新型蛋白质氧化折叠助剂的分子设计和复性方法研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
蛋白质复性是应用基因重组技术在菌体中生产贵重蛋白质的瓶颈。本文针对蛋白质复性过程的两个关键因素,二硫键形成和蛋白质聚集抑制,开展蛋白质氧化折叠助剂的分子设计和复性方法研究。
     针对二硫键形成问题,开展蛋白质氧化折叠动力学研究,揭示二硫键形成是折叠过程中色氨酸包埋的前提条件,发现混合二硫键的形成是控制第一折叠相折叠速率的关键因素。
     在此基础上开展氧化折叠助剂的分子设计。首先考察蛋白质二硫键异构酶(PDI)和DsbA的结构特性,发现其活性位点周围具有疏水性表面。因此,设计具有疏水尾的小分子折叠酶模拟物酰基胱胺作为新型氧化折叠助剂分子。研究表明,酰基胱胺能够在强还原环境下有效地促进蛋白质氧化复性,这是其它任何氧化剂所不能做到的。并且,酰基胱胺能够极大地提高蛋白质氧化复性速率,在较低浓度下依然有效,仅需胱胺浓度的一半即可获得相同的复性效果。其作用机理研究发现,酰基胱胺通过疏水烷基链介导与变性蛋白质之间形成疏水相互作用,获得其促进复性的显著效果。
     其次,研究发现PDI的模拟分子氧化型寡肽CGC作为氧化折叠助剂可显著提高蛋白质氧化复性的速率和收率。以此为基础设计新型氧化折叠助剂分子RKCGC。研究表明,RKCGC能够与二硫苏糖醇很好地耦合,促进二硫键的形成和异构,具有较宽的适用pH范围。且与CGC相比,RKCGC能够更好地提高氧化复性的速率和收率。其作用机理研究发现,RKCGC具有更低的pKa值和更高的还原势,因而获得较好的辅助复性效果。
     针对蛋白质聚集问题,开展抑制聚集方法研究,发现与蛋白质带有同种电荷的离子交换介质能够很好地抑制折叠中间体聚集从而促进蛋白质复性。通过四种带不同电荷的离子交换介质对三种带不同电荷的蛋白质辅助复性研究,证实同电荷离子交换介质能够有效促进蛋白质复性,且在高蛋白质浓度(4 mg/mL溶菌酶;2 mg/mL牛血清白蛋白)条件其促进复性效果依然非常显著,而且不会降低复性速率,优于其它化学添加剂。其作用机理研究发现,同电荷离子交换介质通过静电斥力诱导蛋白质在其带电表面形成定向排列,从而抑制聚集。进而将该方法应用到重组增强型绿色荧光蛋白(EGFP)的复性中,发现EGFP包含体的复性收率提高近一倍,并且在辅助复性的同时还具有纯化的效果。
     在同电荷离子交换介质辅助复性的基础上拓展研究工作,提出同电荷聚电解质辅助蛋白质复性方法。通过比较聚电解质与离子交换介质的作用机制,揭示溶液中的存在形态是影响同电荷物质促进蛋白质复性效果的关键因素。
Disulfide fromation and protein aggregation are the two key factors influencing protein refolding. Herein, a series of agents and methods that can facilitate disulfide formation and inhibit protein aggregation were developed to facilitate protein refolding.
     To investigate the disulfide bond formation, protein oxidative folding kinetics were examined. It was shown that disulfide bond formation was a prerequisite for tryptophan burial. Besides, the results elucidated that the rate of the first folding phase is determined by the formation of mixed disulfide bond.
     Based on these results, novel oxidative folding aids were designed. Firstly, structural characteristic of protein disulfide isomerase (PDI) and DsbA were investigated. Hydrophobic regions were observed around their active sites. Based on this finding, hydrophobic alkyl tails were linked to cystamine to design a novel small molecular foldase mimics, acyl cystamine. Acyl cystamine was proven very effective to facilitate oxidative protein refolding at strong reducing environments, which can not be achieved by any other oxidants. Besides, Acyl cystamine can greatly increase protein folding rate and effectively facilitate protein folding at much lower concentration. To achieve the same refolding effect, the concentration of n-hexanoyl cystamine was only about half of cystamine. Further investigation was performed to explore the mechanism. It was shown that the specific hydrophobic interaction between the alkyl tail and unfolded peptide or folding intermediates make acyl cystamine strikingly better oxidative folding aids.
     Moreover, the disulfide form of a small peptide mimic of PDI, CGC, was explored to facilitate oxidative protein refolding. It was found that CGC increased both the folding rate and refolding yield. Based on this finding, a new pentapeptide, RKCGC was designed. It was confirmed that RKCGC can effectively facilitate protein folding with DTT, and assist both disulfide formation and isomerization. It was effective in a wide pH range. Meanwhile, RKCGC has better effect to increase the folding rate and yield as compared with CGC. Further investigation was perfomed to explore the mechanism. It was shown that the better effect of RKCGC than CGC was contributed by its lower pKa and higher reduction potential.
     The inhibition of protein aggregation during refolding was investigated. Herein we found that electrostatic repulsion between like-charged protein and ion exchange gel beads can greatly suppress the aggregation of folding intermediates, leading to the significant increase of native protein recovery. This finding was extensively demonstrated with three different proteins and four kinds of ion-exchange resins when the protein and ion-exchange gel are either positively or negatively charged at the refolding conditions. It is remarkable that the enhancing effect was significant at very high protein concentrations, such as 4 mg/mL lysozyme (positively charged) and 2 mg/mL bovine serum albumin (negatively charged). Moreover, the folding kinetics was not compromised by the presence of the resins, so fast protein refolding is realized at high protein concentrations. This was not realistic by any other approaches. The working mechanism of the like-charged resin is considered due to the charge repulsion that could induce oriented alignment of protein molecules near the charged surface, leading to the inhibition of protein aggregation. The method was further applied to the renaturation of enhanced green fluorescent protein (EGFP) inclusion bodies. Like-charged exchanger increased the refolding yield by almost 100%. Moreover, it has purification effect at the same time.
     Based on the research in protein refolding assisted by like-charged ion-exchange resin, a protein refolding method with like-charged polyelectrolyte as additive was developed. The working mechanisms of like-charged polyelectrolytes and ion exchangers revealed that the structure was the key factor influencing the effectiveness of like-charged agents in facilitating protein refolding.
引文
[1] Ventura S. Oxidative protein folding: from the test tube to in vivo insights. Antioxid Redox Signal, 2008, 10: 51-54.
    [2] Hartl FU, Hayer-Hartl M. Converging concepts of protein folding in vitro and in vivo. Nat Struct Mol Biol, 2009, 16: 574-581.
    [3]王君.表面活性剂辅助蛋白质复性过程机理研究[博士学位论文].北京:清华大学, 2004.
    [4] De Bernardez Clark E. Protein refolding for industrial processes. Curr Opin Biotechnol, 2001, 12: 202-207.
    [5] Goldberg ME, Guillou Y. Native disulfide bonds greatly accelerate secondary structure formation in the folding of lysozyme. Protein Sci, 1994, 3: 883-887.
    [6] Kucharski Timothy J, Huang Z, Yang Q-Z et al. Kinetics of thiol/disulfide exchange correlate weakly with the restoring force in the disulfide moiety. Angewandte Chemie International Edition, 2009, 48: 7040-7043.
    [7] van den Berg B, Chung EW, Robinson CV et al. Characterisation of the dominant oxidative folding intermediate of hen lysozyme. J Mol Biol, 1999, 290: 781-796.
    [8] Kiefhaber T, Rudolph R, Kohler H-H et al. Protein aggregation in vitro and in vivo: a quantitative model of the kinetic competition between folding and aggregation. Nat Biotech, 1991, 9: 825-829.
    [9] Goldberg ME, Rudolph R, Jaenicke R. A kinetic study of the competition between renaturation and aggregation during the refolding of denatured-reduced egg white lysozyme. Biochemistry, 1991, 30: 2790-2797.
    [10] Hevehan DL, Clark EDB. Oxidative renaturation of lysozyme at high concentrations. Biotechnol Bioeng, 1997, 54: 221-230.
    [11] Gruber CW, Cemazar M, Heras B et al. Protein disulfide isomerase: the structure of oxidative folding. Trends Biochem Sci, 2006, 31: 455-464.
    [12] Morjana NA, Gilbert HF. Effect of protein and peptide inhibitors on the activity of protein disulfide-isomerase. Biochemistry, 1991, 30: 4985-4990.
    [13] Quan H, Fan G, Wang C-c. Independence of the chaperone activity of protein disulfide isomerase from its thioredoxin-like active site. J Biol Chem, 1995, 270: 17078-17080.
    [14] Klappa P, Hawkins HC, Freedman RB. Interactions between protein disulphide isomerase and peptides. Eur J Biochem, 1997, 248: 37-42.
    [15] Cai H, Wang CC, Tsou CL. Chaperone-like activity of protein disulfide isomerase in the refolding of a protein with no disulfide bonds. J Biol Chem, 1994, 269: 24550-24552.
    [16] Puig A, Gilbert HF. Protein disulfide isomerase exhibits chaperone and anti-chaperone activity in the oxidative refolding of lysozyme. J Biol Chem, 1994, 269: 7764-7771.
    [17] Zheng W-d, Quan H, Song J-l et al. Does DsbA have chaperone-like activity? Arch Biochem Biophys, 1997, 337: 326-331.
    [18] Maskos K, Huber-Wunderlich M, Glockshuber R. DsbA and DsbC-catalyzed oxidative folding of proteins with complex disulfide bridge patterns in vitro and in vivo. J Mol Biol, 2003, 325: 495-513.
    [19] Chen J, Song J-l, Zhang S et al. Chaperone activity of DsbC. J Biol Chem, 1999, 274: 19601-19605.
    [20] Heras B, Kurz M, Shouldice SR et al. The name's bond......disulfide bond. Curr Opin Struct Biol, 2007, 17: 691-698.
    [21] Horst R, Bertelsen EB, Fiaux J et al. Direct NMR observation of a substrate protein bound to the chaperonin GroEL. Proc Natl Acad Sci USA, 2005, 102: 12748-12753.
    [22]张佳艺,关怡新,姚善泾.分子伴侣及其在蛋白质复性中的应用.化学通报, 2002, 65: 4-8.
    [23] Stan G, Brooks BR, Lorimer GH et al. Residues in substrate proteins that interact with GroEL in the capture process are buried in the native state. Proc Natl Acad Sci USA, 2006, 103: 4433-4438.
    [24] Kubota S, Kubota H, Nagata K. Cytosolic chaperonin protects folding intermediates of Gβfrom aggregation by recognizing hydrophobicβ-strands. Proc Natl Acad Sci USA, 2006, 103: 8360-8365.
    [25] Jakob U, Gaestel M, Engel K et al. Small heat shock proteins are molecular chaperones. J Biol Chem, 1993, 268: 1517-1520.
    [26] Haslbeck M, Franzmann T, Weinfurtner D et al. Some like it hot: the structure and function of small heat-shock proteins. Nat Struct Mol Biol, 2005, 12: 842-846.
    [27] Kim R, Kim KK, Yokota H et al. Small heat shock protein of Methanococcus jannaschii, a hyperthermophile. Proc Natl Acad Sci USA, 1998, 95: 9129-9133.
    [28]王志珍.蛋白质折叠和分子伴侣.生物学通报, 2004, 39: 1-6.
    [29] Menzella HG, Gramajo HC, Ceccarelli EA. High recovery of prochymosin from inclusion bodies using controlled air oxidation. Protein Expr Purif, 2002, 25: 248-255.
    [30] Lees WJ. Small-molecule catalysts of oxidative protein folding. Curr Opin Chem Biol, 2008, 12: 740-745.
    [31] Madar DJ, Patel AS, Lees WJ. Comparison of the oxidative folding of lysozyme at a high protein concentration using aromatic thiols versus glutathione. J Biotechnol, 2009, 142: 214-219.
    [32] Gurbhele-Tupkar MC, Perez LR, Silva Y et al. Rate enhancement of the oxidative folding of lysozyme by the use of aromatic thiol containing redox buffers. Bioorg Med Chem, 2008, 16: 2579-2590.
    [33] Beld J, Woycechowsky KJ, Hilvert D. Selenoglutathione: efficient oxidative protein folding by a diselenide. Biochemistry, 2007, 46: 5382-5390.
    [34] Beld J, Woycechowsky KJ, Hilvert D. Catalysis of oxidative protein folding by small-molecule diselenides. Biochemistry, 2008, 47: 6985-6987.
    [35] Woycechowsky KJ, Wittrup KD, Raines RT. A small-molecule catalyst of protein folding in vitro and in vivo. Chem Biol, 1999, 6: 871-879.
    [36] Cabrele C, Fiori S, Pegoraro S et al. Redox-active cyclic bis(cysteinyl)peptides as catalysts for in vitro oxidative protein folding. Chem Biol, 2002, 9: 731-740.
    [37] Ignatova Z, Gierasch LM. Inhibition of protein aggregation in vitro and in vivo by a natural osmoprotectant. Proc Natl Acad Sci USA, 2006, 103: 13357-13361.
    [38] Meng F-G, Park Y-D, Zhou H-M. Role of proline, glycerol, and heparin as protein folding aids during refolding of rabbit muscle creatine kinase. The International Journal of Biochemistry & Cell Biology, 2001, 33: 701-709.
    [39] Cleland JL, Wang DIC. Refolding and aggregation of bovine carbonic anhydrase B: quasi-elastic light scattering analysis. Biochemistry, 1990, 29: 11072-11078.
    [40] Cleland JL, Hedgepeth C, Wang DI. Polyethylene glycol enhanced refolding of bovine carbonic anhydrase B. Reaction stoichiometry and refolding model. J Biol Chem, 1992, 267: 13327-13334.
    [41] Cleland JL, Randolph TW. Mechanism of polyethylene glycol interaction with the molten globule folding intermediate of bovine carbonic anhydrase B. J Biol Chem, 1992, 267: 3147-3153.
    [42] Yasuda M, Murakami Y, Sowa A et al. Effect of additives on refolding of a denatured protein. Biotechnol Prog, 1998, 14: 601-606.
    [43] Arakawa T, Tsumoto K. The effects of arginine on refolding of aggregated proteins: not facilitate refolding, but suppress aggregation. Biochem Biophys Res Commun, 2003, 304: 148-152.
    [44] Arakawa T, Ejima D, Tsumoto K et al. Suppression of protein interactions by arginine: A proposed mechanism of the arginine effects. Biophys Chem, 2007,127: 1-8.
    [45] Rozema D, Gellman SH. Artificial chaperones: protein refolding via sequential use of detergent and cyclodextrin. J Am Chem Soc, 1995, 117: 2373-2374.
    [46] Rozema D, Gellman SH. Artificial chaperone-assisted refolding of denatured-reduced lysozyme: modulation of the competition between renaturation and aggregation. Biochemistry, 1996, 35: 15760-15771.
    [47] Rozema D, Gellman SH. Artificial chaperone-assisted refolding of carbonic anhydrase B. J Biol Chem, 1996, 271: 3478-3487.
    [48] Daugherty DL, Rozema D, Hanson PE et al. Artificial chaperone-assisted refolding of citrate synthase. J Biol Chem, 1998, 273: 33961-33971.
    [49] Lu DN, Zhang K, Liu Z. Protein refolding assisted by an artificial chaperone using temperature stimuli responsive polymer as the stripper. Biochem Eng J, 2005, 25: 141-149.
    [50] Sivakama Sundari C, Raman B, Balasubramanian D. Artificial chaperoning of insulin, human carbonic anhydrase and hen egg lysozyme using linear dextrin chains - a sweet route to the native state of globular proteins. FEBS Lett, 1999, 443: 215-219.
    [51] Lanckriet H, Middelberg APJ. Operational regimes for a simplified one-step artificial chaperone refolding method. Biotechnol Prog, 2004, 20: 1861-1867.
    [52] Nomura Y, Ikeda M, Yamaguchi N et al. Protein refolding assisted by self-assembled nanogels as novel artificial molecular chaperone. FEBS Lett, 2003, 553: 271-276.
    [53] Nomura Y, Sasaki Y, Takagi M et al. Thermoresponsive controlled association of protein with a dynamic nanogel of hydrophobized polysaccharide and cyclodextrin: heat shock protein-like activity of artificial molecular chaperone. Biomacromolecules, 2004, 6: 447-452.
    [54] Morimoto N, Endo T, Iwasaki Y et al. Design of hybrid hydrogels with self-assembled nanogels as cross-linkers: interaction with proteins and chaperone-like activity. Biomacromolecules, 2005, 6: 1829-1834.
    [55] Werner MH, Clore GM, Gronenborn AM et al. Refolding proteins by gel filtration chromatography. FEBS Lett, 1994, 345: 125-130.
    [56]杨坤.蛋白质离子交换色谱的吸附动力学研究[博士学位论文].天津:天津大学, 2007.
    [57] Batas B, Chaudhuri JB. Protein refolding at high concentration using size-exclusion chromatography. Biotechnol Bioeng, 1996, 50: 16-23.
    [58] Batas, Batas B, Chaudhuri et al. The influence of operational parameters on lysozyme refolding using size-exclusion chromatography. Bioprocess Biosyst Eng, 2001, 24: 255-259.
    [59] Fahey EM, Chaudhuri JB, Binding P. Refolding and purification of a urokinase plasminogen activator fragment by chromatography. Journal of Chromatography B: Biomedical Sciences and Applications, 2000, 737: 225-235.
    [60] Gu Z, Su Z, Janson J-C. Urea gradient size-exclusion chromatography enhanced the yield of lysozyme refolding. J Chromatogr A, 2001, 918: 311-318.
    [61] Li M, Poliakov A, Danielson UH et al. Refolding of a recombinant full-length non-structural (NS3) protein from hepatitis C virus by chromatographic procedures. Biotechnol Lett, 2003, 25: 1729-1734.
    [62] Chen Y, Leong SSJ. Adsorptive refolding of a highly disulfide-bonded inclusion body protein using anion-exchange chromatography. J Chromatogr A, 2009, 1216: 4877-4886.
    [63] Langenhof M, Leong SSJ, Pattenden LK et al. Controlled oxidative protein refolding using an ion-exchange column. J Chromatogr A, 2005, 1069: 195-201.
    [64] Li M, Zhang G, Su Z. Dual gradient ion-exchange chromatography improved refolding yield of lysozyme. J Chromatogr A, 2002, 959: 113-120.
    [65]白泉,耿信笃.用疏水色谱复性并同时纯化蛋白质的机理及其应用.中国科学B辑, 2002, 32: 460-471.
    [66] Geng X, Chang X. High-performance hydrophobic interaction chromatography as a tool for protein refolding. J Chromatogr A, 1992, 599: 185-194.
    [67] Wang C, Geng X, Wang D et al. Purification of recombinant bovine normal prion protein PrP(104-242) by HPHIC. J Chromatogr B, 2004, 806: 185-190.
    [68] Gong B, Wang L, Wang C et al. Preparation of hydrophobic interaction chromatographic packings based on monodisperse poly(glycidylmethacrylate-co-ethylenedimethacrylate) beads and their application. J Chromatogr A, 2004, 1022: 33-39.
    [69]黄亚渝,童卫,郭立安. RNase等10种标准蛋白质在高效疏水作用色谱上的复性.第四军医大学学报, 2005, 26: 682-684.
    [70] Li JJ, Liu YD, Wang FW et al. Hydrophobic interaction chromatography correctly refolding proteins assisted by glycerol and urea gradients. J Chromatogr A, 2004, 1061: 193-199.
    [71] Berdichevsky Y, Lamed R, Frenkel D et al. Matrix-assisted refolding of single-chain Fv- cellulose binding domain fusion proteins. Protein Expr Purif, 1999, 17: 249-259.
    [72] Panagabko C, Morley S, Neely S et al. Expression and refolding of recombinant human [alpha]-Tocopherol transfer protein capable of specific[alpha]-Tocopherol Binding. Protein Expr Purif, 2002, 24: 395-403.
    [73] Rehm BH, Qi Q, Beermann BB et al. Matrix-assisted in vitro refolding of Pseudomonas aeruginosa class II polyhydroxyalkanoate synthase from inclusion bodies produced in recombinant Escherichia coli. Biochem J, 2001, 358: 263-268.
    [74] Saini DK, Pant N, Das TK et al. Cloning, overexpression, purification, and matrix-assisted refolding of DevS (Rv 3132c) histidine protein kinase of Mycobacterium tuberculosis. Protein Expr Purif, 2002, 25: 203-208.
    [75] Dong X-Y, Chen L-J, Sun Y. Effect of operating conditions on the refolding of his-tagged enhanced green fluorescent protein by artificial chaperone-assisted metal affinity chromatography. Biochem Eng J, 2009, 48: 65-70.
    [76] Dong X-Y, Yang H, Sun Y. Lysozyme refolding with immobilized GroEL column chromatography. J Chromatogr A, 2000, 878: 197-204.
    [77] Teshima T, Kohda J, Kondo A et al. Protein refolding system using holo-chaperonin from the thermophilic bacterium Thermus thermophilus. J Ferment Bioeng, 1998, 85: 564-570.
    [78] Zahn R, Buckle AM, Perrett S et al. Chaperone activity and structure of
    [79] m1A9lo9tna6mo, m9ir3ear:n i1oc5 pM0o2Ml4y-p, 1eG5pa0tri2dc9iea. bCin, dPionsgs adnoim LaDin es to afl?G. OroxEidLa.t iPvreo cre Nfoaltdl iAngc ad Sci USA, chromatography: folding of the scorpion toxin Cn5. Nat Biotech, 1999, 17: 187-191.
    [80] Yoshimoto M, Shimanouchi T, Umakoshi H et al. Immobilized liposome chromatography for refolding and purification of protein. J Chromatogr B, 2000, 743: 93-99.
    [81] Li J-J, Venkataramana M, Sanyal S et al. Immobilized [beta]-cyclodextrin polymer coupled to agarose gel properly refolding recombinant Staphylococcus aureus elongation factor-G in combination with detergent micelle. Protein Expr Purif, 2006, 45: 72-79.
    [82] Misawa S, Kumagai I. Refolding of therapeutic proteins produced in Escherichia coli as inclusion bodies. Peptide Science, 1999, 51: 297-307.
    [83] Creighton TE, Zapun A, Darby NJ. Mechanisms and catalysts of disulphide bond formation in proteins. Trends Biotechnol, 1995, 13: 18-23.
    [84] Tu BP, Weissman JS. Oxidative protein folding in eukaryotes: mechanisms and consequences. J Cell Biol, 2004, 164: 341-346.
    [85] Markovic I, Stantchev TS, Fields KH et al. Thiol/disulfide exchange is a prerequisite for CXCR4-tropic HIV-1 envelope-mediated T-cell fusion during viral entry. Blood, 2004, 103: 1586-1594.
    [86] Ryser HJP, Flückiger R. Keynote review: Progress in targeting HIV-1 entry. Drug Discov Today, 2005, 10: 1085-1094.
    [87] Raman B, Ramakrishna T, Rao CM. Refolding of denatured and denatured/reduced lysozyme at high concentrations. J Biol Chem, 1996, 271: 17067-17072.
    [88] Wetlaufer DB, Saxena VP. Formation of three-dimensional structure in proteins. I. Rapid nonenzymic reactivation of reduced lysozyme. Biochemistry, 1970, 9: 5015-5023.
    [89] Konishi Y, Ooi T, Scheraga HA. Regeneration of ribonuclease A from the reduced protein. 1. Rate-limiting steps. Biochemistry, 1982, 21: 4734-4740.
    [90] Konishi Y, Ooi T, Scheraga HA. Regeneration of RNase A from the reduced protein: models of regeneration pathways. Proc Natl Acad Sci USA, 1982, 79: 5734-5738.
    [91] Scheraga HA, Konishi Y, Ooi T. Multiple pathways for regenerating ribonuclease A. Adv Biophys, 1984, 18: 21-41.
    [92] Kibria FM, Lees WJ. Balancing conformational and oxidative kinetic traps during the folding of bovine pancreatic trypsin inhibitor (BPTI) with glutathione and glutathione disulfide. J Am Chem Soc, 2008, 130: 796-797.
    [93] Anderson WL, Wetlaufer DB. The folding pathway of reduced lysozyme. J Biol Chem, 1976, 251: 3147-3153.
    [94] Arolas JL, Aviles FX, Chang J-Y et al. Folding of small disulfide-rich proteins: clarifying the puzzle. Trends Biochem Sci, 2006, 31: 292-301.
    [95] Shioi S, Imoto T, Ueda T. Analysis of the early stage of the folding process of reduced lysozyme using all lysozyme variants containing a pair of cysteines. Biochemistry, 2004, 43: 5488-5493.
    [96] Perraudin JP, Torchia TE, Wetlaufer DB. Multiple parameter kinetic studies of the oxidative folding of reduced lysozyme. J Biol Chem, 1983, 258: 11834-11839.
    [97] Roux P, Delepierre M, Goldberg ME et al. Kinetics of secondary structure recovery during the refolding of reduced hen egg white lysozyme. J Biol Chem, 1997, 272: 24843-24849.
    [98] Roux P, Ruoppolo M, Chaffotte AF et al. Comparison of the kinetics of S-S bond, secondary structure, and active site formation during refolding of reduced denatured hen egg white lysozyme. Protein Sci, 1999, 8: 2751-2760.
    [99] van den Berg B, Chung EW, Robinson CV et al. The oxidative refolding of hen lysozyme and its catalysis by protein disulfide isomerase. EMBO J, 1999, 18: 4794–4803.
    [100] Jarrett NM, Djavadi-Ohaniance L, Willson RC et al. Immunochemicalpulsed-labeling characterization of intermediates during hen lysozyme oxidative folding. Protein Sci, 2002, 11: 2584-2595.
    [101] Rothwarf DM, Scheraga HA. Equilibrium and kinetic constants for the thiol-disulfide interchange reaction between glutathione and dithiothreitol. Proc Natl Acad Sci USA, 1992, 89: 7944-7948.
    [102] Royer CA, Mann CJ, Matthews CR. Resolution of the fluorescence equilibrium unfolding profile of trp aporepressor using single tryptophan mutants. Protein Sci, 1993, 2: 1844-1852.
    [103] Dong XY, Wu XY, Sun Y. Refolding of denatured lysozyme assisted by artificial chaperones in reverse micelles. Biochem Eng J, 2006, 31: 92-95.
    [104] Morgan CJ, Miranker A, Dobson CM. Characterization of collapsed states in the early stages of the refolding of hen lysozyme. Biochemistry, 1998, 37: 8473-8480.
    [105] Baldwin RL. Making a network of hydrophobic clusters. Science, 2002, 295: 1657-1658.
    [106] Gerstman BS, Chapagain PP. Self-organization in protein folding and the hydrophobic interaction. The Journal of Chemical Physics, 2005, 123: 054901-054906.
    [107] Chang JY, Li L. The unfolding mechanism and the disulfide structures of denatured lysozyme. FEBS Lett, 2002, 511: 73-78.
    [108] (?)ema(?)ar M, Zahariev S, Lopez JJ et al. Oxidative folding intermediates with nonnative disulfide bridges between adjacent cysteine residues. Proc Natl Acad Sci USA, 2003, 100: 5754-5759.
    [109] Gough JD, Lees WJ. Effects of redox buffer properties on the folding of a disulfide-containing protein: dependence upon pH, thiol pKa, and thiol concentration. J Biotechnol, 2005, 115: 279-290.
    [110] Baneyx F, Mujacic M. Recombinant protein folding and misfolding in Escherichia coli. Nat Biotechnol, 2004, 22: 1399-1408.
    [111] Mannall GJ, Myers JP, Liddell J et al. Ultra scale-down of protein refold screening in microwells: Challenges, solutions and application. Biotechnol Bioeng, 2009, 103: 329-340.
    [112] Kersteen EA, Raines RT. Catalysis of protein folding by protein disulfide isomerase and small-molecule mimics. Antioxid Redox Signal, 2003, 5: 413-424.
    [113] Freedman RB, Klappa P, Ruddock LW. Protein disulfide isomerases exploit synergy between catalytic and specific binding domains. EMBO Rep, 2002, 3: 136–140.
    [114] Hu S-H, Peek JA, Rattigan E et al. Structure of TcpG, the DsbA protein foldingcatalyst from Vibrio cholerae. J Mol Biol, 1997, 268: 137-146.
    [115] Kemmink J, Darby NJ, Dijkstra K et al. Structure determination of the N-terminal thioredoxin-like domain of protein disulfide isomerase using multidimensional heteronuclear 13C/15N NMR spectroscopy. Biochemistry, 1996, 35: 7684-7691.
    [116] Tian G, Xiang S, Noiva R et al. The crystal structure of yeast protein disulfide isomerase suggests cooperativity between its active sites. Cell, 2006, 124: 1085-1088.
    [117] Martin JL, Bardwell JCA, Kuriyan J. Crystal structure of the DsbA protein required for disulphide bond formation in vivo. Nature, 1993, 365: 464-468.
    [118] Guddat LW, Martin JL, Bardwell JCA et al. The uncharged surface features surrounding the active site of Escherichia coli DsbA are conserved and are implicated in peptide binding. Protein Sci, 1997, 6: 1148-1156.
    [119] DeCollo TV, Lees WJ. Effects of aromatic thiols on thiol-disulfide interchange reactions that occur during protein folding. J Org Chem, 2001, 66: 4244-4249.
    [120] Gough JD, Barrett EJ, Silva Y et al. ortho- and meta-substituted aromatic thiols are efficient redox buffers that increase the folding rate of a disulfide-containing protein. J Biotechnol, 2006, 125: 39-47.
    [121] Gough JD, Williams RH, Donofrio AE et al. Folding disulfide-containing proteins faster with an aromatic thiol. J Am Chem Soc, 2002, 124: 3885-3892.
    [122] Moroder L, Besse D, Musiol H-J et al. Oxidative folding of cystine-rich peptides vs regioselective cysteine pairing strategies. Peptide Science, 1996, 40: 207-234.
    [123] Cabrele C, Cattani-Scholz A, Renner C et al. Photomodulation of the redox and folding adjuvant properties of bis(cysteinyl) peptides. Eur J Org Chem, 2002, 2002: 2144-2150.
    [124] Woycechowsky KJ, Raines RT. The CXC motif: a functional mimic of protein disulfide isomerase. Biochemistry, 2003, 42: 5387-5394.
    [125] Hansen RE, Ostergaard H, Norgaard P et al. Quantification of protein thiols and dithiols in the picomolar range using sodium borohydride and 4,4 '-dithiodipyridine. Anal Biochem, 2007, 363: 77-82.
    [126] Eyer P, Worek F, Kiderlen D et al. Molar absorption coefficients for the reduced Ellman reagent: reassessment. Anal Biochem, 2003, 312: 224-227.
    [127] Lundstr(o|¨)m-Ljung J, Holmgren A. Glutaredoxin accelerates glutathione-dependent folding of reduced ribonuclease A together with protein disulfide-isomerase. J Biol Chem, 1995, 270: 7822-7828.
    [128] Wang G-Z, Dong X-Y, Sun Y. The role of disulfide bond formation in the conformational folding kinetics of denatured/reduced lysozyme. Biochem EngJ, 2009, 46: 7-11.
    [129] De Vendittis E, Palumbo G, Parlato G et al. A fluorimetric method for the estimation of the critical micelle concentration of surfactants. Anal Biochem, 1981, 115: 278-286.
    [130] Wunderlich M, Otto A, Seckler R et al. Bacterial protein disulfide isomerase: Efficient catalysis of oxidative protein folding at acidic pH. Biochemistry, 1993, 32: 12251-12256.
    [131] Furukawa Y, O'halloran TV. Posttranslational modifications in Cu,Zn-superoxide dismutase and mutations associated with amyotrophic lateral sclerosis. Antioxid Redox Signal, 2006, 8: 847-867.
    [132] Bulaj G, Kortemme T, Goldenberg DP. Ionization-reactivity relationships for cysteine thiols in polypeptides. Biochemistry, 1998, 37: 8965-8972.
    [133] Grauschopf U, Winther JR, Korber P et al. Why is DsbA such an oxidizing disulfide catalyst? Cell, 1995, 83: 947-955.
    [134] Pleasants JC, Guo W, Rabenstein DL. A comparative study of the kinetics of selenol/diselenide and thiol/disulfide exchange reactions. J Am Chem Soc, 1989, 111: 6553-6558.
    [135] Tajc SG, Tolbert BS, Basavappa R et al. Direct determination of thiol pKa by isothermal titration microcalorimetry. J Am Chem Soc, 2004, 126: 10508-10509.
    [136] Schulman BA, Lindstrom DL, Harlow E. Substrate recruitment to cyclin-dependent kinase 2 by a multipurpose docking site on cyclin A. Proc Natl Acad Sci USA, 1998, 95: 10453-10458.
    [137] Knowles JR, Parsons CA. Proximity effect in catalysed systems: a dramatic effect on ester hydrolysis. Nature, 1969, 221: 53-54.
    [138] Knowles JR, Blyth CA. Kinetic consequences of intermolecular attraction. I. Aminolysis of p-nitrophenyl decanoate and acetate by n-decylamine and ethylamine. J Am Chem Soc, 1971, 93: 3017-3021.
    [139] Knowles JR, Blyth CA. Kinetic consequences of intermolecular attraction. II. Hydrolysis of a series of fatty acid p-nitrophenyl esters catalyzed by a series of N-n-alkylimidazoles. A very simple esterase model. J Am Chem Soc, 1971, 93: 3021-3027.
    [140] Wang J, Lu D, Lin Y et al. How CTAB assists the refolding of native and recombinant lysozyme. Biochem Eng J, 2005, 24: 269-277.
    [141] Bazarsuren A, Grauschopf U, Wozny M et al. In vitro folding, functional characterization, and disulfide pattern of the extracellular domain of human GLP-1 receptor. Biophys Chem, 2002, 96: 305-318.
    [142] Ejima D, Ono K, Tsumoto K et al. A novel "reverse screening" to identifyrefolding additives for activin-A. Protein Expr Purif, 2006, 47: 45-51.
    [143] Liang SP, Shu Q, Wang XC et al. Oxidative folding of reduced and denatured huwentoxin-I. J Protein Chem, 1999, 18: 619-625.
    [144] Shimizu H, Fujimoto K, Kawaguchi H. Renaturation of reduced ribonuclease A with a microsphere-induced refolding system. Biotechnol Prog, 2000, 16: 248-253.
    [145] Huang Z, Ni C, Zhou X et al. Mechanism of pH-sensitive polymer-assisted protein refolding and its application in TGF-beta1 and KGF-2. Biotechnol Prog, 2009, 25: 1387-1395.
    [146] Zhang Y, Ma Y, Yang M et al. Expression, purification, and refolding of a recombinant human bone morphogenetic protein 2 in vitro. Protein Expr Purif, 2010: DOI: 10.1016/j.pep.2010.1007.1014.
    [147] Han Y-G, Liu H-L, Zheng H-J et al. Purification and refolding of human [alpha]5-subunit (PSMA5) of the 20S proteasome, expressed as inclusion bodies in Escherichia coli. Protein Expr Purif, 2004, 35: 360-365.
    [148] Singh S, Pandey K, Chattopadhayay R et al. Biochemical, biophysical, and functional characterization of bacterially expressed and refolded receptor binding domain of Plasmodium vivax Duffy-binding Protein. J Biol Chem, 2001, 276: 17111-17116.
    [149] Wei C, Tang B, Zhang Y et al. Oxidative refolding of recombinant prochymosin. Biochem J, 1999, 340: 345-351.
    [150] Rothwarf DM, Scheraga HA. Regeneration and reduction of native bovine pancreatic ribonuclease A with oxidized and reduced dithiothreitol. J Am Chem Soc, 1991, 113: 6293-6294.
    [151] Rothwarf DM, Scheraga HA. Regeneration of bovine pancreatic ribonuclease A. 3. Dependence on the nature of the redox reagent. Biochemistry, 1993, 32: 2690-2697.
    [152] Bach RD, Dmitrenko O, Thorpe C. Mechanism of thiolate?disulfide interchange reactions in biochemistry. J Org Chem, 2007, 73: 12-21.
    [153] Zhang S, Sun Y. Ionic strength dependence of protein adsorption to dye-ligand adsorbents. AIChE J, 2002, 48: 178-186.
    [154] Krishnamurthy VM, Kaufman GK, Urbach AR et al. Carbonic anhydrase as a model for biophysical and physical-organic studies of proteins and protein?ligand binding. Chem Rev, 2008, 108: 946-1051.
    [155] Boyer PM, Hsu JT. Experimental studies of restricted protein diffusion in an agarose matrix. AIChE J, 1992, 38: 259-272.
    [156] Chang C, Lenhoff AM. Comparison of protein adsorption isotherms and uptake rates in preparative cation-exchange materials. J Chromatogr A, 1998,827: 281-293.
    [157] To BCS, Lenhoff AM. Hydrophobic interaction chromatography of proteins: III. Transport and kinetic parameters in isocratic elution. J Chromatogr A, 2008, 1205: 46-59.
    [158] Qu J-B, Wan X-Z, Zhai Y-Q et al. A novel stationary phase derivatized from hydrophilic gigaporous polystyrene-based microspheres for high-speed protein chromatography. J Chromatogr A, 2009, 1216: 6511-6516.
    [159] Du K-F, Yan M, Wang Q-Y et al. Preparation and characterization of novel macroporous cellulose beads regenerated from ionic liquid for fast chromatography. J Chromatogr A, 2010, 1217: 1298-1304.
    [160] Bushmarina NA, Kuznetsova IM, Biktashev AG et al. Partially folded conformations in the folding pathway of bovine carbonic anhydrase II: a fluorescence spectroscopic analysis. Chembiochem, 2001, 2: 813-821.
    [161] Baynes BM, Wang DIC, Trout BL. Role of arginine in the stabilization of proteins against aggregation. Biochemistry, 2005, 44: 4919-4925.
    [162] Lin DQ, Zhong LN, Yao SJ. Zeta potential as a diagnostic tool to evaluate the biomass electrostatic adhesion during ion-exchange expanded bed application. Biotechnol Bioeng, 2006, 95: 185-191.
    [163] Seyrek E, Dubin PL, Tribet C et al. Ionic strength dependence of protein-polyelectrolyte interactions. Biomacromolecules, 2003, 4: 273-282.
    [164] Dong X-Y, Huang Y, Sun Y. Refolding kinetics of denatured-reduced lysozyme in the presence of folding aids. J Biotechnol, 2004, 114: 135-142.
    [165] Kuehner DE, Engmann J, Fergg F et al. Lysozyme net charge and ion binding in concentrated aqueous electrolyte solutions. J Phys Chem B, 1999, 103: 1368-1374.
    [166] Machold C, Schlegl R, Buchinger W et al. Matrix assisted refolding of proteins by ion exchange chromatography. J Biotechnol, 2005, 117: 83-97.
    [167] Trivedi VD, Raman B, Rao CM et al. Co-refolding denatured-reduced hen egg white lysozyme with acidic and basic proteins. FEBS Lett, 1997, 418: 363-366.
    [168] N(o|¨)ppert A, Gast K, Zirwer D et al. Initial hydrophobic collapse is not necessary for folding RNase A. Fold Des, 1998, 3: 213-221.
    [169] Tessier PM, Johnson HR, Pazhianur R et al. Predictive crystallization of ribonuclease A via rapid screening of osmotic second virial coefficients. Proteins: Struct, Funct, Genet, 2003, 50: 303-311.
    [170] Middelberg APJ. Preparative protein refolding. Trends Biotechnol, 2002, 20: 437-443.
    [171] Lu D, Liu Z, Zhang M et al. The mechanism of PNIPAAm-assisted refoldingof lysozyme denatured by urea. Biochem Eng J, 2005, 24: 55-64.
    [172] Kiefhaber T. Kinetic traps in lysozyme folding. Proc Natl Acad Sci USA, 1995, 92: 9029-9033.
    [173] Valente JJ, Verma KS, Manning MC et al. Second virial coefficient studies of cosolvent-induced protein self-interaction. Biophys J, 2005, 89: 4211-4218.
    [174] Johnson EM, Berk DA, Jain RK et al. Diffusion and partitioning of proteins in charged agarose gels. Biophys J, 1995, 68: 1561-1568.
    [175] Liu Y, Fratini E, Baglioni P et al. Effective long-range attraction between protein molecules in solutions studied by small angle neutron scattering. Phys Rev Lett, 2005, 95: 118102.
    [176] Majhi PR, Ganta RR, Vanam RP et al. Electrostatically driven protein aggregation:β-lactoglobulin at low ionic strength. Langmuir, 2006, 22: 9150-9159.
    [177] Grahame DC. The electrical double layer and the theory of electrocapillarity. Chem Rev, 1947, 41: 441-501.
    [178] DePhillips P, Lenhoff AM. Pore size distributions of cation-exchange adsorbents determined by inverse size-exclusion chromatography. J Chromatogr A, 2000, 883: 39-54.
    [179] Tokuriki N, Kinjo M, Negi S et al. Protein folding by the effects of macromolecular crowding. Protein Sci, 2004, 13: 125-133.
    [180] Ai X, Zhou Z, Bai Y et al. 15N NMR spin relaxation dispersion study of the molecular crowding effects on protein folding under native conditions. J Am Chem Soc, 2006, 128: 3916-3917.
    [181] Du F, Zhou Z, Mo Z-Y et al. Mixed macromolecular crowding accelerates the refolding of rabbit muscle creatine kinase: implications for protein folding in physiological environments. J Mol Biol, 2006, 364: 469-482.
    [182] Sasahara K, McPhie P, Minton AP. Effect of dextran on protein stability and conformation attributed to macromolecular crowding. J Mol Biol, 2003, 326: 1227-1237.
    [183] Zhou B-R, Liang Y, Du F et al. Mixed macromolecular crowding accelerates the oxidative refolding of reduced, denatured lysozyme. J Biol Chem, 2004, 279: 55109-55116.
    [184] Mukherjee S, Waegele MM, Chowdhury P et al. Effect of macromolecular crowding on protein folding dynamics at the secondary structure level. J Mol Biol, 2009, 393: 227-236.
    [185] van den Berg B, Wain R, Dobson CM et al. Macromolecular crowding perturbs protein refolding kinetics: implications for folding inside the cell. EMBO J, 2000, 19: 3870-3875.
    [186] van den Berg B, Ellis RJ, Dobson CM. Effects of macromolecular crowding on protein folding and aggregation. EMBO J, 1999, 18: 6927-6933.
    [187] Perham M, Stagg L, Wittung-Stafshede P. Macromolecular crowding increases structural content of folded proteins. FEBS Lett, 2007, 581: 5065-5069.
    [188] Sela M, Steiner LA. Inhibition of lysozyme by some copolymers of amino acids. Biochemistry, 1963, 2: 416-421.
    [189] Domurado D, Moreaué, Chotard-Ghodsnia R et al. in Biomedical Polymers and Polymer Therapeutics. (eds. E. Chiellini, J. Sunamoto, C. Migliaresi, R.M. Ottenbrite & D. Cohn) 159-176 (Springer US, 2002).
    [190] ten Brink T, Exner T. pKa based protonation states and microspecies for protein–ligand docking. J Comput Aided Mol Des, 2010, 24: 935-942.
    [191]吴沛桥,巴晓革,胡海et al.绿色荧光蛋白GFP的研究进展及应用.生物医学工程研究, 2009, 28: 83-86.
    [192] Cabanne C, Noubhani AM, Hocquellet A et al. Purification and on-column refolding of EGFP overexpressed as inclusion bodies in Escherichia coli with expanded bed anion exchange chromatography. J Chromatogr B, 2005, 818: 23-27.
    [193]陈丽君.金属螯合色谱耦合人工伴侣辅助EGFP包含体蛋白质复性[硕士学位论文].天津:天津大学, 2008.
    [194]肖文楚.重组核糖核酸酶A的表达和复性研究[硕士学位论文].天津:天津大学, 2008.
    [195]冯旭东.新型金属螯合亲和反胶团对EGFP萃取特性的研究[硕士学位论文].天津:天津大学, 2009.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700