肾移植急性排斥反应尿液中早期标志物研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
急性排斥反应(acute rejection,AR)是影响移植肾长期存活的一个关键因素。也是引起早期移植物损害和长期的移植物功能障碍以及慢性移植物肾病的主要原因,而目前通常诊断肾移植术后发生急性排斥反应方法包括:1.病人临床体征,2.血肌酐、尿素氮等非特异性生化指标检查,3.移植肾影像学检查,4.移植肾穿刺病理活检。前3项检查指标只有当移植肾功能发生明显损伤后才能出现改变,并且这些改变都是非特异性的。组织活检虽然是确诊急性排斥反应的“金标准”,但穿刺活检可能导致严重的并发症,而由于活检取材部位的限制,并不能及时、全面的反应出移植肾的损伤情况。因此,目前临床急需筛选具有前瞻性、非侵入性和特异性的早期诊断AR的方法,以指导临床用药,提高移植肾的长期存活率
     尿液可以及时准确地反映整个肾脏的生理状态和功能水平,而且收集简单方便、是非侵入性诊断肾脏疾病的理想标本。器官移植排斥反应的发生是一种复杂的、动态的、多因素的病理过程,很难用传统单一的指标来准确反映。而蛋白质组是动态的,有它的时空性,能够在生命有机体整体水平上阐明生命现像的本质和活动规律。因此本课题以肾移植术后发生AR前、中、后各期的尿液作为研究对象,应用二维凝胶电泳-质谱技术(2-DE-MS)及Westernblot技术等,以期找到一组具有前瞻性和特异性的蛋白分子作为早期诊断的候选标志物。
     首先,我们根据尿蛋白的含盐高的特点优化了2-DE过程中等电聚焦程序,与Bio-rad操作手册推荐的等电聚焦方法相比,获得的尿液2-DE图谱点数由476个增加到564个。为充分去除尿液中的盐分,我们建立了尿蛋白丙酮沉淀三步法处理样品,结果显示尿蛋白丙酮沉淀三步法处理得到的样本在上述优化的等电聚焦条件下所获得的分离蛋白点更加清晰且圆,水平条带更少。说明本次研究建立的2-DE技术平台适用于尿蛋白研究。
     然后,为了消除尿蛋白样品的个体差异,本研究采取的是移植排斥反应前后患者自身样品的比较,即:排斥前3天相对光密度值与排斥后21天相对光密度值相比,选择表达量变化大于3倍(P﹤0.05),并根据两个体取6个时相点即:-3/-2/-1/7/14/21天相对光密度值做趋势分析,应同时具有明显下降趋势的30个蛋白点进行鉴定。经MADIL-TOF-MS/MS成功鉴定出其中的16个蛋白,经文献复习、生物信息学和蛋白功能检索,从中筛选了与免疫排斥反应相关的3个蛋白,分别为α-1抗胰凝乳蛋白酶(alpha-1-antichymotrypsin ,AACT),锌-α2糖蛋白(Zn-Alpha-2-Glycoprotein,ZAP)和肿瘤排斥抗原gp96(tumor rejection antigen gp96,gp96),进行进一步临床样本免疫印记验证。
     对肾移植术后发生AR患者临床确诊排斥前3~5天(n=8)和未发生AR患者术后7天(n=10)尿液蛋白进行Westrenblot验证发现:以肉眼观察印记条带出现有无为判断标准,三个蛋白判定的准确率、敏感性和特异性分别为:AACT, 77.78%/100%/60%;ZAG, 77.78%/100%/60%;gp96, 83.33%/87.5%/80%。将三个蛋白各组的测定值与同组对照的比值,运用LINGO8.0软件进行统计学处理,得到三个蛋白的判断理论临界值[0.15, 0.35, 0.50],以此界值作为判别标准时,单一的ZAG可以将准确率、敏感性和特异性提高到100%/100%/100%,AACT准确率、敏感性和特异性分别达到94.4%/100%/90.00%;gp96准确率、敏感性和特异性分别达到88.9%/100%/90.00%。最后应用统计学建立优化模型对三个蛋白联合进行判别时,与原始病例实际结果的符合率达100%,但其确切的诊断价值还有待多中心大样本的临床试验确认。
     综上所述,我们的研究结果提示,联合应用AACT、ZAG和gp96三个蛋白临界值可作为早期诊断肾移植急性排斥反应的指标。
Acute rejection, a key point to the long-term survive of renal grafts, is the major reason for the early damage and long-term dysfunction of the grafts and chronic allograft nephropathy. General diagnosis of acute rejection after renal transplantation includes four aspects as follows: (1)clinical manifestation;(2)laboratory test, some nonspecific biochemical indicator such as serum creatinine and urea nitrogen; (3)imageolgy examination, for example, color Doppler ultrasonography; (4)pathological biopsy. The first three tests, which are not characteristic for diagnosis, are obviously hysteretic so that the precise diagnosis would be made after the allografts had already suffered from acute rejection and been severely damaged. Biopsy, although thought to be the golden rule for the acute rejection final diagnosis, is an invasive way which probably causes terrible complications. Owing to the limited location for acquiring the sample, the whole status of the allografts can not be detected. Thus, early diagnosis biomarkers of acute rejection, which are prospective, noninvasive and specific are emergency requested to guide the clinical medication for the sake of increasing the survival rate of grafts.
     Urine reflects the physiological function and can be conveniently and safely collected, which is a perfect sample for non-invasive diagnosis of renal disease. Organ transplantation rejection is a complicated, dynamic and multiple factor procedure, which can not be accurately diagnosed by traditional unitary index. Proteomics which is dynamic and various in different time and status can systemically reflect the essence and rule of biological phenomenon. Thus we took the urine of pre-, mid- and post-AR as sample; find a series of candidate protein which might be meaningful for the diagnosis of acute rejection via two-dimensional gel electrophoresis mass spectrometry and initially validated these proteins.
     Firstly, we optimized the ampholine electrophoresis procedure since the urine protein had high electrolytes by which the protein dots number in 2-DE map was increased from 476 to 564 and the boosting pressure procedure was simple in contrast to the bio-rad recommended protocol. Meanwhile, in order to minimize the influence of electrolytes on 2-DE, we constructed a new way to acquire urine protein, namely acetone tripartite precipitation, by which the protein dots number in 2-DE pattern was average 761 which was obviously more than average 521 by acetone one step precipitation and the shape of the dots were distinct and circular, not to mention the level strap was apparently reduced.
     Secondly, in order to eliminate the individual difference between the urine protein samples, we choose two victims who suffered AR postoperative and compared the 2-DE map on different time points, which was three days, two days and one day before the clinical diagnosed AR (-3d,-2d,-1d)and the 7th,14th ,21st day after clinical diagnosed AR. Then we applied image analysis software to quantitate the 2-DE map and from the comparison of the 2-DE pattern on three days and two days before and the 21st day after the clinical diagnosed AR, we picked up the protein which was three times changed (P﹤0.05)and had the similar diversity trend in the two victims and identified them via mass spectrum.
     A total of 16 differential protein spots were successfully identified by MALDI-TOF-MS. Analyzed by means of bioinformatics and protein function index, alpha-1-antichymotrypsin (AACT), Zn-Alpha-2-Glycoprotein (ZAP) and tumor rejection antigen gp96 (gp96) were considered as candidate proteins for the further verification via immunoblotting. Four groups were established: urine from both of AR patients (n=8, 3-5days before clinical definition of AR) and stable patients (n=10, 7days after transplantation) after renal transplantations, urine from normal people (n=2) and operative patients of other diseases (n=2, 3days after operation) as control. Judgment from the imprinting straps by macroscopic observation, the accuracy rate, sensibility and specificity of these three proteins were: AACT, 77.78%/100%/60%; ZAG, 77.78%/100%/60%; gp96, 83.33%/100/80%. In view of these unsatisfied standers, we argued that protein existed or not is improper diagnosis stander, and the notable change of protein expression is also significant for diagnosing the disease. Therefore, the quantitative change of the aim proteins were analyzed by LINGO8.0 software, the accuracy rate, sensibility and specificity of the proteins were increased to 94.4%/ 100%/90.00%; 100%/100% 100%/ and 88.9%/100%/90.00%, respectively. Due to the limitation by sample volume, the study of multi-center clinical trials should be further carried out.
     In summary, our data suggest that early diagnosis of kidney graft acute rejection based on supposed criteria of AACT, ZAG and gp96 could be the promising in future.
引文
1. Kyo M, Toki K, Nishimura K, et al. Differential diagnosis of kidney transplant rejection and cyclosporin/tacrolimus nephropathy using urine cytology. Clin Transplant, 2002,16 Suppl 8:40-4.
    2. Jose MD, Ikezumi Y, van Rooijen N, et al. Macrophages act as effectors of tissue damage in acute renal allograft rejection. Transplantation, 2003,76:1015-22.
    3. Sezer S, Ozdemir FN, Akcay A, et al. Contribution of late acute rejection to long-term renal allograft survival in patients with chronic allograft nephropathy. Transplant Proc, 2003,35:2637-8.
    4. Savikko J, Kallio EA, Taskinen E, et al. The effect of acute rejection and cyclosporin A-treatment on induction of platelet-derived growth factor and its receptors during the development of chronic rat renal allograft rejection. Transplantation, 2002,73:506-11.
    5. Colombat M, Groussard O, Lautrette A, et al. Analysis of the different histologic lesions observed in transbronchial biopsy for the diagnosis of acute rejection. Clinicopathologic correlations during the first 6 months after lung transplantation. Hum Pathol, 2005,36:387-94.
    6. Bhalodolia R, Cortese C, Graham M, et al. Fulminant acute cellular rejection with negative findings on endomyocardial biopsy. J Heart Lung Transplant, 2006,25:989-92.
    7. Lasmar EP, Lasmar MF, Lasmar LF, et al. Progression of the renal graft: treatment of acute rejection based on a biopsy against a presumptive diagnosis. Transplant Proc, 2005,37:2775-6.
    8. Traum AZ, Wells MP, Aivado M, et al. SELDI-TOF MS of quadruplicate urine and serum samples to evaluate changes related to storage conditions. Proteomics, 2006,6:1676-80.
    9. Rabilloud T, Strub JM, Luche S, et al. A comparison between Sypro Ruby and ruthenium II tris (bathophenanthroline disulfonate) as fluorescent stains for protein detection in gels. Proteomics, 2001,1:699-704.
    10. Lopez MF, Berggren K, Chernokalskaya E, et al. A comparison of silver stain and SYPRO Ruby Protein Gel Stain with respect to protein detection in two-dimensional gels and identification by peptide mass profiling. Electrophoresis, 2000,21:3673-83.
    11. Kondo T, Hirohashi S. Application of highly sensitive fluorescent dyes (CyDye DIGE Fluor saturation dyes) to laser microdissection and two-dimensional difference gel electrophoresis (2D-DIGE) for cancer proteomics. Nat Protoc, 2006,1:2940-56.
    12. Hoorn EJ, Hoffert JD, Knepper MA. The application of DIGE-based proteomics to renal physiology. Nephron Physiol, 2006,104:p61-72.
    13. Choi NS, Yoo KH, Yoon KS, et al. Nano-scale proteomics approach using two-dimensional fibrin zymography combined with fluorescent SYPRO ruby dye. J Biochem Mol Biol, 2004,37:298-303.
    14. White IR, Pickford R, Wood J, et al. A statistical comparison of silver and SYPRO Ruby staining for proteomic analysis. Electrophoresis, 2004,25:3048-54.
    15. Lanne B, Panfilov O. Protein staining influences the quality of mass spectra obtained by peptide mass fingerprinting after separation on 2-d gels. A comparison of staining with coomassie brilliant blue and sypro ruby. J Proteome Res, 2005,4:175-9.
    16. Felder T, Schalley CA, Fakhrnabavi H, et al. A combined ESI- and MALDI-MS(/MS) study of peripherally persulfonylated dendrimers: false negative results by MALDI-MS and analysis of defects. Chemistry, 2005,11:5625-36.
    17. Hsu J, Chang SJ, Franz AH. MALDI-TOF and ESI-MS analysis of oligosaccharides labeled with a new multifunctional oligosaccharide tag. J Am Soc Mass Spectrom, 2006,17:194-204.
    18. McIlroy SP, Vahidassr MD, Savage DA, et al. Association of serum AACT levels and AACT signal polymorphism with late-onset Alzheimer's disease in Northern Ireland. Int J Geriatr Psychiatry, 2000,15:260-6.
    19. Hachulla E, Laine A, Hedouin V, et al. Variations in the glycoforms of serum alpha 1-antichymotrypsin in liver diseases and after liver transplantation. Clin Sci (Lond), 1992,82:439-46.
    20. Talbot C, Houlden H, Craddock N, et al. Polymorphism in AACT gene may lower age of onset of Alzheimer's disease. Neuroreport, 1996,7:534-6.
    21. Singh-Jasuja H, Scherer HU, Hilf N, et al. The heat shock protein gp96 induces maturation of dendritic cells and down-regulation of its receptor. Eur J Immunol, 2000,30:2211-5.
    22. Goyos A, Cohen N, Gantress J, et al. Anti-tumor MHC class Ia-unrestricted CD8 T cellcytotoxicity elicited by the heat shock protein gp96. Eur J Immunol, 2004,34:2449-58.
    23. Basu S, Binder RJ, Ramalingam T, et al. CD91 is a common receptor for heat shock proteins gp96, hsp90, hsp70, and calreticulin. Immunity, 2001,14:303-13.
    24. Robert J, Menoret A, Basu S, et al. Phylogenetic conservation of the molecular and immunological properties of the chaperones gp96 and hsp70. Eur J Immunol, 2001,31:186-95.
    25. Sanchez LM, Chirino AJ, Bjorkman P. Crystal structure of human ZAG, a fat-depleting factor related to MHC molecules. Science, 1999,283:1914-9.
    26. Sanders PM, Tisdale MJ. Effect of zinc-alpha2-glycoprotein (ZAG) on expression of uncoupling proteins in skeletal muscle and adipose tissue. Cancer Lett, 2004,212:71-81.
    27. McDermott LC, Freel JA, West AP, et al. Zn-alpha2-glycoprotein, an MHC class I-related glycoprotein regulator of adipose tissues: modification or abrogation of ligand binding by site-directed mutagenesis. Biochemistry, 2006,45:2035-41.
    1. Righetti PG, Boschetti E, Lomas L, et al. Protein Equalizer Technology : the quest for a "democratic proteome". Proteomics, 2006,6:3980-92.
    2. Saad R, Gritsch HA, Shapiro R, et al. Clinical significance of renal allograft biopsies with "borderline changes," as defined in the Banff Schema. Transplantation, 1997,64:992-5.
    3. Emovon OE, King JA, Smith SR, et al. Clinical significance of eosinophils in suspicious or borderline renal allograft biopsies. Clin Nephrol, 2003,59:367-72.
    4. Sezer S, Ozdemir FN, Akcay A, et al. Contribution of late acute rejection to long-term renal allograft survival in patients with chronic allograft nephropathy. Transplant Proc, 2003,35:2637-8.
    5. Thongboonkerd V, Malasit P. Renal and urinary proteomics: current applications and challenges. Proteomics, 2005,5:1033-42.
    6. Clarke W, Silverman BC, Zhang Z, et al. Characterization of renal allograft rejection by urinary proteomic analysis. Ann Surg, 2003,237:660-4; discussion 664-5.
    7. Schaub S, Wilkins J, Weiler T, et al. Urine protein profiling with surface-enhanced laser-desorption/ionization time-of-flight mass spectrometry. Kidney Int, 2004,65: 323-32.
    8. Schaub S, Wilkins JA, Antonovici M, et al. Proteomic-based identification of cleaved urinary beta2-microglobulin as a potential marker for acute tubular injury in renal allografts. Am J Transplant, 2005,5:729-38.
    9. O'Riordan E, Orlova TN, Mei J J, et al. Bioinformatic analysis of the urine proteome of acute allograft rejection. J Am Soc Nephrol, 2004,15:3240-8.
    1. Sijpkens YW, Doxiadis II, Mallat MJ, et al. Early versus late acute rejection episodes in renal transplantation. Transplantation, 2003,75:204-8.
    2. Sorof JM, Vartanian RK, Olson JL, et al. Histopathological concordance of paired renal allograft biopsy cores. Effect on the diagnosis and management of acute rejection. Transplantation, 1995,60:1215-9.
    3. Li B, Hartono C, Ding R, et al. Noninvasive diagnosis of renal-allograft rejection by measurement of messenger RNA for perforin and granzyme B in urine. N Engl J Med, 2001,344:947-54.
    4. Roberti I, Reisman L. Serial evaluation of cell surface markers for immune activation after acute renal allograft rejection by urine flow cytometry--correlation with clinical outcome. Transplantation, 2001,71:1317-20.
    5. Woodle ES, Cronin D, Newell KA, et al. Tacrolimus therapy for refractory acute renal allograft rejection: definition of the histologic response by protocol biopsies.Transplantation, 1996,62:906-10.
    6. Beckingham IJ, Nicholson ML, Bell PR. Analysis of factors associated with complications following renal transplant needle core biopsy. Br J Urol, 1994,73:13-5.
    7. Huraib S, Goldberg H, Katz A, et al. Percutaneous needle biopsy of the transplanted kidney: technique and complications. Am J Kidney Dis, 1989,14:13-7.
    8. Benfield MR, Herrin J, Feld L, et al. Safety of kidney biopsy in pediatric transplantation: a report of the Controlled Clinical Trials in Pediatric Transplantation Trial of Induction Therapy Study Group. Transplantation, 1999,67:544-7.
    9. Gaber LW, Moore LW, Gaber AO, et al. Correlation of histology to clinical rejection reversal: a thymoglobulin multicenter trial report. Kidney Int, 1999,55:2415-22.
    10. Curtis JJ, Julian BA, Sanders CE, et al. Dilemmas in renal transplantation: when the clinical course and histological findings differ. Am J Kidney Dis, 1996,27:435-40.
    11. Yamada A, Laufer TM, Gerth AJ, et al. Further analysis of the T-cell subsets and pathways of murine cardiac allograft rejection. Am J Transplant, 2003,3:23-7.
    12. Kreisel D, Krupnick AS, Gelman AE, et al. Non-hematopoietic allograft cells directly activate CD8+ T cells and trigger acute rejection: an alternative mechanism of allorecognition. Nat Med, 2002,8:233-9.
    13.田普川,刘雅峰,薛武军.急性细胞性排斥反应时移植肾组织中B7—1及相关蛋白的表达.中华器官移植杂志,2003. 186.
    14. Shah S, Divekar AA, Hilchey SP, et al. Increased rejection of primary tumors in mice lacking B cells: inhibition of anti-tumor CTL and TH1 cytokine responses by B cells. Int J Cancer, 2005,117:574-86.
    15. Bacso Z, Bene L, Damjanovich L, et al. INF-gamma rearranges membrane topography of MHC-I and ICAM-1 in colon carcinoma cells. Biochem Biophys Res Commun, 2002,290:635-40.
    16. Nicholson ML, Wheatley TJ, Doughman TM, et al. A prospective randomized trial of three different sizes of core-cutting needle for renal transplant biopsy. Kidney Int, 2000,58:390-5.
    17. Gupta RK, Jain M, Sharma RK. Serum & urinary interleukin-2 levels as predictors in acute renal allograft rejection. Indian J Med Res, 2004,119:24-7.
    18. Yoshimura R, Watanabe Y, Kasai S, et al. Hepatocyte growth factor (HGF) as a rapiddiagnostic marker and its potential in the prevention of acute renal rejection. Transpl Int, 2002,15:156-62.
    19. Le Meur Y, Jose MD, Mu W, et al. Macrophage colony-stimulating factor expression and macrophage accumulation in renal allograft rejection. Transplantation, 2002,73:1318-24.
    20. Brown FG, Nikolic-Paterson DJ, Chadban SJ, et al. Urine macrophage migration inhibitory factor concentrations as a diagnostic tool in human renal allograft rejection. Transplantation, 2001,71:1777-83.
    21. Sabek O,Dorak MT,Kotb M,et al. Quantitative detection of T-cell activation markers by real-time PCR in renal transplant rejection and correlation with histopathologic evaluation. transplatation, 2002,74:701-707.
    22. Li B,Hartono C,Ding RC, et al. Noninvasive diagnosis of renal-allograft rejection by measurement of messenger RNA for perforin and granzyme B in urine. N Engl J Med, 2006,344:947-954.
    23. Plezl S, Opelz G, Daniel V, et al. Evaluation of posttransplantation solube CD30 for diagnosis of acute renal allograft rejection. Transplantation, 2003,75:421-423.
    24. Ishida H, Koyama I, Sawada T, et al. Sialyl Iewis(x)(CD15) monitoring as a means to select antirejection therapy in patients with rejection after renal transplantation. transplantation, 2000,69:2208-2211.
    25. Zhang Z, Ohkohchi N, Sakurada M, et al. Analysis of urinatry donor-derived DNA in renal transplant recipients with acute rejection. Clin Transplant, 2002,16:45-50.
    26. Balshaw R ,Machnicki G,Carreno CA ,et al. Two-hour post-dose cy closporine levels in renal transplantation in Argentina :a cost - effective strategy for reducing acute rejection. Transplant Proc, 2005,37:871-874.
    27. Lap in A, Kop sa H, Smetana R, et al. Modified two-dimensional electrophoresis of urinary p roteins for monitoring early stages of kidney transp lantation. Transplant Proc, 1989,21:1880-1888.
    28. Hampe DJ, Schnoelzer M, Reinke P, et al. Proteomic urinalysis of renal transplant patients with acute rejection. Am Soc Nephrol, 2003,14:66-74.
    29. Schaub S,Wilkins JA, Antonovici M, et al. Proteomic-based identification of cleaved urinaryβ2-microglobulinβ2-microglobulin as a potential marker of acute tubularinjury in renal allograft. Am J Transplant, 2005,5:729-738.
    30. Sciascia N,Di scioscio V,Zompatori M, et al. Evaluation of the resistive index(RI) for the diagnosis of acute renal rejection in renal allografts from the same donor. Rsdiol Med, 2004,103:225-232.
    31. Chow L,Sommer FG,Huang J,et al. Power doppler imaging and resistance index measurment in the evaluation of acute renal transplant rejection. Clin Ultrasound, 2001,29:483-490.
    32. Wollenberg K,Waibel B,Pisarski P,et al. Careful clinincal monitoring in comparison to sequential Doppler sonography for the detection of acute rejection in the early phase after renal transplantation. Transpl Int, 2000,13:45-51.
    33. Zhang Y,Dodd SJ,Hendrich KS,et al. Magnetic resonance imaging dectection of rat renal transplant rejection by monitoring macrophage infiltration. Kidney Int, 2000,58:1300-1310.
    34. Ye Q, Yang D, Williams M, et al. In vivo detection of acute rat renal allograft rejection by MRI with USPIO particles. Kidney Int, 2002,61:1124-35.
    1. Thongboonkerd V, Chutipongtanate S, Kanlaya R. Systemic evaluation of sample preparation methods for gel-based human urinary proteomics:quantity, quality, and variability. J Proteome Res 2006;5:183–91.
    2. Oh J, Pyo JH, Jo EH, et al. Establishment of a near-standard twodimensional human urine proteomic map. Proteomics 2004;4:3485–97.
    3. Hortin GL, Jortani SA, Ritchie JC, Valdes R, ChanDW. Proteomics: A new diagnostic frontier. Clin Chem 2006;52:1223–37.
    4. Righetti PG, castagna A, Antonioli P, Boschetti E. Prefractionation techniques in proteome analysis: The mining tools of the third millennium.Electrophoresis 2005;26:297–319.
    5. G?rg A, Weiss W, Dunn MJ. Current two-dimensional electrophoresis technology for proteomics. Proteomics 2004;4:3665–85.
    6. Chakravarti DN, Chakravarti B, Moutsatsos I. Informatic tools for proteome profiling. BioTechniques 2002;32:S4–S15 (Comput. Proteoics Supp).
    7. Van den Bergh G, Arckens L. Fluorescent two-dimensional difference gel electrophoresis unveils the potential of gel-based proteomics. Curr Opin Biotechnol 2004;15:38–43.
    8. Stroink T, Castillo Ortiz M, Bult A, Lingeman H, de Jong GJ, Undenberg WJM. On-line multidimensional liquid chromatography and capillary electrophoresis systems for peptides and proteins. J Chromatogr B 2005;817:49–66.
    9. Aebersold R, Mann M. Mass-spectrometry-based proteomics. Nature 2003;422: 198–207.
    10. Domon B, Aebersold R. Mass spectrometry and protein analysis. Science 2006;312: 212–7.
    11. Cottrell JS. Protein identification by peptide mass fingerprinting. Pept Res 1994;7:115–24.
    12. Fenjo D, Qin J, Chait BT. Protein identification using mass spectrometric information. Electrophoresis 1998;19:998–1005.
    13. Cretich M, Damin F, Pirri G, Chiari M. Protein and peptide arrays: Recent trends andnew directions. Biomol Eng 2006;23:77–88.
    14. Issaq HJ, Veenstra TD, Conrads TP, Felschow D. The SELDI-TOF MS approach to proteomics: protein profiling and biomarker identification.Biochem Biophys Res Commun 2002;292:587–92.
    15. Anderson NG, Anderson NL, Tollaksen SL. Proteins of human urine: I. Concentration and analysis by two-dimensional electrophoresis. Clin Chem 1979;25:1199–210.
    16. Edwards JJ, Tollaksen SL, Anderson NG. Proteins of human urine: II. Identification and two-dimensional electrophoretic map positions of some major urinary proteins. Clin Chem 1982;28:941–8.
    17. Bueler MR, Wiederkher F, Vonderschmitt DJ. Electrophoretic, chromatographic, and immunological studies of human urinary proteins. Electrophoresis 1995;16:124–34.
    18. Marshall T, Williams K. Two-dimensional electrophoresis of human urinary proteins following concentration by dye precipitation. Electrophoresis 1996;17:1265–72.
    19. Heine G, Raida M, Forssmann WG. Mapping of peptides and protein fragments in human urine using liquid chromatography-mass spectrometry.J Chromatogr A 1997;776:117–24.
    20. Spahr CS, Davis MT, McGinley MD, et al. Towards defining the urinary proteome using liquid chromatography-tandem mass spectrometry: I.Profiling an unfractionated tryptic digest. Proteomics 2001;1:93–107.
    21. Thongboonkerd V, McLeish KR, Arthus JM, Klein JB. Proteomic analysis of normal human urinary proteins isolated by acetone precipitation or ultracentrifugation. Kidney Int 2002;62:1461–9.
    22. Pieper R, Gatling CL, McGrath AM, et al. Characterization of the human urinary proteome: a method for high-resolution display of urinary proteins on two-dimensional electrophoresis gels with a yield of nearly 1400 distinct proteins spots. Proteomics 2004;4:1159–74.
    23. Wittke S, Fliser D, Haubitz M, et al. Determination of peptides and proteins in human urine with capillary electrophoresis-mass spectrometry, a suitable tool for the establishment of new diagnostic markers. J Chromatogr A 2003;1013:173–81.
    24. Schaub S, Wilkins J, Weiler T, Sangster K, Rush D, Nickerson P. Urine protein profiling with surface-enhanced laser-desorption/ionization timeof-flight massspectrometry. Kidney Int 2004;65:323–32.
    25. Sun W, Li F, Wu S, et al. Human urine proteome analysis by three separation approaches. Proteomics 2005;5:4994–5001.
    26. CastagnaA, CecconiD, Sennels L, et al. Exploring the hidden human urinary proteome via ligand library beads. J Proteome Res 2005;4:1917–30.
    27. Thongboonkerd V, Klein JB, Jevans AW, McLeish KR. Urinary proteomics and biomarker discovery for glomerular diseases. Contrib Nephrol 2004;141:292–307.
    28. Meier M, Kaiser T, Herrmann A, et al. Identification of urinary protein pattern in Type 1 diabetic adolescents with early diabetic nephropathy by a novel combined proteome analysis. J Diabetes Complications 2005;19:223–232.
    29. Haubitz M, Wittke S, Weissinger EM, et al. Urine protein patterns can serve as diagnostic tools in patients with IgA nephropathy. Kidney Int 2005;67:2313–20.
    30. Park MR, Wang EH, Jin DC, et al. Establishment of a 2-D human urinary proteomic map in IgA nephropathy. Proteomics 2006;6:1066–76.
    31. Clarke W, Silverman B, Zhang Z, Chan DW, Klein AS, Molmenti EP. Characterization of renal allograft rejection by urinary proteomic analysis.Ann Surg 2003;5:660–5.
    32. O'Riordan E, Orlova TN, Mei JJ, et al. Bioinformatic analysis of the urine proteome of acute allograft rejection. J Am Soc Nephrol 2004;15:3240–8.
    33. Schaub S, Rush D, Wilkins J, et al. Proteomic-based detection of urine proteins associated with acute renal allograft rejection. J Am Soc Nephrol 2004;15:219–27.
    34. Schaub S,Wilkins JA, Antonovici M, et al. Proteomic-based identification of cleaved urinaryβ2-microglobulin as a potential marker of acute tubular injury in renal allograft. Am J Transplant 2005;5:729–38.
    35. Rasmussen HH, Orntoff TF, Wolf H, Celis JE. Towards a comprehensive database of proteins from the urine of patients with bladder cancer. J Urol 1996;155:2113–9.
    36. Vlahou A, Schellhammer PF, Mendrinos S, et al. Development of a novel proteomic approach for the detection of transitional cell carcinoma of the bladder in urine. Am J Pathol 2001;158:1491–502.
    37. Saito M, Kimoto M, Araki T, et al. Proteome analysis of gelatin-bound urinary proteins from patients with bladder cancers. Eur Urol 2005;48:865-71.
    38. Rogers MA, Clarke P, Noble J, et al. Proteomic profiling of urinary proteins in renalcancer by surface enhanced laser desorption ionization of key issues affecting potential clinical utility. Cancer Res 2003;63:6971–83.
    39. Theodorescu D, Wittke S, Ross M, et al. Discovery and validation of new protein biomarkers for urothelial cancer: a prospective analysis. Lancet Oncol 2006;7:230–40.
    40. Edwards JJ, Anderson NG, Tollaksen SL, von Eschenbach AC, Guevara J. Proteins of human urine: II. Identification by two-dimensional electrophoresis of a new candidate marker for prostatic cancer. Clin Chem 1982;28:160–3.
    41. Rehman I, Azzouzi AR, Catto JW, et al. Proteomic analysis of voided urine after prostatic massage from patients with prostate cancer: a pilot study. Urology 2004;64:1238–43.
    42. Cadieux PA, Beiko DT, Watterson JD, et al. Surface-enhanced laser desorption/ionization- time of flight-mass spectrometry (SELDI-TOF MS):a new proteomic urinary test for patients with urolithiasis. J Clin Lab Anal 2004;18:170–5.
    43. Tantipaiboonwong P, Sinchaikul S, Sriyam S, Phutrakul S, Chen ST. Different techniques for urinary protein analysis of normal and lung-cancer patients. Proteomics 2005;5:1140–9.