Role of Autophagy in Breast Cancer
CXCL12

CXCL12/CXCR4轴通过调节自噬促进乳腺癌细胞对多柔比星的化疗抗性①李霞梁海珊程学②段哲③(海南省中医院输血科,海口 570100)中图分类号R737.9 文献标志码 A 文章编号1000-484X(2023)07-1446-06[摘要]目的:探究CXCL12/CXCR4轴在乳腺癌细胞多柔比星(Dox)化疗抗性中的作用及相关机制。
方法:体外培养乳腺癌细胞系MCF-7,通过MTT实验检测在CXCL12作用下乳腺癌细胞对Dox的敏感性变化,RT-PCR和Western blot检测上述细胞中CXCL12受体CXCR4与CXCR7的mRNA及蛋白的表达水平;采用siRNA干扰技术靶向沉默乳腺癌细胞中CXCR4表达,RT-PCR和Western blot检测转染效率后,依次使用MTT、Annexin V-FITC/PI流式细胞术及Western blot明确在CXCL12作用下沉默CXCR4表达对Dox介导的乳腺癌细胞的抑制率,凋亡、自噬及PI3K/Akt信号通路表达的影响。
结果:CXCL12能够通过促进CXCR4的表达,提高乳腺癌细胞对Dox的化疗抗性;转染siRNA能够显著抑制乳腺癌细胞中CXCR4的mRNA及蛋白的表达水平;而沉默CXCR4能显著改善CXCL12作用下的乳腺癌细胞对Dox的敏感性,提高Dox诱导的细胞凋亡率,并通过抑制自噬相关蛋白LC3B与Beclin1表达,下调乳腺癌细胞中的自噬水平;同时,沉默CXCR4能通过抑制PI3K/Akt信号通路中PI3K蛋白表达及Akt蛋白磷酸化水平,下调抗凋亡蛋白BCL-2,促进凋亡相关蛋白Bax与cleaved Caspase-3表达。
结论:CXCL12/CXCR4在乳腺癌细胞Dox耐药性中具有重要作用,而沉默CXCR4表达能通过下调MCF-7细胞的自噬水平,提高Dox诱导的细胞凋亡,从而改善肿瘤细胞对Dox的敏感性。
[关键词]趋化因子12(CXCL12);CXCR4;乳腺癌;多柔比星;自噬CXCL12/CXCR4 axis promotes chemotherapy resistance of breast cancer cells to doxorubicin by regulating autophagyLI Xia, LIANG Haishan, CHENG Xue, DUAN Zhe. Department of Blood Transfusion, Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou 570100, China[Abstract]Objective:To explore the role of CXCL12/CXCR4 axis in anti-doxorubicin (Dox) resistance of breast cancer cells and its related mechanism. Methods:Breast cancer cell line MCF-7 was cultured in vitro. The sensitivity of breast cancer cells to Dox was detected by MTT assay. The mRNA and protein expression levels of CXCL12 receptor CXCR4 and CXCR7 were detected by RT-PCR and Western blot. siRNA interference was used to silence CXCR4 expression in breast cancer cells. RT-PCR and Western blot were used to detect the transfection efficiency. MTT, Annexin V-FITC/PI flow cytometry and Western blot were used to determine the effects of CXCR4 silencing on DOX mediated proliferation inhibition, apoptosis, autophagy and PI3K/Akt signaling pathway expres‐sion in breast cancer cells. Results:CXCL12 could enhance the chemoresistance of breast cancer cells to Dox by promoting the expres‐sion of CXCR4. siRNA could significantly inhibit the expression of CXCR4 mRNA and protein in breast cancer cells. Silencing CXCR4 can significantly improve the sensitivity of breast cancer cells to Dox and increase the apoptosis rate mediated by Dox, and down-regu‐lation the autophagy level in breast cancer cell by inhibiting the expression of autophagy related proteins LC3B and Beclin1. At the same time, silencing CXCR4 could inhibit the phosphorylation level of PI3K and Akt protein in PI3K/Akt signaling pathway, reduced the anti-apoptotic protein Bcl-2,promoted apoptosis related protein Bax and cleaved caspase-3 expression. Conclusion:CXCL12/ CXCR4 played an important role in Dox resistance of breast cancer cells. Silencing CXCR4 expression could improve the sensitivity of tumor cells to Dox by down regulating autophagy level and increasing Dox induced apoptosis.[Key words]Chemokine 12 (CXCL12);CXCR4;Breast cancer;Doxorubicin;Autophagy乳腺癌是世界范围内最常见的肿瘤之一,也是女性癌症死亡的首要原因[1-2]。
雷公藤红素通过ROS-JNK信号通路诱导T98G细胞发生凋亡

雷公藤红素通过ROS-JNK信号通路诱导T98G细胞发生凋亡丁成国【摘要】目的探讨雷公藤红素对人胶质母细胞瘤细胞系T98G的生物效应及其机制. 方法将人胶质母细胞瘤细胞系T98G用各种不同浓度的雷公藤红素处理后,采用MTT法检测雷公藤红素对T98G细胞活力的抑制作用;采用流式细胞术检测T98G细胞用雷公藤红素处理后细胞的凋亡情况及ROS的产生;Western blot检测T98G细胞用雷公藤红素处理后cleaved caspase-9、cleaved caspase-3和磷酸化JNK的表达水平. 结果雷公藤红素对T98G细胞活力的抑制效应呈剂量依赖性,1μmol/L、2μmol/L、3μmol/L、4μmol/L、5μmol/L 雷公藤红素组细胞活力抑制率分别为(15.3±1.1)%,(28.2±1.8)%,(37.4±2.6)%,(55.6±3.8)%,(71.3±5.1)%. 雷公藤红素可显著诱导T98G细胞发生凋亡,对照组的凋亡率为(2.1±0.3)%,1μmol/L雷公藤红素组凋亡率为(10.4±0.9)%,3μmol/L 雷公藤红素组凋亡率为(22.5± 1.7)%. 雷公藤红素可显著诱导T98G细胞ROS的产生,并使cleaved caspase-9、cleaved caspase-3和磷酸化JNK的表达水平显著上升. 结论雷公藤红素在体外有良好的抗神经胶质母细胞瘤的生物活性,诱导T98G细胞凋亡的机制可能与激活ROS-JNK信号通路有关.【期刊名称】《浙江实用医学》【年(卷),期】2015(020)005【总页数】4页(P325-328)【关键词】雷公藤红素;T98G;凋亡;ROS;JNK【作者】丁成国【作者单位】杭州市第一人民医院,浙江杭州 310006【正文语种】中文近年的研究发现,雷公藤红素除了有抗炎抗氧化作用外,对离体培养的肿瘤细胞有较强的抑制作用[1-2]。
Lyso-Tracker Red (溶酶体红色荧光探针) 产品说明书

Lyso-Tracker Red (溶酶体红色荧光探针)产品简介:Lyso-Tracker Red 是一种溶酶体(lysosome)红色荧光探针,能通透细胞膜,可以用于活细胞溶酶体特异性荧光染色。
Lyso-Tracker Red 为采用Molecular Probes 公司的DND-99进行了荧光标记的带有弱碱性的荧光探针,其中仅弱碱可部分提供质子,以维持pH 在中性,可以选择性地滞留在偏酸性的溶酶体中,从而实现对于溶酶体的特异性荧光标记。
中性红(Neutral Red)和吖啶橙(Acridine Orange)也都可以对溶酶体进行荧光染色,但中性红和吖啶橙的染色缺乏特异性。
Lyso-Tracker Red 适用于活细胞溶酶体的荧光染色,但不适合用于固定细胞溶酶体的荧光染色。
Lyso-Tracker Red 分子的化学结构式参考图1。
图1. Lyso-Tracker Red 的化学结构式。
Lyso-Tracker Red 的分子式为C 20H 24BF 2N 5O ,分子量为399.25,最大激发波长为577nm ,最大发射波长为590nm 。
Lyso-Tracker Red 的激发光谱和发射光谱参考图2。
图2. Lyso-Tracker Red的激发光谱和发射光谱。
Lyso-Tracker Red 是嗜酸性荧光探针,用于活细胞内酸性细胞器的标记和示踪。
这些探针具有几个重要特征,包括高度选择靶向酸性细胞器和在纳摩尔浓度有效标记活细胞。
Lyso-Tracker Red 必须在极低浓度(通常约50nM)下才能获得优异的选择性。
这些探针的滞留(retention)机制虽然没有被研究清楚,但很可能与酸性细胞器的质子化和滞留性有关,Lyso-Tracker Red 探针的内吞作用动力学研究显示染料进入活细胞的摄入时间仅几秒即可。
然而,这些溶酶体探针会导致溶酶体被碱化,长期孵育会诱使溶酶体pH 值的增加。
桔梗-姜半夏配伍促进小鼠NK细胞在肺脏募集并抑制肿瘤肺脏转移

网络出版时间:2024-01-1010:47:13 网络出版地址:https://link.cnki.net/urlid/34.1086.R.20240108.1833.058桔梗-姜半夏配伍促进小鼠NK细胞在肺脏募集并抑制肿瘤肺脏转移王 悦1,张 癑2,孔令婉3,张 珊2,杨雯越2,姚成芳1,2[1.山东中医药大学中医药创新研究院,山东济南 250355;2.山东第一医科大学(山东省医学科学院)临床与基础医学院,山东济南 250117;3.山东中医药大学中医学院,山东济南 250355]收稿日期:2023-08-16,修回日期:2023-12-16基金项目:国家自然科学基金资助项目(No82074088);山东省重点研发计划(重大科技创新工程)(No2022CXGC020514)作者简介:王 悦(1992-),女,硕士生,研究方向:中医药方剂免疫药理,E mail:w362677253@163.com;姚成芳(1966-),女,博士,研究员,博士生导师,研究方向:中医药免疫药理,通信作者,E mail:yaochengfang@sdfmu.edu.cnCombinationofPlatycodonGrandiflorumandPinel liaRernatapromotesrecruitmentofNKcellsinmouselungandinhibitslungmetastasisoftumorWANGYue1,ZHANGYue2,KONGLing wan3,ZHANGShan2,YANGWen yue2,YAOCheng fang1,2(1.InnovativeInstituteofChineseMedicineandPharmacy,ShandongUniversityofTraditionalChineseMedicine,Jinan 250355,China;2.SchoolofClinicalandBasicMedicine,Shan dongFirstMedicalUniversity(ShandongAcademyofMedicalSciences),Jinan 250117,China;3.SchoolofTraditionalChineseMedicine,ShandongUniversityofTraditionalChineseMedicine,Jinan 250355,China)doi:10.12360/CPB202305022文献标志码:A文章编号:1001-1978(2024)01-0199-02中国图书分类号:R 332;R289 1;R322 35;R392 11;R734 5关键词:桔梗-姜半夏配伍;肿瘤肺脏转移;自然杀伤细胞;CXCR3;CXCL9;募集Keywords:combinationofPlatycodonGrandiflorumandPinel liaTernata;lungmetastasisoftumor;NKcells;CXCR3;CX CL9;recruitment开放科学(资源服务)标识码(OSID): 肺脏肿瘤是一种高发病率、高致死率疾病[1],在发展早期主要依赖NK细胞等淋巴细胞发挥抗肿瘤作用[2],其中CXCR3+NK细胞可依赖CXCL9/10等趋化因子的招募作用[3]而快速迁移,并大量分泌IFN-γ和穿孔素等效应因子,发挥免疫监视等作用[4]。
红景天甙抗肿瘤作用的研究进展

红景天甙抗肿瘤作用的研究进展叶应琴;李惠新【摘要】肿瘤是一类常见病、多发病,据世界卫生组织报告,恶性肿瘤是经济发达国家的主要死因,是发展中国家的第二大死因[1],因此攻克肿瘤是人类所面临的巨大挑战.估计2008年全球癌症新发例数为1 270万,癌症致死例数为760万,其中56%的新发病例和64%的致死数发生在经济发展中国家[2],严重威胁人类生命健康.目前早期肿瘤以手术治疗为主,中晚期肿瘤以放、化疗为主辅以生物治疗、热疗等综合治疗手段.【期刊名称】《临床荟萃》【年(卷),期】2012(027)007【总页数】3页(P642-644)【关键词】细胞凋亡;红景天甙;抗肿瘤药(中药)【作者】叶应琴;李惠新【作者单位】兰州大学第二临床医学院,甘肃兰州730000;兰州大学第二医院妇科,甘肃兰州730000【正文语种】中文【中图分类】R286.91肿瘤是一类常见病、多发病,据世界卫生组织报告,恶性肿瘤是经济发达国家的主要死因,是发展中国家的第二大死因[1],因此攻克肿瘤是人类所面临的巨大挑战。
估计2008年全球癌症新发例数为1 270万,癌症致死例数为760万,其中56%的新发病例和64%的致死数发生在经济发展中国家[2],严重威胁人类生命健康。
目前早期肿瘤以手术治疗为主,中晚期肿瘤以放、化疗为主辅以生物治疗、热疗等综合治疗手段。
但放、化疗的毒副作用、耐药性,热耐受以及高费用等成为肿瘤治疗的主要阻碍,很有必要寻找疗效好且毒副作用小的治疗方法。
近年来中药治疗受到广泛关注,具有调节机体内环境、改善临床症状、提高生活质量和低毒性等特点。
红景天甙(Salidroside),又名红景天苷,为红景天的主要活性成分[3],是代表红景天属植物药用价值的主要质量性状指标。
近年来国内外研究发现红景天及红景天甙不仅具有抗疲劳、抗氧化、抗衰老、心脑血管及神经保护、调节血压及三大物质代谢、增强免疫等生理调节作用,还具有抗炎、抗肿瘤、抗辐射作用[4-8],其开发利用为神经精神、心血管、血液内分泌系统以及肿瘤防治等提供一类新型药物的设想。
NOD样受体介导的信号转导通路及其与肿瘤关系的研究进展

223欢迎关注本刊公众号·综 述·《中国癌症杂志》2019年第29卷第3期 CHINA ONCOLOGY 2019 Vol.29 No.3基金项目:国家自然科学基金(81770137)。
通信作者:陆维祺 E-mail:***********************.cn 先天性免疫应答是机体抗感染免疫的第一道防线,相对于适应性免疫应答来说具有出现早、应答发生速度快等特点。
其主要识别病原体相关分子模式(pathogen-associated molecular patterns,PAMPs)和损伤相关的分子模式(damage-associated molecular patterns,D A M P s )。
其通过模式识别受体(p a t t e r n recognition receptors,PRR)[1]来非特异地识别各种致病物质,PRR主要有以下两类受体:一类是位于细胞膜表面或内体膜上的Toll样受体(Toll-like receptor,TLR),另一类是位于细胞质内的核苷酸结合寡聚化结构域(nucleotide- binding oligomerization domain,NOD)样受体及视黄酸诱导基因(retinoic acid inducible gene,RIG )样受体。
TLR在抗感染与抗肿瘤方面的作用已经被广泛研究,近年来关于同属于PRR的NOD样受体的研究主要集中于其介导的信号通路及其在抗微生物感染中的作用,而关于其与肿瘤关系的研究却很少。
NOD样受体可以分为NLRA、NLRB、NLRC、NLRP和NLRX 5个亚家族,其中NLRC和NLRP亚家族是NOD样受体主要的两种类型,而NOD1和NOD2是NLRC亚家族中的主要代表,也是NOD样受体中研究最多的2个成员[2],本文对NOD1和NOD2受体的分子组成、介导的信号转导通路及其与肿瘤关系的最新NOD样受体介导的信号转导通路及其与肿瘤 关系的研究进展林巧卫1,张 思2,陆维祺11.复旦大学附属中山医院普外科,上海 200032;2.复旦大学上海医学院生物化学与分子生物学系,上海 200032[摘要] 核苷酸结合寡聚化结构域(nucleotide-binding oligomerization domain ,NOD )样受体是一类位于细胞质的模式识别受体,在先天性免疫应答中起着十分重要的作用。
Function of Aurora kinase A in Taxol--resistant breast cancer and its correlation with P--gp
Me i l c n e n e igUno dcl olg ,B in 0 0 0 dc i csa dP kn inMe i l e e ig1 0 5 aS e aC e j
Br a tc n e so eo h o tc mm o ai n n ie s sa n me . n e ry a d m e e s a c ri n ft em s o n m l a tds a e mo g wo n I a l n — g
tsai bes a cr a a e( x 1 (swie sda najvn n ea vn hr— att rat n e,T x n Tao) i dl ue sa du ata dno  ̄u attea c c y
p e . t o g ra tc n e si iil e p n iet x l ih r n rd v lp d r ss a c o is Alh u h b e s a c ri n t l r s o sv O Ta o , n e e to e eo e e itn et ay
Fu cin o r r ia eA x l r ss a tb e s n t fAu o ak n s i Ta o - e itn r a t o n
c n e n s c r ea in wih P— g a c ra d i o r lto t t p
果显 示 ,与正 常组 比较 ,模 型组实 验 开始后 4 ,血 清 蛋 白 T R 显著 升 高 ( 清浓 度 高 于正 周 NF I 血
常组 2 ) L 1显著 降低 ( 清浓度低 于正常组 12 ,8 后血清 T F 、G— C F 倍 ,I 一2 血 /) 周 N —a S、 T A 、I一1 、LX C 一3 L 5 I 、MI —l P a显著 降低 ;卡介苗免 疫组 4周 后血 清 MI 一 1 、T R 、 P a A C E tx oa i n显著 升高 ,I L一1 显 著降低 ,8周后 血清 B C 5 L 、G—C F S 、K C显著 升高 ,T A一3 C 、 MI 一1 、T F 、M—C F显著降低 ;卡介 苗免疫后接瘤组 4 P a N —a S 周后 T F I oa i、B C N R 、E tx n L 显著升高 ,I 一2 、M—C F显著 降低 ,8周后血 清 T F 、G—C F N R 、T F I L 1 S N —a S 、T F I N R I 、
Immunogenic cell death in cancer therapy
ANNUAL REVIEWSClick here for quick links to Annual Reviews content online, including: • Other articles in this volume • Top cited articles • Top downloaded articles • Our comprehensive searchFurtherImmunogenic Cell Death in Cancer TherapyGuido Kroemer,1, 3, 6−9, ∗ Lorenzo Galluzzi, 5, 8, ∗ Oliver Kepp,1, 5, 9 and Laurence Zitvogel2, 4, 91 3Annu. Rev. Immunol. 2013.31:51-72. Downloaded from by INSERM-multi-site account on 03/27/13. For personal use only.U848, 2 U1015, INSERM, 94805 Villejuif, France; email: kroemer@orange.frMetabolomics Platform, 4 Center of Clinical Investigations, Institut Gustave Roussy, 94805 Villejuif, France Institut Gustave Roussy, 94805 Villejuif, France5 6Equipe 11 Labellis´ ee par la Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France Pole Europ´ een Georges Pompidou, AP-HP, 75015 Paris, France ˆ de Biologie, Hopital ˆ Universit´ e Paris Descartes/V, Sorbonne Paris Cit´ e, 75006 Paris, France Universit´ e Paris Sud/XI, 94805 Villejuif, France7 8 9Annu. Rev. Immunol. 2013. 31:51–72 First published online as a Review in Advance on November 12, 2012 The Annual Review of Immunology is online at This article’s doi: 10.1146/annurev-immunol-032712-100008 Copyright c 2013 by Annual Reviews. All rights reserved∗KeywordsATP, autophagy, calreticulin, damage-associated molecular patterns, HMGB1, TLR4AbstractDepending on the initiating stimulus, cancer cell death can be immunogenic or nonimmunogenic. Immunogenic cell death (ICD) involves changes in the composition of the cell surface as well as the release of soluble mediators, occurring in a defined temporal sequence. Such signals operate on a series of receptors expressed by dendritic cells to stimulate the presentation of tumor antigens to T cells. We postulate that ICD constitutes a prominent pathway for the activation of the immune system against cancer, which in turn determines the long-term success of anticancer therapies. Hence, suboptimal regimens (failing to induce ICD), selective alterations in cancer cells (preventing the emission of immunogenic signals during ICD), or defects in immune effectors (abolishing the perception of ICD by the immune system) can all contribute to therapeutic failure. We surmise that ICD and its subversion by pathogens also play major roles in antiviral immune responses.These authors contributed equally to this work.51INTRODUCTIONICD: immunogenic cell death CTL: cytotoxic CD8+ T lymphocyte Treg: FOXP3+ regulatory T cell DC: dendritic cell TLR: Toll-like receptorEvery second, in the healthy human adult, several millions of cells that succumb to programmed cell death mechanisms are efficiently removed without eliciting local or systemic inflammation. This homeostatic cell death, which often occurs through apoptosis, is considered either as a tolerogenic (promoting tolerance to self) or as a null (exerting no impact on the immune system) event (1, 2). The past few years have witnessed the emergence of the concept of immunogenic cell death (ICD), i.e., a cell death modality that does stimulate an immune response against dead-cell antigens, in particular when they derive from cancer cells (2). This model was first proposed in the context of anticancer chemotherapy, based on clinical evidence indicating that tumor-specific immune responses can determine the efficacy of anticancer therapies with conventional cytotoxic drugs (3). In response to antineoplastic agents, the composition of the tumor immune infiltrate changes, and this can be crucial for the outcomes of therapy. Thus, an increased number of T lymphocytes as well as an increased ratio of cytotoxic CD8+ T lymphocytes (CTLs) over FOXP3+ regulatory T cells (Tregs) within the tumor after chemotherapy predict favorable therapeutic responses in human breast and colorectal cancer patients treated with anthracyclines and oxaliplatin, respectively (4–8). Metaanalyses of multiple clinical studies indicate that severe lymphopenia (<1,000 lymphocytes/μL) negatively affects the response to chemotherapy of multiple distinct solid cancers (9). Accordingly, a collection of murine neoplasms (including transplantable cancers as well as chemically induced primary tumors) respond to chemotherapy with anthracyclines or oxaliplatin much more efficiently when they grow in syngeneic immunocompetent mice than when they develop in immunodeficient hosts (10–13). Anthracycline-killed human tumor cells also appear particularly immunogenic (14) and have been successfully used for the therapeutic vaccination of cancer patients (15). The immune response stimulated by chemotherapy involvesKroemer et al.the obligatory contribution of dendritic cells (DCs), which engulf, process, and present antigen from dying tumor cells, as well as that of several T lymphocyte populations. In mice, the perception of ICD signals by DCs relies on Toll-like receptor 4 (Tlr4) and on the purinergic receptor P2rx7 (10, 16, 17). Along similar lines, in humans, loss-of-function alleles of TLR4 and P2RX7 are negative predictors of the clinical response to adjuvant chemotherapy with anthracyclines or oxaliplatin (10, 16, 17). Based on these premises, we suggest that a restricted panel of chemotherapeutics (some of which are currently associated with considerable rates of success) can induce a combination of tumor cell stress and death that is immunogenic. This means that the patient’s dying cancer cells operate as a vaccine that stimulates a tumor-specific immune response, which in turn can control (and sometimes even eradicate) residual cancer (stem) cells (3, 18, 19). As an operational definition of ICD, we consider that ICD must satisfy the following two criteria (Figure 1). First, cancer cells succumbing to ICD in vitro and administered in the absence of any adjuvant must elicit an immune response that protects mice against a subsequent challenge with live tumor cells of the same type (2). Second, ICD occurring in vivo must drive a local immune response featuring the recruitment of innate and cognate immune effector cells into the tumor bed (20) and hence result in the inhibition of tumor growth via mechanisms that depend (at least in part) on the immune system (10, 13). In preclinical models, multiple alterations of the immune system have been tested for the ability to compromise either or both of these phenomena (Table 1). In this review, we discuss the salient features of ICD, the underlying cell biology, and the pathways through which ICD is perceived by immune effector cells, as well as the general impact of ICD on human pathophysiology.Annu. Rev. Immunol. 2013.31:51-72. Downloaded from by INSERM-multi-site account on 03/27/13. For personal use only.SALIENT FEATURES OF IMMUNOGENIC CELL DEATHDistinct chemotherapeutic agents are not equivalent in their capacity to induce ICD.52aVaccinationTumor cells killed in vitro with ICD inducerRechallengeLive tumor cellsOutcomeNo tumorTumor cells killed in vitro with non-ICD inducerLive tumor cells Tumor growthAnnu. Rev. Immunol. 2013.31:51-72. Downloaded from by INSERM-multi-site account on 03/27/13. For personal use only.bEngraftmentLive tumor cellsTreatmentIn vivo chemotherapy with ICD inducerOutcomeOptimal responseWild type Live tumor cells In vivo chemotherapy with ICD inducer Suboptimal responseImmunodeficientFigure 1 Operational definition of immunogenic cell death (ICD). From an operational point of view, cancer cell death can be defined as immunogenic based on two major attributes. First, the injection of cancer cells succumbing to ICD into immunocompetent mice must elicit a protective immune response that is specific for tumor antigens, in the absence of any adjuvant (a). Second, chemotherapy with ICD inducers must exert anticancer effects (i.e., reduction in tumor growth rate and/or tumor mass) that depend, at least in part, on the immune system (b).When cancer cells succumbing to 24 distinct cytotoxic chemotherapeutics were tested for their ability to elicit protective anticancer immune responses in vivo (as for the operationaldefinition provided above), this was observed in four instances only (namely, for three anthracyclines and for oxaliplatin), although all agents induced apoptosis in an equivalent fashion (21). • Immunogenicity of Cancer Cell Death 53Table 1 Examples of experimental alterations of the immune system and their effects on the immunogenicity of cell death Genotype/defect B cell depletion Casp1−/− Ccl20 blockade Ccr6−/− Cx3cr1−/− Cxcl9 blockade Cxcr3−/− Fcgr1−/− Gzmb−/− Observations With anti-CD20 antibodies and upon deletion of the Cμ IgM locus Inflammasome component With neutralizing antibodies Receptor expressed on Th17 cells Cx3cl1 (fractalkine) receptor With neutralizing antibodies Receptor for Cxcl9, Cxcl10, and Cxcl11 Fcγ receptor Granzyme B, essential for cytotoxic functions of NK cells and CTLs Type 1 Ifn receptor Type 2 Ifn system Common interleukin-1 receptor With neutralizing antibodies With neutralizing antibodies and upon deletion of the Il12rb1 locus Il-17 system Il-18 system With neutralizing antibodies With neutralizing antibodies Il-23 Il-6 Adaptor for Tlr4 Adaptor for Tlr4 Inflammasome component Athymic mice Purinergic receptor Perforin, essential for cytotoxic functions of NK cells and CTLs Severe immunodeficiency involving T, B, NK, and NKT cells Alternative Tlrs Tlr4 Tnf-α system With neutralizing antibodies Vaccination responsea ND − ND ND + ND − ND + + − − − ND − + ND ND ND + − − − − − − − + − − ND Chemotherapeutic responsea,b + − + + + + − + + + − − − + − + + + + + − − − − − − − ND − + + Reference(s)c Unpublished 16 20 20 101 Unpublished 102 Unpublished 16 Unpublished 10 16 16 16 and unpublished 20 16 20 12, 20 12, 20 Unpublished 10 10 16 11 16 16 16 10 10 80 and unpublished 16Annu. Rev. Immunol. 2013.31:51-72. Downloaded from by INSERM-multi-site account on 03/27/13. For personal use only.Ifnar1−/− Ifng−/− , Ifngr1−/− Il1r1−/− Il-1β blockade Il-12rb blockade Il17a−/− , Il17ra−/− Il18−/− , Il18r1−/− Il-22 blockade Il-23 blockade Il23a−/− Il6−/− Ly96−/− Myd88−/− Nlrp3−/− nu/nu P2rx7−/− Prf1−/− Rag2−/− γc−/− Tlr2−/− , Tlr3−/− , Tlr9−/− Tlr4−/− Tnf−/− , Tnfr1−/− Tnfsf10 blockadeAbbreviations: CTL, cytotoxic CD8+ T lymphocyte; Ifn, interferon; IgM, immunoglobulin M; Il, interleukin; NK, natural killer; Tlr, Toll-like receptor; Tnf, tumor necrosis factor; Tnfsf10, tumor necrosis factor (ligand) superfamily, member 10. a + = normal; − = reduced; ND = not determined. b Most determinations have been obtained in C57/Bl6 mice (wild type or knockout for the indicated genes) transplanted with syngeneic fibrosarcoma MCA205 cells and treated intratumorally with mitoxantrone or doxorubicin. c Unpublished results come jointly from G. Kroemer’s and L. Zitvogel’s lab.54Kroemer et al.Dying tumor cellCD91 CRT P2RX7 ATP TLR4 HMGB1Immature DC Mature DCAntigen engulfment ICD Therapyinduced stress Antigen presentationIL-1βTherapy-sensitive tumor cellsAnnu. Rev. Immunol. 2013.31:51-72. Downloaded from by INSERM-multi-site account on 03/27/13. For personal use only.γδ T cellIL-17 VesselIFN-γCTLsTherapy-resistant tumor cellsFigure 2Tumor cell lysisProperties of immunogenic cell death (ICD). As a result of premortem endoplasmic reticulum stress and autophagy, cancer cells responding to ICD inducers expose CRT on the outer leaflet of their plasma membrane at a preapoptotic stage, and secrete ATP during apoptosis. In addition, cells undergoing ICD release the nuclear protein HMGB1 as their membranes become permeabilized during secondary necrosis. CRT, ATP, and HMGB1 bind to CD91, P2RX7, and TLR4, respectively. This facilitates the recruitment of DCs into the tumor bed (stimulated by ATP), the engulfment of tumor antigens by DCs (stimulated by CRT), and optimal antigen presentation to T cells (stimulated by HMGB1). Altogether, these processes result in a potent IL-1β- and IL-17-dependent, IFN-γ-mediated immune response involving both γδ T cells and CTLs, which eventually can lead to the eradication of chemotherapy-resistant tumor cells. (Abbreviations: ATP, adenosine triphosphate; CRT, calreticulin; CTL, cytotoxic CD8+ T lymphocyte; DC, dendritic cell; HMGB1, high-mobility group box 1; IFN, interferon; IL, interleukin; TLR, Toll-like receptor.)Subsequent biochemical analyses revealed the distinctive properties of ICD (as opposed to nonimmunogenic cell death, non-ICD): the preapoptotic exposure of calreticulin (CRT) and other endoplasmic reticulum (ER) proteins at the cell surface, the secretion of ATP during the blebbing phase of apoptosis, and the cell death–associated release of the nonhistone chromatin protein high-mobility group box 1 (HMGB1) (Figure 2) (10, 16, 17, 21).Importantly, these parameters appear to be sufficient to make accurate predictions about the capacity of drugs to induce ICD. Thus, videomicroscopic measurements of CRT exposure, ATP secretion, and HMGB1 release in human cancer cells identified, among 1,040 distinct Food and Drug Administration–approved drugs, cardiac glycosides as particularly efficient inducers of these alterations (22). Subsequently, cardiac glycosides were found to • Immunogenicity of Cancer Cell Deathnon-ICD: nonimmunogenic cell death CRT: calreticulin ER: endoplasmic reticulum HMGB1: high-mobility group box 155PS: phosphatidylserine MCA: methylcholanthreneimprove the chemotherapeutic effect of nonICD inducers such as cisplatin or mitomycin C in immunocompetent (but not immunodeficient) mice. Most importantly, cardiac glycosides were shown to extend the overall and progression-free survival of (a) breast cancer patients who were treated with agents other than anthracyclines, and (b) colorectal cancer patients receiving agents other than oxaliplatin (22). These data perhaps explain episodic reports on the beneficial effect of cardiac glycosides for cancer patients (23, 24) and underscore the possibility that the salient features of ICD can be taken advantage of to predict the capacity of prospective anticancer drugs to stimulate therapeutic immune responses. We and others have resolved (part of ) the question of why some cytotoxic chemotherapeutics induce exposure of CRT, secretion of ATP, and release of HMGB1 while others do not. ICD is obligatorily preceded by two types of stress, namely ER stress and autophagy (13, 25, 26). As a result of chemotherapy-induced ER stress, CRT, whose largest fraction is normally secluded in the ER lumen, relocates to the outer surface of the plasma membrane, and this occurs well before cells manifest signs of apoptotic cell death such as the exposure of phosphatidylserine (PS) on the cell surface (26). Ecto-CRT operates as a potent engulfment signal, thus allowing DCs to engulf portions of stressed and dying tumor cells (27, 28). The blockade of CRT exposure in tumors negatively affects the efficacy of anthracycline-based chemotherapy in immunocompetent mice (29), suggesting that this ER stress–dependent immunogenic signal is indeed indispensable for the elicitation of therapeutic anticancer immune responses (17, 21). Macroautophagy (autophagy) is also obligatory for cell death to be perceived as immunogenic (13). Autophagy inhibition affects neither the exposure of CRT nor the release of HMGB1, but it does prevent the secretion of ATP from dying cancer cells. Thus, in response to chemotherapy, autophagy-competent, but not autophagydeficient, tumors recruit DCs and (later) T lymphocytes. Such a defect of autophagy56 Kroemer et al.deficient cancers can be corrected by the inhibition of extracellular ATP–degrading enzymes, resulting in increased pericellular ATP concentrations, reestablished recruitment of immune cells, and restored chemotherapeutic responses, but only in immunocompetent hosts (13). Thus, autophagy is essential for the immunogenic release of ATP from dying cells, and maneuvers that increase extracellular ATP concentrations can potentially improve the efficacy of antineoplastic chemotherapies when autophagy is disabled. Of note, the obligatory contribution of the immune system to the success of conventional chemotherapies has been validated in multiple transplantable models of mouse carcinoma, sarcoma, and lymphoma (10, 11, 16, 21, 25). However, spontaneous mouse breast carcinomas (as induced by a rat Neu transgene or, alternatively, by the simultaneous inactivation of Tp53 and Cdh1) do respond to chemotherapy with oxaliplatin or doxorubicin irrespective of the presence of Rag1 and Rag2 recombinases (both of which are required for the generation of B and T cells) (30). Thus, although transplantable neoplasms (10, 11, 16, 21, 25) as well as methylcholanthrene (MCA)-induced tumors (31) are subjected to immunosurveillance (and hence develop more slowly in immunocompetent mice) (31), the aforementioned models of breast carcinomas are not (30). This apparent discrepancy stems perhaps from the fact that oncogene-driven tumorigenesis may have subverted the exposure of class I MHC molecules and/or hijacked the machineries for ER stress and/or autophagy (32). Alternatively, oncogene-driven cancers may acquire the ability to evoke immunoescape mechanisms and/or simply fail to elicit a natural immunosurveillance. In such cases, curtailing oncogene addiction with targeted therapies (e.g., with agents that specifically inhibit ALK, KIT, MYC, or BCR-ABL) might restore antitumor immune responses (33–35). Using oncogene-targeting agents in combination with anthracyclines might further extend the therapeutic benefits of this approach (34).Annu. Rev. Immunol. 2013.31:51-72. Downloaded from by INSERM-multi-site account on 03/27/13. For personal use only.ER STRESS AND CALRETICULIN EXPOSURE ON DYING CELLSCRT represents the most abundant protein of the ER lumen, yet it can be found in other subcellular compartments, including the cytosol (36, 37). In response to multiple ICD inducers (Table 2), a fraction of CRT translocates from the ER lumen to the surface of stressed and dying cancer cells (21, 28). This phenomenon occurs before cells expose PS on the outer leaflet of the plasma membrane and persists in cytoplasts (i.e., enucleated cells) (21), meaning that the immunogenic exposure of CRT (a) does not require purely nuclear components, (b) is independent from the DNA damage response, and (c) does not result from transcriptional reprogramming. Conversely, anthracycline-induced CRT exposure does require the formation of reactive oxygen species (ROS) and nitric oxide (26, 38). When elicited by anthracyclines or oxaliplatin, the exposure of CRT is activated by an ER stress response that involves the phosphorylation of the eukaryotic translation initiation factor eIF2α by the PKR-like ER kinase (PERK). This is generally followed by the caspase-8-mediated proteolysis of the ER-sessile protein BAP31, the activation of the proapoptotic proteins BAX and BAK, the anterograde transport of CRT from the ER to the Golgi apparatus, and the exocytosis of CRT-containing vesicles, eventually resulting in the SNARE-dependent translocation of CRT onto the plasma membrane surface (26). Interruption of this complex pathway at any level (with pharmacological or genetic interventions) abolishes CRT exposure, annihilates the immunogenicity of apoptosis, and reduces the immune response elicited by anticancer chemotherapies (26). There is consensus on the notion that ER stress is required for the exposure of CRT at the cell surface, but the exact molecular mechanisms that link these two phenomena are not clear. Thus, although the activation of PERK is important for CRT exposure as elicited by both anthracyclines and hypericin-based photodynamic therapy (PDT), eIF2α phosphoryla-tion appears to be mandatory for the former and dispensable for the latter (21, 25). Along similar lines, caspase-8 activation is not required for the translocation of CRT to the cell surface in response to PDT (25), although it is necessary for anthracycline-, oxaliplatin-, and docosahexaenoic acid–induced CRT exposure (26, 39). Finally, whereas the translocation of CRT to the plasma membrane surface as induced by mitoxantrone obligatorily relies on another ERsessile protein, ERp57, (29), the same does not hold true for CRT exposure as stimulated by PDT (40). These results suggest that the exposure of CRT at the cell surface may be the net result of heterogeneous signaling pathways that are elicited in a stimulus-dependent manner (Figure 3). The knockdown of CRT (or that of any of the proteins that are required for CRT exposure) abolishes the immunogenicity of cell death as elicited by multiple ICD inducers (e.g., anthracyclines, oxaliplatin, irradiation, and PDT), hence extinguishing the capacity of dying cells to elicit a protective immune response in vaccination experiments (21, 25, 26, 29). Conversely, the absorption of recombinant CRT to the surface of cells that succumb to non-ICD inducers (such as mitomycin C or cisplatin) can restore the immunogenicity of cell death (21, 26, 29). A similar effect can be obtained by manipulating cells to express a CRT variant engineered to cause its automatic transport to the cell surface as a result of fusion with the human herpesvirus 8 G protein–coupled receptor (41). In addition, CRT exposure can be enforced (a) by manipulations that cause ER stress, including the expression of the Ca2+ channel reticulon 1C and the administration of thapsigargin (a pharmacological inhibitor of the sarco/endoplasmic reticulum Ca2+ -ATPase, SERCA), both of which stimulate the efflux of Ca2+ from the ER lumen (42, 43); and (b) by the inhibition of the GADD34/PP1 complex (with chemicals or with peptides that competitively disrupt the GADD34/PP1 interaction), which functionally antagonizes PERK-dependent eIF2α phosphorylation (21, • Immunogenicity of Cancer Cell DeathROS: reactive oxygen species PERK: PKR-like ER kinase PDT: photodynamic therapyAnnu. Rev. Immunol. 2013.31:51-72. Downloaded from by INSERM-multi-site account on 03/27/13. For personal use only.57Table 2 Pharmacological inducers of immunogenic cell death Inducer(s) Anthracyclines Observations Cancer cells succumbing to anthracyclines (e.g., doxorubicin, mitoxantrone) induce protective immune responses when injected subcutaneously into mice, in the absence of any adjuvant. This property is lost upon cell lysis (e.g., by freeze-thawing, sonication, or exposure to strong detergents) and is accompanied by CRT exposure, ATP secretion, and HMGB1 release as a result of ER stress, autophagy, and secondary necrosis, respectively. The mAb 7A7 induces CRT, HSP70, and HSP90 exposure on the surface of dying D122 cells (a derivative of Lewis lung carcinoma cells), which hence become able to elicit protective immune responses in vaccination experiments. The antitumor efficacy of 7A7 is reduced upon depletion of CD8+ T cells. 7A7-treated D122 cells also stimulate the maturation of DCs (upregulation of CD40, CD80, CD86, and class II MHC molecules). BK channel agonists (e.g., phloretin and pimaric acid) kill rat glioma cells while inducing the overexpression of HSP60, HSP70, and HSP90 as well as the release of HMGB1. Such dying cells promote DC maturation (upregulation of CD86 and class II MHC molecules) and elicit protective immune responses in vivo. Bortezomib induces the exposure of CRT, HSP70 and HSP90 on the surface of dying primary effusion lymphoma and myeloma cells, which can stimulate DC maturation (upregulation of CD83 and CD86) in a CD91-dependent fashion. Murine hepatocellular carcinoma BNL cells (which express hemagglutinin) infected with hTert-Ad and treated with bortezomib plus mitomycin C induce antitumor immune responses, as assessed by hemagglutinin-pentamer staining. The vaccine loses its activity if cells are treated with a caspase inhibitor. This intervention can eradicate BNL tumors and lung metastases in wild-type BALB/c mice but not in CD8-depleted BALB/c mice or in athymic nu/nu mice. Cardiac glycosides can induce CRT exposure, ATP release, and HMGB1 release as a result of ER stress, autophagy, and secondary necrosis. When combined with non-ICD inducers (e.g., cisplatin, mitomycin C), cardiac glycosides stimulate ICD, as determined in vaccination experiments and by chemotherapy of established tumors. The cyclophosphamide derivative mafosfamide promotes CRT exposure and HMGB1 release from EG7 lymphoma cells. Mafosfamide-treated EG7 cells stimulate a protective immune response in vaccination experiments. PP1 inhibitors (e.g., calyculin A, okadaic acid) and peptides that disrupt the GADD34/PP1 interaction fail to stimulate ATP and HMGB1 release, although they trigger CRT exposure. When combined with mitomycin C, which promotes ATP and HMGB1 release, however, these agents induce bona fide ICD, as determined in vaccination experiments and by chemotherapy of established tumors. Ionizing irradiation with UVC light or γ rays causes ICD accompanied by CRT exposure, ATP release, and HMGB1 release. This agent causes cell death accompanied by CRT exposure (but not by ATP and HMGB1 release). In vivo, the therapeutic effect of LV-tSMAC depends on T cells. In vitro, LV-tSMAC-killed tumor cells can activate DCs. Measles virus infects and kills CD46-expressing melanoma cells along with the production of type I interferons and the release of HMGB1. DCs exposed to measles virus–infected cells upregulate CD80 and CD86, stimulating cytotoxic CD8+ T cells to produce IFN-γ. Similar to anthracyclines (but not to cisplatin), oxaliplatin induces bona fide ICD, as determined in vaccination experiments and by chemotherapy of established tumors. This manipulation leads to CRT exposure, ATP secretion, and HMGB1 release, as well as to ICD, as determined in vaccination experiments. There are differences in the mechanisms leading to CRT exposure by PDT and anthracyclines.58 Kroemer et al.Reference(s) 10, 11, 13, 16, 21, 26Anti-EGFR mAb 7A7103Annu. Rev. Immunol. 2013.31:51-72. Downloaded from by INSERM-multi-site account on 03/27/13. For personal use only.BK channel agonists104Bortezomib97, 105Bortezomib plus mitomycin C plus hTert-Ad88Cardiac glycosides plus non-ICD inducers Cyclophosphamide22106GADD34/PP1 inhibitors plus mitomycin44, 107Irradiation LV-tSMAC10, 21, 107, 108 109Measles virus89Oxaliplatin PDT with hypericin10, 17 40(Continued )Table 2 (Continued ) Inducer(s) poly(I:C) Observations Poly(I:C) kills primary human epithelial ovarian cancer cells, as they secrete IFN-β, CCL5, and TNF-α and become capable of stimulating monocyte-derived DCs to upregulate class I/II MHC molecules and to secrete IFN-α, CXCL10, and IL-12. Similarly, poly(I:C) causes ED8 mouse epithelial ovarian cancer cells to stimulate NK cell proliferation and IFN-γ production in vivo. The ER stressor thapsigargin alone fails to induce the release of ATP and HMGB1. The combination of thapsigargin and cisplatin, however, induces bona fide ICD, as determined in vaccination experiments. Reference(s) 110Thapsigargin plus cisplatin17, 42Annu. Rev. Immunol. 2013.31:51-72. Downloaded from by INSERM-multi-site account on 03/27/13. For personal use only.Abbreviations: ATP, adenosine triphosphate; BK, large Ca2+ -activated K+ ; CRT, calreticulin; DC, dendritic cell; EGFR, epidermal growth factor receptor; ER, endoplasmic reticulum; GADD34/PP1, growth arrest and DNA damage-inducible protein 34/protein phosphatase 1; HMGB1, high-mobility group box 1; HSP, heat shock protein; hTERT-Ad, human telomerase reverse transcriptase promoter-regulated adenovirus; ICD, immunogenic cell death; IFN, interferon; LV-tSMAC, lentivirus-encoded cytosolic SMAC mimetic; mAb, monoclonal antibody; MHC, major histocompatibility complex; NK, natural killer; non-ICD, nonimmunogenic cell death; PDT, photodynamic therapy; poly(I:C), polyinosinic-polycytidylic acid; TNF-α, tumor necrosis factor α; UVC, ultraviolet C.ModuleAnthracycline-induced CRT exposure• ROS ↑ • [Ca2+]cyt ↑ • PERK • P-eIF2αPDT-induced CRT exposure• ROS ↑ • PERKaER stress modulePiPERK ER eIF2αPi[Ca2+]cyt ↑bApoptotic moduleBAX BAK CASP-8 ROS ↑ BAP31 Mitochondria• CASP-8 • BAP31 • BAX • BAK• BAX • BAKcTranslocation modulePlasma membrane Golgi apparatus ERvSNAREs PI3K CRT/ERp57• PI3K • ER-to-Golgi transport • ERp57 • Lipid rafts• PI3K • ER-to-Golgi transportFigure 3 Comparison of the signaling pathways that underpin calreticulin (CRT) exposure in response to anthracyclines and photodynamic therapy (PDT). Anthracycline-induced CRT exposure requires the cotranslocation of the endoplasmic reticulum (ER) chaperone ERp57 and relies on the sequential activation of three signaling modules: (a) an ER stress module, featuring increased generation of reactive oxygen species (ROS), increased cytosolic Ca2+ concentrations, PERK activation, and phosphorylation of the translation initiation factor eIF2α; (b) an apoptotic module, featuring the caspase-8-mediated cleavage of BAP31 and involving the Bcl-2 family members BAX and BAK; and (c) a translocation module, involving phosphoinositide-3-kinase (PI3K) activity, the molecular machineries for ER-to-Golgi anterograde transport and vSNARE-dependent exocytosis, as well as lipid rafts. Of note, CRT exposure as triggered by other immunogenic cell death (ICD) inducers depends on mechanisms that partially, but not completely, overlap with the molecular cascades elicited by anthracyclines. Thus, whereas CRT exposure as promoted by hypericin-based PDT obligatorily relies on PERK, BAX, BAK, PI3K activity, and the ER-to-Golgi anterograde transport, this process occurs independently of ERp57 cotranslocation, eIF2α phosphorylation, and cytoplasmic Ca2+ waves. In addition, the translocation of CRT at the cell surface following PDT requires the actin cytoskeleton, but not lipid rafts or the machinery for retrograde transport. These observations suggest that CRT exposure occurs via molecular mechanisms that, at least in part, depend on the initiating stimulus. • Immunogenicity of Cancer Cell Death 59。
甲状腺癌的小科普
甲状腺癌的病理类型分为 4 种,分别为乳头状
癌、滤泡状癌、髓样癌及未分化癌。这 4 种类型的
病例数依次减少,总体预后也是依次降低。占比
最大的是乳头状癌及滤泡状癌,合称为分化型甲
状腺癌,患者总体预后良好(约居全身所有部位恶
性肿瘤预后良好程度前三位),且生存期较长(约
95%以上患者超过 10 年)[2]。后两种类型的预后较
[3] Vegliante R, Desideri E, Leo LD, et al. Dehydroepiandrosterone triggers autophagic cell death in human hepatoma
cell line HepG2 via JNK- mediated p62/SQSTM1 expression[J]. Carcinogenesis, 2016, 37(3): 233-244.
Cell, 2016, 30(4): 595-609.
[17] Ye Y, Fang YF, Xu WX, et al. 3,3'-diindolylmethane induces anti- human gastric cancer cells by the miR- 30e- ATG5
modulating autophagy[J]. Biochem Pharmacol, 2016, 115:
瘤,尤其女性常见。在 2015 年,国内流行病学统计
显示,甲状腺癌已经成为女性增长速度最快的一种
恶性肿瘤,
而且新发病例在逐确,但是与情
绪、压力、内环境紊乱、生活环境、遗传等诸多因素
相关,保持良好的心情及稳定的生活习惯,提升自
我抗压能力对甲状腺疾病及甲状腺癌的预防有很
薯蓣皂苷对耐三苯氧胺乳腺癌细胞生长的影响研究
【Abstract】 Background Tamoxifen is a main drug used for endocrine treatment of breast cancer.It is difficult to treat breast cancer resistant to tamoxifen,and new treatments are needed urgently.Diosgenin(Dio) can inhibit the growth of cancer to some extent.Studying the effect of Dio on the growth of tamoxifen-resistant(TAM-R) breast cancer cells can provide a reference for clinical treatment.Objective To study the effects of Dio on the growth of TAM-R breast cancer cells.Methods From January 2017 to June 2018,TAM-R breast cancer cells were cultured.Corresponding experiments were carried out on cell growth and apoptosis,autophagy and apoptotic markers,and anti-cancer proteins.The details are as follows:automated cell counters were used to count the number of TAM-R breast cancer cells in six groups(control,Dio 0.625 μg/ml,Dio 0.800 μg/ml,Dio 1.000 μg/ml,Dio 1.250 μg/ml and Dio 2.500 μg/ml) after being treated by different doses of Dio for five days,and to measure the apoptosis of TAM-R breast cancer cells in 6 groups(control,TAM 10-7 mol/L,Dio 1 μg/ml,Dio 2 μg/ml,TAM 10-7 mol/L +Dio 1 μg/ml,and TAM 10-7 mol/L+Dio 2 μg/ml) treated with different doses of TAM and Dio for three days.Immunoblotting
- 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)。
Co py r ig ht L a nd e sB i o sc i e n c e 2007©2007 L A N D E S B I O S C I E N C E. D O N O T D I S T R I B U T E .610Autophagy2007; Vol. 3 Issue 6AddendumRole of Autophagy in Breast CancerVassiliki Karantza-Wadsworth 1,2Eileen White 2-5,*1Division of Medical Oncology; Department of Medicine; University of Medicineand Dentistry of New Jersey; Robert Wood Johnson Medical School; Piscataway, New Jersey USA2The Cancer Institute of New Jersey; New Brunswick, New Jersey USA 3Center for Advanced Biotechnology and Medicine; Rutgers University; Piscataway,New Jersey USA4University of Medicine and Dentistry of New Jersey; Robert Wood JohnsonMedical School; Piscataway, New Jersey USA5Departmentof Molecular Biology and Biochemistry; Rutgers University;Piscataway, New Jersey USA*Correspondence to: Eileen White; Center for Advanced Biotechnology and Medicine; 679 Hoes Lane; Piscataway, New Jersey 08854 USA; Tel.: 732.235.5329; Fax: 732.235.5795; Email: ewhite@ Original manuscript submitted: 08/03/07Manuscript accepted: 08/14/07Previously published online as an Autophagy E-publication:/journals/autophagy/article/4867KEy Wordsautophagy, breast cancer, beclin1, DNA damage, genomic instability Addendum to:Autophagy Mitigates Metabolic Stress and Genome Damage in Mammary TumorigenesisV. Karantza-Wadsworth, S. Patel, O. Kravchuk, G. Chen, R. Mathew, S. Jin and E. WhiteGenes Dev 2007; 21:1621-35[Autophagy 3:6, 610-613; November/December 2007]; ©2007 Landes BioscienceAbstrActAutophagy is an evolutionarily conserved process of cytoplasm and cellular organ-elle degradation in lysosomes. Autophagy is a survival pathway required for cellular viability during starvation; however, if it proceeds to completion, autophagy can lead to cell death. In neurons, constitutive autophagy limits accumulation of polyubiquitinated proteins and prevents neuronal degeneration. Therefore, autophagy has emerged as a homeostatic mechanism regulating the turnover of long-lived or damaged proteins and organelles, and buffering metabolic stress under conditions of nutrient deprivation by recycling intracellular constituents. Autophagy also plays a role in tumorigenesis, as the essential autophagy regulator beclin1 is monoallelically deleted in many human ovarian, breast, and prostate cancers, and beclin1+/‑ mice are tumor-prone. We found that allelic loss of beclin1 renders immortalized mouse mammary epithelial cells suscep-tible to metabolic stress and accelerates lumen formation in mammary acini. Autophagy defects also activate the DNA damage response in vitro and in mammary tumors in vivo, promote gene amplification, and synergize with defective apoptosis to accelerate mammary tumorigenesis. Thus, loss of the prosurvival role of autophagy likely contributes to breast cancer progression by promoting genome damage and instability. Exploring the yet unknown relationship between defective autophagy and other breast cancer- promoting functions may provide valuable insight into the pathogenesis of breast cancer and may have significant prognostic and therapeutic implications for breast cancer patients.Autophagy is an evolutionarily conserved catabolic process of self-digestion whereby double-membrane vesicles engulfing cellular organelles and cytoplasm form in the cytosol and fuse with lysosomes, where the sequestered contents are degraded and recycled for protein and ATP synthesis.1 The essential components of the autophagic machinery were initially identified in yeast, but several of the corresponding proteins have homologues in higher eukaryotes.2 During starvation or growth factor deprivation of normal cells, autophagy is a temporary survival mechanism, as it provides an alternative energy source.3,4 Autophagy also promotes cancer cell survival under metabolic stress.5 Autophagy is involved in both the recycling of normal cellular constituents and the removal of damaged proteins and organelles, as autophagy defects result in ubiquitinated protein aggregates and deformed organelles that may promote cellular degeneration.6,7 Autophagy is also a form of cell death, when allowed to proceed to excessive levels and when apoptosis-defective cells are triggered to die. Under these circumstances, it is often unclear whether autophagy contributes to cell death actively or represents an exhausted attempt to preserve cell viability under stress. Recent studies indicate that autophagy may play an active role in programmed cell death, but the conditions under which autophagy promotes cell death versus cell survival are still unknown.8AllElic loss of Beclin1 And brEAst cAncErbeclin1 is the mammalian orthologue of the essential yeast ATG 6/VPS 30 gene, which is required for autophagosome formation in a complex with the class III phosphatidylino-sitol-3-kinase (PIK3C3 or Vps34).9 beclin1 complements the autophagy defect present in atg6/vps30-disrupted yeast and in human MCF7 breast cancer cells.10 Originally entered in GenBank as a gene of unknown function during the positional cloning of BRCA1,11 beclin1 was independently rediscovered in a yeast two-hybrid screen of an adult mouse brain library for proteins interacting with the anti-apoptotic protein Bcl-2.12 The mouseCo py r ig ht L a nd e sB i o sc i e n c e 2007©2007 L A N D E S B I O S C I E N C E. D O N O T D I S T R I B U T E Autophagy611and human beclin1 genes share 93% identity at the nucleotide leveland 98% identity at the amino acid level.12 The human beclin1 gene is on chromosome 17q21 within a region commonly alleli-cally deleted in ovarian, breast and prostate cancers.13 Many breast carcinoma cell lines, although polyploid for chromosome 17, exhibit deletions of one or more beclin1 alleles 13 and human breast tumors show decreased Beclin1 levels compared to normal adjacent tissue.10 Restoration of Beclin1 and autophagy in MCF-7 cells is associated with inhibition of MCF7-induced tumorigenesis in nude mice.10 beclin1‑/‑ mice die early in embryogenesis, likely due to loss of the essential role of autophagy during development. Mammary tissuefrom aging beclin1+/‑ mice shows hyperproliferative, preneoplastic changes,14 indicative of a correlation between low Beclin1 levels and mammary tumorigenesis. beclin1+/‑ mice do not have increased inci-dence of mammary tumors, but rather are susceptible to lymphomas and carcinomas of the lung and liver after a long latency.14,15 T umors forming in beclin1+/‑ mice express wild-type beclin1 mRNA and protein, indicating that beclin1 is a haploinsufficient tumor suppressor.15 Clearly other mutations are required to cooperate with allelic loss of beclin1 for tumorigenesis, which has yet to be modeled in mice.thE tWo fAcEs of AutophAgy: cEllulAr surViVAlAnd tumor supprEssionAlthough the studies summarized above clearly implicate allelic loss of beclin1, and thus defective autophagy, in breast cancer pathogenesis, how loss of a survival pathway such as autophagy promotes breast tumorigenesis was initially an enigma. Allelic loss of beclin1 in immortalized mouse mammary epithelial cells (iMMECs) compromises the autophagy potential of these cells and results in decreased iMMEC viability under metabolic stress in two-dimensional (2D) culture, and in accelerated lumen formation in three-dimensional (3D) morphogenesis assays (Fig. 1). For apop-tosis-competent beclin1+/‑ iMMECs, metabolically stressed central acinar cells undergo apoptosis more readily than beclin1+/+ cells due to failure of autophagy, whereas in the case of Bcl-2-expressing beclin1+/‑ iMMECs, accelerated lumen formation involves induction of necrotic cell death due to concurrent autophagy and apoptosis defects.16In contrast to the prediction that autophagy-deficient cancer cells would similarly exhibit decreased survival in tumors in vivo, autophagy defects in iMMECs enhance tumor progression in ortho-topic growth in the mammary fat pad.16 Similarly, allelic loss of beclin1 in immortalized baby mouse kidney (iBMK) cells increases susceptibility of iBMK cells to metabolic stress in vitro, yet promotes tumorigenesis in vivo.5 An explanation reconciling the two intui-tively contradictory roles of autophagy resides with the finding thatdeficient autophagy, in the form of either beclin1 heterozygosity oratg5 deficiency, leads to DNA damage and genomic instability. This genetic instability likely requires inactivation of the p53 and Rb pathways, and thus non-functional checkpoints,16,17 and is most prominently manifested when apoptosis is also disabled, thus leading to an increased mutation rate and accelerated tumorigenesis.The animal studies mentioned above point toward a possible correlation between allelic loss of beclin1 and inactivation of p53 and pRb in human breast cancer. This intriguing possibility is worthy of further investigation, particularly given the high frequency of beclin1 loss, p53 mutant status and Rb pathway inactivation in breasttumors. p53 mutations are present in 30% of sporadic human breast carcinomas and Li-Fraumeni syndrome patients carrying germ line p53 mutations develop breast cancer at an early age.18 Furthermore, p53 mutations are associated with more aggressive and therapeuticallyrefractory disease.19 Similarly, Rb expression is aberrant in 20–30% of breast tumors, as demonstrated by lack of detectable Rb levels or loss-of-heterozygosity, and Rb function is further compromised by cyclin D1 amplification and p16ink4a loss, both commonly observed in breast tumors.20 Also, deregulation of the Rb pathway has thera-peutic and prognostic implications for breast cancer patients.21Investigation of any functional interaction between autophagy defects and mutant p53 status in mammary tumorigenesis can be performed by using two recently developed mouse models for Li-Fraumeni syndrome. These models involve conditional knock-in of two different dominant negative p53 mutants into the endogenous murine p53 locus,22 and mammary gland-specific expression of one of these p53 mutants resulted in spontaneous and carcinogen-in-duced mammary tumors at high frequency.18 Accelerated mammary tumorigenesis in beclin1+/‑ mice with mammary gland-specificexpression of a dominant negative p53 mutant will indicate synergy between deficient autophagy and p53 mutations. Validation of this result by examination of human breast tumors for concurrent p53 mutations and autophagy defects may provide valuable insight in breast cancer pathogenesis.dEficiEnt AutophAgy And gEnE AmplificAtionDeficient autophagy in the form of beclin1 heterozygosity promotes gene amplification,16,17 as demonstrated by a high frequency of PALA resistance and PALA-induced amplification of the CAD gene 23 in beclin1+/‑ iMMECs and iBMK cells. Gene ampli-fication is common in solid tumors 24 and is likely initiated by DNAdouble strand breaks in cells lacking robust checkpoints.25 Breast cancers frequently show genome copy number aberrations, including high-level amplifications that correlate with worse prognosis.26 For example, HER2/neu amplification is found in about 30% of human breast cancers and is associated with aggressive disease, poor clinical Figure 1. Deficient autophagy accelerates lumen formation in mammary acini. In 3D morphogenesis assays, metabolic stress and autophagy localize in the central acinar cells.16 If apoptosis-deficient and autophagy-competent, the central acinar cells are capable of coping with metabolic stress and/oranoikis 33and thus survive, resulting in mammary acini with filled lumens(left panel). Allelic loss of beclin1, and thus deficient autophagy, abrogatesthe survival advantage conferred to the central acinar cells by disabled apoptosis, resulting in induction of necrotic cell death and accelerated lumen formation (right panel).Co py r ig ht L a nd e sB i o sc i e n c e 2007©2007 L A N D E S B I O S C I E N C E. D O N O T D I S T R I B U T E .612Autophagy2007; Vol. 3 Issue 6outcome and chemotherapy resistance,26 whereas c-myc amplification is found in about 15% of breast cancers and is significantly associated with risk of relapse and death.27 Whether autophagy defects segregate with HER2/neu or c-myc or other specific gene amplification in breast cancer is not known. It is possible that the relationship between deficient autophagy and gene amplification is complex, as a vicious cycle may exist whereby autophagy defects promote genome damage and instability in the form of amplification involving genes that posi-tively regulate cellular growth and proliferation. The concomitant increased metabolic demands may in turn lead to higher levels of DNA damage in autophagy-deficient tumor cells and propagation of genomic instability.28,29functionAl stAtus of AutophAgy And brEAst cAncEr trEAtmEntAutophagy-deficient iMMECs accumulate DNA damage inresponse to metabolic, and likely replication, stress,16 raising the possibility that breast tumor cells with autophagy defects may be particularly sensitive to DNA damaging agents. In a similar situa-tion, BRCA1 and BRCA2 mutant cells, which are defective in DNA repair, have been shown to be exquisitely sensitive to certain DNA damaging agents, such as cisplatin, and to agents interfering with DNA repair, such as poly(ADP-Ribose) polymerase (PARP) inhibi-tors.30 It is therefore conceptually possible that autophagy-deficient breast tumor cells may also exhibit high sensitivity to agents causing DNA damage or those that interfere with DNA replication, as well as to drugs that inhibit DNA repair. The outcome of such treatment will likely depend on the functional status of the cell cycle check-points and the apoptotic cell death pathway.For autophagy-deficient tumor cells with intact apoptotic response, high levels of DNA damage will likely lead to prompt activation of apoptosis and cell death. In the case of tumor cells with combined autophagy and apoptosis defects, but functional p53, DNA damage accumulation may result in senescence, which could be therapeutically beneficial, at least for as long as p53 wild-type status is maintained. In the case of tumor cells triply deficient in apoptosis, autophagy and cell cycle checkpoints, and thus similar to Bcl-2-expressing beclin1+/‑ iMMECs, massive DNA damage may trigger cell death by mitotic catastrophe or necrosis. Potential problems with the later scenario include the possibilities that necro-sis-associated inflammatory reaction may promote tumorigenesis 5 and that genomically unstable tumor cells may acquire mutations that enable them to survive, ultimately resulting in disease relapse. Apoptosis-defective beclin1+/‑ iMMECs are prone to gene amplifi-cation under selective pressure such as PALA treatment,16 raising the concern that anticancer agents such as methotrexate, for which gene amplification mediates drug resistance, may be poor treatment choices for breast tumors with combined apoptosis, autophagy and cell cycle checkpoint defects.Alternatively, treatment of breast carcinomas having normal Beclin1 levels and intact autophagic response with an autophagy inhibitor may sensitize tumor cells to a variety of anticancer agents, especially DNA damaging agents and other S-phase-specific drugs. This principle has already been demonstrated in a Myc-induced lymphoma model in which inhibition of autophagy with either chlo-roquine or ATG5 short hairpin RNA (shRNA) enhanced the ability of alkylating drug therapy to induce tumor cell death.31 Similarly, autophagy inhibitors dramatically augmented the antineoplastic effects of the histone deacetylase (HDAC) inhibitor suberoylanilide hydroxamic acid (SAHA) in CML cell lines and primary CML cells expressing wild-type and imatinib-resistant mutant forms of Bcr-Abl, including T315I.32 Taking it one step further, concurrent inhibition of autophagy and reactivation of apoptosis may provide an even more efficient way to augment the therapeutic benefit of several anticancer drugs, as metabolically susceptible autophagy-deficient cells will be diverted to cell death by apoptosis upon treatment with drugs that cause metabolic stress, such as angiogenesis inhibitors, or DNA damage. In support of this hypothesis, inhibition of autophagy enhanced the ability of p53 activation to induce tumor cell death by apoptosis in a Myc-induced lymphoma model.31In conclusion, autophagy defects compromise the ability of mouse mammary, and very likely human breast, tumor cells to cope with metabolic stress resulting in genome damage and instability, which in turn may accelerate tumor progression. The functional interaction between defective autophagy and other breast cancer-promoting functions, in particular those involving gene amplification as a mechanism of tumorigenesis, remains to be investigated and could have significant prognostic and therapeutic implications for breast cancer patients. Furthermore, investigation of the impact that the functional status of autophagy and its pharmacological manipulation have on breast cancer treatment is of utmost interest and importance as a novel therapeutic target may be identified.References1. Yorimitsu T, Klionsky DJ. Autophagy: Molecular machinery for self-eating. Cell DeathDiffer 2005; 12:1542-52.2. Reggiori F , Klionsky DJ. Autophagy in the eukaryotic cell. Eukaryotic Cell 2002; 1:11-21.3. Lum JJ, Bauer DE, Kong M, Harris MH, Li C, Lindsten T, Thompson CB. Growthfactor regulation of autophagy and cell survival in the absence of apoptosis. Cell 2005; 120:237-48.4. Kuma A, Hatano M, Matsui M, Yamamoto A, Nakaya H, Yoshimori T, Ohsumi Y, TokuhisaT, Mizushima N. The role of autophagy during the early neonatal starvation period. Nature 2004; 432:1032-6.5. Degenhardt K, Mathew R, Beaudoin B, Bray K, Anderson D, Chen G, Mukherjee C, ShiY, Gelinas C, Fan Y, Nelson DA, Jin S, White E. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell 2006; 10:51-64.6. Hara T, Nakamura K, Matsui M, Yamamoto A, Nakahara Y, Suzuki-Migishima R,Yokoyama M, Mishima K, Saito I, Okano H, Mizushima N. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature 2006; 441:885-9.7. Komatsu M, Waguri S, Chiba T, Murata S, Iwata J, Tanida I, Ueno T, Koike M, UchiyamaY, Kominami E, Tanaka K. Loss of autophagy in the central nervous system causes neuro-degeneration in mice. Nature 2006; 441:880-4.8. Baehrecke EH. Autophagy: Dual roles in life and death? Nat Rev Mol Cell Biol 2005;6:505-10.9. Kametaka S, Okano T, Ohsumi M, Ohsumi Y. Apg14p and Apg6/Vps30p form a proteincomplex essential for autophagy in the yeast, Saccharomyces cerevisiae . J Biol Chem 1998; 273:22284-91.10. Liang XH, Jackson S, Seaman M, Brown K, Kempkes B, Hibshoosh H, Levine B. Inductionof autophagy and inhibition of tumorigenesis by beclin 1. Nature 1999; 402:672-6.11. Friedman LS, Ostermeyer EA, Lynch ED, Welcsh P , Szabo CI, Meza JE, Anderson LA,Dowd P , Lee MK, Rowell SE, et al. 22 genes from chromosome 17q21: Cloning, sequenc-ing, and characterization of mutations in breast cancer families and tumors. Genomics 1995; 25:256-63.12. Liang XH, Kleeman LK, Jiang HH, Gordon G, Goldman JE, Berry G, Herman B, LevineB. Protection against fatal Sindbis virus encephalitis by beclin, a novel Bcl-2-interacting protein. J Virol 1998; 72:8586-96.13. Aita VM, Liang XH, Murty VV, Pincus DL, Yu W, Cayanis E, Kalachikov S, Gilliam TC,Levine B. Cloning and genomic organization of beclin 1, a candidate tumor suppressor gene on chromosome 17q21. Genomics 1999; 59:59-65.14. Qu X, Yu J, Bhagat G, Furuya N, Hibshoosh H, T roxel A, Rosen J, Eskelinen EL,Mizushima N, Ohsumi Y, Cattoretti G, Levine B. Promotion of tumorigenesis by heterozy-gous disruption of the beclin 1 autophagy gene. J Clin Invest 2003; 112:1809-20.15. Yue Z, Jin S, Yang C, Levine AJ, Heintz N. Beclin 1, an autophagy gene essential for earlyembryonic development, is a haploinsufficient tumor suppressor. Proc Natl Acad Sci USA 2003; 100:15077-82.Co py rig ht L a nd e sB i o sc i e n c e 2007©2007 L A N D E S B I O S C I E N C E. D O N O T D I S T R I B U T E Autophagy61316. Karantza-Wadsworth V , Patel S, Kravchuk O, Chen G, Mathew R, Jin S, White E.Autophagy mitigates metabolic stress and genome damage in mammary tumorigenesis. Genes Dev 2007; 21:1621-35.17. Mathew R, Kongara S, Beaudoin B, Karp CM, Bray K, Degenhardt K, Chen G, Jin S,White E. Autophagy suppresses tumor progression by limiting chromosomal instability. Genes Dev 2007; 21:1367-81.18. Wijnhoven SW , Zwart E, Speksnijder EN, Beems RB, Olive KP , T uveson DA, Jonkers J,Schaap MM, van den Berg J, Jacks T , van Steeg H, de Vries A. Mice expressing a mammary gland-specific R270H mutation in the p53 tumor suppressor gene mimic human breast cancer development. Cancer Res 2005; 65:8166-73.19. Miller LD, Smeds J, George J, Vega VB, Vergara L, Ploner A, Pawitan Y, Hall P , Klaar S,Liu ET, Bergh J. An expression signature for p53 status in human breast cancer predicts mutation status, transcriptional effects, and patient survival. Proc Natl Acad Sci USA 2005; 102:13550-5.20. Bosco EE, Knudsen ES. RB in breast cancer: At the crossroads of tumorigenesis and treat-ment. Cell cycle (Georgetown, Tex) 2007; 6:667-71.21. Bosco EE, Wang Y, Xu H, Zilfou JT, Knudsen KE, Aronow BJ, Lowe SW, Knudsen ES. Theretinoblastoma tumor suppressor modifies the therapeutic response of breast cancer. J Clin Invest 2007; 117:218-28.22. Olive KP , T uveson DA, Ruhe ZC, Yin B, Willis NA, Bronson RT, Crowley D, Jacks T.Mutant p53 gain of function in two mouse models of Li-Fraumeni syndrome. Cell 2004; 119:847-60.23. Otto E, McCord S, Tlsty TD. Increased incidence of CAD gene amplification in tumori-genic rat lines as an indicator of genomic instability of neoplastic cells. J Biol Chem 1989; 264:3390-6.24. Albertson DG. Gene amplification in cancer. T rends Genet 2006; 22:447-55.25. Paulson TG, Almasan A, Brody LL, Wahl GM. Gene amplification in a p53-deficient cellline requires cell cycle progression under conditions that generate DNA breakage. Mol Cell Biol 1998; 18:3089-100.26. Chin K, DeVries S, Fridlyand J, Spellman PT , Roydasgupta R, Kuo WL, Lapuk A, NeveRM, Qian Z, Ryder T, Chen F , Feiler H, Tokuyasu T, Kingsley C, Dairkee S, Meng Z, Chew K, Pinkel D, Jain A, Ljung BM, Esserman L, Albertson DG, Waldman FM, Gray JW. Genomic and transcriptional aberrations linked to breast cancer pathophysiologies. Cancer Cell 2006; 10:529-41.27. Deming SL, Nass SJ, Dickson RB, T rock BJ. C-myc amplification in breast cancer: Ameta-analysis of its occurrence and prognostic relevance. British Journal of Cancer 2000; 83:1688-95.28. Jin S, DiPaola RS, Mathew R, White E. Metabolic catastrophe as a means to cancer celldeath. J Cell Sci 2007; 120:379-83.29. Jin S, White E. Role of autophagy in cancer: Management of metabolic stress. Autophagy2007; 3.30. Farmer H, McCabe N, Lord CJ, T utt AN, Johnson DA, Richardson TB, Santarosa M,Dillon KJ, Hickson I, Knights C, Martin NM, Jackson SP , Smith GC, Ashworth A. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 2005; 434:917-21.31. Amaravadi RK, Yu D, Lum JJ, Bui T , Christophorou MA, Evan GI, Thomas-TikhonenkoA, Thompson CB. Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma. J Clin Invest 2007; 117:326-36.32. Carew JS, Nawrocki ST , Kahue CN, Zhang H, Yang C, Chung L, Houghton JA, Huang P ,Giles FJ, Cleveland JL. Targeting autophagy augments the anticancer activity of the histone deacetylase inhibitor SAHA to overcome Bcr-Abl-mediated drug resistance. Blood 2007; 110:313-22.33. Debnath J, Mills KR, Collins NL, Reginato MJ, Muthuswamy SK, Brugge JS. The role ofapoptosis in creating and maintaining luminal space within normal and oncogene-express-ing mammary acini. Cell 2002; 111:29-40.。