删除或更新信息,请邮件至freekaoyan#163.com(#换成@)

蛋白质组学在环境毒理学中的研究进展

本站小编 Free考研考试/2021-12-30

耿柠波1,
任晓倩1,2,
张海军1,,,
曹蓉1,
宋肖垚1,
罗云1,2,
张保琴1,
陈吉平1,,
1. 中国科学院大连化学物理研究所, 中国科学院分离分析化学重点实验室, 大连 116023;
2. 中国科学院大学, 北京 100049
作者简介: 耿柠波(1985-),女,博士,副研究员,研究方向为环境毒理学,E-mail:gengningbo@dicp.ac.cn.
通讯作者: 张海军,hjzhang@dicp.ac.cn ; 陈吉平,chenjp@dicp.ac.cn
基金项目: 国家自然科学基金委联合重大研究计划资助项目(91543201);国家自然科学基金资助项目(21607152)


中图分类号: X171.5


A Review on the Application of Proteomic Approaches in Environmental Toxicology

Geng Ningbo1,
Ren Xiaoqian1,2,
Zhang Haijun1,,,
Cao Rong1,
Song Xiaoyao1,
Luo Yun1,2,
Zhang Baoqin1,
Chen Jiping1,,
1. CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China;
2. University of Chinese Academy of Sciences, Beijing 100049, China
Corresponding authors: Zhang Haijun,hjzhang@dicp.ac.cn ; Chen Jiping,chenjp@dicp.ac.cn

CLC number: X171.5

-->

摘要
HTML全文
(0)(0)
参考文献(69)
相关文章
施引文献
资源附件(0)
访问统计

摘要:蛋白质组学是系统生物学的重要组成部分,它以整体、动态和定量的原则来研究各种蛋白质的功能,系统地分析生物体内蛋白质表达,蛋白-蛋白相互作用和翻译后修饰等特征,已成为后基因组时代不可或缺的分析工具。蛋白质组学技术具有高通量的特征,能实现对蛋白质的高效快捷的定量分析。将蛋白质组学技术应用于环境毒理学研究,可从蛋白水平上研究外源性化合物对机体的毒性作用机制,并从中筛选出具有较高特异型和高灵敏度的蛋白标志物,为污染物的健康风险评估提供新的技术手段。本文综述了蛋白质组学技术的主要研究方法、研究策略和在环境毒理学研究中的应用,重点讨论了蛋白质组学技术在重金属和有机化合物尤其是持久性有机污染物(POPs)毒性评估中的应用。
关键词: 重金属/
有机污染物/
空气污染/
蛋白质组学/
环境毒理学

Abstract:Proteomics is an important component of system biology. It studies the functions of proteins using holistic, dynamic and quantitative approaches, and systematically analyzes the protein expressions, protein-protein interactions and post-translational modifications in living organisms. The high throughput characteristics of proteomics technology make the efficient and rapid quantitative analysis of proteins possible. Proteomics has become an indispensable analytical tool in the post-genome era. The application of proteomics in environmental toxicology study can reveal the toxic mechanism of exogenous compounds on organism and screen highly sensitive protein markers, provide a new technology for assessment of environmental pollutants. In this review, the analytical methods, research strategies and applications of proteomics in environmental toxicology were introduced. The problems and future perspective of proteomic research were also discussed. This review focused on the application of proteomic approaches in risk assessment of toxicants such as heavy metals and organic compounds, especially persistent organic pollutants (POPs).
Key words:heavy metals/
organic pollutants/
air pollution/
proteomics/
environmental toxicology.

加载中
Schmidt C W. TOX21:New dimensions of toxicity testing[J]. Environmental Health Perspectives, 2009, 117(8):A348-A353
Andersen M E, Krewski D. Toxicity testing in the 21st Century:Bringing the vision to life[J]. Toxicological Sciences, 2009, 107(2):324-330
Kim M S, Pinto S M, Getnet D, et al. A draft map of the human proteome[J]. Nature, 2014, 509(7502):575-581
Pandey A, Mann M. Proteomics to study genes and genomes[J]. Nature, 2000, 405(6788):837-846
Mallick P, Kuster B. Proteomics:A pragmatic perspective[J]. Nature Biotechnology, 2010, 28:695
Wilkins M R, Pasquali C, Appel R D, et al. From proteins to proteomes:Large scale protein identification by two-dimensional electrophoresis and amino acid analysis[J]. Biotechnology, 1996, 14(1):61-65
Gorg A, Weiss W, Dunn M J. Current two-dimensional electrophoresis technology for proteomics[J]. Proteomics, 2004, 4(12):3665-3685
Wellner D, Panneerselvam C, Horecker B L. Sequencing of peptides and proteins with blocked N-terminal amino acids:N-acetylserine or N-acetylthreonine[J]. Proceedings of the National Academy of Sciences of the United States of America, 1990, 87(5):1947-1949
Fenn J B, Mann M, Meng C K, et al. Electrospray ionization for mass spectrometry of large biomolecules[J]. Science, 1989, 246(4926):64-71
Hillenkamp F, Karas M. Mass Spectrometry of Peptides and Proteins by Matrix-Assisted Ultraviolet Laser Desorption/Ionization[M]//Methods in Enzymology. Academic Press, 1990:280-295
Steen H, Mann M. The abc's (and xyz's) of peptide sequencing[J]. Nature Reviews Molecular Cell Biology, 2004, 5:699-711
Aebersold R, Mann M. Mass spectrometry-based proteomics[J]. Nature, 2003, 422(6928):198-207
Han X, Aslanian A, Yates J R. Mass spectrometry for proteomics[J]. Current Opinion in Chemical Biology, 2008, 12(5):483-490
Gallien S, Duriez E, Crone C, et al. Targeted proteomic quantification on quadrupole-Orbitrap mass spectrometer[J]. Molecular & Cellular Proteomics, 2012, 11(12):1709-1723
Chait B T. Mass spectrometry:Bottom-up or top-down?[J]. Science, 2006, 314(5796):65-66
Catherman A D, Skinner O S, Kelleher N L. Top down proteomics:Facts and perspectives[J]. Biochemical and Biophysical Research Communications, 2014, 445(4):683-693
Doerr A. Mass spectrometry-based targeted proteomics[J]. Nature Methods, 2012, 10:23
Company R, Ant ú nez O, Cosson R P, et al. Protein expression profiles in Bathymodiolus azoricus exposed to cadmium[J]. Ecotoxicology and Environmental Safety, 2019, 171:621-630
Lu Z, Wang S, Shan X, et al. Differential biological effects in two pedigrees of clam Ruditapes philippinarum exposed to cadmium using iTRAQ-based proteomics[J]. Environmental Toxicology and Pharmacology, 2019, 65:66-72
Luo L, Zhang Q, Kong X, et al. Differential effects of zinc exposure on male and female oysters (Crassostrea angulata) as revealed by label-free quantitative proteomics[J]. Environmental Toxicology and Chemistry, 2017, 36(10):2602-2613
Le V Q A, Ahn J Y, Heo M Y, et al. Proteomic profiles of Daphnia magna exposed to lead (Ⅱ) acetate trihydrate and atrazine[J]. Genes & Genomics, 2017, 39(8):887-895
Wang H, Wang S, Cui D, et al. iTRAQ-based proteomic technology revealed protein perturbations in intestinal mucosa from manganese exposure in rat models[J]. RSC Advances, 2017, 7(50):31745-31758
Lee E K, Shin Y J, Park E Y, et al. Selenium-binding protein 1:A sensitive urinary biomarker to detect heavy metal-induced nephrotoxicity[J]. Archives of Toxicology, 2017, 91(4):1635-1648
Zhao W J, Zhang Z J, Zhu Z Y, et al. Time-dependent response of A549 cells upon exposure to cadmium[J]. Journal of Applied Toxicology, 2018, 38(11):1437-1446
Kumar V, Karri V, Edwin M, et al. A system-based comparative proteomics approach to investigate heavy metals mixtures toxicity mechanism relates to the neurodegeneration on hippocampal cell line[J]. Toxicology Letters, 2018, 295(1):S203-S203
Karri V, Ramos D, Martinez J B, et al. Differential protein expression of hippocampal cells associated with heavy metals (Pb, As, and MeHg) neurotoxicity:Deepening into the molecular mechanism of neurodegenerative diseases[J]. Journal of Proteomics, 2018, 187:106-125
Chasapis C T, Andreini C, Georgiopolou A K, et al. Identification of the zinc, copper and cadmium metalloproteome of the protozoon Tetrahymena thermophila by systematic bioinformatics[J]. Archives of Microbiology, 2017, 199(8):1141-1149
Son J, Lee Y S, Lee S E, et al. Bioavailability and toxicity of copper, manganese, and nickel in Paronychiurus kimi (Collembola), and biomarker discovery for their exposure[J]. Archives of Environmental Contamination and Toxicology, 2017, 72(1):142-152
Baig M A, Ahmad J, Bagheri R, et al. Proteomic and ecophysiological responses of soybean (Glycine max L.) root nodules to Pb and Hg stress[J]. BMC Plant Biology, 2018, 18(1):283
Gutsch A, Keunen E, Guerriero G, et al. Long-term cadmium exposure influences the abundance of proteins that impact the cell wall structure in Medicago sativa stems[J]. Plant Biology, 2018, 20(6):1023-1035
Wang X H, Wang Q, Nie Z W, et al. Ralstonia eutropha Q2-8 reduces wheat plant above-ground tissue cadmium and arsenic uptake and increases the expression of the plant root cell wall organization and biosynthesis-related proteins[J]. Environmental Pollution, 2018, 242:1488-1499
Georgiadou E C, Kowalska E, Patla K, et al. Influence of heavy metals (Ni, Cu and Zn) on nitro-oxidative stress responses, proteome regulation and allergen production in basil (Ocimum basilicum L.) plants[J]. Frontiers in Plant Science, 2018, 9:862
Zeng F, Wu X, Qiu B, et al. Physiological and proteomic alterations in rice (Oryza sativa L.) seedlings under hexavalent chromium stress[J]. Planta, 2014, 240(2):291-308
Cheng Z W, Chen Z Y, Yan X, et al. Integrated physiological and proteomic analysis reveals underlying response and defense mechanisms of Brachypodium distachyon seedling leaves under osmotic stress, cadmium and their combined stresses[J]. Journal of Proteomics, 2018, 170:1-13
Liu H, Weisman D, Tang L, et al. Stress signaling in response to polycyclic aromatic hydrocarbon exposure in Arabidopsis thaliana involves a nucleoside diphosphate kinase, NDPK-3[J]. Planta, 2015, 241(1):95-107
Shen Y, Du J, Yue L, et al. Proteomic analysis of plasma membrane proteins in wheat roots exposed to phenanthrene[J]. Environmental Science and Pollution Research, 2016, 23(11):10863-10871
Shen Y, Li J, Gu R, et al. Proteomic analysis for phenanthrene-elicited wheat chloroplast deformation[J]. Environment International, 2019, 123:273-281
Skogland Enerstvedt K, Sydnes M O, Pampanin D M. Study of the plasma proteome of Atlantic cod (Gadus morhua):Effect of exposure to two PAHs and their corresponding diols[J]. Chemosphere, 2017, 183:294-304
Filis P, Walker N, Robertson L, et al. Long-term exposure to chemicals in sewage sludge fertilizer alters liver lipid content in females and cancer marker expression in males[J]. Environment International, 2019, 124:98-108
Nam T H, Jeon H J, Mo H H, et al. Determination of biomarkers for polycyclic aromatic hydrocarbons (PAHs) toxicity to earthworm (Eisenia fetida)[J]. Environmental Geochemistry and Health, 2015, 37(6):943-951
Shi X, Yeung L W Y, Lam P K S, et al. Protein profiles in zebrafish (Danio rerio) embryos exposed to perfluorooctane sulfonate[J]. Toxicological Sciences, 2009, 110(2):334-340
Dorts J, Kestemont P, Marchand P A, et al. Ecotoxicoproteomics in gills of the sentinel fish species, Cottus gobio, exposed to perfluorooctane sulfonate (PFOS)[J]. Aquatic Toxicology, 2011, 103(1):1-8
Roland K, Kestemont P, Hénuset L, et al. Proteomic responses of peripheral blood mononuclear cells in the European eel (Anguilla anguilla) after perfluorooctane sulfonate exposure[J]. Aquatic Toxicology, 2013, 128-129:43-52
Roland K, Kestemont P, Loos R, et al. Looking for protein expression signatures in European eel peripheral blood mononuclear cells after in vivo exposure to perfluorooctane sulfonate and a real world field study[J]. Science of the Total Environment, 2014, 468-469:958-967
Zhang Y Y, Tang L L, Zheng B, et al. Protein profiles of cardiomyocyte differentiation in murine embryonic stem cells exposed to perfluorooctane sulfonate[J]. Journal of Applied Toxicology, 2016, 36(5):726-740
Tan F, Jin Y, Liu W, et al. Global liver proteome analysis using iTRAQ labeling quantitative proteomic technology to reveal biomarkers in mice exposed to perfluorooctane sulfonate (PFOS)[J]. Environmental Science & Technology, 2012, 46(21):12170-12177
Huang Q, Zhang J, Peng S, et al. Proteomic analysis of perfluorooctane sulfonate-induced apoptosis in human hepatic cells using the iTRAQ technique[J]. Journal of Applied Toxicology, 2014, 34(12):1342-1351
Cui R, Zhang H, Guo X, et al. Proteomic analysis of cell proliferation in a human hepatic cell line (HL-7702) induced by perfluorooctane sulfonate using iTRAQ[J]. Journal of Hazardous Materials, 2015, 299:361-370
Li M, Huo X, Pan Y, et al. Proteomic evaluation of human umbilical cord tissue exposed to polybrominated diphenyl ethers in an e-waste recycling area[J]. Environment International, 2018, 111:362-371
Huang S, Cui Y, Guo X, et al. 2,2',4,4'-tetrabromodiphenyl ether disrupts spermatogenesis, impairs mitochondrial function and induces apoptosis of early leptotene spermatocytes in rats[J]. Reproductive Toxicology, 2015, 51:114-124
Song J, Li Z H, He Y T, et al. Decabrominated diphenyl ether (BDE-209) and/or BDE-47 exposure alters protein expression in purified neural stem/progenitor cells determined by proteomics analysis[J]. International Journal of Developmental Neuroscience, 2014, 33:8-14
Fong C C, Shi Y F, Yu W K, et al. iTRAQ-based proteomic profiling of the marine medaka (Oryzias melastigma) gonad exposed to BDE-47[J]. Marine Pollution Bulletin, 2014, 85(2):471-478
Ji C, Wu H, Wei L, et al. Proteomic and metabolomic analysis reveal gender-specific responses of mussel Mytilus galloprovincialis to 2,2',4,4'-tetrabromodiphenyl ether (BDE 47)[J]. Aquatic Toxicology, 2013, 140-141:449-457
Ji C, Wu H, Wei L, et al. Proteomic and metabolomic analysis of earth worm Eisenia fetida exposed to different concentrations of 2,2',4,4'-tetrabromodiphenyl ether[J]. Journal of Proteomics, 2013, 91:405-416
Rasinger J D, Carroll T S, Maranghi F, et al. Low dose exposure to HBCD, CB-153 or TCDD induces histopathological and hormonal effects and changes in brain protein and gene expression in juvenile female BALB/c mice[J]. Reproductive Toxicology, 2018, 80:105-116
Miller I, Serchi T, Cambier S, et al. Hexabromocyclododecane (HBCD) induced changes in the liver proteome of eu- and hypothyroid female rats[J]. Toxicology Letters, 2016, 245:40-51
Miller I, Diepenbroek C, Rijntjes E, et al. Gender specific differences in the liver proteome of rats exposed to short term and low-concentration hexabromocyclododecane (HBCD)[J]. Toxicological Research, 2016, 5(5):1273-1283
Rasinger J D, Carroll T S, Lundebye A K, et al. Cross-omics gene and protein expression profiling in juvenile female mice highlights disruption of calcium and zinc signalling in the brain following dietary exposure to CB-153, BDE-47, HBCD or TCDD[J]. Toxicology, 2014, 321:1-12
Senthil Kumar S, Muthuselvam P, Pugalenthi V, et al. Toxicoproteomic analysis of human lung epithelial cells exposed to steel industry ambient particulate matter (PM) reveals possible mechanism of PM related carcinogenesis[J]. Environmental Pollution, 2018, 239:483-492
Zhao C, Zhu L, Li R, et al. Omics approach reveals metabolic disorders associated with the cytotoxicity of airborne particulate matter in human lung carcinoma cells[J]. Environmental Pollution, 2019, 246:45-52
Huang Q, Zhang J, Peng S, et al. Effects of water soluble PM2.5 extracts exposure on human lung epithelial cells (A549):A proteomic study[J]. Journal of Applied Toxicology, 2014, 34(6):675-687
Luyten L J, Saenen N D, Janssen B G, et al. Air pollution and the fetal origin of disease:A systematic review of the molecular signatures of air pollution exposure in human placenta[J]. Environmental Research, 2018, 166:310-323
Hincal F. Effects of exposure to air pollution and smoking on the placental aryl hydrocarbon hydroxylase (AHH) activity[J]. Archives of Environmental Health, 1986, 41(6):377-383
Obolenskaya M Y, Teplyuk N M, Divi R L, et al. Human placental glutathione S-transferase activity and polycyclic aromatic hydrocarbon DNA adducts as biomarkers for environmental oxidative stress in placentas from pregnant women living in radioactivity- and chemically-polluted regions[J]. Toxicology Letters, 2010, 196(2):80-86
Sorkun H C, Bir F, Akbulut M, et al. The effects of air pollution and smoking on placental cadmium, zinc concentration and metallothionein expression[J]. Toxicology, 2007, 238(1):15-22
Kedryna T, Guminska M, Lucyna Z. Pyruvate kinase activity in the placentas of women living in polluted and unpolluted environments[J]. Medical Science Monitor, 2004, 10(12):CR672-CR678
Bahr B L, Price M D, Merrill D, et al. Different expression of placental pyruvate kinase in normal, preeclamptic and intrauterine growth restriction pregnancies[J]. Placenta, 2014, 35(11):883-890
Saenen N D, Vrijens K, Janssen B G, et al. Placental nitrosative stress and exposure to ambient air pollution during gestation:A population study[J]. American Journal of Epidemiology, 2016, 184(6):442-449
Weldy C S, Liu Y, Liggitt H D, et al. In utero exposure to diesel exhaust air pollution promotes adverse intrauterine conditions, resulting in weight gain, altered blood pressure, and increased susceptibility to heart failure in adult mice[J]. PLoS One, 2014, 9(2):e88582

相关话题/技术 环境 中国科学院 系统 副研究员