J Mol Cell Biol-2011-Ollila-jmcb_mjr016
HIF-1α_纳米抗体的制备及其抑制黑素瘤生长的作用
山西农业科学 2023,51(12):1435-1441Journal of Shanxi Agricultural Sciences HIF-1α纳米抗体的制备及其抑制黑素瘤生长的作用李佳敏1,贾琼1,秦蓉芬1,迟志端1,王富明2,范瑞文1(1.山西农业大学动物医学学院,山西太谷 030801;2.晋中市庄子乡综合便民服务中心,山西晋中 030600)摘要:缺氧诱导因子1α(Hypoxia inducible factor 1α,HIF-1α)参与低氧微环境相关疾病的发生等过程,具有控制肿瘤生长和发展的功能。
黑色素瘤是一种发生于人和动物恶性程度较高的肿瘤。
为探明HIF-1α纳米抗体对黑色素瘤的影响,研究利用前期保存的羊驼源黑色素瘤细胞噬菌体文库筛选HIF-1α纳米抗体,经原核表达与纯化后,通过Western Blot和免疫组织化学验证HIF-1α纳米抗体与抗原的结合性;分别通过CCK-8法、划痕试验以及Western Blot法检测其对B16黑素瘤细胞的增殖和迁移能力及其相关分子表达的影响。
结果表明,经表达和纯化获得的HIF-1α纳米抗体分子质量约为16 ku,没有跨膜结构,具有亲水性。
通过Western Blot和免疫组织化学验证了其具有良好的抗原结合性。
在增殖试验和划痕试验中,与对照组相比,HIF-1α纳米抗体抑制了B16细胞的增殖和迁移,下调了靶基因VEGF的表达,并使细胞增殖和迁移相关蛋白Ras、ERK、RAC和RAF的表达量下调。
预测HIF-1α纳米抗体进入B16细胞内,与抗原结合,通过下游靶基因VEGF下调RAs、ERK、RAC、RAF的表达,从而对细胞增殖和迁移起抑制作用,可作为黑色素瘤治疗的新靶点。
关键词:HIF-1α;纳米抗体;B16细胞;Western Blot法;CCK-8法;细胞增殖;细胞迁移中图分类号:R739.5 文献标识码:A 文章编号:1002‒2481(2023)12‒1435‒07Effect on Preparation of HIF-1α Nano-Antibody and ItsInhibition of Melanoma GrowthLI Jiamin1,JIA Qiong1,QIN Rongfen1,CHI Zhiduan1,WANG Fuming2,FAN Ruiwen1(1.College of Veterinary Medicine,Shanxi Agricultural University,Taigu 030801,China;2.Jinzhong City Zhuangzi Integrated Convenient Service Center,Jinzhong 030600,China)Abstract:The hypoxia inducible factor 1α(HIF-1α) is involved in the occurrence of diseases related to hypoxia microenvironment and has the function of controlling tumor growth and development. As we known, melanoma is a highly malignant tumor occurring in animals and humans. To explore the effect of HIF-1α nano-antibody on melanoma, in this study, the phage library of alpaca-drived melanoma cells previously preserved in our laboratory was used to screen HIF-1α nano-antibodies. After prokaryotic expression and purification, the binding of HIF-1α nano-antibody and its antigen was verified by Western blot and immunohistochemistry. The effects of HIF-1α nano-antibody on the proliferation and migration of B16 melanoma cells and the expression of related molecules were detected by CCK-8, wound healing test, and Western blot methods. The results showed that HIF-1α nano-antibody obtained by expression and purification was hydrophilic protein without transmembrane structure and had a molecular weight of about 16 ku. Western blot and immunohistochemistry results showed that it had good antigenic binding. In the proliferation and wound healing experiments, HIF-1α nano-antibody inhibited the proliferation and migration of B16 cells, down-regulated the expression of target gene VEGF and the proliferation and migration related proteins Ras, ERK, RAC, and RAF, comparing with the control group. In Conclusion, it was predicted that HIF-1α nano-antibody entered B16 cells and combined with antigens and down-regulated the expression of RAs, ERK, RAC, RAF through the downstream target gene VEGF, which inhibited cell proliferation and migration, and could be used as a new target for melanoma treatment.Key words:HIF-1α; nano-antibody; B16 cells; Western Blot method; CCK-8 method; cell proliferation; cell migration氧是生命活动中所必需的物质,且在其中起重要作用[1]。
骨髓间充质干细胞源性外泌体促进小胶质细胞
学 报Journal of China Pharmaceutical University 2023,54(5):599 - 606599骨髓间充质干细胞源性外泌体促进小胶质细胞/巨噬细胞M2极化抑制急性期脑缺血大鼠炎症反应孙逸梅,毛诗慧,李琳,江伟锋,储利胜*(浙江中医药大学基础医学院,杭州 310053)摘 要 为探究骨髓间充质干细胞(bone marrow meaenchymal stem cells, BMSCs)源性外泌体(exosomes, Exos)对急性期脑缺血大鼠小胶质细胞/巨噬细胞M1/M2极化的影响,采用超高速离心法分离提取外泌体并鉴定;采用线栓法制备大鼠大脑中动脉阻塞(middle cerebral artery occlusion, MCAO)模型;采用Longa评分和角实验评价大鼠神经功能,2,3,5-氯化三苯基四氮唑(2, 3, 5-triphenyltetrazole chloride, TTC)染色检测大鼠脑梗死体积;采用CD16/32/Iba1、CD206/Iba1免疫荧光双标法检测小胶质细胞/巨噬细胞M1/M2表型;采用RT-qPCR检测大鼠脑缺血周边区CD86、诱导型一氧化氮合酶(inducible nitricoxide synthase, iNOS)、肿瘤坏死因子-α(tumor necrosis factor, TNF-α)、精氨酸酶1(arginase-1, Arg-1)、白细胞介素-10(interleukin-10, IL-10)和转化生长因子β(transforming growth factor beta, TGF-β)的mRNA表达。
实验结果表明,BMSC-Exos减少缺血周边区CD16/32+/Iba1+阳性细胞数量(P < 0.01),增加CD206+/Iba1+阳性细胞数量(P < 0.01),减少iNOS、CD86和TNF-α的mRNA表达,增加Arg-1、TGF-β和IL-10的mRNA表达(P < 0.05或P < 0.01)。
聚乙二醇化重组人粒细胞集落刺激因子用于多发性骨髓瘤自体造血干细胞动员的研究
聚乙二醇化重组人粒细胞集落刺激因子用于多发性骨髓瘤自体造血干细胞动员的研究丁筱1黄文阳2刘雪莲'杨艳萍1樊红琼1岳婷婷1邹德慧2邱录贵2靳凤艳1 '吉林大学第一医院肿瘤中心血液科,长春130021;2中国医学科学院血液病医院(中国 医学科学院血液学研究所)实验血液学国家重点实验室国家血液系统疾病临床研究 中心,天津 300020通信作者:斩凤艳,Email:fengyanjin@【摘要】目的探讨聚乙二醇化重组人粒细胞集落刺激因子(PEG-rhG-CSF)用于多发性骨髓瘤(MM)患者外周血造血干细胞动员(PBSCM)的效果及药物经济学价值。
方法回顾性分析2015年1月至2017年10月在吉林大学第一医院和中国医学科学院血液病医院住院治疗的9丨例初治MM患者资料。
根据患者意愿,采用大剂量化疗结合皮下注射PEG-rhG-CSF或重组人粒细胞集落刺激因子UhG-CSF)进 行干细胞动员,分别为42、49例。
分析两组动员后采集单个核细胞(MNC)数、采集物CD34+细胞数、动员 中最高中性粒细胞(mANC)数、动员的费用以及移植后白细胞和血小板植人时间,,结果PEG-rhG-CSF 组和rhG-CSF组的中位采集MNC 数分别为 5.86x10s / kg[ ( 1.08 ~ 24.54)x l〇8 / kg]和 6.61x l〇« / kg [(0.83 ~ 33.80)x l〇V kg],差异无统计学意义(t/= 883.00, P= 0.245); PEG-rhG-CSF组的中位采集物CD34+细胞数高于rhG-CSF组,分别为5.56 x l〇6/kg[(0.94~ 19.90) x l〇V kg]和4.82x l〇6/kg[(丨.12~ 14.61) x l〇V kg],差异有统计学意义((7= 732.00, P= 0.038)。
PEG-rhG-CSF组动员期间中位mANC数 较 rhG-CSF组低,分别为 20.50x l09/L[(7.26~61.30)x l0V L;^32.08x l0V L[(6.92~69.99)x l0V L],差异有统计学意义(i/= 490.00, P= 0.001)。
ACT001通过STAT1/CIITA/MHC-Ⅱ通路发挥抗炎抗氧化活性治疗脓毒症引起的急性肺损伤
网络出版时间:2023-11-3016:13:56 网络出版地址:https://link.cnki.net/urlid/34.1086.R.20231130.1319.012◇呼吸药理学◇ACT001通过STAT1/CIITA/MHC Ⅱ通路发挥抗炎抗氧化活性治疗脓毒症引起的急性肺损伤盛 磊,周杰诗,韩 旭,李伊楠,刘慧娟,孙 涛(南开大学药学院,天津 300350)收稿日期:2023-05-27,修回日期:2023-08-26基金项目:国家自然科学基金面上项目(No82272934);国家级大学生创新创业训练计划(No202210055112)作者简介:盛 磊(2002-),男,研究生,研究方向:药理学,E mail:2010402@nankai.edu.cn;孙 涛(1982-),男,博士,教授,博士生导师,研究方向:药理学,通信作者,E mail:tao.sun@nankai.edu.cndoi:10.12360/CPB202305082文献标志码:A文章编号:1001-1978(2023)12-2231-09中国图书分类号:R 332;R284 1;R322 35;R364 5;R631;R563 8摘要:目的 评价含笑内酯衍生物ACT001对脓毒症进程中急性肺损伤的治疗作用并探究其药理机制。
方法 动物水平上,小鼠腹腔注射LPS建立急性肺损伤模型,腹腔注射ACT001进行治疗,从小鼠个体存活状况、肺部炎症损伤及水肿情况等方面评价ACT001的药效;细胞水平上,以LPS刺激RAW264 7细胞构建模型,通过检测炎症反应和氧化应激水平探究其药理机制,并通过蛋白质组学结果分析其相关分子机制。
结果 动物水平上,ACT001可改善急性肺损伤小鼠生存率、减轻肺部炎症、降低血清中炎症因子水平;细胞水平上,ACT001通过抑制MHC-Ⅱ相关通路,促进RAW264 7细胞向抗炎表型极化,抑制NO和相关炎症因子产生的同时提高SOD含量并清除ROS。
原发性胆囊癌的早期诊断
原发性胆囊癌的早期诊断殷保兵【摘要】原发性胆囊癌是胆道常见恶性肿瘤,恶性程度高,发现时往往伴有肝脏转移,预后极差。
掌握原发性胆囊癌的高危因素,对胆囊癌高危人群进行密切随访和筛选,可提高原发性胆囊癌的早期诊断率,对改善胆囊癌的预后有重大意义。
%Primary gallbladder carcinoma (PGC) is the common malignant tumour in biliary tract and is associated with poor prognosis in patients with liver metastasis. It is important to understand the high risk factors of PGC, and closely follow up and screen the high-risk populations in order to improve the early diagnosis of gallbladder cancer and its prognosis.【期刊名称】《上海医药》【年(卷),期】2014(000)014【总页数】3页(P12-14)【关键词】胆囊癌;早期诊断;高危人群【作者】殷保兵【作者单位】复旦大学附属华山医院外科上海 200040【正文语种】中文【中图分类】R735.8原发性胆囊癌是最常见的胆道恶性肿瘤,约占消化道恶性肿瘤的第6位[1-2]。
2007年上海市胆囊癌发病率男性为5.90/10万,女性为10.22/10万,而同期全国胆囊癌死亡率高达4.07/10万[3-4]。
邹声泉等[5]报道2000年我国大陆原发性胆囊癌发病率占同期胆道疾病的0.4%~3.8%。
近年来,胆囊癌的发病率有上升趋势,而发病年龄则呈下降趋势。
胆囊癌的早期诊断非常困难,而其恶性程度极高,且易转移复发,因此预后极差,进展期胆囊癌的中位生存期仅6个月,5年生存率仅为5%[2]。
J Cell Physiol
J Cell PhysiolJ Cell Physiol is a scientific journal that focuses on the field of cellular physiology. It covers a wide range of topics related to the functioning and behavior of cells, with a particular emphasis on their physiological processes. The journal publishes high-quality original research papers, reviews, and commentaries that contribute to our understanding of cell physiology.Cell physiology is an interdisciplinary field that studies the functions and activities of cells, including their metabolism, communication, and response to environmental stimuli. Understanding cell physiology is crucial for understanding the basic processes that underlie complex biological systems and for developing new approaches for the diagnosis and treatment of diseases.Research published in J Cell Physiol encompasses a variety of topics, including cellular metabolism, cell signaling, cell cycle regulation, cell growth and differentiation, cellular responses to stress, and cell communication. The journal also welcomes studies investigating the impact of various factors on cell physiology, such as genetic and epigenetic changes, environmental factors, and therapeutic interventions.One area of interest in J Cell Physiol is cellular metabolism. This field aims to understand the complex network of metabolic pathways that cells utilize to generate energy and synthesize biomolecules. Elucidating the regulation of cellular metabolism can provide insights into various diseases, such as cancer and metabolic disorders, and can help identify potential therapeutic targets.Cell signaling is another key area covered in J Cell Physiol. Cells communicate with each other through a variety of signaling pathways, including cell surface receptors, intracellular signaling cascades, and gene expression regulation. Understanding how cells transmit and process these signals is essential for deciphering the molecular mechanisms underlying normal cellular functioning and for identifying aberrant signaling events associated with disease.The regulation of the cell cycle is vital for cell growth and tissue homeostasis. J Cell Physiol publishes studies investigating the control and coordination of the cell cycle, including the role of key regulatory proteins and the mechanisms involved in cell cycle checkpoints. Dysregulation of the cell cycle is a hallmark of many diseases, including cancer, and understanding its regulation can lead to the development of novel therapeutic strategies.Cell growth and differentiation are also areas of interest in J Cell Physiol. These processes are fundamental for tissue development, repair, and regeneration. Research published in the journal examines the molecular and cellular mechanisms governing cell growth and differentiation, as well as the factors that influence these processes, such as growth factors, signaling pathways, and the cellular microenvironment.Cellular responses to stress, both physiological and pathological, are investigated in J Cell Physiol. Cells have evolved intricate mechanisms to respond to various forms of stress, including oxidative stress, DNA damage, and proteotoxic stress. Understanding these adaptive responses can provide insights into the pathogenesis of diseases and may lead to the development of therapeutic interventions.Finally, J Cell Physiol also covers studies on cell communication. Cells communicate through various mechanisms, including direct cell-cell contact, secretion of signaling molecules, and extracellular vesicles. The journal publishes research on the molecular mechanisms underlying cell communication and its role in physiological and pathological processes, such as immune responses, tissue regeneration, and cancer metastasis.In conclusion, J Cell Physiol is a scientific journal dedicated to advancing our understanding of cellular physiology. It covers a wide range of topics related to the functioning and behavior of cells, with the aim of unraveling the intricate mechanisms underlying normal cellular processes and disease pathology. By publishing high-quality research papers, reviews, and commentaries, the journal contributes to the field of cell physiology and provides a platform for scientific exchange and collaboration.。
抗体药物工艺开发需要考虑的因素
有一个完整的了解。 如何选择最佳的候选药, 是建 立一个稳定的工艺的首要步骤。 目前, 生物制品以
2363
中国新药杂志 2015 年第 24 卷第 20 期
C h in e se Jou rn a l of N e wD ru g s 2015, 24(20)
所以本文讨 单抗类药物 ( 包括 Fc 融合蛋白 ) 为主, 论的工艺开发也以单抗类分子为主。 在中国, 抗体 药物主要分为生物类似药和生物创新药两大类 。生 物类似药是已经被临床和市场证明的药物 , 有现成 的产品做参照, 不存在选择候选药的问题。但是, 企 业要考虑根据是否填补国内市场空白, 并对产品自 身优势进行分析, 预期的市场份额进行分析来选择 项目。因此, 选择最佳候选药主要是针对生物创新 药。以创新性单抗为例, 不管用何种技术 ( 包括人 、 、 源性单抗 鼠源性单抗 兔源性单抗、 鸡源性单抗, 以 及用 单 细 胞 技 术 分 离 出 来 的 单 抗 ) 产 生 的 抗 [6 - 11 ] , 体 同一靶点的候选单抗数量越多, 可供选择 的范围越大。除了满足候选药有效性的基本要素, 如足够阻断靶点的亲和力 ( Kd ) 、高度特异的靶点 IC80 ) 以及 结合力、 有效抑制靶点功能效应 ( IC50 , 有知识产权方面的可操控空间外, 作为一个适合启 动工艺开发的候选药, 还必须具备工艺开发和放大 生产的可行性要素, 如高稳定性 ( Tm 值 ) 、 高可溶 性、 低免疫原性、 有利于生产过程和血液内循环的等 电点( pI, 如 pI = 5 6 或 pI = 8 9 ) 以及在关键结合 部 位 ( 如, complementarity determining regions, CDRs) 不存在潜在的不利因素 ( 包括 N糖、 去甲基 [12 - 14 ] 。 只有充分考虑上述 化、 脱酰氨化等位点 ) 等 综合因素后, 工艺开发才能顺利进行。此外, 必须根 , 据药物的适应症 对抗体依赖的细胞介导的细胞毒 dependent cellmediated cytotoxicity , 作 用 ( antibodyADCC ) / 补 体 介 导 的 细 胞 毒 作 用 ( complement dependent cytotoxicity , CDC ) 活性的需求, 选择合适的 [15 - 16 ] Ig) 的类型 。 免疫球蛋白( immunoglobulin, 2 表达系统和稳定表达细胞株的建立 一旦确立候选抗体药物后, 需要确定用于表达
Klotho_蛋白在子痫前期患者胎盘外泌体中表达及其对血管内皮细胞氧化应激的影响
第 49 卷第 6 期2023年 11 月吉林大学学报(医学版)Journal of Jilin University(Medicine Edition)Vol.49 No.6Nov.2023DOI:10.13481/j.1671‑587X.20230616Klotho蛋白在子痫前期患者胎盘外泌体中表达及其对血管内皮细胞氧化应激的影响薛筱蕾, 胥保梅(新疆医科大学第五附属医院产科,新疆乌鲁木齐830011)[摘要]目的目的:探讨Klotho在子痫前期(PE)患者来源的胎盘外泌体(Exo)中的表达,阐明其对血管内皮细胞氧化应激的影响。
方法方法:收集40例孕产妇的临床资料,其中正常妊娠(NP)者20名,PE患者20例,设为NP组和PE组,分离2组研究对象外周血中胎盘Exo。
将oe-Klotho和oe-NC质粒转染入人绒毛膜滋养层细胞HTR-8/SVneo中,作为oe-Klotho组和oe-NC组,收集HTR-8/ SVneo细胞Exo。
采用实时荧光定量PCR(RT-PCR)法和Western blotting法检测2组研究对象胎盘Exo和2组HTR-8/SVneo细胞Exo中Klotho mRNA及蛋白表达水平。
取生长状态良好的人脐静脉内皮细胞(HUVECs),按Exo来源不同将HUVECs分为PE-Exo组(与PE患者胎盘Exo共培养)、NP-Exo组(与NP胎盘Exo共培养)、oe-Klotho-Exo组(与转染oe-Klotho的HTR-8/SVneo细胞Exo共培养)和oe-NC-Exo组(与转染oe-NC的HTR-8/SVneo细胞Exo共培养)。
采用透射电子显微镜(TEM)观察Exo形态表现,Western blotting法检测Exo标记分子CD63、TSG101和胎盘Exo标记分子PLAP蛋白表达水平以鉴定Exo,荧光显微镜观察HUVECs对Exo的摄取情况。
酶联免疫吸附试验(ELISA)法检测各组HUVECs中一氧化氮(NO)、活性氧(ROS)和丙二醛(MDA)水平及超氧化物歧化酶(SOD)活性,RT-qPCR法检测各组HUVECs中内皮型一氧化氮合酶(eNOS)mRNA 表达水平,Western blotting法检测各组HUVECs中eNOS蛋白表达水平。
人参皂苷Rg1改善小鼠肢体缺血后血管新生
人参皂苷Rg1改善小鼠肢体缺血后血管新生王晓丽;金跃;刘鹭;张凯;杨乃全【摘要】目的:探讨人参皂苷Rg1是否具有促进小鼠缺血后肢血管新生的作用.方法:C57BL/6J小鼠随机分为两组:对照组(n=10)腹腔注射生理盐水0.1 mL,人参皂苷Rg1治疗组(n=10)腹腔注射Rg1 (10 mg· kg-1).每天1次,共2周.所有小鼠均结扎右后肢股动脉制作后肢缺血模型,手术后2周用多普勒血流成像评估双下肢血流,免疫荧光检测缺血肢体毛细血管密度,TUNEL法检测凋亡细胞,采用比色法检测缺血组织中一氧化氮浓度.结果:人参皂苷Rg1治疗能够改善小鼠缺血肢体的血流,且能明显地促进血管新生,提高缺血组织一氧化氮分泌,抑制缺血组织细胞的凋亡.结论:人参皂苷Rg1改善了小鼠缺血肢体血管新生及血流灌注.【期刊名称】《江苏大学学报(医学版)》【年(卷),期】2018(028)002【总页数】4页(P126-128,134)【关键词】人参皂苷Rg1;肢体缺血;血管新生;凋亡【作者】王晓丽;金跃;刘鹭;张凯;杨乃全【作者单位】徐州医科大学附属淮安第二人民医院检验科,江苏淮安223002;徐州医科大学附属淮安第二人民医院检验科,江苏淮安223002;徐州医科大学附属淮安第二人民医院检验科,江苏淮安223002;徐州医科大学附属淮安第二人民医院检验科,江苏淮安223002;徐州医科大学附属淮安第二人民医院心脏内科,江苏淮安223002【正文语种】中文【中图分类】R285.5下肢动脉疾病(peripheral arterial disease,PAD)在2型糖尿病患者常见,常常是预后不好的标志[1-2]。
有研究发现内皮型一氧化氮合酶(eNOS)功能受损在小鼠血管新生功能降低中发挥重要作用[3]。
人参具有很高的医用价值,目前已经从人参中分离鉴定出大量的活性成分,其中人参皂苷Rg1是发挥人参功效的主要成分[4]。
近来大量的数据提示Rg1能够促进血管新生以及人脐静脉内皮细胞增殖、趋化、血管发生[5-8]。
黄连素调节PI3K
黄连素调节PI3K/AKT/NF-κB信号通路对慢性湿疹大鼠皮肤损伤的影响秦宗碧,徐爱琴,蔡翔,邱百怡,王首帆,李伶华,朱立宏摘要:目的探讨黄连素对慢性湿疹大鼠皮肤损伤及磷脂酰肌醇-3-激酶(PI3K)/蛋白激酶B(AKT)/核因子-κB (NF-κB)信号通路的影响。
方法60只SD大鼠分为对照组,慢性湿疹组,黄连素低、中、高剂量组和泼尼松组,每组10只。
除对照组外,其余组大鼠背部涂抹2,4-二硝基氯苯(DNCB)构建慢性湿疹模型。
进行湿疹面积及严重度指数(EASI)评分;酶联免疫吸附试验(ELISA)检测血清组胺、胃泌素释放肽(GRP)、免疫球蛋白E(IgE)、白细胞介素(IL)-4、IL-6、肿瘤坏死因子α(TNF-α)、γ干扰素(IFN-γ)水平;苏木素-伊红(HE)染色观察皮损组织病理学改变;Western blot法检测皮损组织PI3K/AKT/NF-κB信号通路相关蛋白表达。
结果与对照组比较,慢性湿疹组大鼠皮损组织受损严重,血清组胺、GRP、IgE、IL-4、IL-6、TNF-α水平以及皮损组织IL-4、IL-6、TNF-α蛋白表达和p-PI3K/PI3K、p-AKT/AKT、p-NF-κB p65/NF-κB p65、p-NF-κB抑制蛋白α(IκBα)/IκBα比值升高,血清和皮损组织IFN-γ降低(P<0.05)。
与慢性湿疹组比较,黄连素各剂量组和泼尼松组大鼠皮损组织病理损伤有所改善,EASI评分下降,血清组胺、GRP、IL-4、IL-6、TNF-α水平以及皮损组织IL-6、TNF-α蛋白表达和p-PI3K/PI3K、p-AKT/AKT、p-NF-κB p65/ NF-κB p65、p-IκBα/IκBα比值降低,血清和皮损组织IFN-γ升高(P<0.05),同时黄连素中、高组和泼尼松组大鼠血清IgE和皮损组织IL-4降低(P<0.05),且黄连素高剂量组效果更好;黄连素高剂量组和泼尼松组上述指标比较差异无统计学意义(P>0.05)。
博士复试英文PPT
2015.09-
Master degree
****
Awards
Professional Third-class Scholarship from 2015-2018 academic years
Education Experience
• Traditional Chinese Medicine Bureau of Guangdong Province (NO. 20141065), • Natural Science Foundation of Guangdong Province (NO. 2017A32217007) • National University Student Innovation Program.
MenaINV for invasive feature Mena11a for epithelial-specific feature
Background
Two main AS regulators heterogeneous nuclear ribonucleoprotein (hnRNPs) and serine/arginine-rich (SR) proteins. Polypyrimidine Tract-Binding Protein 1 (PTBP1, also known as hnRNP I) binds preferentially to pyrimidine-rich sequences.
Objectives
Regulated by PTBP1 ?
The exact mechanism mediated Mena AS has not been elucidated. The purpose of this study is to explore the molecular mechanism of PTBP1 in regulating Mena alternative splicing and role of PTBP1 in lung carcinoma cells metastasis.
儿童急性髓系白血病的遗传基因异常及其意义
中国小儿血液与肿瘤杂志 2021年 6月第 26卷第 3期 JChinaPediatrBloodCancer,June2021,Vol26,No.3
·131·
2 异常基因在 MRD监测中的作用 21 融 合 基 因 RUNX1RUNX1T1/t(8;21)和 CBFBMYH11/inv(16)/t(16;16)均可用 于 治 疗 后 的 MRD监测 。 [1,811] 诱导或巩固结束后,RTqPCR 检测其转录产物 mRNA拷贝数(ABL做内参),较初 诊时降低 >3个 log者预后良好。究竟在诱导后还 是巩固后降低 >3个 log较有预后意义,视不同方案 而定。如果诱导结束后降低不足 3个 log,在巩固期 间继续降低,直到治疗完全结束后 <初诊时的 4个 log者预后仍好,提示连续监测的重要性。但如果将 RUNX1RUNX1T1和 CBFBMYH11分开统计,可 能 有不同 的 结 果,因 此 需 后 续 更 多 的 研 究[10]。关 于 KMT2A(MLL)基 因 重 排,在 AML 只 有 MLLT3 KMT2A/t(9;11)有详细研究[4],结果显示诱导缓解 后 RTqPCR <10-3和后续监测仍 <10-3复发率低。 BCRABL融合基因在 AML发生率低,用于 MRD监 测的意义尚未有足够的临床验证 。 [11] 以上研究结 果提示在用融合基因检测 MRD方面,连续监测的 重要性,治疗结束后仍低度阳性不影响预后。 22 突变基因 不是所有与 AML相关的突变基因 都适用于 MRD监 测 。 [1,45,1011] 造 血 干 细 胞 基 因 突 变后,获得竞争优势导致克隆造血,但仍有多系分化 和成熟的造血功能不属于白血病细胞,只有在继发 其他突变后才出现分化阻滞和无限增殖,发展为白 血病。随年龄增长干细胞的突变率增加,因此克隆 造血主要见于老年人,儿童少见。较常见的克隆造 血突变基因是编码表观遗传修饰因子的基因 DNMT3A、TET2、IDH、ASXL1,以 及 RNA 剪 接 子 和 Cohesins蛋 白 的 基 因 等。胚 系 突 变 基 因 RUNX1、 GATA2、CEBPA、DDX41和 ANKRD26也 不 适 用 于 MRD检测。与治疗前比较,缓解后突变基因的表达 水平仍没有明显下降,需考虑是克隆造血突变或胚 系突变基因。胚系突变基因可通过检测胚系组织 / 细胞(例如口腔黏膜等)DNA发现。
Am J Physiol Regul Integr Comp Physiol-2011-Olson-R297-312
You might find this additional info useful...195 articles, 48 of which you can access for free at:This article cites /content/301/2/R297.full#ref-list-1 5 other HighWire-hosted articles: This article has been cited by/content/301/2/R297#cited-by including high resolution figures, can be found at:Updated information and services /content/301/2/R297.full can be found at:and Comparative Physiology American Journal of Physiology - Regulatory, Integrativeabout Additional material and information /publications/ajpregu This information is current as of April 17, 2013.Copyright © 2011 the American Physiological Society. ISSN: 0363-6119, ESSN: 1522-1490. Visit our website at 12 times a year (monthly) by the American Physiological Society, 9650 Rockville Pike, Bethesda MD 20814-3991. levels of biological organization, ranging from molecules to humans, including clinical investigations. It is published investigations that illuminate normal or abnormal regulation and integration of physiological mechanisms at all publishes original American Journal of Physiology - Regulatory, Integrative and Comparative Physiology by guest on April 17, 2013/Downloaded fromThe therapeutic potential of hydrogen sulfide:separating hype from hopeKenneth R.OlsonIndiana University School of Medicine-South Bend,South Bend,IndianaSubmitted 21January 2011;accepted in final form 28April 2011Olson KR.The therapeutic potential of hydrogen sulfide:separating hype fromhope.Am J Physiol Regul Integr Comp Physiol 301:R297–R312,2011.Firstpublished May 4,2011;doi:10.1152/ajpregu.00045.2011.—Hydrogen sulfide(H 2S)has become the hot new signaling molecule that seemingly affects all organsystems and biological processes in which it has been investigated.It has also beenshown to have both proinflammatory and anti-inflammatory actions and proapop-totic and anti-apoptotic effects and has even been reported to induce a hypometa-bolic state (suspended animation)in a few vertebrates.The exuberance overpotential clinical applications of natural and synthetic H 2S-“donating”compoundsis understandable and a number of these function-targeted drugs have beendeveloped and show clinical promise.However,the concentration of H 2S in tissuesand blood,as well as the intrinsic factors that affect these levels,has not beenresolved,and it is imperative to address these points to distinguish between thephysiological,pharmacological,and toxicological effects of this molecule.Thisreview will provide an overview of H 2S metabolism,a summary of many of itsreported “physiological”actions,and it will discuss the recent development of anumber of H 2S-donating drugs that show clinical potential.It will also examinesome of the misconceptions of H 2S chemistry that have appeared in the literatureand attempt to realign the definition of “physiological”H 2S concentrations uponwhich much of this exuberance has been established.hydrogen sulfide-donating drugs;vasoactivity;ischemia reperfusion injury;sulfurcycle;gasotransmitterTHE INITIAL DISCOVERY by Hideo Kimura’s group that hydrogen sulfide (H 2S)1was a biologically relevant signaling molecule(reviewed in Ref.74)has heightened interest in the physiology and pharmacology of gaseous mediators.Unlike the first gas-eous signaling molecule,nitric oxide (NO),whose introduction was met with initial skepticism,H 2S has more or less been enthusiastically embraced by the scientific community,and there has been considerable effort to expeditiously imbue this obnoxious smelling gas into medical applications.This wave of exuberance has reheightened interest in the dietary sources of H 2S,and it has spawned the development of a number ofH 2S-“donating”drugs,many of which are in various stages ofclinical trials.However,it is becoming increasingly evident that there is still much to be learned about the basic properties of H 2S measurement,metabolism,and signaling mechanisms.This review will provide an overview of the effects of H 2S on physiological systems,summarize the new H 2S-donating drugs that are showing clinical potential,and take a critical look at the some of the remaining uncertainties surrounding H 2S chemistry and tissue concentrations.Hydrogen Sulfide as a Toxic GasThe toxic effects of H 2S have been known for centuries,and it remains second only to carbon monoxide as the most com-mon cause of gas-related fatalities in the workplace (46,190).H 2S has even gained notoriety in a recent spate of 220suicide cases in less than 3mo in Japan (107).Less is known of the effects of low-level ambient H 2S that are often associated with sewage plants,waste lagoons,natural gas/oil wells,and oil refineries,as well as a variety of other industrial applications.Recent studies on residents of Southeastern New Mexicoexposed to these environments have shown positive correla-tions with H 2S exposure and impaired neurobehavioral func-tions compared with controls (73).This suggests that even “therapeutic”use of H 2S is not without potential hazards.Thresholds for the major effects of H 2S exposure are shown inTable 1.The inhibitory effects of H 2S on mitochondrial cyto-chrome-c oxidase have been well characterized and this is generally assumed to be the focus of H 2S toxicity (34).How-ever,the clinical presentation of poisoning by H 2S and cya-nide,another well-known inhibitor of oxidative phosphoryla-tion that also inhibits cytochrome-c oxidase,are so distinct asto suggest different modes of toxicity (46).Another ratherunusual feature of H 2S toxicity is an extremely steep dose-effect response.Early studies in dogs (47)and other mammals (38,25),and more recent anecdotal information from human cases (46)have shown that H 2S toxicity is closely correlated with H 2S concentration and considerably less dependent upon the duration of exposure.This suggests that animals can 1Unless otherwise noted,H 2S refers to the sum of dissolved H 2S gas andHS Ϫ,often referred to as “sulfide”.At physiological pH,S 2Ϫis assumed to benegligible.Address for reprint requests and other correspondence:K.R.Olson,Indiana Univ.School of Medicine-South Bend,1234Notre Dame Ave.,South Bend,IN 46617(e-mail:kolson.1@).Am J Physiol Regul Integr Comp Physiol 301:R297–R312,2011.First published May 4,2011;doi:10.1152/ajpregu.00045.2011.Review by guest on April 17, 2013/Downloaded fromrapidly metabolize H 2S up to a critical level and,as a corollary,this efficient metabolic capacity should keep free H 2S at very low levels.These studies should,but have not often,raisedquestions regarding “physiological”concentrations of H 2S intissues and blood.This point is discussed in detail in a later section.Hydrogen Sulfide Biosynthesis and Metabolism Biosynthesis.Much of the metabolism of sulfides,including H 2S,passes through cysteine (Cys)metabolism (Fig.1).Cys-teine can be oxidized to cysteinesulfinate (Csa),or it can be desulfurated by reducing reactions that generate either H 2S or sulfane sulfur (a persulfide;149).In the oxidative—and gen-erally assumed catabolic—pathway for cysteine,cysteine di-oxygenase (CDO)catalyzes the addition of molecular oxygen to cysteine producing Csa,which may be further oxidized to sulfite or taurine (149).As perhaps a general indication of a broad-spectrum of sulfur-mediated effects on biological sys-tems,both Csa and its metabolites have themselves beenshown to affect a variety of physiological processes (68,100).CDO is found in liver,adipose,intestine,pancreas,and kidney.Because activity of CDO is highly regulated by dietary cys-teine,CDO is a regulator,if not the primary one,of cysteineavailability in vivo.By oxidizing excess and presumably toxiccysteine,CDO provides a constant and low-level background of cysteine for H 2S and sulfane sulfur biosynthesis.This may be important in preventing excessive H 2S production (33).H 2S can be produced from cysteine via a variety of biochem-ical pathways.Early studies indicated that cystathionine -syn-thase (CBS)was the predominant enzymatic pathway for H 2S production in the brain,whereas cystathionine ␥-lyase (CSE,also known as CGL)was responsible for H 2S production inthe Fig.1.Potential pathways for H 2S production and metabolism.Inset shows potential H 2S production from carbonyl sulfide.CA,carbonic anhydrase;CAT,cysteine aminotransferase;CBS,cystathionine -synthase;CDO,cysteine dioxygenase;CLY,cysteine lyase;CSD,cysteine sulfinate decarboxylase;CSE,cystathionine ␥-ligase;MST,3-mercaptopyruvate sulfurtransferase;R-SH,thiol.[Modified from Julian et al.(65),Kabil et al.(66),Singh et al.(143),and Stipanuk and Ueki (149).]Table 1.The effects of H 2S exposureAmbient H 2S,ppm Equivalent Total Plasma Sulfide,M a Effects0.01–0.30.003–0.1Threshold for detection1–30.3–1offensive odor,headaches10 3.38-h occupational exposure limit in Alberta,Canada15 4.915-min exposure limit in Alberta,Canada20–50 6.5–16.2eye and lung irritation10032.5olfactory paralysis250–50081.1–162.3pulmonary edema500162.3sudden unconsciousness (“knockdown”),death within 4to 8h 1000324.5immediate collapse,breathing ceases within several breaths All except “Equivalent Total Plasma Sulfide”column modified from Guidotti (46).a Equivalent plasma sulfide calculated after Whitfield et al.(186,supplemental information),assuming H 2S equilibrates across the alveolar membranes (169),Henry’s Law constant for H 2S at 37°C,140mM NaCl is 0.0649M·atm Ϫ1(27),and 20%of total sulfide exists as H 2S gas (115).ReviewR298THERAPEUTIC POTENTIAL OF H 2Sby guest on April 17, 2013/Downloaded fromvasculature (75).Recent studies have shown that CBS is present in the endothelium and two enzymes acting in tandem,cysteine aminotransferase (CAT)and 3-mercaptopyruvate sulfurtrans-ferase (MST),are also present in vascular endothelium and brain,whereas MST,but not CAT,is found in vascular smooth muscle(75,119).CAT transfers the amine group from cysteine to an acceptor,such as ␣-ketoglutarate,resulting in 3mercaptopyru-vate,which is then desulfurated by MST.In addition to H 2S,reduced sulfur in the form of sulfane sulfur can also be generated and,in fact,sulfane sulfur appears to be the only product of the CAT-MST pathway (66).Kimura’s group found relatively high levels of CAT-MST in the brain,and they proposed that this is a major pathway for H 2S production in the brain,but they alsosuggested that the H 2S is immediately “stored”as sulfane sulfur,the latter serving as a less labile form of H 2S that may be readilyaccessible during appropriate physiological conditions (60,141).However,reducing conditions and an alkaline environment are necessary for cleavage of this RS-S bond to form H 2S and becausethese conditions may not be routinely encountered intracellularly,the significance of the CAT/MST pathway in H 2S synthesisremains questionable.Both CBS and CSE have recently been shown to circulate in human plasma and to generate H 2S fromcysteine or homocysteine plus cysteine (13).This generation of H 2S has been proposed not only to reduce circulating homocysteine,but also to protect the endothelium from oxidative stress (12).Both CBS and CSE are cytosolic,pyridoxyl-5=-phosphate-dependent,enzymes.CBS activity appears to be controlled by a number of factors.S -adenosylmethionine (AdoMet)is an alloste-ric activator of CBS and when AdoMet levels are low,CBS activity decreases to direct sulfur flow through the transmethyla-tion pathway,thereby conserving methionine.Elevated AdoMet increases CBS activity to produce cysteine via the transsulfuration pathway (148).CBS contains a heme group that,when it binds with carbon monoxide (CO),inhibits the enzyme.CBS is also inhibited by reducing conditions,but contrary to a number of earlier reports,neither NO nor calmodulin appears to be physio-logical regulators of CBS activity (8).Using physiologically relevant substrate concentrations and kinetic simulations,Banerjee’s group (cf.23,66,143)con-cluded that 1)H 2S generation from cysteine is primarily catalyzed by CSE,2)H 2S production by CBS is throughcondensation of cysteine and homocysteine and depending onthe level of AdoMet activation,this may account for 25–70%of the H 2S generated under resting conditions,3)H 2S biosyn-thesis can occur independent of cysteine;condensation of twomolecules of homocysteine,catalyzed by CSE,yields homol-anthionine and H 2S,and may account for as much as 30%ofthe total H 2S biosynthesis,4)CSE activity is substantiallyincreased by elevated homocysteine,whereas CBS activity isunaffected.Condensation of two homocysteine molecules,along with the condensation of homocysteine and cysteine,appear to be important clearance pathways in hyperhomocys-teinemia.It has been proposed that during severely elevatedhomocysteine (200M),as seen in hyperhomocysteinemia,␣,␥-elimination and ␥-replacement of homocysteine,catalyzed by CSE,may produce excessive amounts of H 2S and thereby contribute to the cardiovascular pathology associated with this condition (23).Commonly used inhibitors of CSE include propargyl glycine (PPG)and -cyanoalanine.Aminooxyacetate (AOA)is com-monly used to inhibit CBS and hydroxylamine to inhibit both enzymes (although a number of studies erroneously claim this is a specific inhibitor of CBS).Unfortunately,none of these inhib-itors are specific for sulfur metabolism and H 2S production;furthermore,they are often poorly absorbed by tissues (153).Other Potential Biosynthetic PathwaysThere are numerous other potential metabolic pathways forH 2S generation that have been described in invertebrates (Fig.1;Ref.65),but these have not been systematically evaluated in mammalian tissues.The resurgent interest in H 2S will undoubt-edly lead to reevaluation of these,heretofore,overlooked biosyn-thetic pathways and identification of novel ones.Indeed,theliterature is replete with studies that show that many of thebiological effects produced by H 2S can also be affected by avariety of other sulfur-donating molecules.One potentially novel pathway that needs to be investigated is H 2S production from carbonyl sulfide (COS;chemical structure:O ϭC ϭS).Like H 2S,COS is a gas that has both natural (volcanoes,hot springs,oils and trees)and man-made (biomass and fossil fuel consumption,wastewater treatment,etc.)origins and it is the most prevalent sulfur gas in the atmosphere (152).COS is the only volatile sulfurthat is increased in exhaled air of patients with cystic fibrosis (69)or of lung transplant patients during the acute rejection phase (150).COS is also exhaled by patients with chronic liver disease (135).COS has been demonstrated to be produced by porcine coronary arteries in vitro,and the rate of COS production is enhanced by stimulating the vessels with ACh or the calcium ionophore,A23187(7).In solution,COS slowly decomposes to H 2S,but this reaction is greatly accelerated by the enzyme carbonic anhydrase.In fact,CO 2and COS may be the primary substrates of this enzyme (134).Whether or not the biosynthesis of COS is related to H 2S production and subsequent signaling events remains to be determined.Metabolism (Inactivation)Oxidation of H 2S occurs in the mitochondria (53).As shown in Fig.2,two membrane-bound sulfide:quinone oxidoreducta-ses (SQR)oxidize sulfide to the level of elemental sulfur,simultaneously reducing cysteine disulfide,and resultingin Fig.2.Mitochondrial oxidation of H 2S.Two sulfide:quinone oxidoreductase (SQR)in the mitochondrial membrane (stippled box)oxidize sulfide to the level of elemental sulfur,simultaneously reducing a cysteine disulfide,andresulting in the formation of a persulfide group at one of the SQR cysteines (SQR-SSH).Sulfur dioxygenase (SDO)then oxidizes one persulfide to sulfite(H 2SO 3),consuming molecular oxygen and water in the process.The second persulfide is transferred from the SQR to sulfite by sulfur transferase (ST)producing thiosulfate (H 2S 2O 3).Electrons from H 2S are fed into the respiratory chain via the quinone pool (Q),and ultimately transferred to oxygen bycytochrome-c oxidase (complex IV).ReviewR299THERAPEUTIC POTENTIAL OF H 2S by guest on April 17, 2013/Downloaded fromformation of persulfide groups at one of the SQR cysteines.Sulfur dioxygenase (SDO)then oxidizes one of the persulfides to sulfite (H 2SO 3),consuming molecular oxygen and water in the process.Sulfur from the second persulfide is transferred from the SQR to sulfite by sulfur transferase producing thio-sulfate (H 2S 2O 3).Most thiosulfate is further metabolized to sulfate by thiosulfate reductase and sulfite oxidase.Electrons from H 2S are fed into the respiratory chain via the quinone pool (Q),and finally transferred to oxygen at complex IV.Oxygen consumption is obligatory during H 2S metabolism,and 1mol of oxygen is consumed for every mol of H 2S oxidized along the electron transport chain (53).Oxidation of H 2S to thiosulfate requires additional oxygen at the level of SDO,resulting in a net utilization of 1.5mol of oxygen per mol of H 2S (or 0.75mol of O 2per mol H 2S;Ref.82).Metabolism of H 2S through SQR appears ubiquitous in tissues,a notable exception being brain (82).It is important to note that sulfide oxidation in the mitochondria appears to take priority over oxidation of other carbon-based substrates,ensuring its effi-cient removal (24).This plus the fact that the capacity of cells to oxidize sulfide appears to be considerably greater than the estimated rate of sulfide production (24)ensures that intracel-lular H 2S concentrations are very low.Interestingly,the statin,atorvastatin,increases H 2S production in perivascular adipose tissue by producing coenzyme Q 9deficiency and thereby inhibiting mitochondrial oxidation (189).The relationship between H 2S and O 2consumption is clas-sical hormesis;at low concentrations,H 2S stimulates oxygen consumption (and may even result in net ATP production),whereas it is inhibited by elevated H 2S.This was originally shown in invertebrates and lower vertebrates and more recently demonstrated in the mammalian colon (45).At higher concen-trations,H 2S inhibits the respiratory chain by directly inhibit-ing cytochrome-c oxidase (24).The exact H 2S concentration at which this occurs is unclear;purified cytochrome-c oxidase is inhibited by Ͻ1M H 2S,whereas progressively greater (2or 3orders of magnitude)higher H 2S concentrations are needed to inhibit the enzyme in intact mitochondria and then whole cells.Cytochrome-c oxidase is half maximally inhibited byϳ20M H 2S and may not be fully inhibited until H 2S concentrations reach 40–50M (6,24).This may reflect diffusion limitation as the enzyme becomes further removed from the exogenously administered H 2S.It also should provide a cautionary note in interpreting studies that routinely employ 100M–1mM H 2S to demonstrate a “physiological”effect.The converse,i.e.,the effect of O 2on H 2S consumption,is discussed in H 2S and oxygen sensing .H 2S Biology Interest in H 2S biology has spawned nearly as many reviews (at latest count,32in 2010alone)as original articles.Reviews have even appeared where,at the time,the effects of H 2S on a particular system were unknown (87,196).The following sections provide a brief overview of H 2S biology.For further details,the reader is referred to a few of the most recent reviews following each section.H 2S and the nervous system.Potentiation of the N -methyl-D -aspartate (NMDA)receptor with the resultant alteration of long-term potentiation (LTP)in the hippocampus was the first biological effect ascribed to H 2S (1).Not long thereafter,it was noted that patients with Down syndrome had elevated concen-trations of H 2S in cerebral spinal fluid.This would be predicted from the fact that chromosome 21encodes CBS (which may be the major H 2S-producing enzyme in the brain)and is overex-pressed in these patients (70).It has also been suggested that deficiencies in H 2S biosynthesis are associated with Alzhei-mer’s disease (see Ref.37,reviewed in Ref.130)and that exogenous H 2S may have therapeutic potential by reducing amyloid beta protein plaques (201).H 2S has been proposed to modulate nociception (40,144),induce opioid receptor-depen-dent analgesia (30),prevent neurodegeneration and movement disorders in mouse models of Parkinson’s disease (55,72),and may reduce the stress response of the hypothalamic-pituitary-adrenal axis (102).It has also been proposed to antagonize homocysteine-induced neurotoxicity (162).The protective effects of H 2S have been demonstrated in a number of neurological systems.H 2S has been shown to protect neurons against hypoxic injury (165),inhibit hypochlo-rous acid-mediated oxidative damage (183),and increase glu-tathione production and suppress oxidative stress in mitochon-dria (76).Conversely,H 2S has been shown to mediate cerebral ischemic damage (129)and produce vanilloid receptor 1-me-diated neurogenic inflammation in airways (170).H 2S increases cAMP production in neurons and subsequent activation of PKA may account portion of the LTP.Other functions of H 2S include upregulation of GABA B receptor and neuronal hyperpolarization via K ATP channel activation and induction of calcium waves in astrocytes (130),regulation of intracellular pH in glial cells (98),and the above-mentioned increase in glutathione production.For recent reviews,see Refs.56,130,160and 144.H 2S and the gastrointestinal system.The initial interest in H 2S in the gastrointestinal (GI)system stemmed from the well-known production of H 2S by sulfate-reducing bacteria in the colon and the presumed need to protect tissues from this toxic molecule (133).Today,more is known about the effects of H 2S in the colon than any other segment of the GI tract;however,anti-inflammatory actions of H 2S in the stomach appear to be of important therapeutic value and other areas have received increased attention as well.H 2S is synthesized in the stomach,jejunum,ileum,and colon.CSE immunoreactivity is diffusely distributed through-out the gastrointestinal tract most likely due to its association with the vasculature,whereas CBS staining is predominantly in muscularis mucosa,cell mucosa,and lamina propria but not associated with goblet,crypt,and epithelial cells (105).H 2S relaxes smooth muscle in the stomach (28)intestine(113),and colon (29).The mechanisms of H 2S on GI motilityhave not been fully resolved,and in most instances,we are merely left with a list of factors that do not affect motility.Inthe stomach H 2S acts partly via activation of myosin light-chain phosphatase (28);in the colon,the effects of H 2S areindependent of intracellular calcium and not mediated throughknown K ϩchannels,myosin light-chain phosphatase,or Rho kinase (29),and in the ileum,H2S relaxation is independent ofextrinsic or enteric nerves,NO,KATP ,and KCa ϩchannels(113).H2S inhibits pacemaker activity of mouse small intestine interstitial cells of Cajal by modulating intracellular calcium through mechanisms independent of K ϩchannels (122).Pro-liferation of these interstitial cells is also stimulated by H 2S,which acts via phosphorylation of AKT protein kinase (57).ReviewR300THERAPEUTIC POTENTIAL OF H 2Sby guest on April 17, 2013/Downloaded fromH 2S stimulates chloride secretion in the intestine by targeting vanilloid receptors (transient receptor potential vanilloid 1)on afferent nerves,which,in turn,activate cholinergic secretomo-tor neurons via release of substance P (79).H 2S has both anti-inflammatory and inflammatory effects in the GI tract;however,the former is perhaps better character-ized and appears to be of therapeutic value.In the colon,H 2S is anti-inflammatory and enhances ulcer healing,independent of nitric oxide synthase and K ATP channel involvement (176).H 2S production is increased in experimental models of colitis and H 2S protects against and promotes resolution of this colitis (177).However,H 2S modulates the expression of genes in-volved in cell-cycle progression and may trigger both inflam-matory and DNA repair processes,which may contribute to colorectal cancer (5).In the pancreas,H 2S is a mediator of inflammatory caeru-lein-induced pancreatitis (17,158,159).H 2S acts through ICAM-1expression and stimulates neutrophil adhesion through the NF-B and Src-family kinases (157).However,H 2S has also been shown to protect pancreatic cells from oxidative stress (164).Inhibition of CSE,which is found in both hepatocytes and the bile duct,stimulates biliary bicarbonate secretion,whereas exogenous H 2S inhibits it (39).Bile acids increase liver CSE expression via activation of the farnesoid X receptor,the resultant H 2S production is proposed to maintain vasodilation and minimize the chance for portal hypertension (131).For recent reviews,see Refs.64,71,96,106,133,and 175.H 2S and the cardiovascular system.Collectively,the in-volvement of H 2S on heart and blood vessel physiology has received more attention than any other organ system,even though the therapeutic applications of H 2S are less evident.The vasodilatory effects of H 2S on systemic blood vessels were the first cardiovascular effects of this transmitter de-scribed (54).This has been confirmed repeatedly and even observed in pulmonary arteries of diving mammals (119).H 2S-induced relaxation appears to depend on extracellular Ca 2ϩ(203),and although K ATP channels,are frequently as-sumed to mediate the H 2S relaxation (63,86,203,204),thismechanism typically accounts for no more than half of the relaxation in most vessels.In some animals,such as the mouse,K ATP channels are not involved at all in the response.H 2S mayalso signal via other pathways,such as activation of adenylate cyclase,which,in turn,inhibits superoxide formation,NADPH oxidase,and Rac 1activity (112);it may produce intracellularacidosis and alter intracellular redox status,stimulate an anion exchanger (97),or operate through Ca 2ϩ-dependent K ϩ(K Ca )channels (77,161,206).Relaxation of rat aorta by exogenous H 2S does not depend on vascular prostaglandin synthesis,PKC,or cAMP,nor does it involve superoxide or H 2O 2production (77,78,204).Observations that H 2S sulfhydratesand may regulate biological activity of numerous proteins,including actin (109),suggests that additional key steps in H 2S-mediated vascular signaling are soon to be unraveled.However,even this mechanism has been questioned on the basis of the seemingly nonselectivity and promiscuity of this process (96),and the suggestion that for this to occur,the cysteine residues must be in the oxidized state,and these are rare in the reducing intracellular environment (66).H 2S may also indirectly relax blood vessels in vivo through its ability to inhibit angiotensin-converting enzyme and thus prevent forma-tion of the vasoconstrictor ANG II.Recent evidence has turned to H 2S as the elusive endothe-lium-derived hyperpolarization factor,the third endothelium-derived relaxing factor that,along with NO and prostacyclin,signals vasodilation (180).Crosstalk between H 2S,NO,and CO has been suggested to contribute to vasoactivity and,although CO inhibits CBS (8),interactions between H 2S and NO are far from resolved.NO production has been shown to be directly inhibited by H 2S (81),or indirectly stimulated by it through activating NF-B,which activates the ERK1/2,which,in turn,activates inducible nitric oxide synthase (iNOS)(62).H 2S relaxations have been reported to be independent of NO synthesis or cGMP activation (77,78,203).As described above,NO does not appear to directly affect H 2S production (8).There is also evidence that H 2S and NO may form a simple S-nitrosothiol with vasoactive properties of its own (184).Reports of H 2S-mediated vasoconstrictory responses in mammalian systemic vessels are less common,and many of these show an endothelium-dependent effect that has been attributed to H 2S inactivation of NO.Low concentrations of H 2S (Ͻ200M)produce endothelium-dependent contraction of human internal mammary arteries and rat and mouse aortas (2,81,181),and low-dose H 2S infusion increases blood pres-sure in the rat (2).These contractions have been proposed to result from H 2S inactivation of endothelial NO via production of an inactive nitrosothiol (2,181),whereas Kubu et al.(81)showed that H 2S directly inhibited NO production.Other studies suggest that H 2S may have direct,albeit modest,constrictory effects on systemic vascular smooth muscle.Lim et al.(95)observed 1M H 2S contractions of rat aortas that were partially independent of both the endothelium and K ATP channels and due,in part,to down-regulation of cAMP.Direct H 2S-mediated vasoconstriction has been demonstrated in sys-temic vessels of nonmammalian vertebrates,and H 2S contracts pulmonary vessels in terrestrial mammals in response to hyp-oxia (32,117,118).H 2S has a variety of other effects on the vasculature that arenot directly vasoactive.At times,the findings are contradic-tory,but nevertheless,many are suggestive of therapeutic potential.H 2S has been shown to be both proinflammatory and anti-inflammatory,to reduce leukocyte adhesion,to inhibit platelet aggregation,and although it is proangiogenic,to re-duce deleterious vascular remodeling that often accompanies vascular damage (35,89,155).H 2S is not only a mild antiox-idant,but it also stimulates cysteine uptake and synthesis ofglutathione.H 2S has been implicated in hypotension associatedwith septic and hypovolemic shock,and inappropriate H 2S regulation of insulin secretion in type II diabetes may contrib-ute to macrovascular and microvascular pathologies (85).In-hibition of plasma renin activity by H 2S is antihypertensive inrenin-dependent hypertensive rats (99)and can potentially augment the depressor effect of H 2S vasodilation.While H 2S has been shown to have negative inotropic and chronotropic effects on the heart (207),most interest has centered around its cardioprotective abilities.Numerous stud-ies have shown that transient application of H 2S or H 2S donors can mimic hypoxic preconditioning and postconditioning andthat increased endogenous H2S biosynthesis can also protectthe heart from ischemia/reperfusion injury (reviewed by Refs.35,83,156).Furthermore,the potential for H 2S-mediated ReviewR301THERAPEUTIC POTENTIAL OF H 2S by guest on April 17, 2013/Downloaded from。
阳离子抗菌肽融合细胞穿透肽增强其肿瘤细胞杀伤活性的实验研究_刘珊
第28卷 第1期2011年 2月 生物医学工程学杂志Journal of Biom edical EngineeringV ol.28 No.1February 2011阳离子抗菌肽融合细胞穿透肽增强其肿瘤细胞杀伤活性的实验研究*刘 珊 杨 浩 蔡华伟 万 琳 卢晓风(四川大学华西医院卫生部移植工程与移植免疫重点实验室,成都610041)摘 要:具有肿瘤细胞杀伤活性的膜作用型抗菌肽因不易诱导抗性而在肿瘤治疗中极具潜力。
阳离子抗菌肽的细胞结合能力是影响其细胞杀伤活性的重要因素。
通过偶联有较强细胞结合能力的细胞穿透肽可能增强阳离子抗菌肽的细胞杀伤活性。
本文选择了细胞毒性较弱的抗菌肽M P,与细胞穿透肽A ntp偶联,并观察融合肽细胞杀伤活性的变化。
结果发现,未偶联的抗菌肽M P和细胞穿透肽A ntp对肿瘤细胞无明显杀伤,但两者偶联形成的融合肽M P GA通过破坏细胞膜而显示强烈的肿瘤细胞杀伤作用。
这表明将抗菌肽与细胞穿透肽偶联能够显著提高其细胞毒性,可能成为研发新型抗肿瘤药物的途径之一。
关键词:肿瘤生物治疗;抗菌肽;细胞穿透肽中图分类号 Q-3 文献标识码 A 文章编号 1001-5515(2011)01-0110-05Enhancement of Cytotocixity of Cantionic Antimicrobial Peptide in Tumor Cells by Conjugation to Cel-l penetrating PeptideLiu Shan Yang Hao Cai Huawei Wan Lin Lu Xiaofeng(K ey L ab of Tr ansplant E ngineer ing and I mmunology,M inistry of H ealth,West China H ospital,S ichuan Univ e rsity,Cheng du610041,China)Abstract:Due to their low er r isk fo r induction of r esistance,membrane-act ive antimicr obial peptides wit h anticancer effect are attr active in cancer therapy.Because cell binding co nt ributes to the cytot ox icity o f peptide,it is po ssible to enhance the cyto tox icity o f antim icrobial peptide in tumor cells by conjugat ion t o a cel-l penetrat ing peptide(CPP).In this paper,a fusion peptide M PGA by conjug atio n of antimicro bial peptide M P to CPP A ntp at its N-ter minus w as constructed.Aft er compared the cy toto xicity o f unconjugat ed M P with that o f the fusio n peptide,it was fo und that M P GA sho wed higher cy toto xicity than that of unco njug ated M P.And the fusion pept ide M P GA induced cell death in tumor cells by membr ane disruption.T hese results demo nstr ated that the cytot ox icity o f antimicr io bial peptide can be significantly enhanced by co njugation t o CPP,which might be an effective w ay to develo p nov el ant icancer drug s. Key words:Cancer bio therapy;Antimicrobia l peptide;Cel-l penetr ating peptide(CPP)引言恶性肿瘤是导致现代人类死亡的主要疾病之一。
靶向T细胞免疫治疗急性髓性白血病的策略
doi:10.3971/j.issn.1000-8578.2024.24.0016李扬秋 研究员,二级教授,博士生导师,暨南大学血液学研究所所长,广东省医学领军人才。
中国病理生理学会实验血液学委员会副主委,Blood Science 副主编、J Immuno Ther Cancer (Clinical/Translational Cancer Immunotherapy 专题副主编)、J Hematol Oncol 、Exp Hematol Oncol 、Exp Hematol 、《中华血液学杂志》《中国免疫学杂志》《肿瘤防治研究》等杂志编委。
主要从事血液肿瘤分子发病机制和肿瘤T 细胞免疫及靶向治疗研究。
在Nat Med, Mol Cancer, J Hematol Oncol 和Adv Sci 等国内外期刊发表论文200多篇。
靶向T细胞免疫治疗急性髓性白血病的策略李扬秋Strategy of Targeted T Cell Immunotherapy for Acute Myeloid Leukemia LI Yangqiu Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, ChinaAbstract: T cell dysfunction is a common feature in patients with acute myeloid leukemia (AML). The up-regulation of immune checkpoint (IC) proteins resulting in T cell exhaustion is a key reason for T cell dysfunction. Immunotherapy with IC inhibitors exerts a remarkable effect on AML. However, due to the heterogeneity of T cell exhaustion and other factors that impair T cell function in patients with AML, the optimization of targeted T cell immunotherapy strategy for AML might be based on the multidimensional investigation of immune deficiency with different T cell subtypes.Key words: Acute myeloid leukemia; T cell; Exhaustion; Immune checkpoint protein; Immunotherapy Funding: Natural Science Foundation of China (No. 82293630, 82293632, 82070152)Competing interests: The author declares that she has no competing interests.摘 要:急性髓性白血病(AML )常伴有T 细胞免疫功能不全,由于免疫检查点(IC )分子表达上调引起T 细胞耗竭是其重要原因之一。
Cell Mol Life Sci.-2011-Regulation of flowering time_ all roads lead to Rome
REVIEWRegulation of flowering time:all roads lead to RomeAnusha Srikanth •Markus SchmidReceived:19November 2010/Revised:8February 2011/Accepted:17March 2011/Published online:6April 2011ÓSpringer Basel AG 2011Abstract Plants undergo a major physiological change as they transition from vegetative growth to reproductive development.This transition is a result of responses to various endogenous and exogenous signals that later inte-grate to result in flowering.Five genetically defined pathways have been identified that control flowering.The vernalization pathway refers to the acceleration of flower-ing on exposure to a long period of cold.The photoperiod pathway refers to regulation of flowering in response to day length and quality of light perceived.The gibberellin pathway refers to the requirement of gibberellic acid for normal flowering patterns.The autonomous pathway refers to endogenous regulators that are independent of the photoperiod and gibberellin pathways.Most recently,an endogenous pathway that adds plant age to the control of flowering time has been described.The molecular mecha-nisms of these pathways have been studied extensively in Arabidopsis thaliana and several other flowering plants.Keywords Arabidopsis thaliana ÁFlowering time ÁPhotoperiod ÁVernalization ÁGibberellic acid ÁPathway integrators AbbreviationsGA Gibberellic acid SD Short day LD Long dayIntroductionLand plants have evolved increasingly complex modes of reproduction.While today’s mosses and ferns still reproduce using motile spores/sperm,nonflowering (gymnosperms;with the notable exceptions of Ginkgo biloba and Cycada-ceae)and flowering seed plants (angiosperms)do not.Instead,angiosperms have evolved specialized organs (the flower and components thereof)to further reproduction.The earliest fossil of a flowering plant,Archaefructus liaoning-ensis ,dates back about 125million years.Nowadays fruits of angiosperm flowers form a major source of the staple diet of people and livestock.Flowers are also appreciated for their aesthetic value,their fragrance and their medicinal proper-ties.The formation of flowers is a prerequisite for successful sexual reproduction and as such,the correct timing of this event has adaptive value,in particular in non-self-fertile species in which flowering has to be synchronized between individuals.Even in self-fertile plants,the induction of flowering is tightly controlled by environmental and endogenous cues such as day length,temperature,and hor-monal status.Because of their importance,plants and their flowers have attracted a lot of interest not only from breeders,but scientists as well.Flowering time research in the pre-molecular biology era Regulation of flowering time has been studied for more than 100years [1].These early studies established that plant flowering is regulated by factors that include day length (photoperiod).Subsequently,plants can be classified as long day (LD)plants that induce flowering when day length exceeds a certain threshold,short day (SD)plants that flower when days are short and nights are long,and day-neutral plants whose flowering is not dependent on theA.Srikanth ÁM.Schmid (&)Department of Molecular Biology,Max Planck Institute for Developmental Biology,Spemannstrasse 37-39/VI,72076Tu¨bingen,Germany e-mail:Markus.Schmid@tuebingen.mpg.deCell.Mol.Life Sci.(2011)68:2013–2037DOI 10.1007/s00018-011-0673-yCellular and Molecular Life Scienceslength of the day.This logically led to the question of how and where plants determine photoperiod.Over the years,several hypotheses were put forward to explain how plants perceive photoperiod[1],but it was not until the1930s that a more elaborate solution was sug-gested by Erwin Bu¨nning.As a result of his investigations into‘‘circadian oscillations,’’Bu¨nning proposed the exis-tence of a‘‘biological clock’’that was entrained by the day-night cycle.Bu¨nning further hypothesized that the24h day was divided into two phases,a light sensitive(photophile) and a dark sensitive(scotophile)phase,and that a circadian oscillator regulated the shift from one phase to the other.In this scenario,light behaved as an external signal because its presence during either phase would indicate to the plant if the day was long or short.The Bu¨nning hypothesis was later expanded by Pittendrigh[2]into the‘‘external coin-cidence’’model.In contrast to earlier models,the external coincidence model depends on the presence of light at specific times during the24-h cycle.Pittendrigh[3]later proposed an alternative mechanism,the‘‘internal coinci-dence’’model,in which two different circadian rhythms were entrained by dusk and dawn.As seasons progressed, one of these rhythms would shift phase relative to the other,resulting in(partial)overlap of the two oscillations, which would trigger downstream events,in this case the induction offlowering.Around the same time that the works mentioned above established the basic principles that enable plants(or any other organism)to measure day length,others followed up on the question of where in the plant photoperiod is per-ceived.Knott[4],for example,exposed different parts of plants to light and found that the cue toflower required that the leaves,but not the shoot apex,be exposed to light.This suggested that under inductive photoperiod,plants produce in their leaves aflower-triggering substance that is trans-ported to the shoot apex,an idea that was formalized in the ‘‘florigen hypothesis’’by the Russian botanist Mikhail Chailakhyan.Subsequent experiments such as grafting leaves fromflowering plants onto scions grown under noninductive photoperiod and exposing individual leaves to inductive day length soon confirmed theflorigen hypothesis[1,5,6].Interestingly,the velocity and pattern of movement of theflorigen was found to match those of photosynthetic assimilates,indicating that theflorigen might move through the phloem from the leaf to the apex [7,8].While the presence of aflorigenic substance was confirmed in many experiments,its nature has been a matter of debate for a very long time.Apart from day length,the quality of light also plays a significant role in the transition toflowering.Plants grown at high density or under a dense canopy experience a shift in the red to far-red ratio of the incoming light and respond by stem elongation and precocious induction offlowering,a process known as shade avoidance syndrome[9].Apart from the red/far-red ratio,blue light is also known to regulate the transition toflowering.For example,it has been demonstrated that a day-neutral response can be induced in SD plants upon exposure to high intensities of blue or white lights[10].While these experiments clearly show that various aspects of light(in particular day length, light quantity and quality)control thefloral transition in many plant species,light is by no means the only envi-ronmental factor involved.Other than the various aspects of light,temperature is probably the next most important external cue that affects flowering because plants need a conducive temperature to survive and propagate.In the context offlowering regula-tion,one can distinguish between the effects of the ambient growth temperature and those of a prolonged period of cold.Gassner[11]was among thefirst to describe the requirement of long periods of cold forflowering among different species of plants.He found a marked difference in cold requirements between biennials or winter annuals and spring plants or summer annuals.In1928,the Russian scientist Lysenko coined the name‘‘jarovization’’to describe this response of plants.This was later translated into‘‘vernalization.’’The now accepted definition of ver-nalization as‘‘the acquisition or acceleration of the ability toflower by a chilling treatment’’was suggested in1960by Chourd.In general,summer annuals have a facultative vernalization response while the winter annuals have an obligate vernalization requirement and cannotflower without a prior cold treatment.The normal vernalization parameters range between1and7°C for a period of 1–3months,depending on the species.Furthermore,breaks of warm temperature were shown to disrupt the effect of vernalization in rye[12].An interesting aspect of vernali-zation is thatflowering does not necessarily commence immediately after plants experience normal growth tem-peratures.Instead,an extended period of time can pass beforeflowering is actually induced.However,once the vernalized state is achieved,it is mitotically stable.This is referred to as the‘‘memory of winter’’and is due,as we will discuss below,to the epigenetic silencing of certain vernalization responsive genes.The vernalized state is however not passed on from parent to progeny as silencing of these genes is reset during meiosis.The role of temperature in plant development has been studied since the18th century.Being sessile organisms,it is essential for plants to develop a mechanism to identify conducive temperatures for different life processes, includingflowering.An early review of the effects of tem-perature onflowering was supplied by Wang[13].A more recent review on how plants perceive temperature and dif-ferentiate between day-to-dayfluctuations at a molecular level has been provided by Samach and Wigge[14].2014 A.Srikanth,M.SchmidIn1926,the Japanese scientist Kurosawa noticed that rice seedlings that were infected with the fungus Gibber-ella fujikuroi grew so tall that they were unable to stand upright.In1938,gibberellic acid(GA),the chemical that caused this effect on the rice seedlings,was isolated.In 1952,Anton Lang applied GA to rosettes of Samolus parviflorus and Crepis tectorum and noticed that the plants responded by bolting andflowering.Subsequently,GA was often referred to as theflowering ng however was able to distinguish between theflorigen and GA and concluded that while GA was not theflorigen,it somehow regulated theflorigen[15,16].Conflicting results on the role of GA inflowering were observed in different species. While GA enhancedflowering in some plants,it sup-pressedflowering in others.Exogenous application of GA resulted inflowering in noninductive photoperiods in cer-tain plants,but not in all cases investigated[17].GA was also able to bypass the requirement for vernalization[18].Besides GAs,carbohydrates have also been shown to play an important role in regulating thefloral transition[19].Sugars are produced through photosynthesis and playa vital role in plant development.The major plant sugar is sucrose,which has been shown to accumulate at the shoot apex just prior to transition toflowering.For example, S.alba plants grown in short days accumulated sugars in the apex upon increased irradiation[20].Despite the success of these early works,it was not until the advent of modern plant genetics and molecular biology, particularly in Arabidopsis thaliana,that the mechanisms underlying thefloral transition were better understood.Flowering time mutants in Arabidopsis thalianaIt was Laibach[21]who proposed A.thaliana as a model plant for genetics.Its small genome size,the ease with which it could be crossed and cultivated,its short life cycle, and the large number of seeds produced made it an ideal model organism.Since then,A.thaliana has become the paradigm for understanding plant genetics and molecular biology,although several other plants species are also widely used for scientific research.In general,no environmental conditions are known that completely preventflowering of A.thaliana.Also,no A. thaliana mutants have been reported that,like the veg mutant in peas,fail toflower.However,genetic variation in the response to environmental cues clearly exists among natural accessions of A.thaliana[22–24].Most accessions that are commonly used in the laboratory are summer annuals that do not require vernalization.However,winter annuals do exist,and genetic analyses have shown that natural alleles of two genes,FLOWERING LOCUS C and FRIGIDA,to a large extent account for the vernalization requirement of these accessions[25,26].With respect to photoperiod,flowering time in A.thaliana is dependent on the length of the day,with long days(16h light)in general promotingfloral transition compared to short days(8h). However,A.thaliana will eventuallyflower even under SD and has hence been classified as a facultative LD plant.Re´dei[27]used X-ray irradiation to identify co,gi,and ld as loci that are involved inflter,Koornneef et al.[28]identified11loci that resulted in lateflowering time when mutated in the Landsberg erecta(L er)accession of A.thaliana.These loci included fd,fwa,fe,fpa,fy,fve, ft,fha,fca,and two of the loci(gi and co)that Re´dei had previously identified.Most of the mutations were recessive, although co was intermediate and fwa was almost com-pletely dominant.Flowering time of these mutants was assayed under different photoperiods and in response to vernalization.fca,fve,fy,and fpa were found toflower late under both SD and LD,butflowering could be accelerated by vernalization treatment.These genes define the core elements of what is now known as the autonomous path-way offlowering in A.thaliana.In contrast,mutations in gi,co,and fha delayedflowering specifically under LD suggesting that these genes are involved in a photoperiod-sensing pathway.The last couple of years have seen tremendous progress in our understanding of the molecular regulation offlow-ering time.Numerous genes involved in this process have been identified,and we are beginning to understand how these genes integrate various endogenous and environ-mental cues to control the onset offlowering.Here we present a comprehensive overview of the current state of research on the different pathways that facilitateflowering and the different factors that regulate the transition from vegetative to reproductive growth.Environmental control offloweringAs outlined above,flowering time is under the control of diverse environmental stimuli such as temperature and photoperiod.Photoperiod is perceived in the leaves from which the long distance signal dubbed theflorigen is transmitted to the shoot apex to induceflowering.In the following sections,we will review the genetic and molecular mechanisms that allow plants to regulateflow-ering time in response to the environment.Regulation offlowering by day lengthThe photoperiod pathway—or how to measure day length? As one moves away from the equator,the length of the day varies significantly between summers and winters.Plants have developed the ability to sense this distinction and useFlowering time regulation in Arabidopsis2015it as an indicator to control the onset of flowering.The cascade of events responsible for measurement of day length and the subsequent initiation of flowering is referred to as the photoperiod pathway.Light is perceived by plants at different wavelengths by specialized photoreceptors.Phototropins (blue),crypto-chromes (blue),and phytochromes (red/far-red)are the three main classes of plant photoreceptors [29–31].Several models have been proposed regarding how plants (or organisms in general)might measure day length (see above).Common to all of these hypotheses is that they require internal oscillators,i.e.,genes regulated by the circadian clock,and environmental changes such as the day-night cycle to synchronize these rhythms.Interest-ingly,phytochromes and cryptochromes themselves have been shown to be regulated by the circadian clock,indi-cating the existence of a regulatory loop that modulates gating and resetting of the circadian clock [32].Re´dei [27]was the first to describe mutants that were insensitive to inductive day length.Among them was the constans (co )mutant.a putative zinc finger transcription factor [33],the temporal and spatial regulation of which turned out to be keyto thephotoperiod-dependent induction of flowering (Table 1)[34].CO expression is under the control of the circadian clock,which causes a basic oscillation of CO expression with a phase of 24h,and a maximum approximately 20h after dawn under SD conditions [35].This phasing of CO expression is further modified under LD by the activity of three other proteins:these three genes are themselves regulated by the circadian clock.In long days,both the FKF1and GI proteins follow the same phase with maximum levels being reached 13h after dawn [38,39].In contrast,under SD conditions,GI peaks at 7h after light onset,but FKF1peaks 10h after light onset [36].Interaction assays in yeast showed that FKF1physically interacts with GI [38].Using truncated FKF1protein constructs,the regions of interaction were narrowed down to the LOV (light,oxygen,or voltage)domain of FKF1and the N terminus of GI.Interestingly,FKF1protein binds GI only in the presence of blue light,which it perceives through its flavin-binding domain.As a result of this,FKF1-GI complexes are formed much more efficiently during long days when there is sufficient exposure of the FKF1protein to blue light and FKF1and GI proteins peak at the same time,unlike under short days,where the pro-teins are in different phase and the light,which is required for FKF1-GI complex formation,is lacking [38](Fig.1).FKF1and GI do not regulate CO expression directly but through interactions of FKF1-GI with CDFs [36,37].The CDFs are a family of transcription factors that play an important role in maintenance of CO mRNA levels.The quadruple cdf mutant accumulates CO mRNA both during the day and night and flowers early both in short and long days.CDF1has been shown to directly bind to the CO regulatory regions and act as a repressor of CO transcrip-tion [37].Chromatin immunoprecipitation (ChIP)using tagged versions of the GI protein also showed enrichment of 17different amplicons distributed throughout the CO promoter [38].In addition,ChIP using tagged versions of FKF1showed that this protein binds to similar regions on the CO promoter as GI and CDF1[38].Finally,analysis of the abundance of the three proteins showed that CDF1peaks first,followed by GI,and then finally FKF1peaks in the afternoon in long days [37,38].Together these studies suggest that CDF1protein first binds to the CO promoter in the morning.As soon as there is sufficient GI,the CDF1-GI complex is formed that represses CO transcription.Once FKF1protein peaks,it interacts with the CDF1-GI complex and targets CDF1for degradation through its F-Box domain to finally activate transcription of the CO gene (Fig.1)[38].While CDF1and CDF2are both targets of the FKF1-GI ubiquitination pathway,it is unknown whe-ther the other members of this family follow the same mode of degradation [36].Taken together,the activity of FKF1/GI/CDFs results in a second peak of CO expression towards the end of the subjective LD at approximately 16h after dawn (Fig.1).CO ,however,is not only regulated at a transcriptional level,but also at the level of its protein stability and accumulation.Central to the posttranslational regulation of CO are CONSTITUTIVELY PHOTOMORPHOGENIC ).COP1functions as an E3ubiquitin ligase and has been shown to act downstream of the cryptochrome signalling but upstream of CO .The flowering phenotype of the cop1co double mutant resembled the co single mutant in both long and short days,placing CO genetically downstream of COP1.Similarly,expression of FLOWERING LOCUS T (FT ),a major target of CO (see below),was upregulated in the cop1single mutants in both short and long days but not in cop1co double mutants,suggesting that COP1acts as a negative regulator of CO function,possibly by directing CO for degradation by the 26S proteasome-dependent pathway.This was later shown to be the case,when Liu et al.[40]reported that CO-GST was ubiquitinated specifically by COP1.Furthermore,constitutive overexpression of the CO protein fused to luciferase in cop1mutants resulted in a drastic increase in luciferase signal when compared to wild type,providing evidence that degradation of CO by COP1also occurred in vivo .Finally,yeast-2-hybrid analysis and in vitro protein interaction studies also verified that COP12016 A.Srikanth,M.SchmidFlowering time regulation in Arabidopsis2017 Table1List of importantflowering time regulators2018 A.Srikanth,M.Schmid Table1continuedinteracted with CO.The interaction domain was further narrowed down to the WD repeat domain of the COP1protein.These interactions were also confirmed in vivo by fusing COP1and CO to the yellow and cyan fluorescence proteins and observing their co-localization in nuclear bodies [40].Besides COP1,SPA proteins have also been shown to regulate CO [41,42].In A.thaliana ,the SPA protein family consists of four members that have a WD domain similar to COP1.The spa1mutant flowered early in short days but was indistinguishable from wild type in long days.The early flowering phenotype of the spa1mutant was completely suppressed by mutations in co .The other three spa single mutants did not show any difference in flower-ing in short or long days.The spa1spa3spa4triple mutant,however,flowered earlier than the spa1single mutant in short days but was only slightly earlier than wild type in long days.This indicates that the SPA3and SPA4proteins act redundantly with SPA1to de-repress flowering spe-cifically in SD conditions [42].While CO mRNA levels were found to be unaltered in the spa triple mutants,CO protein levels were strongly elevated in the triple mutants when compared to wild type,suggesting that SPA proteins were regulating the CO protein posttranslationally [42].In agreement with this hypothesis,co-immunoprecipitation studies established that all four SPA proteins indeed interacted with CO through its CCT domain.Further,the SPA1,SPA3,and SPA4proteins were shown to physically interact with the coiled coil domain of COP1[41,43].These results suggest that SPA proteins enable degradation of the CO protein by the COP1-mediated ubiquitination [42].Analysis of CO protein accumulation was also per-formed under different light conditions usingCO:GFPFig.1a ,b Regulation of CONSTANS at a transcriptional and protein level.a In short days,FKF1and GI proteins peak at different times and hence are not able to efficiently repress CDF1,a transcriptional inhibitor of CO.CO protein levels are very low to start with in SD as indicated by the graph.PHYB plays a vital role in maintaining this low level of CO in the early hours of the day.Another protein,DNF,is important for maintaining low levels of CO between 4and 7h after dawn.Active CRY protein represses COP1,a ubiquitin ligase that marks CO for degradation.In the dark,the inactive CRY is no longer able to repress COP1resulting in almost no CO protein being present.b In long days,both FKF1and GI peak at approximately 13h after dawn,resulting in active repression of CDF1,and thereby,COtranscription.The protein levels are regulated by PHYB in the early morning hours,while active CRY and PHYA repress PHYB during the rest of the day.Active CRY protein also binds to and inhibits transport of COP1into the nucleus,hence preventing it from efficiently ubiquitinating the CO protein.Genes are represented in green ,and proteins in orange .Dull colors represent inactive genes/proteins,while bold colors indicate active genes/proteins.Dashed box shows weak complex formation,and the grey box shows efficient complex formation.The clock is a 24h clock.The graph represents expression of CO protein through the day (SD/LD),with the day length represented on the x -axisFlowering time regulation in Arabidopsis 2019fusions.GFPfluorescence was detectable in plants grown under white,blue and far red but not in plants that had been exposed to red or were kept in the dark.This indicated that the accumulation of the CO protein was influenced by a photoreceptor[44].Subsequently,phyB mutants were shown to exhibit increased levels of CO in the red light and early morning hours,indicating that PHYB plays a major role in regulation of CO in the early hours of the day (Fig.1)[44,45].Another interesting protein that has been shown to repress CO independently of GI/FKF1/CDF is DAY NEUTRAL FLOWERING(DNF)[46].In dnf mutants, the circadian rhythm of CO is disturbed,resulting in precocious expression of CO and earlyflowering,in pho-toperiods as short as6h.The molecular mechanism by which DNF regulates CO expression is currently unknown. However,DNF encodes a functional membrane-bound E3 ligase,suggesting that DNF targets a repressor of CO for degradation by the proteasome pathway.In the end,the complex regulation of CO enables the plant to discriminate SD,where CO protein is not being stably produced,from LD,where CO protein accumulates towards the end of the day.The mechanisms involved turn out to be a mix of both the internal and external coinci-dence mechanisms originally proposed by Pittendrigh [2,3].The former is implemented in the synchronized expression of GI and FKF1,which ensures a boosted CO expression by timed degradation of the CDFs specifically under LD.The latter is enacted in the regulation of FKF1 and COP1/SPA activity through light,which leads to the accumulation of CO protein specifically towards the end of a long day.An important aspect of this is that regulation of CO happens in the leaves and not at the shoot apex where flowers will eventually be formed[34].The photoperiod pathway—or what good is knowing day length anyway?Forflowering to occur,the information that a plant expe-riences in the inductive photoperiod needs to be transferred from the leaves to the apex.The question arose as to whether CO itself might constitute a long distance signal (florigen).However,expression of CO mRNA from vari-ous tissue-specific promoters suggested that CO regulates production of a systemicflower-promoting signal in the leaves,but does not act as aflorigen[34,47].Instead,several lines of evidence now indicate that a protein called FLOWERING LOCUS T(FT)is contribut-ing to thefloral induction by acting as a long distance signal between leaves and the shoot meristem.FT was simultaneously cloned by two independent groups using an activation tagging approach[48,49]and a large chromo-somal deletion mutant caused by a T-DNA insertion [50,51].The FT gene encodes a protein with similarities to Raf kinase inhibitory protein(RKIP)and phosphatidyl-ethanolamine binding protein(PEBP).These proteins are known to inhibit Raf,and thereby result in signal trans-duction through the MAP kinase pathway.However,since FT lacks certain key residues conserved in all PEBP and RKIP proteins[52],the molecular function of FT is not entirely clear.Analysis of FT expression revealed not only that its expression is much higher in long days,but also that it follows a circadian pattern,peaking in the evening [35,53].Promoter GUS constructs showed that the FT gene is transcribed in the phloem companion cells,where CO is also present[54].Temporal and spatial expression of FT in the vasculature is controlled by a complex orches-tration of activating and repressive inputs.The latter include proteins that regulate chromatin structure[55]and thus accessibility of FT locus for transcription factor binding.Several studies have demonstrated that trimethy-lation of lysine27in the amino terminus of histone H3 (H3K27me3)provides an assembly platform for repressive complexes.In this context it is interesting to note that recent genome-wide surveys indicate that allflowering time genes but CO are H3K27me3targets[56–58].H3K27 trimethylation is carried out by the polycomb repressive complex2(PRC2)and mutants in a number of PRC2genes [i.e.CURLY LEAF(CLF),EMBRYONIC FLOWER2,etc.]flower early[59–61].In these mutants,earlyflowering was shown to be at least in part due to ectopic expression of FT, suggesting that PRC2complexes play a major role in repressing FT during vegetative growth.Chromatin-immunoprecipitation experiments revealed that CLF in fact bound FT chromatin,establishing a direct link between PRC2and FT repression[62].While PRC2components can be identified rather easily in plants,proteins homolo-gous to PRC1are more elusive.However,it has been suggested that LIKE HETEROCHROMATIN PROTEIN1 (LHP1)might act as a PRC1-like corepressor[63].lhp1 mutantsflower somewhat earlier than wild type and,sim-ilar to mutants in PRC2components,this earlyflowering has been attributed to increased FT expression.Further-more,LHP1is directly associated with the FT locus[64], indicating that,like PRC2,LHP1(PRC1)contributes to FT repression.FT mRNA is not readily detected in short days,but mRNA levels rise rapidly in the leaves upon transfer from short to long days and are detectable even after exposure to a single long day[37,65,66].Several lines of evidence place FT genetically down-stream of CO.In the phloem of SUC2::CO plants,FT mRNA abundance was increased and ft mutations strongly suppressed the earlyflowering of SUC2::CO[34].Over-expression of CO in ft-10plants did not rescue the late flowering phenotype,but FT,when expressed from the2020 A.Srikanth,M.Schmid。
光谱法研究尼美舒利与牛血清白蛋白的相互作用
光谱法研究尼美舒利与牛血清白蛋白的相互作用刘里;成飞翔【摘要】利用荧光、同步荧光和紫外可见光谱来研究牛尼美舒利(Nime)与血清白蛋白(BSA)的相互作用.结果表明:Nime能猝灭BSA的荧光,遵循静态猝灭过程.通过分析同步荧光光谱可知,Nime改变了BSA的二级结构,使BSA腔内疏水环境的极性减弱.BSA的亚螺旋域ⅡA是主要结合位置,离酪氨酸残基更近,有微弱的药物负协同作用.BSA能运输Nime,是一个自发过程,其作用力类型主要为静电作用力.结合位置实验表明Nime与BSA的结合主要发生在亚螺旋域ⅡA.有微弱的药物负协同作用.对于Nime,有一个结合位点.焓为负值,熵为正值,表明在结合过程中静电作用力起了主要作用.另外,它是一个自发放热过程.我们的研究结果可能对尼美舒利的临床研究和疗效具有参考价值.【期刊名称】《南京师大学报(自然科学版)》【年(卷),期】2016(039)002【总页数】6页(P50-55)【关键词】尼美舒利;荧光猝灭;相互作用【作者】刘里;成飞翔【作者单位】曲靖师范学院化学化工学院,云南曲靖655011;曲靖师范学院化学化工学院,云南曲靖655011【正文语种】中文【中图分类】O657.3尼美舒利(Nime)是一种非甾体抗炎药,因抗炎、解热镇痛作用比对乙酰氨基酚和布洛芬起效更快,而且几乎全部通过尿液排泄,即使多次服用也不会出现积累现象[1],所以被一致认为一个具有良好发展前途的药物[2].目前已在50多个国家使用,其市场规模超过10亿美元.但该药对中枢神经和肝脏造成损伤的案例时常出现,因此被禁止用于12岁以下儿童.牛血清白蛋白(简称BSA)因价格低廉,在分子结构和氨基酸序列上与人血清白蛋白极其相似,被广泛地用于与药物相互作用的研究[3].至今还未见光谱法研究Nime与BSA的结合特征的报道.本文优化了体系的实验条件,探讨了Nime与BSA的相互作用机理,测定了3个温度下的结合位点数、热力学常数,分析了Nime与BSA结合对蛋白质构象的影响、体内常见共存物质的影响、药物的协同作用、作用力类型以及结合位置等.这些研究为临床上尼美舒利的用药安全提供理论依据.1.1 仪器与试剂上海虹益仪器仪表有限公司pHS-3C型精密酸度计,上海一恒科技有限公司HWS12型超级恒温水浴,日本日立公司F-4600型荧光光谱仪,美国瓦里安技术中国有限公司Cary 50型紫外-可见光谱仪.牛血清白蛋白(98%,上海楷样生物技术有限公司),尼美舒利(99%,百灵威科技有限公司),其它试剂均为分析纯,实验用水为超纯水.1.2 试验方法依次加入 Nime溶液(0、3.243、4.865、6.486、8.108、9.729、11.351、12.972、14.594、16.215、17.837、19.458、21.079 5)×10-5mol·L-(1编号依次为1~13),1.0×10-5mol·L-1的BSA 1.5 mL,0.5 mol·L-1NaCl溶液2.0 mL和0.1 mol·L-1pH=7.4的缓冲溶液1.5 mL,于10.0 mL比色管中,定容摇匀,分别在291.6 K、301.6 K、311.6 K温度下,孵育40 min后扫描荧光光谱和同步荧光光谱.记录不含Nime的空白溶液的荧光强度为F0和含有Nime溶液的荧光强度为F.按照上述方法扫描Nime-BSA体系的吸收光谱.2.1 反应条件的影响2.1.1 缓冲溶液缓冲溶液的选择不同,对Nime与BSA相互作用的影响不大(图1).其中,Tris-HCl缓冲溶液效果对其相互作用最佳.2.1.2 pH值在Tris-HCl缓冲范围内,体系的F0/F随溶液pH值变化而改变(图2A).pH=7.4时,达到最佳.由图2B可知,Tris-HCl用量为1.5 ml时,ΔF达到最大值.最终确定pH=7.4,0.015 mol·L-1Tris-HCl作为Nime与BSA相互作用的缓冲溶液.2.1.3 BSA的浓度固定其它实验条件,改变BSA的浓度,测定BSA溶液分别为:1.5×10-6mol·L-1至1.5×10-5mol·L-1时对体系的荧光强度的影响(图3).7.5×10-6mol·L-1BSA 作为反应的浓度最佳.2.1.4 试剂加入顺序图4是不同的加入顺序对Nime与BSA相互作用的影响.加入顺序影响Nime与BSA结合,Nime→BSA→NaCl→Tris-HCl的加入顺序最佳.2.1.5 孵育时间考察225 min内孵育时间对体系的影响(图5).实验表明,Nime与BSA的相互作用在299.6 K温度下需要25 min才能完成并稳定.2.2 荧光光谱由于BSA分子中存在着酪氨酸(Tyr)、色氨酸(Trp)等能够发射荧光的芳香性氨基酸残基,所以它是一种典型的内源荧光物[3].其最大激发和发射波长(λex/λem)位于280 nm/340 nm处.从图6可知,随着Nime浓度的增加,λem(移到344 nm)处BSA的荧光强度逐渐降低,说明Nime对BSA的荧光有明显的猝灭作用.2.3 猝灭机理荧光猝灭机理通常可分为动态猝灭和静态猝灭[3-5].在静态猝灭中,生成了新物质,起主要作用的是新物质的稳定性,温度越高,稳定性越差,Ksv越小[3-5].在动态猝灭中Ksv会随着温度的升高而增大,因为分子扩散起主导.猝灭过程遵循Stern-Volmer方程[3-5]:F0/F=1+Kqt[0Nime]=1+Ksv[Nime],式中[Nime]为Nime浓度,Kq为速率常数;t0为荧光寿命,10-8s数量级左右[3-5].在291.6 K,301.6 K,311.6 K时作Stern-Volmer方程(表1),3个温度下的Kq值比最大动态猝灭速率常数2.0×1010L·mol-1·s-1)[3-5]大2个数量级,与动态猝灭机理相违背.由图7可知,随着温度的升高,直线斜率即Ksv降低,与静态猝灭机理吻合.Lineweaver-Burk双倒数方程[6-8]:(F0-F)-1=F0-1+(KLBF[0Nime])-1,其中:KLB常用来分析静态猝灭过程.用(F0-F)-1对[Nime]-1作不同温度下的L-B曲线(图8),计算KLB值列于表1中,KLB值都在104数量级以上,表明Nime与BSA形成的复合物稳定性较好.随着温度的升高,KLB略有下降,这与因静态猝灭方式而形成的复合物随温度升高而越不稳定的作用机理正好相符合. 另一种推断猝灭机理的重要方法是紫外吸收光谱法[4-6],由图9的紫外吸收光谱可知,Nime-BSA体系的吸收峰从280 nm蓝移到265 nm,吸收峰的强度也增加了,表明推断Nime与BSA静态猝灭机理是合理的.2.4 结合常数Kb和结合位点数n尼美舒利与BSA的结合常数Kb以及结合位点数n双对数方程[6-8]lg[(F0-F)/F]=lgKA+nlg[Nime][6-8].由lg[(F0-F)/F]对lg[Nime]作图,由直线截距可得结合常数Kb,斜率可求n(表1).n值接近于1,表明尼美舒利与BSA可形成1个结合位点.Kb为106数量级,表明Nime与BSA之间有很强的结合作用.当温度由291.6 K到301.6 K时,Kb变化不大;但当温度升高到311.6 K 时,Kb值减少1个数量级,表明尼美舒利与BSA的相互作用对温度变化比较敏感,高温不利于血清白蛋白在体内运转、贮存和分配尼美舒利.2.5 作用力类型根据热力学公式ΔG=ΔH-TΔS=-RTlnK和ln(K2/K1)=(1/T1-1/T2)ΔH/R [8-10]计算291.6 K,301.6 K,311.6 K温度下Nime与BSA结合反应的吉布斯自由能变ΔG,焓变ΔH及熵变ΔS(表2).由表2可得,ΔG<0,ΔH<0且ΔS>0,表明BSA与Nime的结合是自发进行的放热反应,主要作用力为静电作用力.2.6 结合位置的确定亚螺旋域IIA(含有酪氨酸和色氨酸)或IIIA(含有酪氨酸)是大多数药物在BSA 上结合位置[11-13].比较激发波长为280 nm、295 nm时Nime-BSA体系荧光程度的变化便可知道.由图10可知,λex=280 nm,295 nm的Nime-BSA光谱曲线平行,表明色氨酸和酪氨酸残基都参与其中;λex=280 nm激发曲线比λex=295 nm的降低程度更大,表明亚螺旋域ⅡA是主要结合位置[8-10]. 2.7 药物协同作用药物的协同作用常用Hill方程[11-13]进行分析:E=(F0-F)/F0,1/E对1[/Nime]作图,截距为1/Em,lgE(Em-E)=lgK+nHlg[Nime],式中,nH 为Hill系数,K为结合常数,E为饱和分数.由表2可知,各温度下的nH值略小于1,表明Nime与BSA结合时有微弱的药物负协同作用,即Nime分子结合到BSA位点上后,对后继药物分子与蛋白质的结合有微弱的阻碍作用.这种负协同作用可能是由于药物分子结构决定的,Nime浓度的加大,导致后续Nime对BSA 的亲和性减弱.随着温度的升高,nH值变化很小,表明Nime的药物协同作用对温度的改变不敏感.2.8 Nime对BSA构象的影响同步荧光光谱法是分析药物小分子对影响蛋白质的构象的常用方法,在Δλ=15nm(显示酪氨酸特征)和Δλ=60 nm(显示色氨酸特征)[6-8]条件下绘制Nime-BSA体系的同步荧光光谱(图11).由图可知,随Nime浓度的增大,λem 发生红移,酪氨酸和色氨酸的荧光强度逐渐降低,表明BSA腔内疏水环境的极性增强,疏水性减弱.酪氨酸残基的猝灭程度大于色氨酸残基,表明Nime与BSA相结合的位点偏向于酪氨酸.2.9 共存物的影响实验中对常见金属离子、有机物和糖类等共存物质对Nime与BSA相互作用的影响,结果列于3中.从表3中可以看出,当相对误差≤±5.0%,Nime浓度为3.243×10-5mol·L-1时,K+、NH4+、Na+、Zn2+、Ba2+、NO3-、Cl-、SO42-、糖类和有机物等几乎不影响Nime对BSA的作用强度,但是Fe3+和Cu2+等对体系的干扰较大,可通过加入EDTA和硫脲将其掩蔽.因此,BSA可作为荧光探针对Nime的含量进行测定,这一方法具有良好的选择性.应用紫外和荧光光谱法研究推断出尼美舒利与BSA的相互作用是静态猝灭过程,两者通过静电作用力相互作用,因有1个结合位点,药物能被蛋白质转运和储存;有微弱的药物负协同作用,结合位置在BSA的亚螺旋域ⅡA中,靠近酪氨酸残基,Nime与BSA相互作用对BSA构象产生影响.探究了常见金属离子、有机物和糖类等共存物质对Nime与BSA相互作用的影响,其中Fe3+和Cu2+对其相互作用影响较大.BSA可作为荧光探针对Nime进行含量测定.这些重要信息对尼美舒利的临床研究具有参考价值,并为后续非甾体抗炎药的研发提供了理论依据.[1]岑彦艳,覃容欣,李小丽,等.尼美舒利解热镇痛抗炎作用比较研究[J].中国现代应用药学,2014,31(8):933-934.[2]陈沫,卢永翎,杭太俊,等.尼美舒利分散片在健康人体内的药代动力学及生物等效性的LC-MS/MS研究尼美舒利分散片制备的处方工艺优选[J].药物分析杂志,2013,33(1):30-32.[3] BOGDAM S.Fluorescence study of sinapic acid interaction with bovine serum albumin and egg albumin[J].J flurescence,2003,13(4):349-356.[4] LAKOWICZ J R.Principles of fluorescence spectroscopy[M].3rd ed.New York:Springer Press,2006:285-292.[5]许金钩,王尊本.荧光分析法[M].3版.北京:科学出版社,2006:23-49. [6]刘里,成飞翔.光谱法研究头孢替唑钠与牛血清白蛋白相互作用[J].江西师范大学学报(自然科学版),2014,38(6):639-644.[7]刘里,成飞翔.光谱法研究洛索洛芬钠与牛血清白蛋白的相互作用及共存金属离子的影响[J].中国生化药物杂志,2014,35(1):21-24.[8]刘保生,杨超,王晶.硫酸头孢匹罗与牛血清白蛋白结合反应的发光机理[J].发光学报,2011,32(3):293-299.[9] CYRIL L,EARL J K,SPERRY W M.Biochemists handbook[M].London:Epon Led Press,1961:84-96.[10]ROSS D P,SUBRAMANTAN S.Thermodynamics of protein association reactions:forces contributing to stability[J].Biochemistry,1981,20(11):3 096-3 102.[11]SULKOWSKA A,MACIAZEK-JURCZYK M,BOJKO B,etpetitive binding of phenylbutazone and colchicine to serum albumin in multidrug therapy:a spectroscopic study[J].Journal of molecular structure,2008,881(1):97-106.[12]MACIAZEK-JURCZYK M,SULKOWSKA A,BOJKO B,etal.Fluorescence analysis of competition of phenylbutazone and methotrexate in binding to serum albumin in combination treatment in rheumatology[J].Journal of molecular structure,2009,919(1):334-338.[13]XU H,GAO S L,LÜ J B,et al.Spectroscopic investigations on the mechanism of interaction of crystal violet with bovine serum albumin [J].Journal of molecular structure,2009,919(1):334-338.。
- 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)。
ReviewThe tumor suppressor kinase LKB 1:lessons from mouse modelsSaara Ollila and Tomi P.Ma¨kela ¨*Institute of Biotechnology,University of Helsinki,Viikki Biocenter,Viikinkaari 9B,FIN-00014,Helsinki,Finland*Correspondence to:Tomi P.Ma¨kela ¨,E-mail:tomi.makela@helsinki.fiMutations in the tumor suppressor gene LKB 1are important in hereditary Peutz–Jeghers syndrome,as well as in sporadic cancersincluding lung and cervical cancer.LKB 1is a kinase-activating kinase,and a number of LKB 1-dependent phosphorylation cascades regulate fundamental cellular and organismal processes in at least metabolism,polarity,cytoskeleton organization,and prolifer-ation.Conditional targeting approaches are beginning to demonstrate the relevance and specificity of these signaling pathways in development and homeostasis of multiple organs.More than one of the pathways also appear to contribute to tumor growth fol-lowing Lkb 1deficiencies based on a number of mouse tumor models.Lkb 1-dependent activation of AMPK and subsequent inacti-vation of mammalian target of rapamycin signaling are implicated in several of the models,and other less well characterized pathways are also involved.Conditional targeting studies of Lkb 1also point an important role of LKB 1in epithelial–mesenchymal interactions,significantly expanding knowledge on the relevance of LKB 1in human disease.Keywords:LKB 1,tumor suppressor,mouse model,AMPKIntroductionCancer arises as a result of accumulating genetic and epige-netic changes,which compromise the cell’s ability to control its identity and proliferation.Many identified tumor suppressors play a well-established role in regulation of cell growth and div-ision (e.g.Rb,APC,p 21,PTEN)and genome maintenance (e.g.p 53,BRCA 1-2,ATM,ATR,MLH 1,MSH 2),providing a logical link between the loss of gene product and promotion of carcinogen-esis.An interesting exception is the serine /threonine kinase gene LKB 1(also known as STK 11),which has in recent years taken a prominent position among tumor suppressors.Heterozygous germline mutations in LKB 1predispose to Peutz–Jeghers syndrome (PJS)where patients develop benign polyps in the gastrointestinal (GI)tract and are in high risk of developing malignant tumors in GI tract,breast,and gyneco-logical organs (Giardiello et al .,2000).Importantly,somatic LKB 1mutations are found at least in lung (Ji et al.,2007)and cervical cancer (Wingo et al .,2009).Through phosphoryl-ation of several cellular kinases LKB 1has been implicated in control of cellular and organismal metabolism,cell polarity,and a variety of other functions ranging from proliferation and migration to senescence,apoptosis,DNA damage responseand differentiation (Vaahtomeri and Ma¨kela ¨,2011).Despite these many functions attributed to LKB 1,their specific contri-butions to the maintenance of tissue homeostasis in vivo and tumor growth are only sketchily appearing with thedevelopment of LKB 1mouse models.This work is important to enable rational treatment strategies to LKB 1-deficient tumors.The LKB 1kinase acts in a trimer with a pseudokinase STRAD and the scaffold protein MO 25to phosphorylate at least 14kinases with conserved activation sites (Katajisto et al.,2007).A well-known substrate of LKB 1is AMPK,which is the master reg-ulator of cellular and organismal metabolism,providing a putative downstream pathway to LKB 1-mediated tumor suppression (Shackelford and Shaw,2009).In mouse studies,AMPK requires LKB 1for activation in vivo in most tissues (Sakamoto et al .,2005;Shaw et al .,2005;Contreras et al .,2008;Hezel et al .,2008).AMPK senses the energy state of cells through monitoring AMP levels as a sensitive readout for ATP.AMPK is activated following exercise,hypoxia,or glucose deprivation,after which it phosphor-ylates multiple targets to increase energy uptake and catabolic processes such as glucose uptake and fatty acid oxidation,and suppress anabolic processes such as lipogenesis and cholesterol synthesis (Hardie et al.,2003).AMPK is the potential candidate to mediate LKB 1’s effects in cell growth via the mammalian target of rapamycin (mTOR)signal-ing (Corradetti et al .,2004;Shaw et al .,2004),which is the pathway monitoring the availability of nutrients in regulation of cell size and protein synthesis as well as proliferation (Zoncu et al.,2011).Increased mTOR signaling is common in cancer (Guertin and Sabatini,2007)and also present in at least some Lkb 1-deficient tumors (Shaw et al.,2004;Ji et al.,2007;Contreras et al.,2008;Hezel et al.,2008;Shackelford et al.,2009).An additional link between LKB 1and mTOR pathway#The Author (2011).Published by Oxford University Press on behalf of Journal ofMolecular Cell Biology ,IBCB,SIBS,CAS.All rights reserved.doi:10.1093/jmcb /mjr 016Journal of Molecular Cell Biology (2011),Vol no.0,1–11|1Journal of Molecular Cell Biology Advance Access published September 15, 2011 at Shihezi University on September 27, 2011 Downloaded frommay lie in regulation of PI 3K-Akt pathway inhibitor PTEN by LKB 1(Mehenni et al .,2005).Loss of cell polarity is commonly noted in cancer,and LKB 1is an important factor for cell polarity in different organisms.In C.elegans ,the orthologs for LKB 1(par-4)and MARK s (par-1)were identified in a panel of six partitioning (par )mutants which disrupted the polarity of the early embryos (Kemphues et al.,1988).In Drosophila ,Lkb 1is required for proper oocyte polarity (Martin and St Johnston,2003).In mammalian cells,in both 2D and 3D cell culture models and in vivo ,LKB 1is known to regulate polarity (Baas et al .,2004;Partanen et al .,2007;Hezel et al .,2008).Polarity defects are,however,not seen in all Lkb 1-deficient tumors (Contreras et al.,2008,2010).Several of the LKB 1substrates have been reported to mediate the regulation of cell polarity through regulating the cytoskeleton and formation of cell–cell junctions.MARK kinases are implicated in the stability of microtubules by phosphorylating and thereby dissociation microtubule-associated proteins (MAPs),for example the tau protein,from microtubules (Drewes et al .,1997;Stoothoff and Johnson,2005).Neuronal polarity and axon formation are regu-lated by LKB 1at least partially via BRSK kinases (Kishi et al.,2005;Barnes et al.,2007).To what extent LKB 1acts as a polarity protein in mammalian non-neuronal cells still remains to be deter-mined,although at least in both exo-and endocrine pancreas Lkb 1loss leads to polarity defects in vivo (Hezel et al .,2008;Granot et al .,2009).As formation of stress fibers is essential incell contractility,recent studies associate LKB 1with cell motility via NUAK 1and NUAK 2,which have been implicated in regulation of myosin light chain phosphorylation (Vallenius et al.,2010;Zagorska et al.,2010).For detailed information of the molecular signaling pathways of Lkb 1,the reader is recommended recent reviews more focused on that topic (Katajisto et al.,2007;Hezel and Bardeesy,2008;Vaahtomeri and Ma¨kela ¨,2011).Role of Lkb 1in development and tissue homeostasis in miceAlthough LKB 1is a tumor suppressor,inactivation of Lkb 1through homologous recombination or ‘knock-out’(KO)does not always lead to tumors.This is due partly to essential functions of Lkb 1in development and partly demonstrates the tissue-specificity of Lkb 1functions,where in some cell types biallelic deletion is detrimental to cells or affects specific functions in metabolism as summarized in Figure 1and discussed below.Role of Lkb 1in embryogenesisGeneration of full KO revealed that Lkb 1is essential for embry-ogenesis;no viable Lkb 12/2embryos were seen after E 11.Analysis of the E 8.5–E 9.5embryos revealed severe developmen-tal defects including impaired neural tube closure and somitogen-esis,mesenchymal tissue cell death,and defective vasculature.The extra-embryonic tissues (yolk sac and placenta)were also deformed.VEGF signaling was highly upregulated in theKOFigure 1Non-tumorigenic phenotypes following Lkb 1targeting in mice.Phenotypes (green)are grouped according to tissue type,cell typeaffected /analyzed (blue),and alleles used for targeting.When appropriate,activator of deletor is indicated in purple.Noted signaling change(s)indicated in red.Alleles as displayed in original publications except for Lkb 1flox 2h /flox 2h hypomorphic Lkb 1(Sakamoto et al,2005).(1)Londesborough et al.,2008;(2)Ohashi et al.,2010;(3)Cao et al.,2010;Tamas et al.,2010;(4)Shorning et al.,2009;(5)Woods et al.,2011;(6)Shaw et al.,2005;(7)Sun et al.,2010a ;(8)Sun et al.,2011;(9)Granot et al.,2009;Fu et al.,2009;(10)Koh et al.,2006;(11)Sakamoto et al.,2005;(12)Sakamoto et al.,2006;Jessen,et al.,2010;(13)Ikeda et al.2009;(14)Gurumurthy et al.,2010;Nakada et al.,2010;(15)Gan et al.,2010;(16)Barnes et al.,2007;(17)Ylikorkala et al.,2001.tam,tamoxifen;b -NF,b -naphtoflavone;pIpC,polyinosinic–polycytidylic acid;iv,intravenous.2|Journal of Molecular Cell Biology Ollila and Ma¨kela ¨ at Shihezi University on September 27, 2011 Downloaded fromembryos,possibly relating to the vascular phenotype (Ylikorkala et al .,2001).Embryonic lethality,no embryonic turning,and small somites were also shown in another report of Lkb 1full KO (Jishage et al .,2002).The severe developmental defect was not a result of the abnormal extra-embryonic tissues,since epiblast-specific conditional inactivation of Lkb 1using Mox 1-Cre resulted in very similar embryonic lethal phenotype to full KO (Londesborough et al .,2008).The important role of Lkb 1in development and maintenance of neurons,mesenchymal cells,and vascularization has been recapitulated in tissue-specific Lkb 1KOs.Role of Lkb 1in angiogenesisLondesborough et al .(2008)further dissected the role of Lkb 1in endothelia by deleting Lkb 1in vascular endothelial cells using Tie 1-Cre (Figure 1).The mice died at E 12.5and displayed dilated embryonic vessels and pericardial swelling.The vessels were irre-gular and distorted and suffered from inadequate supportive vas-cular smooth muscle cell layer.Since Tgf b signaling was reduced both in Lkb 1-deficient mouse yolk sacs and human umbilical vein endothelial cells (HUVECs)where LKB 1expression was silenced by siRNA,the vascular phenotype was suggested to result from a loss of supporting vascular smooth muscle cells as a conse-quence of attenuated Tgf b signaling from endothelial cells (Londesborough et al .,2008).Another report also described mice lacking Lkb 1in endothelial cells,deleted using Tie 2-Cre driver (Ohashi et al.,2010)(Figure 1).This study repeated the finding that endothelial Lkb 1is essential for proper embryonic development and no homozygous mutants were born.Analysis of heterozygous Tie 2-Cre;Lkb 1flox /+mice revealed that the mice,including vasculature,seemed phenotypically normal,but displayed reduced revascularization after hind-limb ischemia.Studies in mouse tissues,primary mouse endothelial cells,and HUVECs implemented that the phenotype was mediated via AMPK (Ohashi et al.,2010).In this study,the authors did not address the contribution of Tgf b signaling to the observed phenotype.In the Tie 2-Cre model,Lkb 1–AMPK axis seemed to mediate proangiogenetic signaling as Lkb 1heterozygosity resulted in reduction of revascularization in adult mice (Ohashi et al.,2010).In developing embryo,increased VEGF signaling upon Lkb 1loss would suggest the opposite,antiangiogenic role for Lkb 1(Ylikorkala et al .,2001).Also in the context of PJS polyps where a loss of Lkb 1leads to increased HIF 1a and vasculature,Lkb 1seems to be rather antiangiogenic (Shackelford et al .,2009).However,reduced capillary density was reported in mice where Lkb 1was conditionally deleted from the heart (Ikeda et al .,2009).In 3D culture system where endothelial cells are embedded in Matrigel,both over-expression (Xie et al .,2009)and inhibition (Ohashi et al.,2010)of Lkb 1have been reported to inhibit network formation,suggesting proper expression of LKB 1is essential for angiogenesis.Thus,the precise role of Lkb 1in angiogenesis seems to be dependent on the tissue type and /or the developmental phase,varying from inhibition to promotion.Role of Lkb 1in liverThe finding that Lkb 1functions upstream of AMPK (Shaw et al .,2004)led to interest to study its effects in liver,where many path-ways of carbohydrate and lipid metabolism,including glycogen-esis,glycogenolysis,gluconeogenesis,lipogenesis,and cholesterol synthesis take place.Tail-vain injection of Adeno-Cre to mice carrying conditional Lkb 1allele led to hepatocyte-specific Lkb 1deletion since Adeno-Cre has high tropism for hepatocytes (Shaw et al .,2005)(Figure 1).Lkb 1loss resulted in nearly complete abolishment of AMPK activation in liver,and the glucose metabolism of the mice was impaired demonstrated by elevated blood glucose.CRTC 2phosphorylation was reduced in the livers of the mice,leading to elevated CREB-mediated transcription,including expression of PGC 1a and other gluconeogenetic genes.Also lipogenetic genes were over-expressed.Metformin,the diabetes drug which reduces blood glucose levels via AMPK pathway (Zhou et al .,2001),did not lower blood glucose in the liver-specific Lkb 1KO mice,demonstrating that AMPK activity induced by Lkb 1in liver is required for the effects of metformin in vivo .In another report of liver-specific Lkb 1knockout using Alb-Cre driver,Woods et al.(2011)reported defective bile ducts in liver,leading to accumulation of bile in liver and serum (Figure 1).Bile salt export pump was not located in canalicular membrane of the bile canaliculi,indicating possible defects in cell polarity.The mice also suffered from cholestasis (Woods et al.,2011).These reports of liver-targeted deletions of Lkb 1demonstrate the critical requirement of Lkb 1in glucose,lipid,bile,and cholesterol metab-olism.Furthermore,they show that in liver,Lkb 1is the main acti-vator of AMPK,and its activity is required for the AMPK-mediated suppression of lipogenesis and gluconeogenesis to take place.Role of Lkb 1in muscleMuscles are highly energy-consuming tissues whose glucose homeostasis needs to be regulated both in response to insulin after blood sugar increase,and to exercise-mediated deficiency of glucose storage.Sakamoto et al.(2005)provided the first genetic evidence that Lkb 1is required for AMPK activation in vivo in skeletal muscle.They generated conditional Lkb 1mice in which cDNA of Lkb 1exons 5–7fused with neomycin resistance cassette,surrounded by loxP sites,was inserted between exons 4and 8in the genomic Lkb 1locus.The resulting mice were hypomorphic and expressed only 10%–20%of normal levels of Lkb 1in the absence of Cre -mediated ing MCK-Cre driver to create muscle-specific Lkb 1KO,they found that AMPK a 2(one of the two alternative catalytic subunits of AMPK)activation either by the AMP analog AICAR,muscle con-traction or phenformin,a similar blood glucose lowering drug to metformin,was lost and AMPK a 1activation greatly reduced.Upon contraction,glucose transport to muscle cells was abol-ished (Sakamoto et al.,2005).In another study using the same muscle-specific MCK-Cre with another (non-hypomorphic)con-ditional Lkb 1line,effects of Lkb 1loss in muscle to levels of blood glucose were investigated (Koh et al.,2006)(Figure 1).Interestingly,glucose metabolism seemed to be enhanced in these mice,demonstrated by reduced fasting blood glucose and blood insulin concentrations,improved glucose tolerance,and increased muscle glucose uptake.This phenotype,indicating that Lkb 1in muscle functions as a negative regulator of glucose metabolism,was suggested to be resulting from improved muscle glucose uptake,mediated by increased phosphorylation of Akt and reduced the gene expression of the Akt inhibitor TRB 3.Lkb 1loss abolished the activity of AMPK a 2,but notLessons from LKB 1mouse modelsJournal of Molecular Cell Biology |3at Shihezi University on September 27, 2011 Downloaded fromAMPK a 1in muscle cells.Also MARK 4,but not MARK 2/3activitywas significantly reduced.Based on this study,the metabolic effects mediated by Lkb 1in muscle seem to oppose those of the liver,at least in terms of blood glucose levels (Koh et al.,2006).Recently,the Lkb 1substrate NUAK 2was proposed to be a mediator of contraction-stimulated glucose transport by skel-etal muscle (Koh et al.,2010).Also cardiac muscle lacking Lkb 1has been investigated.Sakamoto et al.(2006)studied the effect of Lkb 1deficiency in heart using the MCK-Cre driver,which deletes Lkb 1in both skel-etal and cardiac myocytes and found that Lkb 1inactivation did not lead to overt cardiac dysfunction,although the weight of the heart was reduced and the atria enlarged;however,the study revealed that cardiac Lkb 1is required for activation of AMPK a 2both in basal conditions and in response to ischemia (Figure 1).Also Jessen et al.(2010)used the MCK-Cre driver but the Lkb 1allele was not hypomorphic as in the Sakamoto et al.(2006)study.They showed that ablation of Lkb 1in heart leads to impaired cardiac function both in basic conditions and post-ischemia and suggested that failure to downregulate mTOR sig-naling by AMPK a 2activation underlined the phenotypes.Ikeda et al.(2009)used a -MHC-Cre to delete Lkb 1specifically from the heart,and a more severe phenotype was observed:the mice displayed hypertrophy and impaired function of the heart,reduction of cardiac capillary density,and increased fibrosis and collagen content and died by 6months of age.The differ-ences between these phenotypes may reflect differences in the timing of Cre activity,specificity of the Cre recombination,and /or the conditional Lkb 1allele used.However,it seems clear that Lkb 1is needed for the normal function of heart both in basal and ischemic conditions.Role of Lkb 1in pancreasPancreatic b -cells secrete insulin and are thus important mediators of whole-body glucose metabolism.As Lkb 1–AMPK axis is important in regulation of liver metabolism and muscle glucose homeostasis,it is of interest to study whether Lkb 1has an effect on the insulin release.Granot et al.(2009)used the Pdx 1-CreER driver to delete pancreatic Lkb 1in 6-week-old mice by tamoxifen injection (Figure 1).In response to glucose injection,the mutant mice secreted more insulin than control mice,which carried the conditional Lkb 1allele but were not subjected to tamoxifen injection.Deletion of Lkb 1led to increased size of b -cells together with disrupted polarity.Increased mTOR signal-ing seemed to mediate the cell size increase,while the polarity defect took place at least partially through MARK 2.Increased insulin secretion was partially dependent on AMPK (Granot et al .,2009).Fu et al .(2009)used the same Pdx 1-CreER system to delete Lkb 1in adult b -cells and also found that the mice showed improved glucose tolerance,b -cells mass had increased,and mTOR pathway was activated (Figure 1).These results place Lkb 1as an important regulator of pancreatic b -cell size,polarity,and function,further highlighting its essence in regulation of organismal metabolism.Sun et al.(2010a)investigated pancreatic b -cells with the Rip 2-Cre driver,which activates Cre -mediated recombination in pancreatic b -cells and some hypothalamic neurons,and found that the mice displayed diminished food intake and weight gain,enhanced insulin secretion,and improved glucose tolerance (Figure 1).Also here,mTOR pathway was activated.However,the study by the same group where both AMPK a subunits were deleted in b -cells using the same Rip 2-Cre showed decreased insulin secretion (Sun et al.,2010b ).This suggests that Lkb 1loss regulates mTOR signaling in b -cells partially independent of AMPK,or that the hypothalamic Lkb 1and AMPK have different functions,impacting the feeding behavior and hormonal balance.Role of Lkb 1in immune systemThree recent studies elegantly demonstrated that Lkb 1regu-lates the quiescence and maintenance of hematopoietic stem cells (HSCs)using conditional Lkb 1alleles with Mx 1-Cre followed by injections of polyinosinic–polycytidylic acid (pIpC),or Rosa 26-CreERt 2followed by tamoxifen injections (Gan et al.,2010;Gurumurthy et al.,2010;Nakada et al.,2010)(Figure 1).Both approaches resulted in a similar phenotype:increased pro-liferation followed soon by decline in HSC number,resulting in loss of all immune cell types (pancytopenia)and death.Transplantation experiments demonstrated that Lkb 1-deficient HSCs were not able to reconstitute the bone marrow of irradiated wild-type (wt)mice,nor were they able to compete with wt donor cells,demonstrating that the effect was cell-autonomous;mito-chondrial defects and decreased ATP levels,as well as altered long-chain fatty acid and nucleotide metabolite levels suggested metabolic defects to underlie the phenotypes noted (Gan et al.,2010;Gurumurthy et al.,2010;Nakada et al.,2010).Interestingly,only minor similarities in mitochondrial phenotypes were found when mice defective for both AMPK a subunits were compared with Lkb 1KO mice (Nakada et al.,2010),implicating other Lkb 1substrates in these phenotypes.Consistent with this,rapamycin or AMPK activators AICAR and A 769662did not rescue the phenotype in any of the studies.Immune cell apopto-sis was increased,and Lkb 1-deficient HSCs also demonstrated increased autophagy in bone marrow,and inhibiting this further decreased immune cell survival (Gan et al.,2010;Gurumurthy et al.,2010;Nakada et al.,2010).This would suggest that Lkb 1in this context is suppressing autophagy,whereas previously it has been reported to activate it following elevation of reactive oxygen species (Alexander et al.,2010).Yet another phenotype potentially decreasing HSC viability was the noted increase in supernumerary centrosomes,aberrant mitotic spindles,and aneuploidy (Nakada et al.,2010),which could be due to compro-mised BRSK 2activity (Alvarado-Kristensson et al.,2009).Recently,two groups generated mice where Lkb 1expression is specifically abolished in the T cell progenitors using the proximal p 56lck-Cre promoter.The studies demonstrate severe deficiency in survival and proliferation of T cell progenitors and mature T cells in the absence of Lkb 1(Cao et al.,2010;Tamas et al.,2010)(Figure 1).Also the survival of isolated peripheral T cells in vitro was dependent on Lkb 1(Tamas et al.,2010).Transfection of thymo-cytes with constitutively active AMPK a 2partially rescued the thy-mocytes from cell death,indicating that thymocyte survival is mediated at least via AMPK pathway (Cao et al.,2010).Thus,the common hematopoietic cell precursors and T cell precursors seem to have different requirement for AMPK signaling,although cell sur-vival is defective in both cell types in the absence of Lkb 1.The studies in hematopoietic cells have revealed an interesting aspect4|Journal of Molecular Cell Biology Ollila and Ma¨kela ¨ at Shihezi University on September 27, 2011 Downloaded fromof Lkb 1biology:although being a tumor suppressor in some tissues,in others Lkb 1is required for survival.Role of Lkb 1in nervous systemLkb 1KO embryos exhibit severe deficiencies in development of neuronal tissues (Ylikorkala et al .,2001).Since LKB 1orthologs in nematodes and fruit flies have been identified through their indis-pensable role in establishing polarity (Kemphues et al .,1988;Martin and St Johnston,2003)and LKB 1regulates polarity also in some mammalian cells (Baas et al .,2004;Partanen et al .,2007),it was of interest to generate models which would reveal the in vivo relevance of Lkb 1in establishing the axon-dendrite polarity in neuronal cells.Barnes et al.(2007)deleted Lkb 1in cer-ebral cortex of developing mice using Emx-Cre driver and showed that Lkb 1and its substrates BRSK 1and BRSK 2are required for axon specification in the studied neurons.This finding confirmed the previously described role of BRSK kinases in neuronal polar-ization (Kishi et al .,2005),and placed Lkb 1as the upstream kinase required for the polarization to take place.Lkb 1-activated BRSKs were shown to modify the cytoskeleton by phosphorylating MAPs (Barnes et al.,2007).Studies in rat hip-pocampal neurons in vitro and developing rat cortical neurons in vivo agreed with the finding that Lkb 1is essential in establishing neuronal polarity;there,lack of either Lkb 1or STRAD prevented axon differentiation (Shelly et al.,2007).Interestingly,over-expression of Lkb 1and STRAD resulted in formation of multiple axons.PKA-mediated phosphorylation of Lkb 1Ser 431was shown to be required for the axon specification (Barnes et al.,2007;Shelly et al.,2007).Thus,Lkb 1activity is modulated by upstream factors in a tissue-and context-specific manner.Not only axon specification but also maintenance seems to be regulated via Lkb 1in some systems.Sun et al.(2011)reported,using the pancreatic and hypothalamic Rip 2-Cre ,that the mice developed hind-limb paralysis due to axon degeneration in thor-acic spinal cord neurons at about 7–8weeks of age (Figure 1).The Rip 2-Cre was found to be active also in spinal cord,especially in the thoracic segments.Deleting both AMPK a subunits did not result in axon degeneration or paralysis,and the authors specu-lated that in the absence of Lkb 1,the neuronal polarization and axon degeneration defects might be mediated by BRSK kinase pathways (Sun et al.,2011).PJS and its mouse modelsLKB 1was linked to human disease when its mutations were found to be causative for PJS (Hemminki et al .,1998;Jenne et al .,1998).A major manifestation in PJS is the appearance of large occluding hamartomatous polyps in the GI tract (Giardiello and Trimbath,2006).Mice carrying one inactivated allele of Lkb 1(Lkb 1+/2)recapitulate PJS by developing hamartomatous GI polyps which are indistinguishable from PJS patient polyps (Bardeesy et al .,2002;Jishage et al .,2002;Miyoshi et al .,2002;Rossi et al .,2002)(Figure 2),although in mice polyps appear more in the stomach and less in the small intestine.Polyps appear at 4–6months (Udd et al.,2010),and lead to lethality at an average age of 11months due primarily to obstructions.Biallelic loss of wt Lkb 1is not a prerequisite for polyp formation,indicating that Lkb 1is a haploinsufficient tumor suppressor at least in the context of PJS polyps (Jishage et al .,2002;Miyoshiet al .,2002;Rossi et al .,2002).Strong up-regulation of COX 2has been identified in the mouse and also PJS patient polyps (Rossi et al .,2002),and COX 2inhibitors have been shown to be efficient suppressors of PJS polyps (Udd et al .,2004).PJS is associated with elevated risk of cancer,especially in the GI tract,and also in breast,pancreas and gynecological cancers (Giardiello and Trimbath,2006;Hearle et al .,2006;Mehenni et al .,2006).Lkb 1+/2mice in turn have been reported to have increased frequency of cancer in liver (Nakau et al .,2002),bones (Robinson et al .,2008),and endometrium (Contreras et al .,2008)(Figure 2).Polyposis in Lkb 1+/2mice is accelerated in a p 53-deficient background (our unpublished data)(Wei et al .,2005;Takeda et al .,2006)(Figure 2),and p 53mutations are detected in the GI cancers of PJS patients (Miyaki et al .,2000).Despite these observations,progression of the benign hamarto-matous polyps to dysplasia or carcinoma is not clearly estab-lished possibly due to the rapid growth of the hamartomatous polyps leading to GI occlusions.As haploinsufficiency of Lkb 1is sufficient for polyp initiation (Jishage et al .,2002;Miyoshi et al .,2002;Rossi et al .,2002)though biallelic loss has been noted (Bardeesy et al .,2002),loss of the remaining allele of Lkb 1may represent a progression step,although it has also been suggested that the loss of Lkb 1is associated with the resist-ance to progression in this context (Bardeesy et al.,2002).Mesenchymal Lkb 1loss leads to PJS-type polyposis in mice PJS polyps are classified as hamartomatous polyps thought to contain all the cell types of the surrounding tissue.However,it was recently noted that epithelial differentiation is disrupted in gastric and small intestinal polyps in Lkb 1+/2mice (Udd et al.,2010),but the model did not enable distinguishing whether this was a cell autonomous function of Lkb 1in epithelial cells.Biallelic disruption of Lkb 1in GI epithelia lead to imbalanced differentiation and positioning of epithelial cells (Shorning et al .,2009)(Figure 1),but was not reported to be associated with tumorigenesis.Polyps in both PJS patients and Lkb 1+/2mice harbor a large component of smooth muscle tissue.Remarkably,in a mouse model,where Lkb 1deficiency was restricted to the smooth muscle lineage by using a tamoxifen-inducible SM 22-CreERt 2line,PJS type polyps appeared in stomachs of the mice with the hetero-and homozygous Lkb 1mutants (Katajisto et al .,2008)(Figure 2).The polyps appeared later than those in the Lkb 1+/2mice,suggesting either that tamoxifen-induced Lkb 1loss at 6weeks of age delayed the poly-posis,or that mesenchymal loss of Lkb 1signaling is sufficient to drive hyperproliferation of epithelial tissue,but that coexisting epithelial mutations accelerate the process.This interesting aspect of Lkb 1signaling in tissue interactions is discussed later.Other Lkb 1tumor mouse modelsInactivating LKB 1mutations are associated with the develop-ment of cancer in several tissues.Various strategies of targeted inactivation of Lkb 1in mice,sometimes in combination of other tumorigenic mutations,have led to the development of various types and grades of tumors in multiple tissues,sometimes mod-eling human cancers in very useful ways as discussed below and summarized in Figure 2.Lessons from LKB 1mouse modelsJournal of Molecular Cell Biology |5at Shihezi University on September 27, 2011 Downloaded from。