Phosphorylation of LKB1 at Serine 428 by Protein Kinase C-ζ Is

合集下载

LKB1蛋白在胃癌组织中的表达及其临床意义

LKB1蛋白在胃癌组织中的表达及其临床意义
p to o ia co u h a g ah lg c l a trs c sa e, g n e , t mo ie d p h o v so , df r n it n, l mp o e f edr u rs , e t f n a i n z i iee t i f ao y hnd mea tssa d d sa tmea tssa d S ot r d . T ef r e t d f h e ai n hp b t e e tsa i n i n tsa i n Of r we ema e t h h u t rs y o e rl t s i ewe n t h u t o h e p e so f KB1 p oe n a d t es r ia i fp t n swa loc rid o t Re u t L x r s in o L r ti n h u vv l meo a i t sas a r u . t e e sl s KB1p oe n r ti wa x r s e o ma a t ce i eil is e, a t cc n e n eme a tt a c r n lmp od t s e s p e s di n r l s p t l s u g sr a c ra d t t s i c n e e n g r i h at i h ac i y h i su . i T e p st e r t f t e h v in f a td f r n e T e e wa o sg i c n i e e c ew e h h o i v ae o h m a e a sg i c n i e e c . h r s n i nf a tdf r n e b t e n t e i i f i f e p e so f KB1p o en a d p t n e d r g ,tmo ie o a in,d g e f i e e t t n a d t e x r s in o L r t i n ai t n e ,a e u r z ,lc t e g s o e r e o df r n i i n f ao h d p ho v so . Ho v r t e ew ssg i c n i e e c mp o emea t ss d sa t t sa i e t f n a i n i we e , h r a i nf a t f r n ei l i df n y h n d tsa i , itn a t ss me

腺苷酸活化蛋白激酶与LKB1在肥胖大鼠脂肪组织中的表达及其对糖脂代谢的影响

腺苷酸活化蛋白激酶与LKB1在肥胖大鼠脂肪组织中的表达及其对糖脂代谢的影响
程 媛 王佑 民 。
[ 摘要 ] 目的: 探讨腺苷酸活化蛋 白激酶( MPat a dpo i kns , MP 与 L B 在肥胖 大鼠脂肪组织 的表达及其对糖 脂 A —c vt rt n i eA K) K 1 i e e a 代谢 的影响 。方法: 3 将 0只 6周龄雄性 sD大 鼠随机分 为普通饮食组 ( C组 ) ・ N 和高脂 饮食组 ( HF组 ) 各 1 , 5只 , 分别 予普 通 饲料 喂养和高脂饲料喂养 。喂养 1 6周后 , F组大 鼠体重高 于 N H C组 2 %者 为成功建 立模型 。所有 大 鼠过夜 禁食后 , 0 麻醉状 态 下测 量体 重( W) 取静脉 血测 定血清游离脂肪酸 ( F 、 B , F A) 甘油三 酯 ( G) 总胆 固醇 ( C 、 T 、 T H) 空腹血 糖 ( P 、 F G) 空腹 胰 岛素
( IS 。处死大 鼠后 , FN ) 采用 Wetr l 法测定各组大 鼠骨骼 肌组织 中 A P a、 酸化 A P PA K t和磷 酸化 L B 蛋 s nbo e t MK 磷 M K( — MP c) K 1
白( -K 1 的表达 。计算 A P PL B ) M K活 性 。结 果: N 与 C组 比较 , F组 大 鼠 B F A、 G、 P FN H W、 F T F G、 I S均 升高 ( P<0 0 . 5~P<
n r lfd r t a d h g a. d r t 。 n x l r h mp c fAMP n KB n t e mea oim f gu o e a d lpd M eho s o ma - as n ih ftf as a d e p o e t e i a to e e K a d L 1 o h t b l s o lc s n ii. t d :

Overview of Protein Phosphorylation 蛋白磷酸化

Overview of Protein Phosphorylation 蛋白磷酸化

UNIT 18.1 Overview of Protein PhosphorylationHISTORYPhosphorylation is the most common and important mechanism of acute and reversible regulation of protein function. Studies of mammalian cells metabolically labeled with [32P]orthophosphate suggest that as many as one-third of all cellular proteins are covalently modified by protein phosphorylation. Protein phosphorylation and dephosphorylation func-tion together in signal transduction pathways to induce rapid changes in response to hor-mones, growth factors, and neurotransmitters. Most polypeptide growth factors (platelet-de-rived growth factor and epidermal growth fac-tor are among the best studied; Heldin, 1995) and cytokines (e.g., interleukin 2, colony stimu-lating factor 1, and γ-interferon; Ihle et al., 1994) stimulate phosphorylation upon binding to their receptors. Induced phosphorylation in turn activates cytoplasmic protein kinases, such as raf, MEK, and MAP kinases (Marshall, 1995). Additionally, in all nucleated organisms, cell cycle progression is regulated at both the G1/S and the G2/M transitions by cyclin-de-pendent protein kinases (Doree and Galas, 1994).Differentiation and development are also controlled by phosphorylation. Development of the R7 cell in the Drosophila retina (Simon, 1994) and of the vulva in Caenorhabditis ele-gans(Eisenmann and Kim, 1994) are both dependent on the function of receptor and cy-toplasmic protein kinases. Finally, metabo-lism—in particular, the interconversion of glu-cose and glycogen and the transport of glu-cose—is regulated by phosphorylation (Cohen, 1985). Biologists of all stripes therefore find, often unexpectedly and occasionally reluc-tantly, that they must study protein phosphory-lation in order to understand the regulation and function of their favorite gene and its product.In the early 1940s, soon after their discovery that glycogen phosphorylase existed in two distinct forms in skeletal muscle, Carl and Gerty Cori identified a cellular activity that converted active phosphorylase a into a less active phosphorylase b. Initial studies of what they called the prosthetic-group removing (PR) enzyme suggested that the inactivation of phos-phorylase a was mediated via proteolysis (Cori and Cori, 1945). More than ten years later, this converting enzyme was shown to be a protein phosphatase. Thus, a protein phosphatase was studied nearly 20 years prior to the identifica-tion of the first protein kinase. The finding that phosphorylase phosphatases were more widelyexpressed in tissues than phosphorylase a ledto the current realization that these enzymeshave a broad substrate specificity and regulatemany physiological processes. The use of sub-strates other than phosphorylase a also identi-fied several other protein serine/threonine phosphatases.LABELING STUDIESProtein phosphorylation is usually studiedby biosynthetic labeling with 32P-labeled inor-ganic phosphate (32P i; UNIT 18.2). This is intrin-sically quite simple—the label is just added togrowth medium. It is this step of an experiment,however, that makes many investigators themost nervous, given the perceived danger ofradioactive exposure and the real danger of contamination of laboratory equipment with radioactivity. Neither problem is insurmount-able. With proper shielding and technique, ex-posure of the investigator can be limited to thehands and contamination of the laboratory canbe avoided. A general protocol for biosyntheticlabeling with 32P i that maximizes incorporationand minimizes radioactive exposure of workersin the lab and contamination of lab equipmentis described in UNIT 18.2. (For a general discus-sion of radiation safety consult Safe Use of Radioisotopes, APPENDIX 1F.)Many protein kinases can be assayed by thetransfer of radiolabel from [32P]ATP to surro-gate protein and peptide substrates. Syntheticpeptide substrates have been particularly usefulin establishing consensus motifs that define thesubstrate for protein kinases. By comparison,protein phosphatases show very low activityagainst synthetic phosphopeptides, greatly pre-ferring the phosphoprotein substrates (Shenoli-kar and Ingebritsen, 1984). Therefore, attemptsto define structural requirements for protein dephosphorylation using synthetic phos-phopeptides have failed to establish the mo-lecular basis for substrate specificity of themajor cellular protein phosphatases.The rate of 32P incorporation into proteinsin vivo, at either basal or hormone-stimulatedlevels, depends heavily on the rate of turnoverof phosphate in the protein. For example, lowphosphatase activity against specific sites orproteins will reduce the turnover of protein-bound phosphates and hinder their metaboliclabeling in the intact cell. Conversely, physi-Supplement 33Contributed by Bartholomew M. Sefton and Shirish Shenolikar Current Protocols in Molecular Biology (1996) 18.1.1-18.1.5Copyright © 2000 by John Wiley & Sons, Inc.18.1.1 Analysis of Protein Phosphorylationological stimuli that increase phosphatase ac-tivity may facilitate metabolic labeling of pro-teins, allowing rapid replacement of endo-genous “cold” phosphate with the radiolabel.Thus, 32P incorporation by itself gives no indi-cation about the extent of phosphorylation of a particular protein. As most phosphoproteins are phosphorylated on multiple sites, this also makes it difficult to correlate changes in protein function with 32P labeling of a given protein in the intact cell.SITES OF PHOSPHORYLATIONMost proteins are found to be phosphory-lated at serine or threonine residues, and many proteins involved in signal transduction are also phosphorylated at tyrosine residues. These three hydroxyphosphoamino acids exhibit suf-ficient chemical stability at acidic pH that they can be recovered after acid hydrolysis and iden-tified in a straightforward manner. Proteins that contain covalently bound phosphate at histid-ine, cysteine, and aspartic acid residues, either as phosphoenzyme intermediates or as stable modifications, have also been described. Each of these phosphoamino acids is chemically labile and impossible to study with the standard techniques used for the acid-stable phos-phoamino acids. Indeed, they are often identi-fied by inference or elimination. A technique for identifying phosphoserine, phosphothreon-ine, and phosphotyrosine by acid hydrolysis and two-dimensional thin-layer electrophore-sis is described in UNIT 18.3. Techniques for analyzing acid-labile forms of protein phos-phorylation are described in Ringer, 1991;Kamps, 1991; and Duclos et al., 1991.Phosphotyrosine is not an abundant phos-phoamino acid. Therefore, its detection in sam-ples labeled with 32P i is often difficult, espe-cially if the samples contain large quantities of proteins phosphorylated at serine residues or are contaminated with RNA. Detection of phos-photyrosine, as well as of phosphothreonine,can be enhanced considerably by incubation of gel-fractionated samples in alkali. This hydro-lyzes RNA and dephosphorylates phosphoser-ine, allowing visualization of minor tyrosine-and threonine-phosphorylated proteins. A sim-ple procedure for alkaline treatment is de-scribed in the alternate protocol of UNIT 18.3.DETECTION OF UNLABELED PHOSPHOAMINO ACIDSIf a protein is modified by phosphorylation,identification of the phosphoamino acid canoften be accomplished without resorting to bio-synthetic labeling. For example, tyrosine phos-phorylation can be studied because proteins containing this rare phosphoamino acid can be detected with great specificity and sensitivity by antibodies to phosphotyrosine (UNIT 18.4). By comparison, the quantitation of phosphoserine and phosphothreonine is more difficult and requires partial acid hydrolysis of the phospho-protein and subsequent separation of phos-phoamino acids by high-voltage electrophore-sis on thin-layer cellulose acetate plates. At-tempts to generate antibodies that recognize phosphoserine or phosphothreonine have failed to produce reagents with the required specific-ity and/or sufficient sensitivity to be useful.However, once the primary sequence around a phosphorylation site containing phosphoserine or phosphothreonine has been determined, it is possible to make antibodies against synthetic phosphopeptides modeled on these phosphory-lation sites (Czernik et al., 1991). Such anti-phosphopeptide antibodies have been very use-ful tools for monitoring the phosphorylation of the parent protein at specific sites. The phos-pho-specific antibodies have also been used to inhibit the dephosphorylation of individual sites and demonstrate their role in protein func-tion. Several recent studies have produced phospho-specific antibodies against phospho-rylation sites predicted solely by the primary sequence of a protein. These reagents have provided new insights into phosphorylations that control protein function.More generally, because phosphorylation often alters the mobility of a protein during SDS–polyacrylamide gel electrophoresis and almost always alters its isoelectric point, the presence of phosphorylated residues in an unlabeled protein can be deduced from al-tered gel mobility after incubation of the protein with a phosphatase. Most phosphory-lation sites are thought to reside near the surface of phosphoproteins. Therefore, they are equally accessible to phosphatases and proteases. In-deed, limited proteolysis often produces func-tional changes in phosphoproteins that closely mimic their in vitro dephosphorylation by phosphatase. As monitoring the release of ra-dioactivity from a 32P-labeled phosphoprotein does not distinguish between a phosphatase and a protease reaction, additional steps must to be taken to differentiate orthophosphate from small phosphopeptides. This is particularly im-portant when dephosphorylation reactions are carried out in crude extracts (UNIT 18.5).Supplement 33Current Protocols in Molecular Biology18.1.2Overview ofProteinPhosphorylationPROTEIN KINASESProtein kinases exhibit a strict specificity for phosphorylation of either serine/threonine or tyrosine residues. Yet these enzymes share structural homology in several domains such as the ATP-binding and catalytic sites, emphasiz-ing their common function as phosphotrans-ferases. A third group of protein kinases more closely resembles the serine/threonine kinases in their primary sequence but phosphorylate both serine/threonine and tyrosine residues.MEK, a MAP kinase activator (Crews and Erickson, 1992) and wee1, an inhibitor of cdc2kinase (Parker et al., 1992) are examples of such dual-specificity protein kinases that phospho-rylate threonine and tyrosine residues which are closely located in substrate proteins.Kinases have been used in a number of ways to analyze protein phosphorylation. Mutations that abrogate ATP-binding inactive protein ki-nases. Such inactive kinases retain the ability to recognize substrates and therefore act as dominant-negative reagents to analyze the physiological function of kinases in the intact cell (Thorburn et al., 1994; Alberola-Ila et al.,1995). Cell-permeable compounds modeled on ATP also inhibit selected kinases in the intact cell.Growth factors promote dimerization and activation of transmembrane receptor kinases.Mutant receptors that can dimerize in the ab-sence of the natural ligand offer a novel ap-proach to activate receptor kinases and initiate the cellular responses to growth factors (Spencer et al., 1993). Several cytoplasmic ki-nases are also activated by binding of intracel-lular second messengers. The allosteric activa-tion of these kinases results from conformation changes that eliminate internal autoinhibitory interactions which silence the kinases in the absence of ligand. Deletion of the autoinhibi-tory sequences generates a constitutively active kinase that provides important insights into its physiological role (Planas-Silva and Means,1992). Synthetic peptides modeled on the autoinhibitory sequences can also be used to selectively inhibit these kinases and reveal their role in controlling protein phosphorylation.Still other kinases, which are not directly regulated by ligand binding, are activated by phosphorylation. Some of these kinases partici-pate in cascades of sequential phosphorylations that amplify physiological signals. Substitution of the amino acids phosphorylated during the activation of these kinases with nonphosphory-lated residues generates dominant-negative en-zymes that can block the functions of suchkinases in intact cells. Alternately, acidic amino acids may be substituted for phosphoamino acids to produce a constitutively active kinase (Brunet et al., 1994). Such reagents have been used to validate the role of protein kinases in specific phosphorylation events.PROTEIN PHOSPHATASESMany protein phosphatases also show a strict specificity for either phosphoserine/phos-phothreonine or phosphotyrosine residues.However, unlike kinases, the serine/threonine and tyrosine phosphatases are not evolutionar-ily related and exhibit no primary sequence homology (Shenolikar and Nairn, 1991; Char-bonneau and Tonks, 1992). Acid and alkali phosphatases can also dephosphorylate phos-phoproteins in vitro but share no structural homology with protein phosphatases. A new family of phosphatases, such as cdc25 and CL100, are distantly related to the tyrosine phosphatases but dephosphorylate both phos-photyrosine and phosphothreonine in their tar-get substrates. Mutation of an essential cysteine in tyrosine phosphatases has produced domi-nant-negative enzymes that have been shown to shield their substrates from dephosphoryla-tion by endogenous phosphatases, thereby pro-longing the biological effects of protein phos-phorylation (Sun et al, 1993). Mutating several conserved amino acids can inactivate a ser-ine/threonine phosphatase (Zhou et al, 1994),but the ability of such mutant phosphatases to modulate serine/threonine phosphorylation in cells has not yet been demonstrated.A number of potent phosphatase inhibitors have been identified in recent years. Okadaic acid and several other toxins inhibit protein (serine/threonine) phosphatase 1 and 2A, and vanadate and phenylarsenoxide inhibit tyrosine phosphatase. These compounds have impli-cated reversible phosphorylation as a regula-tory mechanism in many physiological proc-esses (Cohen, 1989; Hardie et al., 1991; She-nolikar and Nairn, 1991; Shenolikar, 1994);under certain conditions, they may be the domi-nant regulators of these cellular processes.These phosphatase inhibitors can also be used to distinguish protein dephosphorylation from proteolysis in crude tissue extracts (Cohen,1991). However, by far the most important contribution of these reagents has been that they have allowed assessment of the role played by phosphorylation in cellular processes where neither the identity of the phosphoprotein in-volved nor that of the kinase(s) that regulates its function are known.Current Protocols in Molecular BiologySupplement 3318.1.3Analysis of ProteinPhosphorylationThe units in this chapter describe techniques that detect protein phosphorylation and identify amino acids that have been covalently modified (UNITS 18.2, 18.3, & 18.4). The next step is to use phosphatases to dephosphorylate the substrate protein in vitro and address the functional rele-vance of the covalent modification (UNIT 18.5).Once the regulatory role of phosphorylation has been clearly established, the more sophisticated approaches discussed above can be used to to identify the specific kinases and phosphatases involved and thereby begin to elucidate the physiological mechanism that regulates the substrate in the intact cell.LITERATURE CITEDAlberola-Ila, J., Forbush, K.A., Seger, R., Krebs,E.G., and Perlmutter, R.M. 1995. Selective re-quirement for MAP kinase activation in thymo-cyte differentiation. Nature 373:620-623.Brunet, A., Pages, G., and Poussegur, J. 1994. Con-stitutively active mutants of MAP (MEK1) in-duce growth factor-relaxation and oncogenicity when expressed in fibroblasts. Oncogene 9:3379- 3387.Charbonneau, H. and Tonks, N.K. 1992. 1002 pro-tein phosphatases? Annu. Rev. Cell Biol. 8:463-493.Cohen, P . 1985. The role of protein phosphorylation in the hormonal control of enzyme activity. Eur .J. Biochem. 15:439-448.Cohen, P. 1989. The structure and regulation of protein phosphatases. Annu. Rev. Biochem.58:453-508.Cohen, P. 1991. Classification of protein ser-ine/threonine phosphatases: Identification and quantitation in cell extracts. Methods Enzymol.201:389-398.Cori, G.T. and Cori, C.F. 1945. Enzymatic conver-sion of phosphorylase a to b . J. Biol. Chem.158:321-345.Crews, C.M. and Erickson, R.L. 1992. Purification of a murine protein-tyrosine/threonine kinase that phosphorylates and activates the erk-1 gene product: Relationship to the fission yeast byr1gene product. Proc. Natl. Acad. Sci. U.S.A.89:8205-8209.Czernik, A.J., Girault, J.A., Nairn, A.C., Chen, J.,Snyder, G., Kebabian, J., and Greengard, P . 1991.Production of phosphorylation state–specific an-tibodies. Methods Enzymol. 201:264-283.Doree, M. and Galas, S. 1994. The cyclin-dependent protein kinases and the control of cell division.F ASEB J. 85:1114-1121.Duclos, B., Marcandier, S., and Cozzone, A.J. 1991.Chemical properties and separation of phos-phoamino acids by thin-layer chromatography and/or electrophoresis. Methods Enzymol.201:10-21.Eisenmann, D.M. and Kim, S.K. 1994. Signal transduction and cell fate specification during Caenorhabditis elegans vulval development.Curr . Opin. Genet. Dev. 4:508-516.Hardie, D.G., Haystead, T.A.J., and Sim, A.T.R.1991. Use of okadaic acid to inhibit protein phosphatases in intact cells. Methods Enzymol.201:469-477.Heldin, C.-H. 1995. Dimerization of cell surface receptors in signal transduction. Cell 80:213-223.Ihle, J.N., Witthuhn, B.A., Quelle, F.W., Yamamoto,K., Thierfelder, W.E., Kreider, B., and Silven-noinen, O. 1994. Signalling by the cytokine re-ceptor superfamily: JAKS and STA TS. Trends Biochem. Sci. 19:222-227.Kamps, M.P . 1991. Determination of phosphoamino acid composition by acid hydrolysis of protein blotted to Immobilon. Methods Enzymol.201:21-27.Marshall, C.J. 1995. Specificity of receptor tyrosine kinase signalling: Transient versus sustained ex-tracellular signal-regulated kinase activation.Cell 80:179-185.Parker, L.L., Atherton-Fessler, S., and Piwinica-Worms, H. 1992. p107 wee1 is a dual-specificity kinase that phosphorylates p34cdc2 on tyrosine 15. Proc. Natl. Acad. Sci. U.S.A. 89:2917-2921.Planas-Silva, M.D. and Means, A.R. 1992. Expres-sion of a constitutive form of calcium/cal-modulin-dependent protein kinase II leads to arrest of the cell cycle in G2. EMBO J. 11:507-517.Ringer, D.P . 1991. Separation of phosphotyrosine,phosphoserine, and phosphothreonine by high-performance liquid chromatography. Methods Enzymol. 201:3-10.Shenolikar, S. 1994. Protein serine/threonine phos-phatases: New avenues for cell regulation. Annu.Rev. Cell Biol. 10:55-86.Shenolikar, S. and Ingebritsen, T.S. 1984. Protein (serine, threonine) phosphate phosphatases.Methods Enzymol. 107:102-130.Shenolikar, S. and Nairn, A.C. 1991. Protein phos-phatases: Recent progress. Adv. Second Messen-ger Phosphoprotein Res. 23:1-121.Simon, M. 1994. Signal transduction during the development of the Drosophila R7 photorecep-tor. Dev. Biol. 166:431-442.Spencer, D.M., Wandless, T.J., Schreiber, S.L., and Crabtree, G.R. 1993. Controlling signal transduction with synthetic ligands. Science 262:1019-1024.Sun, H., Charles, C.H., Lau, L.F., and Tonks, N.K.1993. MKP-1 (3CH134), an immediate early gene product, is a dual-specificity phosphatase that dephosphorylates MAP kinase in vivo. Cell 75:487- 493.Supplement 33Current Protocols in Molecular Biology18.1.4Overview ofProteinPhosphorylationThorburn, J., McMahon, M. and Thorburn, A. 1994.Raf-1 kinase activity is necessary and sufficient for gene expression changes but not sufficient for cellular morphology changes associated with cardiac myocyte hypertrophy. J. Biol. Chem.269: 30580-30586.Zhou, S., Clemens, J.C., Stone, R.L., and Dixon, J.E.1994. Mutational analysis of a ser/thr phos-phatase. Identification of residues important in phosphatase substrate binding and catalysis. J.Biol. Chem. 269:26234-26238.Contributed by Bartholomew M. Sefton The Salk Institute San Diego, CaliforniaShirish ShenolikarDuke University Medical Center Durham, North CarolinaCurrent Protocols in Molecular BiologySupplement 3318.1.5Analysis of ProteinPhosphorylation。

四组细胞瘤块切片进行免疫组化分析...

四组细胞瘤块切片进行免疫组化分析...

图i克隆的鉴定A:RT--PCR鉴定结果B:westernblot鉴定结果I:阳性对照办2:MDA--MB--435/Ikbl(L),3:MDA一船一435几KBl(H)。

4:MDA一蛐--4352,5:MDA一船--435/vec图2各组细胞transwell细胞侵袭实验数据为4株肿瘤细胞3次独立实验的均数±标准差。

误差线代表标准误。

2图3各细胞MMP一2、删P一9、VEGF、BFGFmRNA的表达隋况A:RT—PCR检测结果l、2、3,4分别代表为MDA-MB-435细胞,MDA一蛐一435/vec细胞-叭—姗一435/LI(B1(H)细胞、D为MDA-MB-435/LKBl(L)细胞3图5MMP一2,删P一9活性的测定A:凝胶酶谱MMP-2,姗P一9检测结果1、2、3.4分别代表为MDA-惦一435细胞、MI)A-MB一435/vec细胞MI)A—MB-435/LKBl(H)细胞,D为~f1)A-14B一435/LKBl(L)细胞图4各细胞MMP一2、姗P一9、VEGF、BFGF蛋白的表达情况A:westernblot检测结果l、2,3,4分别代表为MDA-MB-435细胞、l¥)A-MB-435/vec细胞盼A一邺一435几I(Blm)细胞、D为如A-船一435/LKBl(L)细胞图6人乳癌裸鼠原位移植瘤情况MDA·MB-435/LKBl(L)MDA·MB·435/LKBl(H)MDA—MB一435NECMDA-MB-435图7各组鼠肺转移灶比较PuIm。

narymelastaslstum。

阽(HE’100)图8各移植瘤MMP一2、MMP--9、VEGF、BFGF蛋白的表达情况A:westernblot检测结果1、2、3代表MDA-MB-435细胞、4,5,6代表d4DA-MB-435/vec细胞7,8,9代表MDA-MB-435/LKBl(L)细胞、10,11.12代表hⅢA—m一435/u(Bl(H)细胞图9各组瘤组织微血管密度变化图10LKBl基因与微血管密度变化的关系LKB1基因与人乳腺癌细胞生长和侵袭的相关性研究作者:庄志刚学位授予单位:复旦大学1.Westerman AM.Entius MM.de Baar E Peutz-Jeghers syndrome:78-year follow-up of the original family 19992.Tiainen M.Ylikorkala A.Makela TP Growth arrest by the LKB1 tumor suppressor:induction ofp21(WAFl/CIP1) 2002(13)3.Shen Z.Wen n F The tumor suppressor gene LKB1 is assoclate with prognosis in human breast carcinoma 2002(07)4.Sapkota GP.Boudeau J.Deak M Phosphorylation of the protein kinase mutated in Peutz-Jeghers cancer syndrome,LKB1/STK11,at Ser431 by p90(RSK)and cAMP-dependent protein kinase,but not its farnesylation at Cys(433),is essential for LKB1 to suppress cell vrowth 2001(22)5.8apkota GP.Boudeau J.Deak M Identification and characterization of four novel phosphorylationsites(Ser31,Ser325,Thr336 and Thr366)on LKB1/STK11,the protein kinase mutated in Peutz-Jeghers cancer syndrome 2002(02)6.Forster LF.Defres S.Goudie DR An investigation of the Peutz-Jeghers gene(LKB1)in sporadic breast and colon cancers 20007.Chen J Lindblom Germline mutation screening of the STK11/LKB1 gene in familial breast cancer with LOH on 19p 20008.Kleiner DE.Stetler-Stevenson WG Matrix metalloproteinases and metastasis 19999.McCawley LJ.Matrisian LM Matrix metalloproteinases:they are not just for matrix anymore! 200110.Hojilla CV.Mohammed FF.Khokha R Matrix metalloproteinases and theirtissue inhibitors direct cell fate during cancer development 2003(10)11.Stamenkovic I Matrix metalloproteinases in tumor invasion andmetastasis 2000(06)12.John A.Tuszynski G The role of matrix metalloproteinases in tumor angiogenesis and tumor metastasis 2001(01)13.Freije JM.Balbin M.Pendas AM Matrix metalloproteinases and tumor progression 200314.Jones JL.Shaw JA.Pringle JH Primary breast myoepithelial cells exert an invasion-suppressor effect on breast cancer celis via paracrine down-regulation of MMP expression in fibroblasts and tumour cells 2003(04)15.Karuman P.Gozani O.Odze RD The Peutz-Jegher gene product LKB1 is a mediator of p53-dependent cell death[外文期刊] 200116.Wang JL.Sun Y.Wu S Gamma-irradiation induces matrix metalloproteinase Ⅱ expression in a p53-dependent manner 2000(04)17.Folkmen J Tumor angiogenesis:Therapeutic implication 197121.Tsutsui S.Kume M.Era S Prognostic value of microvessel density in invasive ductal carcinoma of the breast 2003(04)22.Weidner N.Semple J P.Welch W R.Folkman J Tumor angiogenesis and metastasis-correlat ion in invasive breast carcinoma 199123.Guidi AJ.Schnitt SJ.Fischer L Vascular permeability factor(vascular endothelial growthfactor)expresslon and anglogenesis in patients with ductal carcinoma in situ of the breast 199724.Ramanathan M.Giladi A.Leibovich SJ Regularion of vascularendothelial growth factor gene expression in murine macrophages by nitric oxide and hypoxia 2003(06)25.Ylikorkala A.Rossi DJ.Korsisaari N Vascular abnormalities and deregulation of VEGF in LKB1-deficient mice[外文期刊] 200126.Westerman AM.Entius MM.de Baar E Peutz-Jeghers syndrome:78-year follow-up of the original family 199927.Tiainen M.Ylikorkala A.Makela TP Growth arrest by the LKB1 tumor suppressor:induction ofp21(WAF1/CIP1) 2002(13)28.Jimenez AI.Fernandez P.Dominguez O Growth and molecular profile of lung cancer cells expressing ectopic LKB1:down-regulation of the phosphatidylinositol 3-phosphate kinase/PTEN pathway 2003(06)29.Ylikorkala A.Rossi DJ.Korsisaari N Vascular abnormalities and deregulation of VEGF in Lkb1-deficient mice[外文期刊] 200130.Miyoshi H.Nakau M.Ishikawa TO Gastrointestinal hamartomatous polyposis in lkb1 heterozygous knockout mice 2002(08)31.Rossi DJ.Ylikorkala A.Korsisaari N Induction of cyclooxygenase-2 in a mouse model of Peutz-Jeghers polyposis 2002(19)32.Bardeesy N.Sinha M.Hezel AF Loss of the Lkb1 tumor suppressor provokes intestinal polyposis but resistance to transformation 2002(6903)33.Nakau M.Miyoshi H.Seldin MF Hepatocelhlar carcinoma caused by loss of heterozygosity in lkb1 gene knockout mice 2002(16)34.Watts JL.Morton DG.Bestman j The C.elegans par-4 gene encodes a putative serine-threonine kinase required for establishing embryonic asymmetry35.Martin SG.St Johnston D A role for Drosophila LKB1 in anterior-posterior axis formation and epithelial polarity[外文期刊] 2003(6921)36.Baas AF.Boufeau J.Sapkota GP Activation of the tumour suppressor kinase LKB1 by the STE20-like pseudokinase STRAD 2003(12)37.Sapkota GP.Boudeau J.Deak M Phosphorylation of the protein kinase mutated in Peutz-Jeghers cancer syndrome,LKB1/STK11,at Ser431 by p90(RSK)and cAMP-dependent protein kinase,but not its farnesylation at Cys(433),is essential for LKB1 to suppress cell vrowth 2001(22)38.Sapkota GP.Boudeau J.Deak M Identification and characterization of four novel phosphorylation39.Smith DP.Rayter SI.Niederlander C LIP1,a cytoplasmic proteinfunctionally linked to the Peutz-Jeghers syndrome kinase LKB1 2001(25)40.Marignani PA.Kanai F.Carpenter CL LKB1 associates with Brg1 and is necessary for Brg1-induced growth arrest 2001(35)41.Karuman P.Gozani O.Odze RD The Peutz-Jegher gene product LKB1 is a mediator of p53-dependent cell death[外文期刊] 2001(06)42.Bignell GR.Barfoot R.Seal S Low frequency of somatic mutations in the LKB1/Peutz-Jeghers syndrome gene in sporadic breast cancer 1998(07)43.Forster LF.Defres S.Goudie DR An investigation of the Peutz-Jeghers gene(LKB1)in sporadic breast and colon cancers 2000(10)44.Chen J Lindblom Germline mutation screening of the STK11/LKB1 gene in familial breast cancer with LOH on 19p 2000(05)45.Shen Z.Wen n F The tumor suppressor gene Lkb1 is associate with prognosis in human breast carcinoma 2002(07)1.费菲原肌球蛋白-4在人类乳腺癌高低转移细胞株中的差异性表达及临床意义[学位论文]20052.沈赞抑癌基因LKB1在乳腺癌中的作用研究[学位论文]20023.王振军.严仲瑜.毕郭龙国人黑斑息肉病LKB1基因胚系突变的检测[期刊论文]-中华外科杂志2000,38 (2)4.董慧明上皮钙黏蛋白对人炎性乳腺癌细胞系生物学特性的影响[学位论文]20055.丁锦华微浸润在导管原位癌中的临床意义及MMP-2、Tenascin-C在导管原位癌中的表达[学位论文]20056.刘刚乳腺癌血管、淋巴管生成与转移预后的研究[学位论文]20037.张杰乳腺癌前哨淋巴结活检及骨髓播散肿瘤细胞的检测[学位论文]20058.王劲松Rab27A对人乳腺癌细胞生物学特性的影响及其机制的研究[学位论文]20079.宋科瑛耐药乳腺癌细胞MDR-MCF-7侵袭力增强机制探讨[学位论文]200310.李鹤成ERα和Her-2受体在人乳腺癌细胞株作用通路的实验研究及基质金属蛋白酶在淋巴结阴性乳腺癌的预后意义[学位论文]2005本文链接:/Thesis_Y952108.aspx。

抑癌基因肝激酶B1(LKB1在肿瘤中研究进展

抑癌基因肝激酶B1(LKB1在肿瘤中研究进展

69抑癌基因肝激酶B1(LKB1)在肿瘤中研究进展郑 涛(四川大学华西第二医院,四川 成都 610041)摘 要:肝激酶B1(LKB1),又称为丝氨酸/苏氨酸激酶,已经明确为许多癌细胞中的关键抑癌因子。

它是AMP激活蛋白激酶(AMPK)相关蛋白激酶的主要上游激酶,并具有多种生物学功能。

因此,本综述主要讨论有关LKB1表达、调控、下游信号转导和癌症抑制功能,有助于更好地了解该基因及其癌症中的意义。

关键词:肝激酶B1;LKB1;癌症;AMPK;MARK中图分类号:R73-3 文献标识码:A作者简介:郑涛,四川大学华西第二医院。

肝激酶B1(LKB1)基因,编码约50kD 的丝氨酸/苏氨酸激酶,最早在Peutz-Jeghers 综合征(PJS)中发生突变,主要以皮肤粘膜色素沉着为特征,增加胃肠道错构瘤性息肉病及良性和恶性肿瘤的风险,是罕见的遗传性疾病[1]。

考虑到LKB1在不同癌症中的重要性。

本文总结了有关LKB1在表达、下游信号通路及在癌症抑制中的作用。

1 肝激酶B1(lkb1)分子特征和定位1.1 肝激酶B1(lkb1)分子特征LKB1属于丝氨酸/苏氨酸激酶11(STK11),可以调节细胞的各种生物学过程,如细胞代谢、细胞极性、增殖和迁移。

人源LKB1基因,由10个跨度为23kb 的外显子组成,位于19P13.3染色体上。

人体中几乎所有组织均有LKB1表达,但在上皮和睾丸生精小管中显着表达。

胎儿组织中LKB1表达水平高于成人组织[1]。

1.2 LKB1基因亚细胞分布LKB1在哺乳动物细胞中具有不同的定位。

来自PJS 患者的样本中,野生型LKB1分布在细胞核和细胞质中,而LKB1突变体SL26具有正常的激酶功能,但仅在细胞核中积累。

仅在哺乳动物细胞中过表达时,LKB1大部分位于细胞核,仅小部分位于细胞质。

然而,只有位于细胞质中的LKB1才能发挥主要的生物学功能。

2 LKB1表达的调控2.1 表观遗传修饰尽管已在散发性癌症中发现LKB1基因突变,但其体细胞突变却非常罕见,这表明LKB1基因的失活可能由其他机制调控。

lkb1(thr336)磷酸化多克隆抗体的制备及在细胞中表达的检测

lkb1(thr336)磷酸化多克隆抗体的制备及在细胞中表达的检测

安徽大学硕士学位论文LKB1(Thr336)磷酸化多克隆抗体的制备及在细胞中表达的检测Preparation and Intracellular Expression detection of Phosphorylated LKB1 (Thr336) Polyclonal Antibody姓名:任艳敏学科专业:细胞生物学研究方向:细胞凋亡与肿瘤发生指导教师:黄蓓完成时间: 2008-5LKB1(Thr336)磷酸化多克隆抗体的制备及在细胞中表达的检测摘要PJS综合症是一种遗传性肿瘤综合症,该病患者患肿瘤的几率增加。

致病基因编码的核定位激酶LKB1作为肿瘤抑制子在发挥作用,但是LKB1在细胞中调控的机制及生理基础都不清楚。

研究表明LKB1在细胞中被磷酸化并且Thr336磷酸化位点在LKB1抑制细胞生长中起到重要的作用。

为了研究LKB1在不同细胞中的表达规律及其对细胞增殖的抑制功能,本实验通过生物信息学方法选取LKB1 Thr336位点附近10个氨基酸且使Thr336位点磷酸化,并铰链上BSA,以此为抗原,对兔子进行免疫,制备的抗血清通过Western blot检测抗体效价,并用LKB1抗体和p-LKB1(Thr336)抗体分别对人正常肝脏细胞HL7702、人胚肾细胞HEK293、小鼠肉瘤细胞S180、非洲绿猴肾细胞COS7及人结肠癌细胞Caco2检测。

结果显示,LKB1抗体不仅识别His-LKB1蛋白而且识别p-LKB1(Thr336)肽段,而p-LKB1(Thr336)抗体仅识别p-LKB1(Thr336)抗原;且在正常细胞及肿瘤细胞中LKB1和p-LKB1(Thr336)表达量有差异,在增殖越快的细胞中,p-LKB1(Thr336)表达量越高。

关键词: LKB1; 磷酸化多克隆抗体; 免疫印迹法; 生物信息学AbstractPeutz±Jeghers syndrome is an inherited cancer syndrome, which results in a greatly increased risk of developing tumours in those affected. The causative gene encodes a nuclear-localized protein kinase, termed LKB1, which is predicted to function as a tumour suppressor. The mechanism by which LKB1 is regulated in cells is not known, and nor have any of its physiological substrates been identified. Recent studies have demonstrated that LKB1 is phosphorylated in cells and Its Thr336 phosphorylation site play an important role in cell growth inhibition. To study the expression and the suppression to cell proliferation of LKB1in different cells, the LKB1Thr336 was phosphorylated and the neighboring 10-amino acid region was chosen to hinge on a BSA through Bioinformatics methods. Then the constructed peptide was used as antigen to immunize rabbits and antibody titer was performed by anti-serum western blot detection. In addition, Rabbit anti-LKB1 antibody and the p-LKB1 (Thr336) antibody were also used to assay with several cell lines, including the human liver cells HL7702, embryonic kidney cells HEK293, the mice sarcoma cells S180, the African green monkey kidney cells COS7 and the human colon cancer cells Caco2. The results showed that Rabbit-anti-LKB1 antibodies can interact with both His-LKB1 protein and p-LKB1 (Thr336) peptide, while the p-LKB1 (Thr336) antibody can only interact with p-LKB1 (Thr336) antigen. Moreover, there are quite different expression patterns of LKB1 and p-LKB1 (Thr336) between normal and tumor cells. Our results also suggested that the expression levels of p-LKB1 (Thr336) are in positive correlation with the proliferation of cells.Key words:p-LKB1 (Thr336); Phosphorylated Polyclonal antibody; Western blot;Bioinformatics目录第1章绪论 (7)1.1. LKB1/STK11 的基本结构及分布 (7)1.2 LKB1/STK11的生物学功能 (7)1.3. LKB1磷酸化位点研究现状 (10)1.4.国内外LKB1 Thr336位点磷酸化抗体研究现状 (11)1.5.本实验的目的和意义 (11)第2章LKB1蛋白的生物信息分析及磷酸化抗原的设计 (14)2.1 实验有关数据库 (14)2.2 实验方法 (14)2.2.1 LKB1蛋白序列分析 (14)2.2.2.LKB1磷酸化位点分析 (15)2.2.3. LKB1抗原决定蔟的预测 (17)2.2.4. LKB1疏水性及抗原性分析 (17)2.2.5.合成肽段的设计原则 (19)2.3. LKB1抗原设计合成结果及纯度检测 (19)第3章His-LKB1重组蛋白的诱导表达及分离纯化 (21)3.1实验材料 (21)3.2实验方法 (26)3.3. SDS-PAGE检测实验结果 (30)第4章 LKB1(Thr336)多肽抗原的设计及其多克隆抗体的制备 (33)4.1 实验试剂 (33)4.2 实验方法 (33)4.2.1兔子免疫与血清处理 (33)4.2.2 Western blot方法检测抗体及效价 (34)4.3 实验结果 (36)第5章 P-LKB1(Thr336)抗体的特异性鉴定 (38)5.1 实验材料 (38)5.2 实验方法-Western blot检测抗体特异性 (38)5.3 Western Blot检测抗体特异性结果 (38)第6章 p- LKB1(Thr336)多克隆抗体的分离纯化 (39)6.1 实验材料 (39)6.2 实验方法-抗原条结合法分离纯化抗血清 (40)6.3 实验结果 (40)第7章LKB1在正常细胞及肿瘤细胞中表达的检测 (41)7.1. 实验材料与试剂 (41)7.2 实验方法 (42)7.2.1细胞培养方法 (42)7.2.4 LKB1在细胞中表达规律的检测 (45)7.3 实验结果 (45)总结 (47)1.. 结果与讨论 (47)2. 本实验创新点 (48)3. 存在的问题与展望 (48)致谢 (49)参考文献 (50)第1章绪论LKB1 基因,又名STKⅡ( serinePthreonine proteinkinase 11) 基因, 是由Hemminki 等[1]在1998 年从Peutz Jeghers syndrome ( PJS) 患者的血细胞中克隆出来的一种基因, PJS是一种常染色体显性遗传疾病,患者不但肠道、胃、胰腺发生恶性肿瘤的可能性增加, 而且在乳腺、子宫颈、肺、卵巢、睾丸发生恶性变危险度也增加,初步估计93 %的PJS 患者平均在43 岁时发生恶性肿瘤[2]。

蛋白质磷酸化修饰的研究进展_姜铮

蛋白质磷酸化修饰的研究进展_姜铮

doi:10.3969/j.issn.1009-0002.2009.02.025综述蛋白质磷酸化修饰的研究进展姜铮,王芳,何湘,刘大伟,陈宣男,赵红庆,黄留玉,袁静中国人民解放军疾病预防控制研究所,北京100071[摘要]蛋白质磷酸化是最常见、最重要的一种蛋白质翻译后修饰方式,它参与和调控生物体内的许多生命活动。

通过蛋白质的磷酸化与去磷酸化,调控信号转导、基因表达、细胞周期等诸多细胞过程。

随着蛋白质组学技术的发展和应用,蛋白质磷酸化的研究越来越受到广泛的重视。

我们介绍了蛋白质磷酸化修饰的主要类型与功能、磷酸化蛋白质分析样品的富集及制备、磷酸化蛋白的鉴定及磷酸化位点的预测、蛋白分离后磷酸化蛋白的检测,及蛋白质磷酸化的分子机制,并综述了近年来国内外的主要相关研究进展。

[关键词]磷酸化修饰;磷酸化蛋白鉴定;磷酸化位点检测[中图分类号]Q52[文献标识码]A[文章编号]1009-0002(2009)02-0233-05Progress on Protein/Peptide PhosphorylationJIANG Zheng,WANG Fang,HE Xiang,LIU Da-Wei,CHEN Xuan-Nan,ZHAO Hong-Qing,HUANG Liu-Yu,YUAN JingInstitute of Disease Control and Prevention,Academy of Military Medical Sciences,100071Beijing,China[Abstract]Phosphorylation is one of the most important post-translational modifications of proteins,which is related to many activities of life.By reversible protein phosphorylation eukaryotes control many cellular processes including signal transduction,gene expression,and the cell cycle etc.As the development and application of the proteomics,the studies of the protein phosphorylation have become more important.This article has introduced the main types and functions of the protein phosphorylation,the enrichment and preparation of phosphoproteins and phosphopeptides,the identification of the phosphopeptides,the determination and prediction of the specific-phosphorylation-site,the phosphorelated modifications of the proteins,and the progress on studies above as well.[Key words]phosphorelated modifications;identification of the phosphopeptides;determination of the specific-phosphory-lation-site几乎所有的蛋白质在合成过程中或合成后都要经过某些形式的翻译后修饰,一些不合适的修饰常常与疾病相关,某些特定的翻译后修饰还被作为疾病的生物标志或治疗的靶标。

《在LeLa细胞中研究LKB1和PTEN的相互作用》范文

《在LeLa细胞中研究LKB1和PTEN的相互作用》范文

《在LeLa细胞中研究LKB1和PTEN的相互作用》篇一一、引言近年来,随着生物医学的飞速发展,我们对细胞内信号转导与基因调控的理解越来越深入。

LeLa细胞作为一种广泛应用的细胞模型,在研究各种生物学过程中具有独特的重要性。

本研究将探讨LeLa细胞中LKB1(肝细胞癌激酶B1)和PTEN(磷酸酶与张力蛋白同源物)的相互作用,以揭示其在细胞信号调控和疾病发生发展中的作用。

二、LKB1和PTEN的基本概述LKB1是一种丝氨酸/苏氨酸蛋白激酶,在多种生物过程中发挥着重要作用,包括细胞生长、代谢和凋亡等。

而PTEN则是一种重要的肿瘤抑制基因,具有磷酸酶活性,可负向调节PI3K/AKT信号通路。

这两种基因在多种癌症中表现出异常表达或突变,因此研究它们在LeLa细胞中的相互作用具有重要的科学意义。

三、LKB1和PTEN在LeLa细胞中的相互作用我们的研究显示,LKB1和PTEN在LeLa细胞中存在直接的相互作用。

通过蛋白质互作实验和免疫共沉淀技术,我们证实了LKB1和PTEN在细胞内形成复合物。

进一步的研究表明,这种相互作用可能影响LKB1的激酶活性和PTEN的磷酸酶活性,从而调节细胞的信号转导。

四、LKB1和PTEN相互作用的功能研究我们的研究发现,LKB1和PTEN的相互作用在调节细胞生长、凋亡和代谢等方面具有重要作用。

具体来说,这种相互作用可能通过影响PI3K/AKT信号通路来调节细胞的生长和凋亡。

此外,我们还发现LKB1和PTEN的相互作用可能对细胞的能量代谢产生影响,从而影响细胞的生存和发展。

五、LKB1和PTEN相互作用的潜在应用价值我们的研究结果为进一步探索LKB1和PTEN在癌症等疾病中的作用提供了新的思路。

未来可以通过调节LKB1和PTEN的相互作用来开发新的治疗策略,例如通过药物或基因疗法来激活或抑制LKB1和PTEN的功能,以实现对肿瘤等疾病的干预和治疗。

此外,这一研究还可以为药物筛选提供新的靶点,以寻找能够针对特定癌症类型进行治疗的药物。

《在LeLa细胞中研究LKB1和PTEN的相互作用》范文

《在LeLa细胞中研究LKB1和PTEN的相互作用》范文

《在LeLa细胞中研究LKB1和PTEN的相互作用》篇一一、引言近年来,随着生物医学研究的深入,细胞信号传导机制成为了研究的热点。

LKB1和PTEN作为细胞内重要的信号分子,在多种生物过程中发挥着关键作用。

为了更深入地理解这两种分子在细胞中的功能及其相互作用,本文以LeLa细胞为研究对象,探讨了LKB1和PTEN的相互作用及其在细胞信号传导中的作用机制。

二、LKB1和PTEN简介LKB1(肝激酶B1)是一种丝氨酸/苏氨酸蛋白激酶,主要参与细胞的能量代谢、极性建立和细胞周期调控等过程。

而PTEN (磷酸酯酶失活型张力蛋白)是一种负向调节因子,可以调节细胞内多种信号通路的活性,对肿瘤细胞的生长、增殖及凋亡具有重要影响。

三、研究方法本研究采用LeLa细胞作为研究对象,运用基因编辑技术构建了LKB1和PTEN的敲除及过表达细胞模型。

通过细胞培养、Western blot、PCR等技术手段,探讨LKB1和PTEN在LeLa细胞中的相互作用及其对细胞生长、凋亡等生物过程的影响。

四、实验结果(一)LKB1和PTEN的表达与调控实验结果表明,在LeLa细胞中,LKB1和PTEN的表达水平受到多种因素的调控。

在正常条件下,LKB1和PTEN的表达水平相对稳定。

当细胞受到外界刺激时,如生长因子、肿瘤抑制剂等,LKB1和PTEN的表达水平会发生变化,从而影响细胞的生长和凋亡。

(二)LKB1和PTEN的相互作用通过基因编辑技术构建的敲除及过表达细胞模型显示,LKB1和PTEN之间存在相互作用。

在LeLa细胞中,LKB1可以通过激活下游信号通路来影响PTEN的表达和活性。

而PTEN则可以通过负反馈机制调节LKB1的活性。

这种相互作用对细胞的生长、增殖及凋亡等生物过程具有重要影响。

(三)LKB1和PTEN对细胞生长、凋亡的影响实验结果表明,在LeLa细胞中,LKB1和PTEN的表达水平与细胞的生长、凋亡密切相关。

当LKB1或PTEN的表达水平发生变化时,细胞的生长和凋亡过程也会受到影响。

《HeLa细胞中研究LKB1-NUAK1对PTEN的磷酸化影响》范文

《HeLa细胞中研究LKB1-NUAK1对PTEN的磷酸化影响》范文

《HeLa细胞中研究LKB1-NUAK1对PTEN的磷酸化影响》篇一HeLa细胞中研究LKB1-NUAK1对PTEN的磷酸化影响一、引言在细胞生物学的研究中,蛋白质的磷酸化是一个重要的生物学过程,它对于细胞信号传导、细胞周期调控以及细胞存活等过程起着关键作用。

PTEN(磷酸酶与张力蛋白同源物)作为一种重要的肿瘤抑制基因,其磷酸化状态与多种癌症的发生和发展密切相关。

而LKB1(肝激酶B1)和NUAK1(非催化性腺苷酸激活蛋白激酶1)则是参与磷酸化过程的两个重要基因。

本篇论文旨在探讨HeLa细胞中LKB1/NUAK1对PTEN的磷酸化影响。

二、材料与方法2.1 细胞系与质粒本实验采用HeLa细胞系作为研究对象,同时构建了LKB1和NUAK1的过表达质粒以及PTEN的野生型和突变型质粒。

2.2 实验方法(1)细胞培养与转染:将HeLa细胞培养在适宜的培养基中,并进行LKB1和NUAK1的过表达质粒转染。

(2)蛋白提取与磷酸化检测:提取细胞中的蛋白质,通过Western blot等方法检测PTEN的磷酸化水平。

(3)基因敲除与回复实验:利用CRISPR-Cas9技术对HeLa 细胞中的LKB1或NUAK1进行敲除,并观察PTEN磷酸化水平的变化;同时通过回复实验验证LKB1和NUAK1对PTEN磷酸化的影响。

三、结果与分析3.1 LKB1和NUAK1对PTEN磷酸化的影响通过 Western blot 实验发现,在 HeLa 细胞中过表达 LKB1 和NUAK1 后,PTEN 的磷酸化水平显著提高。

这一结果表明LKB1 和 NUAK1 在 PTEN 的磷酸化过程中起到了重要作用。

3.2 基因敲除实验结果利用CRISPR-Cas9 技术对HeLa 细胞中的LKB1 或NUAK1 进行敲除后,PTEN 的磷酸化水平降低,进一步验证了LKB1 和NUAK1 对 PTEN 磷酸化的促进作用。

3.3 回复实验结果通过回复实验发现,当在基因敲除的HeLa 细胞中再次引入LKB1 和 NUAK1 时,PTEN 的磷酸化水平得以恢复,表明 LKB1 和 NUAK1 能够直接或间接地影响 PTEN 的磷酸化过程。

《HeLa细胞中研究LKB1-NUAK1对PTEN的磷酸化影响》范文

《HeLa细胞中研究LKB1-NUAK1对PTEN的磷酸化影响》范文

《HeLa细胞中研究LKB1-NUAK1对PTEN的磷酸化影响》篇一HeLa细胞中研究LKB1-NUAK1对PTEN的磷酸化影响一、引言在细胞生物学领域,蛋白质磷酸化是一个重要的调控机制,它涉及到多种信号转导途径的激活和失活。

PTEN(磷酸酶和张力蛋白同源物)是一种在多种癌症中起到重要作用的肿瘤抑制基因。

其功能主要通过去磷酸化作用实现,从而调控细胞内多种信号转导过程。

近年来,越来越多的研究表明,LKB1和NUAK1等激酶对PTEN的磷酸化调控作用,对于细胞生长、分化和肿瘤发生发展等具有重要影响。

本篇论文以HeLa细胞为研究对象,探讨了LKB1/NUAK1对PTEN的磷酸化影响及其机制。

二、材料与方法2.1 实验材料HeLa细胞系、LKB1激酶、NUAK1激酶、PTEN蛋白及相关抗体等。

2.2 实验方法(1)细胞培养与处理:培养HeLa细胞,并分别对细胞进行LKB1和NUAK1的处理。

(2)蛋白质提取与纯化:提取处理后的细胞蛋白质,进行纯化处理。

(3)激酶反应:将纯化后的PTEN蛋白与LKB1/NUAK1激酶进行反应,观察磷酸化情况。

(4)数据分析:利用相关软件对实验数据进行处理和分析。

三、实验结果3.1 LKB1对PTEN的磷酸化影响实验结果显示,在HeLa细胞中,LKB1激酶能够显著促进PTEN的磷酸化。

通过激酶反应,我们可以观察到PTEN蛋白在LKB1的作用下,其磷酸化程度明显增强。

这表明LKB1对PTEN 的磷酸化具有正向调控作用。

3.2 NUAK1对PTEN的磷酸化影响同样地,我们在HeLa细胞中观察到NUAK1激酶也能够促进PTEN的磷酸化。

与LKB1类似,NUAK1的作用下,PTEN蛋白的磷酸化程度也有所增强。

这表明NUAK1也对PTEN的磷酸化具有正向调控作用。

3.3 LKB1与NUAK1的协同作用进一步的研究发现,当同时存在LKB1和NUAK1时,它们对PTEN的磷酸化具有协同作用。

《溶血磷脂酰胆碱的促动脉粥样硬化作用及其机制研究》范文

《溶血磷脂酰胆碱的促动脉粥样硬化作用及其机制研究》范文

《溶血磷脂酰胆碱的促动脉粥样硬化作用及其机制研究》篇一一、引言动脉粥样硬化(AS)是一种常见的血管疾病,其特点是动脉壁出现脂质沉积、纤维组织增生和钙化等病理改变。

溶血磷脂酰胆碱(LPC)作为一种生物活性分子,近年来在动脉粥样硬化的发病机制中受到了广泛关注。

本文旨在探讨溶血磷脂酰胆碱的促动脉粥样硬化作用及其机制,为预防和治疗动脉粥样硬化提供新的思路。

二、溶血磷脂酰胆碱与动脉粥样硬化溶血磷脂酰胆碱(LPC)是细胞膜磷脂代谢的产物,在体内具有多种生物活性。

近年来,越来越多的研究表明,LPC在动脉粥样硬化的发病过程中发挥了重要作用。

LPC能够通过多种途径促进动脉粥样硬化的发生和发展,包括影响血管内皮功能、促进平滑肌细胞增殖和迁移、诱导炎症反应等。

三、溶血磷脂酰胆碱促动脉粥样硬化的机制研究1. 影响血管内皮功能:LPC能够破坏血管内皮细胞的完整性,导致内皮功能障碍,使血管通透性增加,进而促进低密度脂蛋白(LDL)等脂质成分进入血管壁,形成脂质沉积。

2. 促进平滑肌细胞增殖和迁移:LPC能够刺激血管平滑肌细胞的增殖和迁移,使平滑肌细胞在血管壁内过度聚集,形成斑块,导致血管狭窄和堵塞。

3. 诱导炎症反应:LPC能够激活血管内皮细胞和巨噬细胞等免疫细胞,释放炎症因子,如细胞因子、氧化应激产物等,进一步促进动脉粥样硬化的发生和发展。

四、研究方法为了深入探讨溶血磷脂酰胆碱促动脉粥样硬化的机制,我们采用了多种研究方法,包括细胞培养、动物实验和分子生物学技术等。

首先,我们通过细胞培养实验观察了LPC对血管内皮细胞和平滑肌细胞的影响;其次,我们利用动物模型研究了LPC在动脉粥样硬化发病过程中的作用;最后,我们通过分子生物学技术探讨了LPC促动脉粥样硬化的相关信号通路和基因表达变化。

五、研究结果通过上述研究方法,我们得出了以下结论:1. 溶血磷脂酰胆碱能够破坏血管内皮细胞的完整性,导致内皮功能障碍,促进低密度脂蛋白等脂质成分进入血管壁。

AMP激活蛋白激酶活性调节研究进展

AMP激活蛋白激酶活性调节研究进展

AMP激活蛋白激酶活性调节研究进展付大波;杨震国;王蕾;张伟;肖飞【摘要】腺苷一磷酸激活蛋白激酶能感知细胞能量代谢状态的改变,维持机体的能量代谢平衡,称为"能量开关",因AMWATP升高而被激活,还能被其上游酶如ATM、TAK1等激活.文章主要探讨了AMPK活性的调控因素.【期刊名称】《饲料博览》【年(卷),期】2010(000)009【总页数】3页(P9-11)【关键词】腺苷一磷酸激活蛋白激酶;AMP依赖通路;非AMP依赖通路;AMPK激酶【作者】付大波;杨震国;王蕾;张伟;肖飞【作者单位】武汉工业学院,动物营养与饲料科学湖北省重点实验室,武汉,430023;武汉工业学院,动物营养与饲料科学湖北省重点实验室,武汉,430023;武汉工业学院,动物营养与饲料科学湖北省重点实验室,武汉,430023;武汉工业学院,动物营养与饲料科学湖北省重点实验室,武汉,430023;武汉工业学院,动物营养与饲料科学湖北省重点实验室,武汉,430023【正文语种】中文【中图分类】Q814.9%Q55腺苷一磷酸激活蛋白酶(AMPK)能感知细胞能量代谢状态的改变,维持机体的能量代谢平衡。

当细胞内能量缺失时,AMPK被磷酸化激活,抑制消耗ATP的合成代谢过程,启动生成ATP的分解代谢过程。

AMPK的活性受到许多因素的调控,本文对AMPK活性的调控因素作一概述。

1 AMPK的分子结构AMPK在真核细胞生物中广泛存在,属丝氨酸/苏氨酸蛋白激酶。

其是由催化亚基α、调节亚基β和γ组成的一个异源三聚体。

3个亚单位在AMPK的稳定性和活性中有各自特殊的作用。

α亚基含有2个功能区:一个N端激酶结构域和一个C 端结构域,两者大小基本相等。

N端是催化核心部位,C端负责与β和γ亚基结合。

β亚基N端区域之后紧跟着两个保守的结构域-KIS和ASC,ASC结构域为形成稳定有活性的α、β、γ复合物所必需,KIS结构域为β亚基上的功能性糖原结合结构域[1-2]。

LKB1蛋白在肺癌组织中的表达及意义

LKB1蛋白在肺癌组织中的表达及意义

LKB1蛋白在肺癌组织中的表达及意义孙万仆【摘要】目的分析LKB1蛋白在肺癌组织中的表达特点,并探讨其临床意义.方法选择本院收治的非小细胞肺癌患者80例,取患者癌肿原发灶组织与距癌肿边缘5cm以上的正常组织为研究对象,应用免疫组化方法检测LKB1蛋白在不同组织中的表达情况.结果 LKB1蛋白在正常肺组织中的阳性率明显比肺癌组织高,差异有统计学意义(P<0.05);LKB1蛋白在鳞癌与腺癌患者肿瘤组织中的阳性表达率比较,差异无统计学意义(P>0.05);高分化肿瘤组织中LKB1蛋白的阳性率比中-低分化肿瘤高,差异具有统计学意义(P<0.05);淋巴结转移阳性的肿瘤组织中LKB1蛋白阳性率明显低于淋巴结转移阴性组织,差异具有统计学意义(P<0.05);LKB1蛋白在Ⅰ~Ⅱ期肺癌中的表达阳性率比在ⅢA~B期肺癌中高,差异有统计学意义(P<0.05).结论LKB1蛋白的表达与非小细胞肺癌的发生、发展关系密切,LKB1蛋白的表达程度随肿瘤分化程度的下降、淋巴结转移阳性率的升高、疾病分期的进展而降低,LKB1蛋白表达的下调可能减弱了对肿瘤发展的抑制作用,其低表达可能是疾病预后不良的危险因素.【期刊名称】《青岛医药卫生》【年(卷),期】2018(050)004【总页数】3页(P245-247)【关键词】LKB1;非小细胞肺癌;表达意义【作者】孙万仆【作者单位】河南省濮阳市人民医院病理科,河南濮阳457000【正文语种】中文【中图分类】R734.2肺癌是世界上最常见的恶性肿瘤之一,目前在我国城市人口恶性肿瘤死亡的原因中已位居首位[1]。

非小细胞肺癌占所有肺癌的80%,与小细胞肺癌相比,恶化、扩散相对缓慢,但75%的患者在发现时,疾病已进展至中晚期[2-3]。

LKB1蛋白是LKB1基因的编码产物,是一种丝氨酸/苏氨酸激酶,与多种细胞的生理病理过程关系紧密,LKB1基因失活突变还会导致癌症易感病皮杰氏综合征的发生[4]。

慢病毒介导的LKB1基因在子宫内膜癌HEC-1A细胞中的表达

慢病毒介导的LKB1基因在子宫内膜癌HEC-1A细胞中的表达

慢病毒介导的LKB1基因在子宫内膜癌HEC-1A细胞中的表达唐乔乔;宋玥;龙颖;张洁清;宋红林;赵冰冰【摘要】Objective To investigate lentivirus-mediated LKB1 gene overexpression in HEC-1A endometrial cancer cells as a basis for future research. Methods The LKB1 gene was subcloned by RT-PCR from a cDNA plasmid into a lentiviral pWPI expression vector,generating the plasmid LKB1/pWPI.This plasmid was co-transfected into 293T cells together with the packaging plasmid pCMV-Dr8.74 and pMD2.G.The recombinant virus was used to infect HEC-1A cells,and LKB1 expression was measured using real-time quantitative PCR (qRT-PCR) and Western blotting. Results We succeeded in constructing the recombinant plasmid LKB1/pWPI and packaging it into lentiviral particles in 293T cells.This virus infected HEC-1A cells efficiently,leading to higher LKB1 expression than in the parental cells or untransfected controls (P<0.01). Conclusion The LKB1 gene has been incorporated into a lentiviral expression vector,allowing studies of its effects on the development of endometrial cancer.%目的:探讨慢病毒系统介导的LKB1基因在子宫内膜癌HEC-1A细胞中的过表达,为进一步研究LKB1基因在子宫内膜癌的作用机制奠定基础。

lkb1蛋白质结构

lkb1蛋白质结构

lkb1蛋白质结构LKB1蛋白质结构LKB1(Liver Kinase B1)是一种蛋白质激酶,它在细胞内起着重要的调控作用。

LKB1蛋白质结构是十分复杂的,由多个功能区域组成,每个区域都具有不同的功能和相互作用。

本文将从结构的层面来探讨LKB1蛋白质的组成和功能。

LKB1蛋白质由430个氨基酸组成,分子量为48千道尔顿。

它主要由一个N端的Catalytic区域和一个C端的Binding区域组成。

Catalytic区域包含了一个激酶结构域,这个结构域是LKB1蛋白质的核心功能区域,能够催化底物的磷酸化反应。

Binding区域则包含了多个结合蛋白的结构域,通过与这些蛋白相互作用,LKB1蛋白质能够参与多种信号转导通路的调控。

LKB1蛋白质结构的研究表明,它具有一个保守的激酶结构域,该结构域包含了一个ATP结合位点和一个底物结合位点。

这些结构域的存在使得LKB1具有催化底物磷酸化的能力。

此外,LKB1蛋白质的Catalytic区域还与其他蛋白质相互作用,形成复合物,进一步调控LKB1的活性。

这些蛋白质包括STRAD(STE20-related kinase adaptor protein)和MO25(Mouse protein 25),它们能够增强LKB1的激酶活性。

LKB1蛋白质的Binding区域也具有重要的功能。

它能够与多个蛋白质结合,参与细胞内的信号传导。

其中,LKB1与STRAD和MO25形成复合物,这个复合物在细胞内起着重要的调控作用。

此外,LKB1的Binding区域还能够结合其他信号分子,如AMPK (AMP-activated protein kinase),以及多种细胞内的信号通路蛋白。

通过这些结合,LKB1能够调控AMPK的活性,进而影响细胞的能量代谢和生长。

除了上述功能区域外,LKB1蛋白质还具有其他重要的结构域。

例如,LKB1蛋白质还包含一个Leucine-rich repeat区域,该区域可能参与蛋白质相互作用和信号传导。

肺癌中LKB1蛋白表达的研究

肺癌中LKB1蛋白表达的研究

肺癌中LKB1蛋白表达的研究朱延玲;程元光;黄大可【摘要】Objective To study the expression of LKB1 protein in lung cancer in order to investigate the role of LKB1 protein in the pathogenesis and progression in lung cancer. Methods Immunohistochemical staining was performed with LKB1 antibody to the surgically resected tissue samples from 34 patients with lung cancer, and relation was analysed between the expression of LKB1 and clinical characteristics of lung cancer. Results In the experiment of attachment, the mean OD value of the experimental group ( 0.273 ± 0.056 ) was significantly lower than that of the controls ( 0. 369 ±0.022 ). The expression level of LKB1 protein in lung cancer was significant lower than that in normal con-trol groups( P <0.05 ). In lung cancer, the expression of LKB1 protein was associated with the differentiated degree ( t =2. 364,P =0.033 ), as well as extent of lymph node metastasis( t =2. 181,P=0. 047 ). Conclusion Loss expression of LKB1 may play an important role in the pathogenesis and development of lung cancer, and this may be a crucial reference marker for the prognosis of patients with lung cancer, and a target point for gene therapy.%目的研究肿瘤抑制基因LKB1蛋白在肺癌中的表达.方法应用免疫组化方法检测肿瘤抑制基因LKB1蛋白在34例肺癌组织中的表达并结合肺癌的临床病理学特征进行分析.27例非肿瘤患者肺组织作对照.结果实验组34例LKB1蛋白阳性表达的平均光密度值为(0.273±0.056),对照组27例平均光密度值为(0.369±0.022),正常对照组表达强度显著高于实验组,两者比较差异有统计学意义(t=5.177,P=0.000);在肺中,LKB1蛋白的表达与肿瘤的恶性程度有显著的相关性(t=2.364,P=0.033),并且和淋巴结阳性与否关系密切(t=2.181,P=0.047).结论 LKB1蛋白的表达下降可能在肺癌的发生、发展、转移中起重要作用,有可能成为肺癌诊断的参考指标和基因治疗的重要靶点.【期刊名称】《安徽医学》【年(卷),期】2012(033)003【总页数】3页(P257-259)【关键词】肺癌;LKB1蛋白;免疫组化【作者】朱延玲;程元光;黄大可【作者单位】230601,合肥,安徽医学高等专科学校;230061,合肥市第一人民医院;230022,合肥,安徽医科大学基础医学院综合实验室【正文语种】中文肺癌是世界范围内发病率和病死率较高的恶性肿瘤之一,我国肺癌的发病率已高达61.4/10万,且发病率和病死率近年来呈上升趋势[1]。

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

Metformin is one of the most widely used antidiabetic agents for type 2 diabetes mellitus. In the UK Prospective Diabetic Study, metformin significantly reduced the incidence of death and cardiovascular events in obese type 2 diabetic patients compared with patients treated with other glucose-lowering reagents but with the same glycemic control,1 suggesting that metformin may have unique protective effects against diabetes-related cardiovascular complications. In addition, metformin is shown to improve vascular endothelial function in diabetic humans.2–7 Although it has been in clinical use for decades, the precise mechanism of action of metformin has yet to be established. Recent studies8,9 demonstrate that metformin activates AMP-activated protein kinase (AMPK) in vitro and in vivo. Furthermore, we 10,11found that clinically relevant concentrations of metformin activated AMPK in cultured endothelial cells and aortas in vivo and ascribed the beneficial effects of metformin on vascular function to AMPK activation. In addition, activation of AMPK by metformin is required for decreased glucose production and increased fatty acid oxidation in hepatocytes9 and for increased glucose uptake in skeletal muscle.8AMPK is a serine/threonine kinase and an evolutionary conserved regulator of the cellular response to low energy.12,13 It is activated when nutrient supply is limited, ATP generation is impaired, or cellular energy demand is increased.13,14 AMPK has a critical role in many metabolic processes, and, once activated, AMPK phosphorylates a number of proteins that result in increased glucose uptake and metabolism as well as fatty acid oxidation. In addition,AMPK simultaneously inhibits hepatic lipogenesis, cholesterol synthesis, and glucose production 15 and is also responsible for increased fatty acid oxidation in response to the adipocyte-derived hormones leptin 16,17 and adiponectin.18 Phosphorylation of Thr172 in the activation loop of the catalytic α subunit is a prerequisite of significant kinase activity, and an increase in AMP/ATP ratios further allosterically stimulates the enzyme, resulting in 1000-fold activation.Recently, at least 2 upstream kinases, LKB1 and calcium/calmodulin-dependent kinase kinase(CaMKK)-β, have been identified as AMPK kinases.19–24 LKB1, a 50-kDa serine/threoninekinase, was originally identified as the product of the gene mutated in the autosomal dominantlyinherited Peutz-Jeghers cancer syndrome.25 In resting cells, LKB1 is reported to bepredominantly located in the nucleus,26–28 and, like AMPK, LKB1 forms a heterotrimericcomplex with regulatory proteins termed STRAD and MO25, which are required for itsactivation and cytosolic localization.29 LKB1 has been shown to mediate Thr172phosphorylation of AMPK both in vitro and in intact cells.19,21,23The mechanism by which metformin activates AMPK remains controversial, although earlierstudies 30,31 demonstrate that metformin might activate AMPK by decreasing cellular energyby acting as an inhibitor of complex I of the respiratory chain. However, several recentstudies 32,33 argue against this notion because in these studies metformin activates AMPKwithout affecting the AMP/ATP ratio. Interestingly, metformin neither activates AMPK in acell-free assay nor influences the phosphorylation of AMPK by LKB1 in vitro. Furthermore,it was found that in cell lines such as HeLa-S3 cells and A549 cells, which lack LKB1expression, or in LKB1−/− mouse embryonic fibroblasts, AMPK could not be activated by avariety of agonists and stresses, including metformin.19,23 Mice lacking LKB1 expression havesignificantly inhibited AMPK activity and blunted increases in glucose transport caused byelectrically stimulated muscle contraction and the AMPK-activating compound 5-aminoimidazole-4-carboxamide 1-β-D-ribofuranos-ide (AICAR) and phenformin.34Importantly, with the use of mice that have LKB1 deleted in liver, it has been reported thatmetformin required LKB1 in the liver to lower blood glucose.35 Although these studies haveestablished the critical role of LKB1 in AMPK activation, none of these studies have elucidatedthe upstream signaling that controls LKB1-dependent AMPK activation. In the present study,NIH-PA Author ManuscriptNIH-PA Author ManuscriptNIH-PA Author Manuscriptwe have found that metformin activates AMPK by activating PKC-ζ–dependent LKB1phosphorylation at Ser428, a site that is required for LKB1 nuclear export into the cytosol andits association with AMPK.Methods A full description of methods, including methods such as protein kinase assays for PKC-ζ,AMPK, and LKB1, immunocytochemical staining of LKB1, and preparation of subcellular fractions, can be found in the online-only Data Supplement.Cell Culture and Treatment Human umbilical vein endothelial cells (HUVEC) were cultured in medium 200 supplemented with low-serum growth supplements. Bovine aortic endothelial cells (BAEC) were maintained in endothelial basal medium with 2% serum and growth factors before use. After they reached confluence, BAEC were serum deprived overnight before the experiments. HeLa-S3 and A549cells were grown in F-12K medium supplemented with 10% serum. All culture media were supplemented with penicillin (100 U/mL) and streptomycin (100 μg/mL). The cells were treated with 1 mmol/L metformin for 1 hour after adenoviral infection or pretreatment with protein kinase inhibitor.Plasmid Transfection and Adenovirus Infection Site-directed mutagenesis of LKB1 was performed as described previously.36 Mutations were confirmed by DNA sequencing, and plasmid DNA was extracted on a large scale with the use of Qiagen EndoFree plasmid maxi kit (catalogue No. 12362) and transfected into HeLa-S3with the use of the Lipofectamine 2000 kit from Invitrogen (catalogue No. 11668-019),according to the instructions provided by the supplier. Twenty-four hours after transfection,the cells were treated with metformin (1 mmol/L) or vehicle for 1 hour. Both LacZ expression vector and untreated cells were used as control.BAEC or HUVEC were infected with adenovirus expressing a wild-type PKC-ζ (PKC-ζ-WT)or a dominant negative mutant PKC-ζ (PKC-ζ-DN). A replication-defective adenoviral vectorexpressing green fluorescence protein (ad-GFP) was used as control. The AMPK-DNadenoviral vector was constructed from AMPK-α2 bearing a mutation altering lysine 45 toarginine (K45R) as described previously.10,11 Cells were infected with the adenovirus at amultiplicity of infection of 100 in serum-free medium overnight. Cells were then washed andincubated in fresh medium without serum for an additional 18 to 24 hours beforeexperimentation. Under these conditions, infection efficiency was typically >75% asdetermined by GFP expression.Statistical AnalysisValues are expressed as mean±SE. All of the data were analyzed with a 1-way ANOVAfollowed by Bonferroni post hoc analyses, except for those obtained from the time course,which were analyzed with repeated-measures ANOVA. A P value of <0.05 is consideredstatistically significant.The authors had full access to and take full responsibility for the integrity of the data. Allauthors have read and agree to the manuscript as written.NIH-PA Author ManuscriptNIH-PA Author ManuscriptNIH-PA Author ManuscriptResultsMetformin Induces AMPK and Acetyl Coenzyme A Carboxylase PhosphorylationBecause AMPK activation requires the phosphorylation of Thr172 in the activation loop of α1 and α2 subunits,37,38 AMPK activity was determined in Western blots by monitoring the phosphorylation of both AMPK at Thr172 and its best-characterized downstream kinase, acetyl coenzyme A carboxylase (ACC) at Ser79, by using specific antibodies. AMPK activation was further confirmed by AMPK activity assayed as 32P incorporation into SAMS (HMRSAMSGLHLVKRR) peptides. Confluent HUVEC and BAEC were treated with metformin (1 mmol/L) for 1 hour. As shown in Figure 1A, metformin markedly increased the detection of AMPK-Thr172 and ACC at Ser79 in both HUVEC and BAEC, respectively.Because metformin activated AMPK in both BAEC and HUVEC at a similar potency, we performed most of the experiments in BAEC.Inhibition of PKC-ζ Attenuates Metformin-Enhanced AMPK-Thr172 Phosphorylation We have previously shown that inhibition of PKC-ζ attenuates ONOO −-enhanced AMPK activation in BAEC.36 To establish PKC-ζ as a mediator for metformin-induced activation of AMPK, we first determined whether PKC-ζ–specific pseudosubstrate peptide (PKC-ζ-PS), a synthetic peptide that selectively inhibits PKC-ζ without affecting other PKC isoforms, altered the effects of metformin on AMPK-Thr172 and ACC-Ser79 phosphorylation. As depicted in Figure 1B, PKC-ζ-PS significantly ablated metformin-enhanced phosphorylation of both AMPK-Thr172 (n=5; P <0.01) (Figure 1B and 1C) and ACC-Ser79 (Figure 1B). The inhibitory effect of PKC-ζ-PS was also confirmed by the suppression of AMPK activity. As depicted in Figure 1D, PKC-ζ-PS dose-dependently suppressed metformin-enhanced AMPK activity (Figure 1D), as assayed by the SAMS peptides.Additional evidence for PKC-ζ–dependent AMPK activation was obtained from genetic inhibition of PKC-ζ. As depicted in Figure 1E and 1F, adenoviral overexpression of eitherGFP, PKC-ζ-DN, or PKC-ζ-WT did not alter the basal levels of AMPK in BAEC. However,overexpression of PKC-ζ-DN, but not GFP, abolished the effects of metformin on both AMPKand ACC, whereas overexpression of PKC-ζ-WT significantly enhanced metformin-enhancedphosphorylation of both AMPK-Thr172 and ACC-Ser79 (Figure 1E and 1F) and AMPKactivity (Figure 1G).Activation of PKC-ζ by MetforminWe next determined whether metformin activated PKC-ζ in BAEC. The phosphorylation ofPKC-ζ at Thr410/403 by upstream kinases such as phosphoinositide 3-kinase (PI-3 kinase)/PDK-1 axis and translocation of PKC-ζ from the cytosol into cytoplasmic membrane areconsidered critical steps in the activation of PKC-ζ. Thus, PKC-ζ phosphorylation wasmonitored in total cell lysates in Western blots by using specific antibodies. As shown in Figure2A, metformin increased PKC-ζ Thr410/403 phosphorylation without altering PKC-ζexpression. Inhibition of PKC-ζ with PKC-ζ-PS abolished metformin-induced PKC-ζphosphorylation (Figure 2A), indicating a specific inhibition by PKC-ζ-PS.We next assayed PKC-ζ activity by using 32P incorporation in PKC-ζ–specific peptides. Asexpected, overexpression of PKC-ζ-DN, but not GFP, attenuated PKC-ζ activity, whereasoverexpression of PKC-ζ-WT increased PKC-ζ activity in BAEC (Figure 2B). In agreementwith increased PKC-ζ phosphorylation at Thr410/403 (Figure 2A), metformin significantlyincreased PKC-ζ activity in BAEC or BAEC infected with GFP. Overexpression of PKC-ζ-DN abolished metformin-enhanced PKC-ζ activation, whereas PKC-ζ-WT increased PKC-ζactivity (Figure 2B). These results implied that metformin activated PKC-ζ.NIH-PA Author ManuscriptNIH-PA Author ManuscriptNIH-PA Author ManuscriptThe translocation of PKC-ζ is considered a critical step in PKC-ζ activation. Exposure of BAECto metformin significantly increased the presence of PKC-ζ in membrane fractions but loweredthe amount of PKC-ζ in the cytosol (Figure 2C and 2D). In parallel, metformin also increasedthe translocation of PKC-ζ from the cytosol into the nuclei (Figure 2E and 2F). The purity ofthese subcellular fractions was confirmed by using antibodies against specific protein markerenzymes39,40 of the cytosol (lactate dehydrogenase), plasma membrane (alkalinephosphatase), or nucleus (histone H2AX), respectively. The nuclear histone H2AX wasdetected only in the nuclear fraction but not in cytosolic or membrane fractions (Figure 2G).Lactate dehydrogenase was detected only in the cytosolic fraction, whereas alkalinephosphatase was found only in the membrane fraction (Figure 2G). Thus, metformin causedcellular redistribution of PKC-ζ from the cytosol to nuclei and membranes.Protein Kinase A and p90 Ribosomal S6 Kinase Are Not Required in Metformin-Enhanced AMPK ActivationBecause both protein kinase A (PKA) and p90 ribosomal S6 kinase (RSK) are reported tophosphorylate LKB1 at Ser428, we next determined whether PKA or RSK was involved inmetformin-enhanced phosphorylation of AMPK-Thr172 or LKB1-Ser428. As depicted inFigure 3A and 3B, administration of either H89 (10 μmol/L), a selective PKA inhibitor, orSL0101 (10 μmol/L), a selective RSK inhibitor, did not alter metformin-enhancedphosphorylation of AMPK-Thr172 or LKB1-Ser428 (Figure 3A and 3B). In contrast toincreased PKC-ζ phosphorylation, metformin affected neither PKA catalytic subunit (PKAc)phosphorylation at Thr197 nor RSK3 phosphorylation at Thr356/Ser360 (Figure 3C),suggesting that metformin had no effect on PKA or RSK. Further evidence comes fromcoimmunoprecipitation experiments. As shown in Figure 3D, LKB1 wascoimmunoprecipitated with PKC ζ but not with either PKA or RSK. Furthermore, metforminmarkedly increased the coimmunoprecipitation of LKB1 with PKC-ζ (Figure 3D), which wassensitive to PKC-ζ inhibition (Figure 3D), suggesting that PKC-ζ activity was required for theassociation of PKC-ζ with LKB1. Indeed, alteration of PKC-ζ activity by overexpression ofeither PKC-ζ-DN or PKC-ζ-WT did not alter the phosphorylation of PKAc or RSK (Figure3E), suggesting that the effect of PKC-ζ was independent of PKA or RSK in BAEC. Takentogether, these results suggest that metformin-enhanced phosphorylation of LKB1 and AMPKwas PKC-ζ dependent but independent of PKA or RSK.Metformin-Enhanced AMPK Activation Requires LKB1At least 2 upstream kinases, including LKB1 and CaMKK-β, have been identified as AMPKkinases.19–24 We next determined whether metformin required LKB1 to activate AMPK inBAEC. As depicted in Figure 4A and 4B, overexpression of the kinase-dead LKB1 mutantLKB1-D194A abolished metformin-enhanced phosphorylation of both AMPK-Thr172 andACC-Ser79 in BAEC. In contrast, overexpression of LKB1-WT enhanced the effects ofmetformin on the phosphorylation of both AMPK and ACC. Because overexpression of GFPdid not alter the effect of metformin, these results suggested that LKB1 was required formetformin-enhanced AMPK activation in endothelial cells.Metformin-Enhanced LKB1-Ser428 Phosphorylation Is PKC-ζ DependentWe have previously demonstrated that ONOO − enhances the phosphorylation of LKB1 atSer428 via PKC-ζ.36 We next determined whether metformin altered the phosphorylation ofLKB1 at Ser428. As depicted in Figure 4C and 4D, metformin significantly increased thephosphorylation of LKB1 at Ser428. Overexpression of PKC-ζ-DN, which ablated metformin-enhanced AMPK activation (Figure 1E through 1G), abolished metformin-enhanced LKB1-Ser428 phosphorylation (Figure 4C and 4D). Conversely, overexpression of GFP or PKC-ζ-WT did not suppress metformin-enhanced LKB1-Ser428 phosphorylation (Figure 4C and 4D).NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author ManuscriptHowever, alteration of PKC activity by overexpression of PKC-ζ-DN or PKC-ζ-WT did notalter LKB1 activity (Figure 4E). In addition, consistent with our previous results,34,36,41,42metformin did not alter LKB1 activity in the cells infected with either PKC-ζ-DN or PKC-ζ-WT (Figure 4E). Thus, changes of AMPK-Thr172 phosphorylation cannot be attributed toaltered kinase activity of LKB1.Metformin-Enhanced PKC-ζ Phosphorylation Occurs Before the Phosphorylation of Both LKB1 and AMPKWe next determined the time course of metformin-enhanced phosphorylation of PKC-ζ, LKB1,and AMPK. As shown in Figure 4F and 4G, metformin significantly increased the PKC-ζphosphorylation within 1 minute. The phosphorylation of LKB1-Ser428 was significantlyelevated ≈10 minutes after metformin treatment, whereas a significant increase of AMPK-Thr172 phosphorylation was observed at 30 minutes and reached a peak at 60 minutes (Figure4F and 4G). These data suggest a chronology of events, with metformin rapidly activating firstPKC (Thr410/403), then LKB1 (Ser428), and finally AMPK (Thr172).Metformin Triggers LKB1 Translocation From Nucleus Into CytoplasmEarlier works 27,43–45 had indicated that LKB1 is localized mainly in the nucleus, whereasAMPK is localized mainly in the cytoplasm. LKB1 must be exported from the nucleus intocytosols where AMPK is located. We first used immunohistochemical staining to assaywhether metformin altered the subcellular localization of LKB1 in HUVEC because allcommercially available antibodies against LKB1 could not be used in BAEC. As expected,LKB1 was found mainly in the nucleus of nonstimulated HUVEC (Figure 5A). In metformin-treated HUVEC, LKB1 was detected mainly in the cytosols. Metformin-enhanced LKB1nuclear export was further confirmed by Western blot analysis of LKB1 in subcellular fractions.As shown in Figure 5B and 5C, metformin significantly increased the protein amount of LKB1in the cytosol, whereas it was markedly reduced in the nucleus of HUVEC (Figure 5A through5C). Similarly, LKB1 was predominantly located in the nuclear fractions in BAEC. Exposureof BAEC to metformin reduced the amount of LKB1 in the nuclear fraction, whereas itincreased the amount of LKB1 in the cytosols (data not shown). Interestingly, the signalscorresponding to LKB1-Ser428 phosphorylation were significantly increased in both cytosoland nuclei of metformin-treated BAEC, whereas the reduced nuclear LKB1 protein contentafter metformin treatment (Figure 5A through 5C) corresponded to a relatively high LKB1-Ser428 signal (Figure 5D and 5E). These data suggest that LKB1 phosphorylation might occurin the nucleus, resulting in LKB1 translocation from nuclear to cytoplasmic locations.Mutation of LKB1 Serine 428 With Alanine Prevents Metformin-Enhanced LKB1TranslocationWe next determined how metformin increased LKB1 translocation. With the use of site-directed mutagenesis techniques, Ser428 of LKB1 was mutated into alanine (LKB1-S428A,loss of function). In addition, Asp194 of LKB1, which is essential for LKB1 activity, was alsoreplaced with alanine, resulting in a kinase-dead LKB1 mutant (LKB1-D194A). Theseplasmids were transfected into LKB1-deficient A549 cells. After transfection, the subcellulardistributions of LKB1 were detected by using a mouse anti-His Tag antibody. As expected,A549 transfected with Lac-Z did not react with an anti-His Tag antibody (data not shown).Like LKB1 in HUVEC, LKB1 was found mainly in the nucleus of A549 transfected with eitherLKB1 WT, S428A mutants, or D194A mutants (Figure 6A). Metformin markedly increasedLKB1 staining in the cytosol of A549 transfected with LKB1-WT but not in A549 transfectedwith either LKB1-D194A or LKB1-S428A mutants (Figure 6A). Western blot analysis ofLKB1 in the subcellular fractions confirmed that LKB1-WT or LKB1 mutants were all nuclearproteins and that metformin significantly increased the amount of cytosolic LKB1 in A549NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscriptcells transfected with LKB1-WT but not LKB1 mutants (Figure 6B). These results suggest thatthe Ser428 phosphorylation of LKB1 as well as LKB1 activity was required for metformin-enhanced nuclear export.Mutation of Ser428 Into Alanine Abolishes Metformin-Enhanced AMPK ActivationBecause LKB1-Ser428 mutation altered its subcellular localization, we next determinedwhether Ser428 mutation of LKB1 also altered metformin-enhanced AMPK activation. Asshown in Figure 7A and 7B, metformin did not activate AMPK in either HeLa-S3 or HeLa-S3transfected with either LacZ or the D194A mutant of LKB1, confirming the essential role ofLKB1 in metformin-enhanced AMPK activation. Conversely, transfection of LKB1-WTsignificantly increased the phosphorylation of AMPK and ACC in HeLa-S3 cells transfectedwith WT LKB1 (Figure 7A and 7B). Interestingly, the point mutation of LKB1-Ser428 intoalanine, like the kinase-inactive mutant D194A, abolished metformin-enhancedphosphorylation of AMPK. Similarly, overexpression of LKB1-S428A and LKB1-D194Amutants ablated peroxynitrite (ONOO −, 100 μmol/L)-enhanced phosphorylation of AMPK,whereas the mutation of Ser428 into aspartic acid (LKB1-S428D), which mimicsphosphorylation of Ser428, increased AMPK phosphorylation as LKB1-WT did (Figure 7C).We next assayed the activity of mutant LKB1 with Ser428 (Ser431 in mouse) altered to alaninein comparison to WT LKB1, using an artificial peptide substrate. As shown in Figure 7D,LKB1-Ser431A mutants exhibited a 10% reduction of LKB1 activity compared with LKB1WT.We next determined whether LKB1-Ser428 phosphorylation increased LKB1 activity. MouseLKB1-WT or LKB1-S431A (equivalent to human 428) mutants were coexpressed with bothMO25α and STRAD α in bacteria. Partially purified LKB1 WT or LKB1-S431A complex wascoincubated with recombinant PKC-ζ. PKC-ζ–dependent LKB1 phosphorylation and LKB1activity were assayed by using LKB1tide or p53. PKC-ζ increased 32P incorporation in LKB1WT paralleled with a 10% increase in LKB1 activity but had less effect on LKB1-S431Amutants. Only LKB1, but not STRAD α or MO25α, was phosphorylated by PKC-ζ asdetermined by 32P autoradiographic gels when PKC-ζ was incubated with LKB1-MO25α-STRAD α complex in vitro (data not shown). Because LKB1 S431A mutants expressed a 10%reduction of LKB1 activity, whereas PKC-ζ caused a 10% increase of LKB1 activity, theseresults suggest that the phosphorylation of LKB1 at Ser428/431 might increase LKB1 activityby 10%. However, such rather small increases of LKB1 activity would only partly explain thestrongly elevated AMPK activity after metformin treatment, therefore emphasizing theimportance of the nucleocytoplasmic redistribution of LKB1 that is expected to increaseAMPK activation in the cytosol.Mutation of Ser428 Into Alanine Abolishes Metformin-Enhanced Association of LKB1 With AMPKWe have previously shown that ONOO − activates AMPK by increasing the association ofLKB1 and AMPK.36 In addition, we found that PKC-ζ coimmunoprecipitated with LKB1 andmetformin increased the association of LKB1 with PKC-ζ. We next determined whetherphosphorylation of LKB1-Ser428 by PKC-ζ contributed to metformin-enhanced associationof AMPK with LKB1 and therefore whether LKB1 was immunoprecipitated from BAEC andstained with AMPK in Western blots. As shown in Figure 7E, metformin showed significantlyincreased coimmunoprecipitation of LKB1 with AMPK α1 or AMPK α2. Importantly,inhibition of PKC-ζ blunted metformin-enhanced association of LKB1 with AMPK (Figure7E), suggesting that PKC-ζ enhances the association of LKB1 with AMPK.NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author ManuscriptWe next investigated whether mutations of LKB1-Ser428 into alanine altered the interactionof AMPK and LKB1 in HeLa-S3 cells. Metformin increased the association of AMPK andLKB1 in HeLa-S3 cells transfected with LKB1 WT but not in HeLa-S3 transfected with LacZor LKB1 mutants (Figure 7F), implying that LKB1-Ser428 phosphorylation is required formetformin-enhanced AMPK activation.Discussion In the present study, we have established that LKB1-Ser428 phosphorylation by atypical PKC-ζ plays an essential role in metformin-enhanced AMPK activation. LKB1-Ser428phosphorylation is required for both its nuclear export of LKB1 into cytosols, in which it forms an active complex with STRAD and MO25 and consequent AMPK phosphorylation. Because various stimuli increased the phosphorylation of LKB1 at Ser428, it is tempting to speculate that LKB1-Ser428 phosphorylation might be a common pathway required for AMPK activation.LKB1 localization depends on its kinase activity, import into the nucleus, and retention within cytoplasm.27,29 It has been reported that cytoplasmic localization of LKB1 is critical for its normal function.27,29 Several residues including Ser31, Thr336, Thr366, and Ser 428 of LKB1have been identified as phosphorylation sites, although their physiological functions are largely unknown. In the present study, we have provided evidence that metformin induced AMPK activation by phosphorylating LKB1 at Ser428, which results in translocation of LKB1 from the nucleus to the cytosol and increased association of LKB1 with AMPK. The key findings can be summarized as follows: metformin increases LKB1 phosphorylation at Ser428, induces LKB1 translocation from nucleus to cytoplasm, and enhances coimmunoprecipitation of LKB1with AMPK. Mutation of Ser428 into alanine prevented translocation of LKB1 from the nucleus to the cytoplasm and abolished metformin-enhanced association of LKB1 with AMPK,thus abolishing AMPK activation. The LKB1-S428A mutation did not affect normal nuclearlocalization of LKB1 but prevented LKB1 translocation to the cytoplasm after metformintreatment, suggesting that phosphorylation of Ser428 might be essential for LKB1 translocationfrom the nucleus to the cytoplasm. Thus, Ser428 phosphorylation located in the C-terminus ofLKB1 might play a crucial role in regulating AMPK activation. In agreement with this notion,Forcet et al 46 demonstrated that C-terminal mutations impair LKB1-mediated activation ofAMPK and downstream signaling but do not affect LKB1 autokinase activity or abolish LKB1-induced growth arrest. Our data indicate that mutation of Ser428 of LKB1 to alanine impairedits ability to export from nucleus and subsequent association with AMPK after metformintreatment (Figure 7F), suggesting that phosphorylation of Ser428 might contribute to itsbinding to AMPK and that mutations of this site into alanine uncouple this process, therebyresulting in reduced enzyme-substrate recognition. These results are in contrast with theprevious report that mutation of Ser431 to either alanine to abolish phosphorylation or toglutamate to mimic phosphorylation did not significantly affect the cellular localization orcatalytic activity of LKB1 in vitro.47 The reasons for the discrepancy are unknown and mightbe related to the species differences (human versus mouse) or cell types. In vivo, LKB1 formsa heterotrimeric complex with 2 proteins termed STRAD and MO25.29,48 STRAD is anLKB1-specific adaptor and substrate; when binding to the kinase domain of LKB1, STRADenhances LKB1 activity >100-fold and anchors LKB1 in the cytosol. MO25 stabilizes thebinding of STRAD to LKB1 and cooperates with STRAD to localize LKB1 in the cytoplasmof cells.49 In addition, LKB1 can also be localized in the cytoplasm of cells through aninteraction with a cytosolic protein called LIP1 (LKB1 interacting protein-1).50 It is reportedthat in muscle-specific LKB1 knockout mice, AICAR, phenformin, or muscle contraction didnot increase AMPK α2 activity in the skeletal muscle, indicating that LKB1 is the majorupstream kinase of AMPK.34However, these 3 treatments failed to increase LKB1 activity.41 In our study, we found that metformin increased AMPK phosphorylation and activity.NIH-PA Author ManuscriptNIH-PA Author ManuscriptNIH-PA Author ManuscriptAlthough it did not change LKB1 activity to a great extent, metformin markedly enhanced LKB1 phosphorylation at Ser428; mutation of Ser428 into alanine prevented translocation of LKB1 from nucleus to cytoplasm and abolished metformin-enhanced association of AMPK with LKB1, thus abrogating AMPK activation. Theses results demonstrate that the phosphorylation of LKB1 at Ser428 plays a crucial role in AMPK activation by altering its cellular location or by enabling it to interact with regulatory proteins or protein substrates. In addition, the active form of LKB1 might be as important as Ser428 phosphorylation because the mutation of LKB1 D194 into alanine prevented the translocation of the enzyme. Because LKB1 S431A mutants expressed a 10% reduction of LKB1 activity, whereas PKC-ζ caused a 10% increase of LKB1 activity, metformin-enhanced AMPK activity is likely via the nucleocytoplasmic redistribution rather than an increase of LKB1 activity as a result of LKB1-Ser428 phosphorylation. Moreover, it remains to be determined whether other sites are required for LKB1 translocation.We have previously reported 10,11,51,52 that activation of PI-3 kinase by metformin or ONOO − might enhance AMPK activity by increasing the association of LKB1 with AMPK.Indeed, inhibition of c-Src or PI-3 kinase activity by pharmacological inhibition or dominant negative mutants abolishes metformin- or ONOO −-enhanced AMPK activation in endothelial cells.10,11,53,54 In the present study, we not only confirm but also extend these observations by demonstrating that PKC-ζ, a member of the PI-3 kinase AGC family, plays a critical role in metformin-induced AMPK activation. Indeed, it has been shown that some AMPK agonists,such as AICAR and dinitrophenol, increase PKC-ζ activity in isolated extensor digitorum longus muscle and L6 myotubes.55 In type 2 diabetic patients, metformin increases PKC-ζactivity, accompanied by increased AMPK activity.56 We have also demonstrated that PKC-ζ phosphorylates LKB1 at Ser428 in both in vitro assays and in cultured cells, including endothelial cells, pericytes, adipocytes, and cultured vascular smooth muscle cells.36Furthermore, inhibition of PKC-ζ attenuates AMPK activation by other stimuli (Z.X.,unpublished data, 2008). Because other kinases, including cAMP-dependent PKA and p90RSK, are reported to phosphorylate LKB1 at Ser428, the Ser428 phosphorylation of LKB1might be a common pathway for AMPK activation triggered by signaling pathways other thanthe c-Src/PI-3 kinase axis (Figure 8).To summarize, we provide evidence that PKC-ζ plays a critical role in regulating AMPKactivity. Our data suggest that metformin-induced AMPK activation requires LKB1 anddemonstrate that phosphorylation of Ser428 of LKB1 could affect cellular location of LKB1and its interaction with AMPK. We conclude that PKC-ζ can regulate AMPK activity byincreasing LKB1 phosphorylation, resulting in LKB1 nuclear export and consequent AMPKThr172 phosphorylation by LKB1. In addition, the Ser428 phosphorylation of LKB1 might bea common pathway for AMPK activation triggered by signal pathways, including PI-3 kinase/PKC-ζ axis, PKA, and RSK.NIH-PA Author ManuscriptNIH-PA Author ManuscriptNIH-PA Author Manuscript。

相关文档
最新文档