留言板

尊敬的读者、作者、审稿人, 关于本刊的投稿、审稿、编辑和出版的任何问题, 您可以本页添加留言。我们将尽快给您答复。谢谢您的支持!

姓名
邮箱
手机号码
标题
留言内容
验证码

基于单颗粒计数的蛋白质定量方法研究进展

张馨艺 米薇 胡志上

张馨艺,米薇,胡志上. 基于单颗粒计数的蛋白质定量方法研究进展[J]. 计量科学与技术,2023, 67(4): 28-36 doi: 10.12338/j.issn.2096-9015.2023.0093
引用本文: 张馨艺,米薇,胡志上. 基于单颗粒计数的蛋白质定量方法研究进展[J]. 计量科学与技术,2023, 67(4): 28-36 doi: 10.12338/j.issn.2096-9015.2023.0093
ZHANG Xinyi, MI Wei, HU Zhishang. Research Progress in Protein Quantification Based on Single Particle Counting in Large-Scale Metrology[J]. Metrology Science and Technology, 2023, 67(4): 28-36. doi: 10.12338/j.issn.2096-9015.2023.0093
Citation: ZHANG Xinyi, MI Wei, HU Zhishang. Research Progress in Protein Quantification Based on Single Particle Counting in Large-Scale Metrology[J]. Metrology Science and Technology, 2023, 67(4): 28-36. doi: 10.12338/j.issn.2096-9015.2023.0093

基于单颗粒计数的蛋白质定量方法研究进展

doi: 10.12338/j.issn.2096-9015.2023.0093
基金项目: 国家重点研发计划项目(2021YFF0600702);中国计量科学研究院重点领域项目(AKYZD2114-1)。
详细信息
    作者简介:

    张馨艺(1998-),中国计量科学研究院在读研究生,研究方向:抗体药物分析技术与测量装置研究,邮箱:karmic_xy@163.com

    通讯作者:

    胡志上(1979-),中国计量科学研究院副研究员,研究方向:蛋白质化学计量,邮箱:huz@nim.ac.cn

  • 中图分类号: TB98

Research Progress in Protein Quantification Based on Single Particle Counting in Large-Scale Metrology

  • 摘要: 蛋白质是执行功能的重要的生物大分子,蛋白质类生物标志物的鉴别与定量和复杂基质中目标蛋白准确含量的测定对疾病的预防、诊断及治疗有重要意义。伴随着生命科学的不断发展,科学家开发出许多种蛋白质定量方法,较为常见的有紫外可见分光光度法、酶联免疫吸附法及基于质谱的定量方法。但是对于高分子量蛋白质,尤其是分子量在200 kDa以上的蛋白质,由于其结构复杂及分子量巨大,导致前处理复杂或反应不专一、不完全,造成定量结果不准确。单颗粒计数法是通过对溶液中粒子逐一计数实现定量,相比于传统定量方法,单颗粒计数法具有灵敏度高、区分度好、外界因素干扰少的特点,加之此类方法样品处理简单,属于非破坏性的分析,适合对高分子量蛋白质进行完整结构水平的定量。总结了蛋白质定量方法,重点关注基于单颗粒计数的3种典型技术,包括单颗粒电感耦合等离子体质谱法(SP-ICP-MS)、单分子阵列技术(Simoa)和电喷雾-差分电迁移率-凝结核颗粒计数法(ES-DMA-CPC),系统阐述了这几种方法的工作原理及在蛋白质定量中的应用,并对未来该领域的发展进行了展望。
  • 图  1  SP-ICP-MS工作示意图

    Figure  1.  Schematic of SP-ICP-MS operation

    图  2  Simoa工作示意图

    Figure  2.  Schematic of Simoa operation

    图  3  ES-DMA-CPC工作示意图

    Figure  3.  Schematic of ES-DMA-CPC operation

    表  1  本文中提到的基于单颗粒计数的蛋白质定量方法及优缺点概述

    Table  1.   Overview of the advantages and disadvantages of protein quantification methods based on single particle counting discussed in this paper

    优点缺点
    SP-ICP-MS多种标签可用于定量,特异性高,信噪比高,分析速度快不同操作者标记效率差异较大,传输效率无法准确测定,杂质峰干扰
    Simoa灵敏度极高,检出限低,分析速度快,线性范围宽交叉反应,非特异性结合,背景干扰
    ES-DMA-CPC无需标记,不破坏样品,准确度高随机误差较大,误差来源研究不充分
    下载: 导出CSV
  • [1] Luo Y, Wu L, Yang B, et al. A novel potential primary method for quantification of enantiomers by high performance liquid chromatography-circular dichroism[J]. Scientific Reports, 2018, 8(1): 7390.
    [2] Phizicky E, Bastiaens P I H, Zhu H, et al. Protein analysis on a proteomic scale[J]. Nature, 2003, 422(6928): 208-215.
    [3] Rafalo-Ulinska A, Piotrowska J, Kryczyk A, et al. Zinc transporters protein level in postmortem brain of depressed subjects and suicide victims[J]. Journal of Psychiatric Research, 2016, 83: 220-229.
    [4] Rifai N, Gillette M A, Carr S A. Protein biomarker discovery and validation: the long and uncertain path to clinical utility[J]. Nature Biotechnology, 2006, 24(8): 971-983.
    [5] Cheng S-S, Yang G-J, Wang W, et al. The design and development of covalent protein-protein interaction inhibitors for cancer treatment[J]. Journal of Hematology & Oncology, 2020, 13(1): 26.
    [6] Marzo L D, Pranata J, Barbano D M. Measurement of casein in milk by Kjeldahl and sodium dodecyl sulfate–polyacrylamide gel electrophoresis[J]. Journal of Dairy Science, Elsevier, 2021, 104(7): 7448-7456.
    [7] Hayes M. Measuring Protein Content in Food: An Overview of Methods[J]. Foods, 2020, 9(10): 1340.
    [8] Huang T, Zhang W, Dai X, et al. Precise measurement for the purity of amino acid and peptide using quantitative nuclear magnetic resonance[J]. Talanta, 2014, 125: 94-101.
    [9] Hokazono E , Ota E , Goto T , et al. Development of a protein assay with copper chelator chromeazurol B, based on the biuret reaction[J]. Analytical Biochemistry, 2021, 630: 114320.
    [10] Deepachandi B, Weerasinghe S, Andrahennadi T P, et al. Quantification of Soluble or Insoluble Fractions of Leishmania Parasite Proteins in Microvolume Applications: A Simplification to Standard Lowry Assay[J]. International Journal of Analytical Chemistry, 2020, 2020: e6129132.
    [11] Li J, Zhao Y, Jiang X. Quantitative analysis of protein in thermosensitive hydroxypropyl chitin for biomedical applications[J]. Analytical Biochemistry, 2020, 599: 113745.
    [12] Ernst O, Zor T. Linearization of the Bradford Protein Assay[J]. JoVE, 2010(38): e1918.
    [13] Giuffrida F, Austin S, Cuany D, et al. Comparison of macronutrient content in human milk measured by mid-infrared human milk analyzer and reference methods[J]. Journal of Perinatology, 2019, 39(3): 497-503.
    [14] Narwal V, Sharma N, Sharma R, et al. Applicability of protein estimation methods for assaying glycomacropeptide[J]. International Journal of Dairy Technology, 2018, 71(2): 539-543.
    [15] Zaguri M, Kandel S, Rinehart S A, et al. Protein quantification in ecological studies: A literature review and empirical comparisons of standard methodologies[J]. Methods in Ecology and Evolution, 2021, 12(7): 1240-1251.
    [16] Gill S C, von Hippel P H. Calculation of protein extinction coefficients from amino acid sequence data[J]. Analytical Biochemistry, 1989, 182(2): 319-326.
    [17] Anthis N J, Clore G M. Sequence-specific determination of protein and peptide concentrations by absorbance at 205 nm: Sequence-Specific Protein Concentration at 205 nm[J]. Protein Science, 2013, 22(6): 851-858.
    [18] Ponzini E, Santambrogio C, De Palma A, et al. Mass spectrometry-based tear proteomics for noninvasive biomarker discovery[J]. Mass Spectrometry Reviews, 2022, 41(5): 842-860.
    [19] Weiner S, Sauer M, Visser P J, et al. Optimized sample preparation and data analysis for TMT proteomic analysis of cerebrospinal fluid applied to the identification of Alzheimer’s disease biomarkers[J]. Clinical Proteomics, 2022, 19(1): 13.
    [20] Repetto O, Caggiari L, De Zorzi M, et al. Quantitative Plasma Proteomics to Identify Candidate Biomarkers of Relapse in Pediatric/Adolescent Hodgkin Lymphoma[J]. International Journal of Molecular Sciences, 2022, 23(17): 9911.
    [21] Dash S, Wu C-C, Wu C-C, et al. Extracellular Vesicle Membrane Protein Profiling and Targeted Mass Spectrometry Unveil CD59 and Tetraspanin 9 as Novel Plasma Biomarkers for Detection of Colorectal Cancer[J]. Cancers, 2023, 15(1): 177.
    [22] Wang H, Shi T, Qian W-J, et al. The clinical impact of recent advances in LC-MS for cancer biomarker discovery and verification[J]. Expert Review of Proteomics, 2016, 13(1): 99-114.
    [23] 樊小雪, 翟睿, 楚占营, 等. 基于液相色谱-质谱联用技术的蛋白质类肿瘤标志物定量方法研究进展[J]. 质谱学报, 2022, 43(3): 263-277.
    [24] Lee H, Lee J. Peptide purity assignment for antibody quantification by combining isotope dilution mass spectrometry and liquid chromatography[J]. Bulletin of the Korean Chemical Society, 2022, 43(5): 704-713.
    [25] 朱文, 倪鑫茹, 徐昇. 同位素稀释质谱法在蛋白质计量比对中的应用[J]. 计量与测试技术, 2022, 49(7): 12-14.
    [26] Hädener M, Weinmann W, van Staveren D R, et al. Rapid quantification of free and glucuronidated THCCOOH in urine using coated well plates and LC–MS/MS analysis[J]. Bioanalysis, 2017, 9(5): 485-496.
    [27] Luis P B, Kunihiro A G, Funk J L, et al. Incomplete Hydrolysis of Curcumin Conjugates by β-Glucuronidase: Detection of Complex Conjugates in Plasma[J]. Molecular Nutrition & Food Research, 2020, 64(6): 1901037.
    [28] Kato M, Kinumi T, Yoshioka M, et al. Development of C-reactive protein certified reference material NMIJ CRM 6201-b: optimization of a hydrolysis process to improve the accuracy of amino acid analysis[J]. Analytical and Bioanalytical Chemistry, 2015, 407(11): 3137-3146.
    [29] Ji M, Cho B, Cho Y S, et al. Development of a Quantitative Sandwich Enzyme-Linked Immunosorbent Assay for Detecting the MPT64 Antigen of Mycobacterium tuberculosis[J]. Yonsei Medical Journal, 2014, 55(3): 746-752.
    [30] Helbing D L, Böhm L, Oraha N, et al. A Ponceau S Staining-Based Dot Blot Assay for Rapid Protein Quantification of Biological Samples[J]. Gels, 2022, 8(1): 43.
    [31] Reichel M, Weitzel V, Klement L, et al. Suitability of GRK Antibodies for Individual Detection and Quantification of GRK Isoforms in Western Blots[J]. International Journal of Molecular Sciences, 2022, 23(3): 1195.
    [32] Rebeski D E, Winger E M, Shin Y-K, et al. Identification of unacceptable background caused by non-specific protein adsorption to the plastic surface of 96-well immunoassay plates using a standardized enzyme-linked immunosorbent assay procedure1Mention of any commercial product in this paper does not constitute an endorsement or other comment on the product by the FAO, IAEA, or Bender Medsystems. 1[J]. Journal of Immunological Methods, 1999, 226(1): 85-92.
    [33] Cohen L, Walt D R. Highly Sensitive and Multiplexed Protein Measurements[J]. Chemical Reviews, 2019, 119(1): 293-321.
    [34] Ellington A A, Kullo I J, Bailey K R, et al. Antibody-Based Protein Multiplex Platforms: Technical and Operational Challenges[J]. Clinical Chemistry, 2010, 56(2): 186-193.
    [35] Lengfeld J, Zhang H, Stoesz S, et al. Challenges in Detection of Serum Oncoprotein: Relevance to Breast Cancer Diagnostics[J]. Breast Cancer: Targets and Therapy, 2021, 13: 575-593.
    [36] 武利庆. 什么是蛋白质计量[J]. 计量技术, 2018(9): 7.
    [37] 刘洋, 胡志上, 李红梅. 计量对我国蛋白质及肽类药物研发生产的重要意义[J]. 中国计量, 2019(8): 103-105.
    [38] 邓凤琳, 黄彦捷, 于婷, 等. 我国临床检验蛋白类标志物标准物质研制的现状与发展[J]. 生命科学仪器, 2023, 21(1): 4-10.
    [39] 董莲华, 刘亚辉, 傅博强, 等. 生物计量研究现状及展望[J]. 计量学报, 2023, 44(3): 317-325.
    [40] Zhang H, Chon C H, Pan X, et al. Methods for counting particles in microfluidic applications[J]. Microfluidics and Nanofluidics, 2009, 7(6): 739.
    [41] Bhumkar A, Magnan C, Lau D, et al. Single-Molecule Counting Coupled to Rapid Amplification Enables Detection of α-Synuclein Aggregates in Cerebrospinal Fluid of Parkinson’s Disease Patients[J]. Angewandte Chemie International Edition, 2021, 60(21): 11874-11883.
    [42] Rajan N K, Rajauria S, Ray T, et al. A simple microfluidic aggregation analyzer for the specific, sensitive and multiplexed quantification of proteins in a serum environment[J]. Biosensors and Bioelectronics, 2016, 77: 1062-1069.
    [43] Zhang L, Yang Y J, Xiong J Y, et al. Absolute quantification of particle number concentration using a digital single particle counting system[J]. Microchimica Acta, 2019, 186(8): 529.
    [44] Horáček M, Engels D J, Zijlstra P. Dynamic single-molecule counting for the quantification and optimization of nanoparticle functionalization protocols[J]. Nanoscale, 2020, 12(6): 4128-4136.
    [45] Strenge I, Engelhard C. Capabilities of fast data acquisition with microsecond time resolution in inductively coupled plasma mass spectrometry and identification of signal artifacts from millisecond dwell times during detection of single gold nanoparticles[J]. Journal of Analytical Atomic Spectrometry, 2015, 31(1): 135-144.
    [46] Zawadowicz M A, Lance S, Jayne J T, et al. Quantifying and improving the optical performance of the laser ablation aerosol particle time of flight mass spectrometer (LAAPToF) instrument[J]. Aerosol Science and Technology, 2020, 54(7): 761-771.
    [47] Fischer S K, Joyce A, Spengler M, et al. Emerging Technologies to Increase Ligand Binding Assay Sensitivity[J]. The AAPS Journal, 2015, 17(1): 93-101.
    [48] He L, Tessier D R, Briggs K, et al. Digital immunoassay for biomarker concentration quantification using solid-state nanopores[J]. Nature Communications, 2021, 12(1): 5348.
    [49] Blömeke L, Pils M, Kraemer-Schulien V, et al. Quantitative detection of α-Synuclein and Tau oligomers and other aggregates by digital single particle counting[J]. npj Parkinson’s Disease, 2022, 8(1): 1-13.
    [50] Lin J, Liu Y, Yang Y, et al. Calibration and correction of LA-ICP-MS and LA-MC-ICP-MS analyses for element contents and isotopic ratios[J]. Solid Earth Sciences, 2016, 1(1): 5-27.
    [51] Galazzi R M, Chacón-Madrid K, Freitas D C, et al. Inductively coupled plasma mass spectrometry based platforms for studies involving nanoparticle effects in biological samples[J]. Rapid Communications in Mass Spectrometry, 2020, 34(S3): e8726.
    [52] Hogeling S M, Cox M T, Bradshaw R M, et al. Quantification of proteins in whole blood, plasma and DBS, with element-labelled antibody detection by ICP-MS[J]. Analytical Biochemistry, 2019, 575: 10-16.
    [53] Ferreira N, Henriques B, Viana T, et al. Validation of a methodology to quantify macro, micro, and potentially toxic elements in food matrices[J]. Food Chemistry, 2023, 404: 134669.
    [54] Lee H-S, Kim S H, Jeong J-S, et al. Sulfur-based absolute quantification of proteins using isotope dilution inductively coupled plasma mass spectrometry[J]. Metrologia, 2015, 52(5): 619.
    [55] Degueldre C, Favarger P-Y. Colloid analysis by single particle inductively coupled plasma-mass spectroscopy: a feasibility study[J]. Colloids and Surfaces A: Physicochemical and Engineering Aspects, 2003, 217(1): 137-142.
    [56] Degueldre C, Favarger P-Y, Bitea C. Zirconia colloid analysis by single particle inductively coupled plasma–mass spectrometry[J]. Analytica Chimica Acta, 2004, 518(1): 137-142.
    [57] Pace H E, Rogers N J, Jarolimek C, et al. Determining Transport Efficiency for the Purpose of Counting and Sizing Nanoparticles via Single Particle Inductively Coupled Plasma Mass Spectrometry[J]. Analytical Chemistry, 2011, 83(24): 9361-9369.
    [58] Dan Y, Zhang W, Xue R, et al. Characterization of Gold Nanoparticle Uptake by Tomato Plants Using Enzymatic Extraction Followed by Single-Particle Inductively Coupled Plasma-Mass Spectrometry Analysis[J]. Environmental Science & Technology, 2015, 49(5): 3007-3014.
    [59] Wang J, Yue L, Zhao J, et al. Uptake and bioaccumulation of nanoparticles by five higher plants using single-particle-inductively coupled plasma-mass spectrometry[J]. Environmental Science: Nano, 2022, 9(8): 3066-3080.
    [60] Hu S, Liu R, Zhang S, et al. A new strategy for highly sensitive immunoassay based on single-particle mode detection by inductively coupled plasma mass spectrometry[J]. Journal of the American Society for Mass Spectrometry, 2009, 20(6): 1096-1103.
    [61] Huang Z, Li Z, Jiang M, et al. Homogeneous Multiplex Immunoassay for One-Step Pancreatic Cancer Biomarker Evaluation[J]. Analytical Chemistry, 2020, 92(24): 16105-16112.
    [62] Li B R, Tang H, Yu R Q, et al. Single-Nanoparticle ICP-MS for Sensitive Detection of Uracil-DNA Glycosylase Activity[J]. Analytical Chemistry, 2021, 93(24): 8381-8385.
    [63] Liu J, Murphy K E, Winchester M R, et al. Overcoming challenges in single particle inductively coupled plasma mass spectrometry measurement of silver nanoparticles[J]. Analytical and Bioanalytical Chemistry, 2017, 409(25): 6027-6039.
    [64] Geiss O, Bianchi I, Bucher G, et al. Determination of the Transport Efficiency in spICP-MS Analysis Using Conventional Sample Introduction Systems: An Interlaboratory Comparison Study[J]. Nanomaterials, 2022, 12(4): 725.
    [65] Mozhayeva D, Engelhard C. A quantitative nanoparticle extraction method for microsecond time resolved single-particle ICP-MS data in the presence of a high background[J]. Journal of Analytical Atomic Spectrometry, 2019, 34(8): 1571-1580.
    [66] Peyneau P-E. Poisson process modelling of spike occurrence in single particle inductively coupled plasma mass spectrometry time scans for very dilute nanoparticle dispersions[J]. Spectrochimica Acta Part B: Atomic Spectroscopy, 2021, 178: 106126.
    [67] Mozhayeva D, Engelhard C. A critical review of single particle inductively coupled plasma mass spectrometry – A step towards an ideal method for nanomaterial characterization[J]. Journal of Analytical Atomic Spectrometry, 2020, 35(9): 1740-1783.
    [68] Hu J, Deng D, Liu R, et al. Single nanoparticle analysis by ICPMS: a potential tool for bioassay[J]. Journal of Analytical Atomic Spectrometry, 2018, 33(1): 57-67.
    [69] O’Connell G C, Alder M L, Smothers C G, et al. Use of high-sensitivity digital ELISA improves the diagnostic performance of circulating brain-specific proteins for detection of traumatic brain injury during triage[J]. Neurological Research, 2020, 42(4): 346-353.
    [70] Rodero M P, Decalf J, Bondet V, et al. Detection of interferon alpha protein reveals differential levels and cellular sources in disease[J]. Journal of Experimental Medicine, 2017, 214(5): 1547-1555.
    [71] Akama K, Shirai K, Suzuki S. Highly sensitive multiplex protein detection by droplet-free digital ELISA[J]. Electronics and Communications in Japan, 2019, 102(2): 43-47.
    [72] Nilsson J, Ashton N J, Benedet A L, et al. Quantification of SNAP-25 with mass spectrometry and Simoa: a method comparison in Alzheimer’s disease[J]. Alzheimer’s Research & Therapy, 2022, 14(1): 78.
    [73] Wu D, Katilius E, Olivas E, et al. Incorporation of Slow Off-Rate Modified Aptamers Reagents in Single Molecule Array Assays for Cytokine Detection with Ultrahigh Sensitivity[J]. Analytical Chemistry, 2016, 88(17): 8385-8389.
    [74] Chang L, Rissin D M, Fournier D R, et al. Single molecule enzyme-linked immunosorbent assays: Theoretical considerations[J]. Journal of Immunological Methods, 2012, 378(1): 102-115.
    [75] Rissin D M, Fournier D R, Piech T, et al. Simultaneous Detection of Single Molecules and Singulated Ensembles of Molecules Enables Immunoassays with Broad Dynamic Range[J]. Analytical Chemistry, 2011, 83(6): 2279-2285.
    [76] Mora J, Chunyk A G, Dysinger M, et al. Next Generation Ligand Binding Assays—Review of Emerging Technologies’ Capabilities to Enhance Throughput and Multiplexing[J]. The AAPS Journal, 2014, 16(6): 1175-1184.
    [77] Kuhle J, Barro C, Andreasson U, et al. Comparison of three analytical platforms for quantification of the neurofilament light chain in blood samples: ELISA, electrochemiluminescence immunoassay and Simoa[J]. Clinical Chemistry and Laboratory Medicine (CCLM), 2016, 54(10): 1655-1661.
    [78] Costa O R, Verhaeghen K, Roels S, et al. An analytical comparison of three immunoassay platforms for subpicomolar detection of protein biomarker GAD65[J]. PLOS ONE, 2018, 13(3): e0193670.
    [79] Woolley C F, Hayes M A, Mahanti P, et al. Theoretical limitations of quantification for noncompetitive sandwich immunoassays[J]. Analytical and Bioanalytical Chemistry, 2015, 407(28): 8605-8615.
    [80] Perez J W, Pantazides B G, Watson C M, et al. Enhanced Stability of Blood Matrices Using a Dried Sample Spot Assay To Measure Human Butyrylcholinesterase Activity and Nerve Agent Adducts[J]. Analytical Chemistry, 2015, 87(11): 5723-5729.
    [81] Guo Y, Yu D, Cupp-Sutton K A, et al. Optimization of protein-level tandem mass tag (TMT) labeling conditions in complex samples with top-down proteomics[J]. Analytica Chimica Acta, 2022, 1221: 340037.
    [82] Weiss V U, Wieland K, Schwaighofer A, et al. Native Nano-electrospray Differential Mobility Analyzer (nES GEMMA) Enables Size Selection of Liposomal Nanocarriers Combined with Subsequent Direct Spectroscopic Analysis[J]. Analytical Chemistry, 2019, 91(6): 3860-3868.
    [83] Kaufman S L, Skogen J W, Dorman F D, et al. Macromolecule Analysis Based on Electrophoretic Mobility in Air: Globular Proteins[J]. Analytical Chemistry, 1996, 68(11): 1895-1904.
    [84] Pease L F, Elliott J T, Tsai D-H, et al. Determination of protein aggregation with differential mobility analysis: Application to IgG antibody[J]. Biotechnology and Bioengineering, 2008, 101(6): 1214-1222.
    [85] Guha S, Wayment J R, Tarlov M J, et al. Electrospray-Differential Mobility Analysis as an Orthogonal Tool to Size-Exclusion Chromatography for Characterization of Protein Aggregates[J]. Journal of Pharmaceutical Sciences, 2012, 101(6): 1985-1994.
    [86] Guha S, Ma X, Tarlov M J, et al. Quantifying Ligand Adsorption to Nanoparticles Using Tandem Differential Mobility Mass Analysis[J]. Analytical Chemistry, 2012, 84(15): 6308-6311.
    [87] Pease L F. Physical analysis of virus particles using electrospray differential mobility analysis[J]. Trends in Biotechnology, 2012, 30(4): 216-224.
    [88] Hutchins P M, Ronsein G E, Monette J S, et al. Quantification of HDL Particle Concentration by Calibrated Ion Mobility Analysis[J]. Clinical Chemistry, 2014, 60(11): 1393-1401.
    [89] Li M, Tan J, Tarlov M J, et al. Absolute Quantification Method for Protein Concentration[J]. Analytical Chemistry, 2014, 86(24): 12130-12137.
    [90] Clouet-Foraison N, Gaie-Levrel F, Coquelin L, et al. Absolute Quantification of Bionanoparticles by Electrospray Differential Mobility Analysis: An Application to Lipoprotein Particle Concentration Measurements[J]. Analytical Chemistry, 2017, 89(4): 2242-2249.
    [91] Tsai D-H, Zangmeister R A, Pease III L F, et al. Gas-Phase Ion-Mobility Characterization of SAM-Functionalized Au Nanoparticles[J]. Langmuir, 2008, 24(16): 8483-8490.
    [92] Brilke S, Resch J, Leiminger M, et al. Precision characterization of three ultrafine condensation particle counters using singly charged salt clusters in the 1-4 nm size range generated by a bipolar electrospray source[J]. Aerosol Science and Technology, 2020, 54(4): 396-409.
    [93] Tseng Y-H, Pease L F. Electrospray differential mobility analysis for nanoscale medicinal and pharmaceutical applications[J]. Nanomedicine: Nanotechnology, 2014, 10(8): 1591-1600.
    [94] Li M, Guha S, Zangmeister R, et al. Method for Determining the Absolute Number Concentration of Nanoparticles from Electrospray Sources[J]. Langmuir, 2011, 27(24): 14732-14739.
  • 加载中
图(3) / 表(1)
计量
  • 文章访问数:  316
  • HTML全文浏览量:  146
  • PDF下载量:  32
  • 被引次数: 0
出版历程
  • 收稿日期:  2023-03-29
  • 录用日期:  2023-04-03
  • 修回日期:  2023-05-21
  • 网络出版日期:  2023-06-29
  • 刊出日期:  2023-04-18

目录

    /

    返回文章
    返回