on homologous chromosomes of a normal diploid cell (Figure 15.34). In inherited retinoblastoma, one defective copy of Rb is genetically transmitted. The loss of this single Rb copy is not by itself sufficient to trigger tumor development, but retinoblastoma almost always develops in these individuals as a result of a second somatic mutation leading to the loss of the remaining normal Rb allele. Noninherited retinoblastoma, in contrast, is rare, since its development requires two independent somatic mutations to inactivate both normal copies of Rb in the same cell.Figure 15.34Mutations of Rb during retinoblastoma development. In hereditary retinoblastoma, a defective copy of the Rb gene (Rb-) is inherited fromthe affected parent. A second somatic mutation, which inactivates the single normal Rb+ copy in a retinal cell, then (more...)The functional nature of the Rb gene as a negative regulator of tumorigenesis was initially indicated by observations of chromosome morphology. Visible deletions of chromosome 13q14 were found in some retinoblastomas, suggesting that loss (rather than activation) of the Rb gene led to tumor development (Figure 15.35). Gene-mapping studies further indicated that tumor development resulted from loss of normal Rb alleles in the tumor cells, consistent with the function of Rb as a tumor suppressor gene. Isolation of the Rb gene as a molecular clone in 1986 then firmly established that Rb is consistently lost or mutated in retinoblastomas. Gene transfer experiments also demonstrated that introduction of a normal Rb gene into retinoblastoma cells reverses their tumorigenicity, providing direct evidence for the activity of Rb as a tumor suppressor.Figure 15.35Rb deletions in retinoblastoma. Many retinoblastomas have deletions of the chromosomal locus (13q14) that contains the Rb gene.Although Rb was identified in a rare childhood cancer, it is also involved in some of the more common tumors of adults. In particular, studies of the cloned gene have established that Rb is lost or inactivated in many bladder, breast, and lung carcinomas. The significance of the Rb tumor suppressor gene thus extends beyond retinoblastoma, apparently contributing to development of a substantial fraction of more common human cancers. In addition, as noted earlier in this chapter, the Rb protein is a key target for the oncogene proteins of several DNA tumor viruses, including SV40, adenoviruses, and human papillomaviruses, which bind to Rb and inhibit its activity (Figure 15.36). Transformation by these viruses thus results, at least in part, from inactivation of Rb at the protein level rather than from mutational inactivation of the Rb gene.Figure 15.36Interaction of Rb with oncogene proteins of DNA tumor viruses. The oncogene proteins of several DNA tumor viruses (e.g., SV40 T antigen) induce transformation by binding to and inactivating Rb protein.Characterization of Rb as a tumor suppressor gene served as the prototype for the identification of additional tumor suppressor genes that contribute to the development of many different human cancers (Table 15.5). Some of these genes were identified as the causes of rare inherited cancers, playing a role similar to that of Rb in hereditary retinoblastoma. Other tumor suppressor genes have been identified as genes that are frequently deleted or mutated in common noninherited cancers of adults, such as colon carcinoma. In either case, it appears that most tumor suppressor genes are involved in the development of both inherited and noninherited forms of cancer. Indeed, mutations of some tumor suppressor genes appear to be the most common molecular alterations leading to human tumor development.Table 15.5Tumor Suppressor Genes.The second tumor suppressor gene to have been identified is p53, which is frequently inactivated in a wide variety of human cancers, including leukemias, lymphomas, sarcomas, brain tumors, and carcinomas of many tissues, including breast, colon, and lung. In total, mutations of p53 may play a role in up to 50% of all cancers, making it the most common target of genetic alterations in human malignancies. It is also of interest that inherited mutations of p53 are responsible for genetic transmission of a rare hereditary cancer syndrome, in which affectedindividuals develop any of several different types of cancer. In addition, the p53 protein (like Rb) is a target for the oncogene proteins of SV40, adenoviruses, and human papillomaviruses.Like p53, the INK4 and PTEN tumor suppressor genes are very frequently mutated in several common cancers, including lung cancer, prostate cancer, and melanoma. Two other tumor suppressor genes (APC and MADR2) are frequently deleted or mutated in colon cancers. In addition to being involved in noninherited cases of this common adult cancer, inherited mutations of the APC gene are responsible for a rare hereditary form of colon cancer, called familial adenomatous polyposis. Individuals with this condition develop hundreds of benign colon adenomas (polyps), some of which almost inevitably progress to malignancy. Inherited mutations of two other tumor suppressor genes, BRCA1and BRCA2, are responsible for hereditary cases of breast cancer, which account for 5 to 10% of the total breast cancer incidence.Additional tumor suppressor genes have been implicated in the development of brain tumors, pancreatic cancers, and basal cell skin carcinomas, as well as in several rare inherited cancer syndromes, such as Wilms' tumor. The number of identified tumor suppressor genes is rapidly expanding, and the characterization of these genes remains an active area of cancer research.Go to:Functions of Tumor Suppressor Gene ProductsIn contrast to proto-oncogene and oncogene proteins, the proteins encoded by most tumor suppressor genes inhibit cell proliferation or survival. Inactivation of tumor suppressor genes therefore leads to tumor development by eliminating negative regulatory proteins. In several cases, tumor suppressor proteins inhibit the same cell regulatory pathways that are stimulated by the products of oncogenes.The protein encoded by the PTEN tumor suppressor gene is an interesting example of antagonism between oncogene and tumor suppressor gene products (Figure 15.37). The PTEN protein is a lipid phosphatase that dephosphorylates the 3 position of phosphatidylinositides, such as). By dephosphorylating phosphatidylinositol 3,4,5-bisphosphate (PIP3PIP, PTEN antagonizes the activities of PI 3-kinase and Akt, which can 3act as oncogenes by promoting cell survival. Conversely, inactivation or loss of the PTEN tumor suppressor protein can contribute to tumordevelopment as a result of increased levels of PIP, activation of Akt,3and inhibition of programmed cell death.Figure 15.37Suppression of cell survival by PTEN. The tumor suppressor protein PTENat the 3 position of is a lipid phosphatase that dephosphorylates PIP3. PTEN thus counters the action of the oncogenes inositol, yielding PIP2PI 3-kinase and Akt, which promote cell (more...)Proteins encoded by both oncogenes and tumor suppressor genes also function in the Hedgehog signaling pathway (see Figure 13.44). The receptor Smoothened is an oncogene in basal cell carcinomas, whereas Patched (the negative regulator of Smoothened) is a tumor suppressor gene. In addition, the Gli proteins (the mammalian homologs of the Drosophila Ci transcription factor activated by Smoothened) were first identified as the products of an amplified oncogene.Several tumor suppressor genes encode transcriptional regulatory proteins. A good example is provided by the product of WT1, which is frequently inactivated in Wilms' tumors (a childhood kidney tumor). The WT1 protein is a repressor that appears to suppress transcription of a number of growth factor-inducible genes. One of the targets of WT1 is thought to be the gene that encodes insulin-like growth factor II, which is overexpressed in Wilms' tumors and may contribute to tumor development by acting as an autocrine growth factor. Inactivation of WT1 may thus lead to abnormal growth factor expression, which in turn drives tumor cell proliferation. Two other tumor suppressor genes, DPC4 and MADR2, encode SMAD family transcription factors that are activated by TGF-βsignaling and lead to inhibition of cell proliferation.The products of the Rb and INK4tumor suppressor genes regulate cell cycle progression at the same point as that affected by cyclin D1 (Figure 15.38).by repressing Rb inhibits passage through the restriction point in G1transcription of a number of genes involved in cell cycle progression and DNA synthesis (see Figure 14.20). In normal cells, passage through the restriction point is regulated by Cdk4/cyclin D complexes, which phosphorylate and inactivate Rb. Mutational inactivation of Rb in tumorsprogression. The INK4also regulates inhibits Cdk4/cyclin D activity. Inactivation of INK4therefore leads toelevated activity of Cdk4/cyclin D complexes, resulting in uncontrolled phosphorylation of Rb.Figure 15.38Inhibition of cell cycle progression by Rb and p16. Rb inhibits. Cdk4/cyclin D complexes progression past the restriction point in G1promote passage through the restriction point by phosphorylating and inactivating Rb. The activity of Cdk4/cyclin D is (more...)The p53gene product regulates both cell cycle progression and apoptosis (Figure 15.39). DNA damage leads to rapid induction of p53, which activates transcription of the Cdk inhibitor p21 (see Figure 14.21). The inhibitor p21 blocks cell cycle progression, both by acting as a general inhibitor of Cdk/cyclin complexes and by inhibiting DNA replication by binding to PCNA (proliferating cell nuclear antigen). The resulting cell cycle arrest presumably allows time for damaged DNA to be repaired before it is replicated. Loss of p53 prevents this damage-induced cell cycle arrest, leading to increased mutation frequencies and a general instability of the cell genome. Such genetic instability is a common property of cancer cells, and it may contribute to further alterations in oncogenes and tumor suppressor genes during tumor progression.Figure 15.39Action of p53. Wild-type p53 is required for both cell cycle arrest and apoptosis induced by DNA damage.In addition to mediating cell cycle arrest, p53is required for apoptosis induced by DNA damage. Unrepaired DNA damage normally induces apoptosisof mammalian cells, a response that is presumably advantageous to the organism because it eliminates cells carrying potentially deleterious mutations (e.g., cells that might develop into cancer cells). Cells lacking p53 fail to undergo apoptosis in response to agents that damage DNA, including radiation and many of the drugs used in cancer chemotherapy. This failure to undergo apoptosis in response to DNA damage contributes to the resistance of many tumors to chemotherapy. In addition, loss of p53 appears to interfere with apoptosis induced by other stimuli, such as growth factor deprivation and oxygen deprivation. These effects of p53 inactivation on cell survival are thought to account for the high frequency of p53 mutations in human tumors.Go to:Roles of Oncogenes and Tumor Suppressor Genes in Tumor DevelopmentAs discussed earlier, the development of cancer is a multistep process in which normal cells gradually progress to malignancy. The complete sequence of events required for the development of any human cancer is not yet known, but it is clear that both the activation of oncogenes and the inactivation of tumor suppressor genes are critical steps in tumor initiation and progression. Accumulated damage to multiple genes eventually results in the increased proliferation, invasiveness, and metastatic potential that are characteristic of cancer cells.The role of multiple genetic defects is best understood in the case of colon carcinomas, which have been studied extensively by Bert Vogelstein and his colleagues. These tumors frequently involve mutation of ras K oncogenes and inactivation or deletion of three distinct tumor suppressor genes—APC, MADR2,and p53. Lesions representing multiple stages of colon cancer development are regularly obtained as surgical specimens, so it has been possible to correlate these genetic alterations with discrete stages of tumor progression (Figure 15.40).Figure 15.40Genetic alterations in colon carcinomas. Inactivation of APC is an early event in tumor development, giving rise to a proliferative cell population. Mutations of ras K then frequently occur and are found in early-stage adenomas. Subsequent mutations of (more...)These studies indicate that inactivation of APC is an early event in tumor development. Genetic transmission of mutant APC genes in patients with familial adenomatous polyposis results in abnormal colon cell proliferation, leading to the outgrowth of multiple adenomas in the colons of affected patients. Mutations of APC also occur frequently in patients with noninherited colon carcinomas and are generally detected at early stages of the disease process. Mutations of ras K genes then appear to occur, and ras K oncogenes are also frequently present in small and intermediate-size adenomas. In contrast, the MADR2 and p53 tumor suppressor genes are inactivated usually at later stages of tumor progression. Mutations in these genes are only rarely found in early-stage adenomas, but they are frequently present in advanced adenomas and malignant carcinomas. The loss of these tumor suppressor genes in colon cancers thus appears to be involved in later stages of progression to malignancy, rather than in the initial stages of tumor formation.Although these genetic alterations most often occur in the order described here, this is not an obligatory sequence of events. For example, mutations in p53are sometimes detected in early adenomas. More important than the order in which mutations occur is the fact that colon cancer ultimately results from accumulated damage to multiple genes. Accumulated damage toboth oncogenes and tumor suppressor genes similarly appears to be responsible for the development of other types of cancer, including breast and lung carcinomas. The progressive loss of growth control that is characteristic of cancer cells is thus thought to be the end result of abnormalities in the products of multiple genes that normally regulate cell proliferation, differentiation, and survival.BoxMolecular Medicine: Herception: Cancer Treatment Targeted against the erbB-2 Oncogene. Breast cancer is the most common cancer among women, accounting for nearly one-third of all cancers diagnosed in women in the United States. In the year 2000, it is (more...)By agreement with the publisher, this book is accessible by the search feature, but cannot be browsed.。
肿瘤基因治疗的基本策略和常用方法
c. 制备肿瘤KNA疫苗 采用编码特异抗原的基因直接注入人体,通过其在机体内的表达从而激发机体对编码抗原的免疫反应。应用癌胚抗原(CEA)制备的DNA疫苗在实验中显示出一定效果,研究的前景可观,进入临床药理实验只是时间问题而毋庸置疑 。
2 恶性肿瘤的病因性基因治疗
肿瘤病因性基因治疗主要针对癌基因和抑癌基因,其治疗策略是抑制、阻断癌基因的表达或替代,恢复抑癌基因的功能。从而达到治疗肿瘤的目的。
02
与基因治疗有关几个概念
c.抑癌基因正常细胞中存在抑制肿瘤发生的基因,称为抑癌基因。抑癌基因的存在从最初的一个发现中得到证实,即一个肿瘤细胞和一个正常细胞融合成的杂交细胞,往往不具有细胞表型,即使两种不同的肿瘤细胞杂交,也可能是非肿瘤性的,只有正常的亲代失去某些基因之后,其子代才会发生肿瘤。后来,通过限制性内切酶片段长度多态分析及分子克隆技术的研究证实,正常细胞中有一类对细胞增殖起负性调节作用的基因,如大肠癌中的p53基因,视网膜母细胞瘤和骨肉瘤中的Rb基因。这两种基因也是目前被确认并研究最为深人的抑癌基因。如将正常的Rb基因转人骨肉瘤细胞,Rb基因表达增高,细胞分裂几乎完全抑制,将Rb基因转人前列腺细胞,对正常细胞的生长无影响,但抑制了细胞的癌变,所 以当这一类的抑癌基因发生丢失,失活或变异时,就会促进细胞恶变。
基因毒癌发生发展的第一步是活化致癌物,改变细胞内的DNA,导致原癌基因的移位及扩增,这些特殊基因的翻译,使带有这些异常基因细胞的某些特性得以专一表达,一个特殊的化学物对一系列细胞有诱变及DNA修复作用者,称为基因毒。目前人类已知的致癌物均为基因毒。
01
原癌基因(前癌基因)细胞癌基因存在于细胞基因组内,激活前的细胞癌基因称为原癌基因(前癌基因),一旦原癌基因激活成癌基因便会引起细胞癌变,癌基因激活后,通过其表达产物改变细胞生长和分化规律,诱发细胞癌变。细胞癌基因被激活的途径很多,如启动子插人,基因重排和扩增,基因点突变和DNA分子中去甲基化等。
2-3 抑癌基因与肿瘤 (tumor suppressor gene)
抑癌基因(tumor suppressor gene)概述:抑癌基因:在正常情况下,细胞内的一类基因产物能抑制细胞生长并能潜在抑制细胞癌变,当通过纯合缺失或失活可引起恶性转化的基因群,又称为隐性癌基因、抗癌基因或肿瘤易感基因。
抑癌基因的作用:在调控细胞生长、增殖及分化过程中起负调控作用,并能潜在地抑制肿瘤的生长。
如果其功能失活或出现基因缺失、突变等异常,可导致细胞恶性转化而发生肿瘤。
抑癌基因的生物学功能与癌基因相反。
抑癌基因理论上的三个基本条件:1.该恶性肿瘤的相应正常组织中该基因必须正常表达。
2.该恶性肿瘤中这种基因应有功能失活或结构改变或表达缺陷。
3.将这种基因的野生型导入基因异常的肿瘤细胞内,可部分或全部改变其恶性表型。
抑癌基因的功能:1、诱导终末分化2、维持基因稳定3、触发衰老,诱导细胞程序性死亡。
4、调节细胞生长5、抑制蛋白酶活性6、改变DNA甲基化酶活性7、调节组织相容性抗原8、调节血管形成9、促进细胞间联系。
第一节抑癌基因研究的历史回顾1.细胞融合试验是抑癌基因存在的最早依据。
当正常细胞与肿瘤细胞融合形成杂交细胞时,能抑制癌细胞的致癌性,表明正常细胞具有抑制细胞癌变的基因成分。
2. 1971年Knudson系统地研究了有明显显性遗传的儿童视网膜母细胞瘤,提出了著名的肿瘤“两次突变”的假说,认为在有遗传倾向的病人体内所有干细胞及体细胞都存在一种突变,在此基础的发育过程中任一视网膜母细胞再出现第二次突变,即可导致肿瘤发生。
Rb基因是源于父母双方的两个视网膜母细胞瘤相关的等位基因。
肿瘤形成时,13号染色体(13q1.4)的两个等位基因常常同时缺乏或失活,表明这类基因具有抑制肿瘤形成作用。
抑癌基因在正常机体中可抑制细胞增殖,当杂合性丢失(loss off heterozygosity,LOH)发生时(即两个等位基因同时改变),就会引起肿瘤细胞无限制增殖。
3.在肿瘤细胞中常发现有等位基因的杂合型丢失,据此以发现新的抑癌基因但是不能把所有能抑制细胞生长作用的因素都称为抑癌基因,如干扰素、生长抑素等。
模块1第9周周测试题1下列哪种为癌基因Aras基因Bp53基因C
模块1第9周周测试题1. 下列哪种为癌基因:A. ras基因B. p53基因C. Rb基因D. Apc基因E. NF-1基因:答案:A2. 下列哪些肿瘤不与病毒感染密切相关有:A. 肝细胞肝癌B. 宫颈癌C. 鼻咽癌D. 皮肤癌E. Burkitt淋巴瘤答案:D3. 关于肿瘤的论述,下列除哪些是正确的:A. 肿瘤的发生是多阶段、多步骤的长期过程B.肿瘤的发生与原癌基因的激活和抑癌基因的失活有关C. 肿瘤的发生是单个基因突变的结果D. 肿瘤的发生是多因素造成的答案:C4. 下列不属于抑癌基因的是:A.bcl-2B.p53C.nm23D.pRbE.p16答案:A5. 下列哪些细胞黏附分子可能与肿瘤转移的器官选择有关A.整合素B.E-钙粘素C.选择素D.免疫球蛋白超家族E.CD44答案:C6. 下列哪些分子在肿瘤中高表达提示预后良好A.MMP和TIMPB.erbB2和P16C.TIMP 和KAI1D.Ras 和MycE.以上都不是答案:C7. 骨肉瘤的好发部位是:A.头面骨B.脊椎骨C.扁骨D.长骨E.指(趾)骨答案:D8. 皮下一肿块,边界清楚,有包膜,质硬,灰白色,切面性质均匀一致,无继发改变,可见纤细的编织状条纹,该肿物可能诊断为:(A)A.纤维瘤B.纤维肉瘤C.平滑肌瘤D.平滑肌肉瘤E.脂肪瘤答案:A9. 肉瘤的肉眼特点通常为:A.结节状、有包膜,切面灰白质硬B.结节状、有包膜,切面灰白质软C.分叶状,可有或无包膜,切面灰红或粉红色,质硬D.形状不定,切面灰白色,质硬,干燥E.不规则结节状,有无完整包膜不定,切面灰红色,湿润质软如鱼肉状答案:E10. 以下哪项是癌与肉瘤的主要区别点:A.发病年龄B.大体特点C.组织结构D.恶性程度E.转移途径答案:C11. 血道转移发生较早的癌是:A.绒毛膜癌B.鼻咽癌C.肝癌D.肺癌E.胃癌答案:A12. 膀胱最常见的恶性肿瘤为:A.腺癌B.鳞状细胞癌C.移行细胞癌D.纤维肉瘤E.平滑肌肉瘤答案:C13. 高分化鳞癌的组织学特点是:A.实质与间质分界清楚B.无细胞间桥C.基底膜完整D.有癌巢形成E.有角化珠形成答案:E14. 胃癌癌细胞穿透浆膜脱落于腹膜上形成的转移瘤称为:A.远处转移B.接触性转移C.肿瘤直接蔓延D.种植性转移E.移植性转移答案:D15. 下列哪项为纤维腺瘤的特征:A.多呈分叶状B.包膜不完整,切除后易复发C.易恶变D.瘤实质由增生的腺上皮和纤维组织组成E.常发生于卵巢答案:D16. 下列哪项不属于细胞异型性:A.核数目不一B.核肿胀,核膜不清C.核染色质分布不均D.核大小不等,核畸形E.病理性核分裂象答案:B17. 最能体现腺癌的特点的是:A.发生于腺上皮B.癌细胞呈腺样排列C.癌巢形成D.呈结节样外观E.异型性明显答案:B18. 下图是哪种疾病的镜下表现:A. 大肠腺癌B. 骨肉瘤C. 肝癌(间变癌)D. 何杰金氏病E. 食管鳞癌答案:E19. 骨肉瘤的主要诊断依据是:A.好发于青少年B.血道转移C.发生于长骨骨干D.出现肿瘤性骨质E.可发生病理性骨折答案:D20. 良性肿瘤的异型性主要表现在:A.瘤细胞的多形性B.细胞核的多形性C.组织结构的异型性D.病理性核分裂E.细胞异型性答案:C21. 乳头状瘤的乳头中心(轴索)的成分是:A.癌细胞B.脉管C.纤维结缔组织D.癌细胞和纤维结缔组织E.纤维结缔组织和脉管答案:E22. 来源于三个胚层组织的肿瘤称:A.癌肉瘤B.混合瘤C.畸胎瘤D.错构瘤E.胚胎瘤答案:C23. Which of the following mechanisms may be involved in rejection of a tumor:A. T cell-mediated cytotoxicityB. ADCCC. Complement-dependent cytotoxicityD. All of the above.E. None of the above.答案:D24. 人HPV与下列哪种肿瘤的发生有关:A. 宫颈癌B.乳腺癌C.黑色素瘤D.肝癌E.前列腺癌答案:A25. 机体的抗肿瘤免疫效应机制中起主导作用的是:A. 体液免疫B.细胞免疫C.巨噬细胞杀伤作用D. NK细胞杀伤肿瘤E. 细胞因子杀瘤作用答案:B26. 以下对NK细胞杀瘤有关叙述,错误的是:A.无特异性B.无需预先活化,即可直接杀瘤C.可依赖抗体通过ADCC方式杀瘤D.依赖补体,通过CDC方式杀瘤E.无MHC限制性答案:D27. 关于Tumor-Associated Antigens (TAA)最准确的描述:A.They are antigens found only in tumor cellsB.They can be found in both tumor and some normal cells, but the level issignificantly higher in the tumor cellsC.They are typically the products of tumor suppressor genes; an example wouldbe RbD.They are the result of genes mutated by DNA-damaging carcinogens such asMNNGE.They are typically encoded by oncogenic viruses such as EBV答案:B28. 什么是抗原调变:A. 肿瘤细胞表面抗原表位减少或丢失,从而使肿瘤细胞避免宿主免疫系统的杀伤B. 肿瘤细胞表面的抗原与正常细胞的膜分子差异很小C. 肿瘤细胞MHC-Ⅰ类抗原表达下降,逃避CTL的攻击D. 肿瘤细胞不表达MHC-II类抗原E. 以上都不是答案:A29. 肿瘤细胞常高表达下述分子:A. MHC I类分子B. MHC II类分子C. CD80D. CD86E. TGF- 、IL-10等免疫抑制分子答案:E30.肿瘤局部的Gr-1+CD11b+髓系来源抑制性细胞主要发挥下述作用:A. 杀伤肿瘤细胞B. 激活树突状细胞C. 参与肿瘤免疫逃逸D. 产生抗体E. 协助CTL抗肿瘤答案:C31.Which of the following statements is most true regarding therapy with anti-tumor antibodies?A. Their use is currently under investigation, but none have yet been approved for clinical useB. They are important reagents in the application of LAK (Lymphokine-activated killers) therapy to tumorsC. Their use requires the prior pulsing of dendritic cells with B7 and/or IL-2.D. They can be conjugated with toxins or radioactive components in order to deliver a toxic dose specifically to tumor cellsE. They are particularly useful in tumors which have been selected to express very high levels of MHC molecules答案:D32.长春新碱使肿瘤细胞较多的处于增殖周期的:A.S期B.G0期C.G1期D.G2期E.M期答案:E33.异型性与分化之间的关系:A.异型性小,分化程度高B.异型性大,分化程度高C.异型性小,分化程度低答案:A34.抑制二氢叶酸还原酶的抗肿瘤药是:A.顺铂B.阿霉素C.环磷酰胺D.5-氟尿嘧啶E.甲氨蝶呤答案:E35.以下属于S期特异性的抗肿瘤药物是:A.喜树碱B.博莱霉素C.甲氨蝶呤D.左旋门冬酰胺酶E.鬼臼毒素答案:C36.以下属于M期特异性抗肿瘤药物是:A.长春新碱B.环磷酰胺C.鬼臼毒素D.左旋门冬酰胺酶E.甲氨蝶呤答案:A37. 博莱霉素主要用于治疗:A.慢性粒细胞白血病B.急性淋巴细胞白血病C.绒毛膜上皮癌D.前列腺癌E.鳞状上皮癌答案:E38.肿瘤的转移方式为:A.血行B.淋巴C.种植性D.A、B和C答案:D39.请问以下大体标本是:A.肝癌B.肺结节C.肺转移性肝癌答案:C40.39 题中肿瘤的转移方式为:A.血行B.淋巴C.种植性D.A、B和C答案:A。
p53讲演(英文修改)
the p53 protein binding to DNA
p53蛋白在避免癌症发生中扮演着重要的角色,例 蛋白在避免癌症发生中扮演着重要的角色, 蛋白在避免癌症发生中扮演着重要的角色 如,细胞凋亡 (apoptosis) 、基因组稳定性 (genetic stability) 、抑制血管新生 (angiogenesis) 。 受损时, 蛋白能活化 蛋白能活化DNA修复蛋白 当DNA受损时,p53蛋白能活化 受损时 修复蛋白 (DNA repair proteins)。 。 p53蛋白能抑制细胞生长周期 蛋白能抑制细胞生长周期(Inhibition of cell cycle) 蛋白能抑制细胞生长周期 停留于G1/S的节律点上,以达成 的节律点上, 损坏辨识。 若能 停留于 的节律点上 以达成DNA损坏辨识。 (若能 损坏辨识 将细胞于此节律点上停留够久, 将细胞于此节律点上停留够久,DNA修护蛋白将有更 修护蛋白将有更 充裕的时间修复DNA损坏部位,并继续细胞生长周 损坏部位, 充裕的时间修复 损坏部位 期。) 若细胞的DNA受损且不能修复,p53蛋白将起 受损且不能修复, 蛋白将起 若细胞的 受损且不能修复 动细胞凋亡程序, 动细胞凋亡程序,避免使不正常的细胞继续分裂生 活化的p53蛋白能接合 蛋白能接合DNA,促使多个基因表 长。活化的 蛋白能接合 , 包括基因WAF1/CIP1,其为 蛋白之编码基 达,包括基因 ,其为p21蛋白之编码基 因。当p21蛋白 (WAF1) 与CDK2形成复合体时,细 蛋白 形成复合体时, 形成复合体时 胞将无法进入到细胞分裂阶段。 而突变后的p53蛋 胞将无法进入到细胞分裂阶段。 而突变后的 蛋 白将可能丧失与DNA形成有效结合的能力,导致 形成有效结合的能力, 白将可能丧失与 形成有效结合的能力 p21蛋白无法形成,不能发出停止细胞分裂的信号, 蛋白无法形成, 蛋白无法形成 不能发出停止细胞分裂的信号, 致使受损细胞不受控制的进行细胞分裂,最终形成 致使受损细胞不受控制的进行细胞分裂, 肿瘤。 肿瘤。
神经胶质瘤的研究意义
1神经胶质瘤的研究意义神经胶质瘤( Glioma)是最常见的颅内肿瘤,约占所有颅内肿瘤的50%左右。
目前临床上 IV 型神经胶质瘤是恶性程度最高的脑部肿瘤,至今无法治愈[1]。
多形性胶质母细胞瘤(Glioblastoma Multiforme, G BM) 起源于脑部神经胶质细胞,成年脑癌患者中约 80%为浸润性的星形细胞瘤。
由于该肿瘤是浸润性生长物,其和正常脑组织没有明显的界限,因此难以被完全切除或根本不能手术。
血脑屏障的存在,又使得化学药物和一般抗肿瘤的中药难以发挥疗效。
该肿瘤细胞对放疗亦不甚敏感,非常容易复发。
因此脑胶质瘤至今仍是全身肿瘤中预后最差的肿瘤之一。
据文献报道神经胶质瘤的中位生存时间和无进展生存时间分别为 14.6个月和 6.9个月, 5 年生存率为 9.8%2。
近年来,原发性恶性脑肿瘤发生率逐年递增,年增长率达到每 100,000人口中有 5.26 个人,而且每年有 17000人被最新诊断出患有该致死性疾病[3, 4]。
因此,积极研究神经胶质瘤的发病原因、寻找恶性胶质瘤治疗切实有效的治疗手段已成为目前亟待解决的关键问题。
和(或)抑癌基因改变的细胞;肿瘤的发展(包括由分化较好的肿瘤2 上皮 -间质转化与神经胶质瘤发生发展的关系人脑胶质瘤发生、发展过程中,多种癌基因、抑癌基因的异常改变牵涉其中。
肿瘤的形成和发展与基因改变间的关系是近年来肿瘤学研究的核心问题之一。
多种基因的改变都参与到肿瘤细胞生长过程中,肿瘤细胞生长是肿瘤最基本的生物学行为,生长异常是肿瘤恶性行为的基础。
大量的研究证实,肿瘤的形成起源于单个获得了癌基因转变成分化差的肿瘤,局部浸润和远处转移)则是更多癌基因和抑癌基因受累后作用叠加,肿瘤细胞克隆选择和扩增的最终结果3 4 5 6。
在胶质瘤发生发展的大量研究中,人端粒酶逆转录酶( hTERT)[6]、内皮生长因子受体( EGFR)7、热休克蛋白( HSP)8 9 10等分子以及 PKA、PKC、ERK[9, 10]等关键信号转导分子均在神经胶质瘤的发生发展过程中发挥重要作用。
癌症
癌症摘要:癌症即恶性肿瘤,是正常细胞生长失控而造成的疾病。
原癌基因被激活,抑癌基因的缺失或失活引起癌症基因的突变导致癌症的发生。
癌症可以通过一些科学合理的方法预防,减少癌症的发生。
关键词:癌肿瘤细胞生长原癌基因预防动物体内因分裂调节失控而无限增殖的细胞称为肿瘤细胞。
肿瘤可分为良性肿瘤与恶性肿瘤,前者是一群仅局限于原有位置、不侵染周围其他组织的肿瘤细胞,后者是一群不局限于原有位置、有转移能力的肿瘤细胞[1]。
肿瘤细胞与正常细胞不同,丧失了正常的结构和功能,形态上趋于一致,表现出某些未分化细胞的特征,是一群失去正常调控的“不死”的永生细胞[2]。
上皮组织的恶性肿瘤称为癌。
目前癌细胞已作为恶性肿瘤细胞的通用名称。
1 癌基因1.1 癌基因癌基因(oncogene) 是指编码产物在体内引起肿瘤、在体外引起细胞转化的一类基因,也称转化基因。
早在1910 年,Rous 就发现鸡肉瘤的无细胞滤液(含肉瘤病毒) 能使鸡体诱发新的肉瘤,证明肉瘤病毒基因组含致癌基因。
此后的研究表明,Rous 肉瘤病毒基因组是一条单链RNA ,只含几个结构蛋白基因和一个癌基因( src 基因) 。
在分类上属于逆转录病毒科。
Rous 肉瘤病毒侵染细胞后,以逆转录方式大量繁殖并诱发细胞癌变。
该病毒基因组中的src 基因编码产物是分子量为60KD 的蛋白质,故称做p60src ,具有酪氨酸蛋白激酶(TPK) 活性,能使多个靶蛋白的酪氨酸残基发生磷酸化,促使细胞癌变。
目前已鉴定了几十种逆转录病毒的癌基因。
现已了解,各种生物细胞的基因组中普遍存在着与病毒癌基因(v - onc) 相似的序列,不过在正常情况下它们不表达或低水平表达,对细胞无害;但在某些情况下被激活而异常表达时,则导致细胞癌变。
由于在正常细胞中的癌基因是以非活化形式存在的,故常称为原癌基因(pro -oncogene) ;当原癌基因被激活后,称细胞癌基因(c - onc) ,不过两者也经常作为同义词出现于文献中。
葡萄糖转运蛋白
人类不同肿瘤和癌细胞系中已发现5种葡萄糖转运子亚型,Glut-1是惟一的几乎存在于所有癌细胞系中的亚型,并已发现在许多人类肿瘤中均有高表达,与肿瘤最为密切相关。
Glut-1的过度表达是恶性细胞在生长转化过程中对其所处的特殊生理微环境的反应。
Chung JK,Lee YJ,Kim C,el a1.Mechanisms related to[ F]fluoro de-oxyglucose uptake of human colon cancers transplanted in nude mice[J].J Nucl Med,1999,40:339.Glut-1过度表达可转运更多的葡萄糖以满足恶性细胞高代谢率和快速生长的需要。
体内外的实验表明,Glut-1对18F-FDG的吸收起主要作用。
Brown RS,Leung JY,Kison PV,et a1.Glucose transporters andFDG up-take in untreated primary hmnan non-small cell lung cancer. J Nucl Med,1999,40 :556一.葡萄糖转运蛋白1(glucose transporter 1,GLUT-1)是一种组织细胞进行跨膜转运葡萄糖的重要载体,在哺乳动物胚胎和成熟组织中低水平表达,但在缺氧及缺血的恶性肿瘤细胞中表达显著增高,且与肿瘤进展、患者预后有着一定关系。
研究发现,不同部位GLUT-1的表达程度与肿瘤的关系存在着一定的差异,例如:在食管癌中,GLUT-1表达的高低和肿瘤细胞的浸润程度、淋巴结转移以及病理分级都有着一定相关性;在肺癌中,GLUT-1表达与其分期有关,越是晚期的肿瘤相应的GLUT-1表达阳性率越高;在眼鳞状细胞癌细胞中,GLUT-1表达强弱与肿瘤的等级及细胞的增殖呈正相关¨;在胰腺导管腺癌中,通过GLUT-1的检测能为其早期判断肿瘤的恶性程度、生存率高低提供重要信息J。
癌基因、抑癌基因与生长因子
RAS MYC FOS、JUN
作用 PDGF-2 FGF同类物,促进细胞增殖 EGF受体,促进细胞增殖 EGF受体类似物,促进细胞增殖 M-CSF受体、SCF受体,促增殖 与受体结合转导信号 在细胞内转导信号 MAPK通路中的重要分子
MAPK通路中的重要分子 促进增殖相关基因表达 促进增殖相关基因表达
P.Rous
目录
•Rous:1911年,从母鸡身上的恶性结缔 组织(肉瘤)中提取滤液,接种到健康 的鸡体内,鸡患上同样的肿瘤。 •表明:滤液中含致病原,可传播肿瘤。 •1932年,Shope发现,野生棉尾兔的皮 肤肿瘤也可借助无细胞滤液传播。
4
目录
•病毒致癌理论:即传播肿瘤的无 细胞滤液中含的是病毒(src)。 •劳斯(Rous),获1966年诺贝尔生 理学和医学奖
目录
CML大致可分为三个阶段:
慢性期: 从初发阶段到暴发期,骨髓内 异常细胞数目一直上升,而且扩散至骨 髓外。慢性期可潜伏4到5年,在这一阶 段,患者症状很轻,往往只在例常的血 检时才被检测发现。
加速期:在6到18个月之间,这时白血球 的扩增往往已难用常规疗法控制。
暴发期:患者只能存活1到6个月。
在某些因素(如放射线、某些化学物质等)作用下, 癌基因一旦被激活,发生数量上或结构上的变化时,就 会形成癌性的细胞转化基因。
目录
功能上相关的癌基因家族分类
1. src家族:产物多具有酪氨酸蛋白激酶活性, 能促进增殖信号的转导。
2. ras家族:编码小G 蛋白p21,参与cAMP水平 的调节。
3. myc家族:编码核内DNA结合蛋白,调节其 他基因转录的作用。
多种肿瘤
Rb、骨肉瘤 胶质瘤、膀胱癌、前列腺癌、子宫内
乳腺病理研究生简况登记表(按入学年份排序)
李妤
牛昀
硕士 2000-2003
博士期间:1.中心体异 常与乳腺癌前病变及 其癌变关系的研究(国 家自然科学基金资助 项目). 2. 中心体调 控因子 Nek2 及其剪接 异构体在乳腺上皮细 胞恶性转化中的作用 (国家自然科学基金 资助项目) 3 Centrosome Amplification in Human Breast Tumors (中心体与乳腺肿瘤) 4 Role of Aurora-A in the development of EMT and metastases in breast cancer (Aurora-A 在乳腺癌 上皮间质转换及远处 转移中的作用)美国 Mayo clinic foundation 细胞周期调控因子与 乳腺细胞癌变的相关 性研究(天津市科委自 然基金项目 9936078110)
1. 刘铁菊 CyclinD1、p16 与乳腺癌相关性的研究进展. 癌 症, 2002,21(3):330-332 2. 刘铁菊,牛昀,于泳,吕阿娟 杨晓东. 乳腺轻、重度乳 头状瘤病和导管内癌中 p16 蛋白表达的比较观察. 中国肿瘤
天津医科大学 附属肿瘤医院
天津医科大学 附属肿瘤医院
2002-2007 河 北唐山市工人 医院病理科 2008.10-至今
韦丽,牛昀 乳腺癌细胞中心体调控研究新进展,中国肿瘤临 床. 2005,32(14):838-840
1. 张媛媛,付丽. p53 基因治疗研究的新进展. 2006, 35(1):48-50.
天津医科大学 总医院肾科
北京人民医院 内科;北京大 学团委书记
博士 2004-2007
博士 2004-2007
王颖, 牛昀 方志沂 中心体在肿瘤发生中的作用. 肿瘤防 治研究(核心期刊),2004,11(5):143-146.
形成肿瘤 英语
形成肿瘤英语Tumor FormationCancer is a complex and multifaceted disease that arises from the uncontrolled growth and division of cells within the body. At its core, the development of a tumor, or neoplasm, is a consequence of genetic and epigenetic alterations that disrupt the normal mechanisms governing cell proliferation, differentiation, and death. These changes can lead to the accumulation of abnormal cells, which can then invade and damage surrounding tissues, and potentially spread to other parts of the body, a process known as metastasis.The initiation of tumor formation is often attributed to the activation of oncogenes, which are genes that promote cell growth and division, and the inactivation of tumor suppressor genes, which normally function to inhibit cell proliferation and promote cell death. Oncogenes can be activated through various mechanisms, such as mutations, amplifications, or chromosomal rearrangements, while tumor suppressor genes can be inactivated by mutations, deletions, or epigenetic silencing.One of the hallmarks of cancer is the ability of tumor cells to evadethe body's natural defense mechanisms, such as the immune system. Tumor cells can develop strategies to avoid detection and destruction by the immune system, allowing them to continue to proliferate and spread. This can involve the production of immunosuppressive factors, the recruitment of regulatory T cells, or the downregulation of molecules that are recognized by the immune system.Another key aspect of tumor formation is the ability of cancer cells to sustain their own growth and proliferation. This can be achieved through various mechanisms, such as the activation of growth factor signaling pathways, the acquisition of self-sufficiency in growth signals, and the evasion of growth-inhibitory signals. Additionally, cancer cells can also develop the capacity to resist cell death, or apoptosis, which is a normal process that eliminates damaged or abnormal cells.The tumor microenvironment, which includes the surrounding stromal cells, blood vessels, and extracellular matrix, also plays a crucial role in the development and progression of cancer. Tumor cells can actively remodel their microenvironment, promoting angiogenesis (the formation of new blood vessels) to ensure a steady supply of nutrients and oxygen, and recruiting various stromal cells, such as fibroblasts and immune cells, to create a supportive and pro-tumorigenic niche.The metastatic potential of cancer cells is another important aspect of tumor formation. Metastasis occurs when cancer cells break away from the primary tumor site, enter the bloodstream or lymphatic system, and establish new tumors in distant organs. This process is facilitated by the acquisition of certain characteristics, such as the ability to degrade the extracellular matrix, to invade through the basement membrane, and to survive in the circulatory system.The development of a tumor is a multistep process that often takes place over an extended period of time. The initial genetic and epigenetic alterations that lead to the transformation of a normal cell into a cancer cell are typically followed by the clonal expansion of these cells, the acquisition of additional mutations, and the gradual evolution of the tumor towards a more aggressive and invasive phenotype.It is important to note that the specific mechanisms underlying tumor formation can vary greatly depending on the type of cancer, the genetic and environmental factors involved, and the individual patient's response to treatment. Advances in our understanding of the molecular and cellular mechanisms driving cancer have led to the development of targeted therapies and personalized treatment approaches, which aim to more effectively combat this complex and devastating disease.In conclusion, the formation of a tumor is a multifaceted process that involves the dysregulation of various cellular pathways and the interplay between the tumor cells and their surrounding microenvironment. By elucidating the intricate mechanisms underlying cancer development, researchers and clinicians can continue to develop more effective strategies for the prevention, early detection, and treatment of this disease, ultimately improving the lives of those affected by it.。
肺腺癌糖酵解过程基因
肺腺癌糖酵解过程基因英文回答:The glycolysis process is a metabolic pathway that converts glucose into pyruvate, generating ATP and NADH in the process. In lung adenocarcinoma, several genes play a role in regulating the glycolysis process.One of the key genes involved in the glycolysis process in lung adenocarcinoma is the glucose transporter 1 (GLUT1) gene. GLUT1 is responsible for transporting glucose into the cancer cells. It is often overexpressed in lung adenocarcinoma, allowing the cancer cells to take up more glucose for energy production through glycolysis.Another important gene in the glycolysis process is pyruvate kinase M2 (PKM2). PKM2 is an isoform of pyruvate kinase, an enzyme that catalyzes the final step of glycolysis. In lung adenocarcinoma, PKM2 is often upregulated, leading to increased glycolytic flux. PKM2also has additional functions beyond glycolysis, including promoting cell proliferation and survival.Hypoxia-inducible factor 1-alpha (HIF-1α) is a transcription factor that regulates the expression of several glycolytic genes, including GLUT1 and PKM2. In lung adenocarcinoma, HIF-1α is often stabilized and upregulated, leading to increased expression of glycolytic genes and promoting glycolysis.In addition to these genes, other factors such as oncogenes and tumor suppressor genes can also affect the glycolysis process in lung adenocarcinoma. For example, mutations in the tumor suppressor gene TP53 can lead to increased glycolysis by upregulating the expression of glycolytic genes.Overall, the dysregulation of genes involved in the glycolysis process contributes to the metabolic reprogramming observed in lung adenocarcinoma. Understanding the role of these genes can provide insights into the development of targeted therapies for this type ofcancer.中文回答:肺腺癌糖酵解过程中的基因有多个。
转录激活因子3在消化系统肿瘤中的研究进展
转录激活因子3在消化系统肿瘤中的研究进展弋东敏;房新辉;韩双印;李健【摘要】转录激活因子3(activating transcription factor,ATF3)属于转录因子ATF/CREB家族的成员,是一种适应反应基因,参与多种细胞活动的调控以适应细胞内外环境的变化.近年来的研究表明,ATF3作为抑癌或致癌基因在消化道系统肿瘤的发生及演变过程中发挥重要的作用.现就ATF3在消化系统肿瘤方面的研究进展作一概述.%Activating transcription factor 3 ( ATF3 ) is a member of the ATF/CREB transcription factor family and a kind of adaptive reaction gene. It involved in a variety of regulating cell activity to adjust to changes in the extra- and intra-cellular environment. It has been identified as either an oncogene or a tumor suppressor and plays an important role in the occurrence and progression of the digestive malignant tumor. The research advances of ATF3 in digestive malignant cancer were reviewed.【期刊名称】《胃肠病学和肝病学杂志》【年(卷),期】2017(026)011【总页数】4页(P1305-1308)【关键词】转录激活因子3;消化系统肿瘤【作者】弋东敏;房新辉;韩双印;李健【作者单位】河南省人民医院郑州大学人民医院消化内科,河南郑州450003;河南省人民医院郑州大学人民医院消化内科,河南郑州450003;河南省人民医院郑州大学人民医院消化内科,河南郑州450003;河南省人民医院郑州大学人民医院消化内科,河南郑州450003【正文语种】中文【中图分类】R735转录激活因子 3 (activating transcription factor3, ATF3)是一种适应反应基因,最初由Hai等[1]在HaLa细胞中分离而得。
- 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)。
Downloaded from on July 26, 2010 - Published by Cold Spring Harbor Laboratory PressThe tumor suppressor Smad4/DPC4 and transcriptional adaptor CBP/p300 are coactivators for Smad3 in TGF- β-induced transcriptional?activationXin-Hua Feng, Ying Zhang, Rui-Yun Wu, et al. Genes Dev. 1998 12: 2153-2163 Access the most recent version at doi:10.1101/gad.12.14.2153ReferencesThis article cites 37 articles, 16 of which can be accessed free at: /content/12/14/2153.full.html#ref-list-1 Article cited in: /content/12/14/2153.full.html#related-urlsEmail alerting serviceReceive free email alerts when new articles cite this article - sign up in the box at the top right corner of the article or click hereTo subscribe to Genes & Development go to: /subscriptionsCold Spring Harbor Laboratory PressDownloaded from on July 26, 2010 - Published by Cold Spring Harbor Laboratory PressThe tumor suppressor Smad4/DPC4 and transcriptional adaptor CBP/p300 are coactivators for Smad3 in TGF- -induced transcriptional activationXin-Hua Feng, Ying Zhang, Rui-Yun Wu, and Rik Derynck1Departments of Growth and Development and Anatomy, and Programs in Cell Biology and Developmental Biology, University of California, San Francisco, California 94143-0640 USASmads regulate transcription of defined genes in response to TGF- receptor activation, although the mechanisms of Smad-mediated transcription are not well understood. We demonstrate that the TGF- -inducible Smad3 uses the tumor suppressor Smad4/DPC4 and CBP/p300 as transcriptional coactivators, which associate with Smad3 in response to TGF- . The association of CBP with Smad3 was localized to the carboxyl terminus of Smad3, which is required for transcriptional activation, and a defined segment in CBP. Furthermore, CBP/p300 stimulated both TGF- - and Smad-induced transcription in a Smad4/DPC4-dependent fashion. Smad3 transactivation and TGF- -induced transcription were inhibited by expressing E1A, which interferes with CBP functions. The coactivator functions and physical interactions of Smad4 and CBP/p300 with Smad3 allow a model for the induction of gene expression in response to TGF- . [Key Words: TGF- signaling; Smad; CBP/p300; transcription; PAI-1 promoter]Received January 23, 1998; revised version accepted May 8, 1998.Members of the transforming growth factor (TGF- ) superfamily regulate cell growth and differentiation through their ability to induce or repress transcription of various genes, including cell-cycle control genes. TGFinduces and/or stabilizes the formation of a cell-surface heteromeric receptor complex consisting of type I and II receptors, both of which are transmembrane serine/ threonine kinases (Derynck and Feng 1997). In response to TGF- binding, the constitutively active type II receptor kinase (T RII) phosphorylates and activates the TGF- type I receptor (T RI) (Wrana et al. 1994; Chen and Weinberg 1995). The intracellular responses such as growth inhibition and extracellular matrix production are specified by the kinase domain of the type I receptor (Feng and Derynck 1997). Activated type I receptors phosphorylate and thus activate the intracellular signaling mediators, named Smads, which relay TGF- signaling into the nucleus (Derynck and Zhang 1996; Wrana and Attisano 1996; Heldin et al. 1997; Massague et al. 1997). There are three ´ subgroups of Smads: ligand responsive (e.g., Smad1, Smad2, Smad3, Smad5, and Smad8), shared signaling (e.g., the tumor suppressor Smad4/DPC4 in vertebrates, Medea in Drosophila, and Sma-4 in Caenorhabditis elegans), and inhibitory (e.g., Smad6 and Smad7 in verte1 Corresponding author. E-MAIL derynck@; FAX (415) 476-1499.brates and Dad in Drosophila) (Heldin et al. 1997). Among ligand-responsive Smads, Smad1 and Smad5 respond to bone morphogenetic proteins (BMPs), whereas Smad2 and Smad3 are activated by TGF- and activin (Heldin et al. 1997). Smad2 and Smad3 are able to associate with the T RI/T RII receptor complex and are carboxy-terminally phosphorylated by activated T RI (Macıas-Silva et al. 1996; Zhang et al. 1996). Activated ´ Smad2/3 forms a heteromeric complex with Smad4 and is then translocated into the nucleus (Macıas-Silva et al. ´ 1996; Nakao et al. 1997; Zhang et al. 1997). In Xenopus, Smad2/4 complexes have been shown to associate through a DNA-binding protein with two different activin-responsive elements (Candia et al. 1997; Chen et al. 1997), whereas in Drosophila Mad binds directly to a promoter sequence (Kim et al. 1997). Although these interactions with a promoter are thought to be of critical importance, the underlying mechanism for transcriptional activation is poorly understood. Transcription from the promoter for plasminogen activator inhibitor type I (PAI-1) is strongly induced by TGF- and is often used as a marker for TGF- responsiveness in mammalian cells (Keeton et al. 1991). Coexpression of the TGF- -responsive Smad2 or Smad3 and Smad4 also induces strongly transcription from the PAI-1 promoter (Zhang et al. 1996, 1997). However, the respective roles of these two Smads in transcriptional activation are unclear and whether and how Smad3 andGENES & DEVELOPMENT 12:2153–2163 © 1998 by Cold Spring Harbor Laboratory Press ISSN 0890-9369/98 $5.00; 2153Downloaded from on July 26, 2010 - Published by Cold Spring Harbor Laboratory PressFeng et al.Smad4 interplay with the transcriptional machinery is unknown. We now show that CBP/p300 and Smad4 act as coactivators for the transcription factor Smad3 through TGF- -inducible direct physical interactions. Results and Discussion The transcription activity of Smad3 is TGFinducible and requires its carboxy-terminal SSXS motif Smad3 synergizes with Smad4/DPC4 to induce a high level transcription from the PAI-1 promoter, and overexpression of carboxy-terminally truncated Smad3 or Smad4 results in dominant-negative inhibition of TGF-induced transcription from this promoter (Zhang et al. 1996). Because Smad3 or Smad2 associates directly with Smad4 in response to TGF- (Nakao et al. 1997) and the heteromeric complex interacts with the promoter to induce transcription (Candia et al. 1997; Chen et al. 1997), we characterized the role of Smad3 and Smad4 in TGF-induced transcription. Smad3 was fused to the GAL4 DNA-binding domain, which confers nuclear localization (Silver et al. 1984), and was accordingly localized in the nucleus (data not shown). The transcriptional activity of GAL–Smad3 from a heterologous GAL4 promoter was low, but increased about 15-fold in response to TGF- (Fig. 1A). Whereas the structurally closely related Smad2 also had a TGF- -dependent transcriptional activity, Smad4 had only minimal activity both in the absence or presence of TGF- . These data are consistent with the ability of Smad3 and the inability of Smad4 to activate transcription in yeast, that is, in the absence of endogenous Smads (Wu et al. 1997). The basal activity of GAL–Smad4 may be caused by functional cooperativity with endogenous Smad2 or Smad3. Receptor activation results in phosphorylation of the two distal serines in the carboxy-terminal SSXS sequence of Smad1, Smad2, or Smad3 (Abdollah et al. 1997; Souchelnytskyi et al. 1997). Replacement of all three serines with alanines renders these Smads biologically inactive, which may be caused by impaired release from the receptor, lack of heteromerization with Smad4, and/ or inability to translocate into the nucleus (Macıas-Silva ´ et al. 1996; Kretzschmar et al. 1997). To investigate the role of ligand-induced phosphorylation in transcriptional activity of Smad3, we replaced the two distal serines in GAL–Smad3 with alanines, thus generating GAL– Smad3(2SA). This mutation abolished the TGF- -induced transcriptional activity of Smad3 (Fig. 1A), even though GAL–Smad3(2SA) was localized in the nucleus (data not shown). In contrast, replacement of the two serines with aspartic acids in Smad3(2SD), thus providing negative charges similar to those of phosphoserines, conferred a high level of constitutive transcriptional activity, which could still be further enhanced by TGF- . Thus, the carboxy-terminal negative charges, resulting from receptor-mediated phosphorylation, may be required for the ligand-responsive Smads to adopt a TGF-induced conformation for their optimal transcriptional activity. The role of the carboxy-terminal phosphoryla-tion in transcriptional activation complements its role in heteromerization with Smad4 and subsequent nuclear translocation. The transactivation activity of Smad3 is potentiated by Smad2 or Smad3 and Smad4/DPC4 The carboxyl domain of Smad4 forms a homotrimer (Shi et al. 1997), thus raising the possibility that Smads function as homotrimers. Homomerization of Smad3 may be required for biological activity (Hata et al. 1997; Wu et al. 1997), and Smad2 and Smad3 interact with each other (Wu et al. 1997) and synergize in their biological activity (Nakao et al. 1997). To evaluate the role of oligomerization in transcriptional activity of Smad3, we coexpressed GAL–Smad3 with Smad2 or Smad3 and measured its transcriptional activity. Smad2 and Smad3 enhanced the ligand-independent and -dependent transcriptional activity of GAL–Smad3 (Fig. 1B), without altering the extent to which GAL–Smad3 was localized in the nucleus (data not shown). In contrast, the BMP-2/4-responsive Smad1 did not affect the transcriptional activity of Smad3. Our data thus suggest that the transcriptional activity of Smad3 is enhanced by oligomerization with TGF- -responsive Smad2 or Smad3. Coexpression of Smad3(2SA) or Smad3(2SD), however, decreased the transcriptional activity of GAL–Smad3 (Fig. 1B). Because Smad3(2SA) was transcriptionally inactive (Fig. 1A), oligomerization of wild-type Smad3 with this mutant may result in an inactive complex; alternatively, the association of Smad3(2SA) with T RI may prevent carboxy-terminal phosphorylation of GAL–Smad3. On the other hand, Smad3(2SD) had a higher transcriptional activity than wild-type Smad3; its inhibitory effect on GAL–Smad3 is therefore less likely caused by the formation of an inactive complex, but could result from sequestration of endogenous Smad4, which interacts with and coactivates Smad3 (see below). Because Smad4 has no transcriptional activity on its own (Fig. 1A; Wu et al. 1997), its presence in the transcription complex (Candia et al. 1997; Chen et al. 1997) raises the possibility that it may act as a transcriptional coactivator for ligand-responsive Smads. As shown in Figure 1, C and D, Smad4 strongly increased the transcriptional activity of GAL–Smad3 in the absence or presence of TGF- . However, Smad4 was not essential for transcriptional activity of GAL–Smad3, as assessed in SW480.7 cells, which lack Smad4 (Goyette et al. 1992) (Fig. 1D). In the absence of Smad4, the transcriptional activity of GAL–Smad3 was still induced by coexpressing an activated T RI receptor (Fig. 1D), consistent with the notion that receptor-mediated carboxy-terminal phosphorylation enhances the transcriptional activity of Smad3 (see above). Reciprocally, the minimal transcriptional activity of GAL–Smad4 was moderately enhanced by coexpressing Smad2 or Smad3, but not Smad1 (data not shown), and the carboxy-terminal serine mutations in Smad3 abolished this synergy (Fig. 1E). Taken together, Smad4 has only minimal transcriptional activity by itself and acts as a coactivator of Smad3 in the tran-2154GENES & DEVELOPMENTDownloaded from on July 26, 2010 - Published by Cold Spring Harbor Laboratory PressTranscriptional cooperation of Smad and CBPFigure 1. Transcriptional activity of Smad3 and effect of interactions with Smad2, Smad3, and Smad4 on Smad3-mediated transcription. (A) Smad3 is a TGF- -inducible transcriptional activator. HepG2 cells were transfected with plasmids encoding the indicated GAL–Smad and the transcriptional activity from the cotransfected GAL4–luciferase reporter plasmid was measured. Assays were performed in the presence or absence of TGF- . (B) Smad2 and Smad3 potentiate the transactivation activity of GAL–Smad3. RI14 cells were cotransfected with pFR–Luc and the plasmid encoding GAL–Smad3, and expression plasmids for the indicated Smads or mutants. (C) Smad4 is a potent transcriptional coactivator for Smad3. RI14 cells were cotransfected with pFR–Luc, pGAL–Smad3, and indicated amounts of a Smad4 expression plasmid. (D) Smad4 is not essential for transcriptional activity of Smad3. Smad4-deficient SW480.7 cells were cotransfected with pFR– Luc and pGAL–Smad3, without (open bars) or with (solid bars) T RI (act.). The transcriptional activity was measured in the absence (open bars) or presence (black bars) of a coexpressed constitutively active T RI. (E) Smad2 and Smad3 stimulate the low transactivation activity of pGAL–Smad4. Mv1Lu cells were cotransfected with pGAL–Smad4 and pFR–Luc, and expression plasmids for the indicated Smads or relevant mutants. (A–C,E) (Open bars) −TGF- ; (solid bars) +TGF- . Note the lower scale of luciferase activity, when compared to A–D.scription complex. This coactivator role of Smad4 may complement a role in ligand-induced Smad nuclear translocation. CBP/p300 functions as a Smad4-dependent transcriptional coactivator for Smad3 CBP (CREB-binding protein) and the structurally closely related p300 act as transcriptional coactivators for andinteract with multiple transcription factors, including CREB, c-Jun, c-Fos, and basal transcription factor TFIIB (Goldman et al. 1997; Shikama et al. 1997). To evaluate whether CBP or p300 plays a role in TGF- -induced transcription and Smad function, we tested whether p300 and CBP could regulate the transcriptional activity of Smad3. In GAL4-based transcription assays, CBP and p300 both increased the transcriptional activity of Smad3 in the presence or absence of TGF- (Fig. 2A).GENES & DEVELOPMENT2155Downloaded from on July 26, 2010 - Published by Cold Spring Harbor Laboratory PressFeng et al.Figure 2. CBP/p300 functions as a transcriptional coactivator for Smad3. (A) CBP and p300 potentiate the transcriptional activity of GAL–Smad3. RI14 cells were cotransfected with pGAL–Smad3 and pFR–Luc, and indicated amounts of expression plasmids for CBP or p300. (B) Smad4 is required for efficient coactivation of Smad3 by CBP. SW480.7 cells were cotransfected with pGAL–Smad3 and pFR–Luc, and indicated amounts of an expression plasmid for Smad4, in the absence (open bar) or presence (hatched bar) of an expression plasmid for CBP. Transfected cells were treated with TGF- and luciferase values were measured. (C) CBP stimulates Smad3/4-induced transcription from the PAI-1 promoter in the absence or presence of TGF- . HepG2 cells were cotransfected with the PAI-1 luciferase reporter p800luc, and indicated combinations of expression plasmids for Smad3, Smad4, and CBP. (D) CBP and p300 stimulate TGF- -induced transcription from the PAI-1 promoter. Mv1Lu cells were cotransfected with the PAI-1 luciferase reporter p800luc, and indicated amounts of expression plasmids for CBP or p300. (E) Smad3 and Smad4 stimulate the transactivation activity of CBP. Mv1Lu and SW480.7 cells were cotransfected with pGAL–CBP(1678–2441), pFR–Luc, and expression plasmids for Smad3 and/or Smad4. (F) The −732 to −635 segment of the PAI-1 promoter mediates TGF- - and Smad3/4-inducible transcription. Mv1Lu cells were cotransfected with the PAI-1 luciferase reporter pGL5P/97, and indicated combinations of expression plasmids for Smad3 and Smad4. (G) p300, Smad3, and Smad4 participate in a complex assembled at the PAI-1 promoter. Nuclear extracts, prepared from 293 cells, were incubated with the 32P-labeled, 97-bp TGF- - and Smad3/4-inducible segment of the PAI-1 promoter. Free DNA, DNA–protein (shift), and supershifted (SS) complexes are marked and the nuclear lysates and antibodies are also shown. (A,C–F) (Open bars) −TGF- ; (solid bars) +TGF- .This coactivation of Smad3 by CBP was not observed in Smad4-deficient SW480.7 cells (Fig. 2B) and expression of Smad4 not only increased the transcriptional activity of Smad3, consistent with its role as coactivator, but alsoallowed CBP to transactivate Smad3 (Fig. 2B). We then examined whether CBP/p300 could also potentiate transcription from the PAI-1 promoter in response to TGF(Fig. 2C,D). Consistent with the ability of CBP to trans-2156GENES & DEVELOPMENTDownloaded from on July 26, 2010 - Published by Cold Spring Harbor Laboratory PressTranscriptional cooperation of Smad and CBPactivate Smad3, CBP expression increased transcription from the PAI-1 promoter in response to Smad3 and Smad4 (Fig. 2C), and in response to TGF- (Fig. 2C,D). In reciprocal experiments, we evaluated the ability of Smad3 and Smad4 to regulate the transcriptional activity of CBP. The carboxy-terminal segment (amino acids 1678–2441) mediates transcriptional activity of CBP (Kwok et al. 1994); thus GAL–CBP(1678–2441)-mediated transcription from the GAL4-promoter (Fig. 2E). In the absence of Smad4, that is, in SW480.7 cells, Smad3 increased the transcription activity of CBP, but this increase was not further enhanced by TGF- , unless Smad4 was expressed (Fig. 2E). Thus, Smad4 provided TGF- inducibility and enhanced the transcription activity of GAL–CBP, presumably in cooperation with endogenous Smad3. Accordingly, the transcriptional activity of CBP was enhanced by TGF- in Mv1Lu cells and by increased Smad3 or Smad4 expression (Fig. 2E). Finally, Smad3 and Smad4 synergized to increase the ligand-independent and -dependent transcriptional activity of CBP (Fig. 2E). Taken together, our data indicate that CBP transactivates Smad3, but efficient transactivation requires Smad4, and that Smad3 and Smad4 cooperate to transactivate CBP and provide TGF- inducibility to its transcriptional activity. The TGF- -dependent transcriptional activity of CBP in Mv1Lu cells is thus likely regulated by endogenous Smad2/3 and Smad4.ported to induce supershifts. The complex formation of Smad3 and CBP/p300 at the PAI-1 promoter was also confirmed in gel supershift analyses using tag-specific antibodies and nuclear extracts of cells transfected with tagged Smad3 and CBP/p300 (lanes 17–20). We also determined that Smad3 and Smad4 interact with the same promoter fragment. Thus, cells coexpressing Smad3 and Smad4 together (lane 10) showed a complex of the same size as the TGF- -inducible, Smad3-dependent complex (lanes 4,17) and this complex, which reacted with antiSmad3 (lanes 5,6,18) or anti-p300 (lanes 7,19), was supershifted using anti-Smad4 antibody (lane 11). The antiSmad4-supershifted complex also reacted with the antip300 antibody (lane 13), although its intensity was decreased and only a slight supershift occurred, which is not surprising considering the large size of the complex of the DNA fragment associated with p300 and the Smad3/4 heterohexamer. The anti-Smad3(N-19) antibody interfered with the formation of the anti-Smad4supershifted complex and the anti-Smad4/anti-p300 complex (lanes 12 vs. 11,14 vs. 13). These data thus suggest that p300 (or CBP), Smad3, and Smad4 interact in a TGF- -dependent manner in a nucleoprotein complex associated with the TGF- - and Smad3/4-responsive 97bp PAI-1 promoter segment.Smad3 and CBP associate with each other in a TGF- -dependent manner The functions of CBP/p300 and Smad4 as coactivators of Smad3 in TGF- -induced transcription and their coexistence at the TGF- - and Smad3/4-responsive PAI-1 promoter segment strongly suggested a pattern of physical interactions. As shown in Figure 3A and in previous studies (Lagna et al. 1996; Nakao et al. 1997), Smad4 associates directly with Smad3 or Smad2 in a TGF- dependent manner. This heteromeric association is mediated by the conserved carboxyl domains of the Smads (Hata et al. 1997; Wu et al. 1997). To investigate whether Smad3 interacts with CBP in response to TGF- , we performed coimmunoprecipitation analyses using transfected cells. No association with CBP was observed without receptor stimulation, but TGF- receptor activation resulted in interaction of Smad3, but not Smad4, with CBP (Fig. 3B). Smad1 did not associate with CBP, which is consistent with its responsiveness to BMP-2/4 and not to TGF- (Hoodless et al. 1996; Liu et al. 1996; Kretzschmar et al. 1997). Furthermore, the carboxy-terminal segment (amino acids 1678–2441) of CBP associated with Smad3 in response to TGF- stimulation, but this interaction was not seen with Smad3(2SA) (Fig. 3C). This lack of association of Smad3(2SA) could be caused by a decreased level of nuclear translocation and/or may reflect the role of the TGF- -induced carboxy-terminal phosphorylation of Smad3 in the interaction with CBP, which is consistent with the role of phosphorylation in the transcriptional activity of Smad3 (Fig. 1A). Mammalian two-hybrid analyses were used to evalu-Smad3, Smad4, and p300 interact in a nucleoprotein complex with the PAI-1 promoter The coactivation of Smad3 by CBP/p300 prompted us to test whether Smad3 and p300/CBP interact with a 97-bp PAI-1 promoter fragment (nucleotides −732 to −635), which confers TGF- and Smad3/4 responsiveness (Fig. 2F). Nuclear extracts from 293 cells provided a cleaner background than those from Mv1Lu or HepG2 cells in gel shift and supershift analyses using the 97-bp probe (data not shown). Whereas untransfected cells did not clearly show a gel-shifted complex (Fig. 2G, lanes 1,2), a TGF- -dependent DNA–protein complex was detected in transfected cells expressing Smad3 (lanes 3,4), suggesting that this complex contained Smad3. This complex was specific for the 97-bp PAI-1 promoter segment, as it competed with a 25-fold excess of cold probe, but not with unrelated DNA (lanes 15,16). An anti-Smad2/3 antibody (N-19) abolished the TGF- -inducible complex (lane 5), whereas another anti-Smad3 antibody (I-20) supershifted the complex (lane 6). Control antibodies did not abolish or supershift the complex (e.g., lane 10). The TGF- -inducible, Smad3-dependent complex was supershifted by an antibody against endogenous p300 (lane 7), which has been validated in supershift experiments (Avantaggiati et al. 1997). Addition of anti-Smad3(N-19) antibody abolished this anti-p300-supershifted complex (lane 8), whereas anti-Smad3(I-20) antibody further decreased the electrophoretic mobility of the supershifted complex (lane 9). No anti-CBP antibodies have been re-GENES & DEVELOPMENT2157Downloaded from on July 26, 2010 - Published by Cold Spring Harbor Laboratory PressFeng et al.Figure 3. TGF- -dependent association of CBP with Smad3 and cooperation of Smad4. (A) TGF- -dependent association of Smad3 and Smad4 . RI14 cells were transfected with Flag-tagged Smad3 and HA-tagged Smad4, and treated with (+) or without (−) TGF- . Cell lysates were immunoprecipitated (IP) with anti-Flag antibody, followed by immunoblotting (IB) with anti-HA antibody to detect Smad3-bound Smad4, or with anti-Flag immunoblotting to demonstrate equal expression of Smad3. (B) Smad3 coimmunoprecipitates with CBP after TGF- -receptor activation. COS-1 cells were transfected with Flag-tagged Smads as shown and HA-tagged CBP, in the absence or presence of a plasmid for an activated T RI. Immunoprecipitation with anti-Flag antibody was followed by anti-HA immunostaining to detect Smad-bound CBP. The control panel shows the expression levels of Smad3 and Smad4. (C) TGF- -dependent CBP–Smad3 interaction requires the carboxy-terminal SSXS site of Smad3. RI14 cells were transfected with the indicated combination of plasmids expressing GAL–CBP(1678–2441) and Flag–Smad3 or Flag–Smad3(2SA). Immunoprecipitation with anti-Flag antibody was followed by immunostaining with anti-GAL4 (BabCO) to detect Smad-bound CBP. The control panel shows the Smad3 expression levels. (D) TGF- -dependent interaction of Smad3 and Smad4 with CBP in mammalian two-hybrid assays. Plasmids for GAL-fused CBP segments, in combination with VP16–Smad plasmids, as indicated, were transfected into RI14 cells, together with the luciferase reporter plasmid pFR–Luc. The interactions were measured by luciferase expression in the absence (−, open bars) or presence (+, solid bars) of TGF- . GAL–DNA-binding domain is the control containing only the GAL4 DNA-binding domain, not fused to a CBP segment. (E) Smad4 enhances the association of Smad3 with CBP in mammalian two-hybrid assays. Smad4-deficient SW480.7 cells were cotransfected with plasmids for GAL-fused CBP segments and VP16–Smd3, in the absence (open bars) or presence (solid bars) of an activated T RI plasmid, and the CBP–Smad3 association was scored in the absence or presence of coexpressed Smad4.ate the ability of defined CBP segments to interact with Smad3 or Smad4 in Mv1Lu cells. As shown in Figure 3D, the interaction of CBP with Smad3 was localized to two segments, a weakly interacting amino-terminal segment (amino acids 1–451) and the much stronger interacting carboxy-terminal segment (amino acids 1891–2441). These interactions were strongly enhanced by TGF- , thus confirming the TGF- -inducible coimmunoprecipitation of CBP and Smad3. Accordingly, when individual CBP domains were coexpressed with Smad3, CBP (1891– 2441), but not the other segments, coimmunoprecipitated with Smad3 in response to TGF- (Fig. 3C; data not shown). Smad4 also showed ligand-inducible interaction with the two CBP segments in two-hybrid assays in Mv1Lu cells (Fig. 3D). This is in contrast with the lack of Smad4–CBP interaction in coimmunoprecipitation (Fig. 3B) and yeast two-hybrid (see below) experiments, suggesting that this interaction is mediated through the ligand-dependent association of Smad4 with endogenous Smad3, which in turn interacts in a ligand-dependent fashion with CBP. In addition, coexpression of Smad4 in SW480.7 cells increased the interaction of Smad3 withthe amino- and carboxy-terminal domains of CBP (Fig. 3E), whereas coexpression of Smad3 promoted the association of Smad4 and CBP in mammalian two-hybrid assays (data not shown). Our results thus suggest a ternary protein complex, whereby the ligand-dependent interaction of Smad3 with CBP (primarily its carboxy-terminal segment) is stabilized by Smad4. This interpretation is consistent with the participation of all three proteins in a nucleoprotein complex at the promoter (Fig. 2G; see above). The stabilization by Smad4 may be required for the ability of CBP to efficiently coactivate Smad3, as illustrated in Fig. 2B. The direct association of Smad3 and CBP is mediated by the carboxy-terminal domains of both proteins The interaction of CBP with Smad3 was also analyzed using yeast two-hybrid assays (Fig. 4A). Smad3 was only observed to interact with the carboxy-terminal segment of CBP that comprised amino acids 1891–2441, but not with amino acids 1992–2441, thus indicating a requirement of amino acids 1891–1991. The interaction of2158GENES & DEVELOPMENTDownloaded from on July 26, 2010 - Published by Cold Spring Harbor Laboratory PressTranscriptional cooperation of Smad and CBPFigure 4. The association of CBP and Smad3 is mediated by carboxy-terminal domains of both proteins. (A) Yeast two-hybrid assays demonstrate the interaction of Smad2 and Smad3, but not Smad1 and Smad4, with carboxy-terminal sequences (aa 1891–2441) of CBP. Smad–CBP interactions were detected by measuring -galactosidase activity. (−) Lack of detectable interaction; (++++) very strong interaction. The structural organization of CBP is shown with the previously characterized location of sequences required for interaction with the proteins shown. Our results now allow the localization of sequences required for association with Smad2 and Smad3. (B) Localization of the CBP-interacting sequences in Smad3 using yeast two-hybrid. Interactions were scored by measuring -galactosidase activity from negative (−) to strongly positive (+++) and not determined (n.d.). The correlation with in vitro binding to GST-fused Smad3 or its fragments is also shown. Besides some previously defined functions shown on the schematic diagram of Smad3, the sequences that mediate interaction with CBP have now been localized to the carboxyl domain and require the carboxyterminal sequence. (C) Direct interaction of 35S-labeled CBP(1678–2441) with GST–Smad3 and GST–Smad3C, but not GST–Smad4. Results are summarized in B.Smad3 with CBP is most likely direct, as no Smad or CBP homologs are encoded by the yeast genome, and CBP(1678–2441) interacted with GST–Smad3 in vitro (see below). Smad2, which is structurally closely related to Smad3, also interacted with the carboxy-terminal segment of CBP. In contrast, Smad4 and the BMP-2/4-responsive Smad1 did not interact with CBP in yeast twohybrid assays. These results allow us to assign a new function to a defined segment of CBP (Fig. 4A). To map the domains in Smad3 that interact with CBP, we carried out yeast two-hybrid assays that were further confirmed using GST-based in vitro binding assays. Smads have highly conserved amino and carboxyl domains, separated by a less conserved, proline-rich linker (L) region. The carboxyl domain of ligand-responsiveSmads mediates transcriptional activation in GAL4 transactivation assays (Liu et al. 1996) and in transcription assays in yeast (Wu et al. 1997). As shown in Figure 4B, the interaction of Smad3 with the carboxy-terminal segment of CBP was mediated by the carboxyl domain and not by the amino domain or L segment. This interaction was most likely direct as the in vitro-synthesized 35 S-labeled CBP(1678–2441) interacted with GST–Smad3 and GST–Smad3C (Fig. 4C). Consistent with the yeast two-hybrid results (Fig. 4A), the carboxy-terminal segment of CBP did not interact with GST–Smad4. Deletion of the carboxy-terminal 35 amino acids (the c deletion), which inactivates the transcriptional activity of Smads (Zhang et al. 1996), abolished the interaction of full-sized Smad3 or its carboxyl domain with CBP in yeast two-GENES & DEVELOPMENT2159。