Volume 38 Issue 2
Mar.  2017
Turn off MathJax
Article Contents
Shi-Hao Wu, Zhi-Xing Liao, Joshua D. Rizak, Na Zheng, Lin-Heng Zhang, Hen Tang, Xiao-Bin He, Yang Wu, Xia-Ping He, Mei-Feng Yang, Zheng-Hui Li, Dong-Dong Qin, Xin-Tian Hu. Comparative study of the transfection efficiency of commonly used viral vectors in rhesus monkey (Macaca mulatta) brains. Zoological Research, 2017, 38(2): 88-95. doi: 10.24272/j.issn.2095-8137.2017.015
Citation: Shi-Hao Wu, Zhi-Xing Liao, Joshua D. Rizak, Na Zheng, Lin-Heng Zhang, Hen Tang, Xiao-Bin He, Yang Wu, Xia-Ping He, Mei-Feng Yang, Zheng-Hui Li, Dong-Dong Qin, Xin-Tian Hu. Comparative study of the transfection efficiency of commonly used viral vectors in rhesus monkey (Macaca mulatta) brains. Zoological Research, 2017, 38(2): 88-95. doi: 10.24272/j.issn.2095-8137.2017.015

Comparative study of the transfection efficiency of commonly used viral vectors in rhesus monkey (Macaca mulatta) brains

doi: 10.24272/j.issn.2095-8137.2017.015
Funds:  This study was supported by the National Program on Key Basic Research Project (973 Programs 2015CB755605) and the National Natural Science Foundation of China (81471312)
More Information
  • Corresponding author: Dong-Dong Qin, Xin-Tian Hu
  • Received Date: 2017-01-09
  • Rev Recd Date: 2017-03-03
  • Publish Date: 2017-03-18
  • Viral vector transfection systems are among the simplest of biological agents with the ability to transfer genes into the central nervous system. In brain research, a series of powerful and novel gene editing technologies are based on these systems. Although many viral vectors are used in rodents, their full application has been limited in non-human primates. To identify viral vectors that can stably and effectively express exogenous genes within non-human primates, eleven commonly used recombinant adeno-associated viral and lentiviral vectors, each carrying a gene to express green or red fluorescence, were injected into the parietal cortex of four rhesus monkeys. The expression of fluorescent cells was used to quantify transfection efficiency. Histological results revealed that recombinant adeno-associated viral vectors, especially the serotype 2/9 coupled with the cytomegalovirus, human synapsin I, or Ca2+/calmodulin-dependent protein kinase II promoters, and lentiviral vector coupled with the human ubiquitin C promoter, induced higher expression of fluorescent cells, representing high transfection efficiency. This is the first comparison of transfection efficiencies of different viral vectors carrying different promoters and serotypes in non-human primates (NHPs). These results can be used as an aid to select optimal vectors to transfer exogenous genes into the central nervous system of non-human primates.
  • loading
  • [1]
    Alexopoulou AN, Couchman JR, Whiteford JR. 2008. The CMV early enhancer/chicken β actin (CAG) promoter can be used to drive transgene expression during the differentiation of murine embryonic stem cells into vascular progenitors. BMC Cell Biology, 9:2.
    [2]
    Arhel N, Munier S, Souque P, Mollier K, Charneau P. 2006. Nuclear import defect of human immunodeficiency virus type 1 DNA flap mutants is not dependent on the viral strain or target cell type. Journal of Virology, 80(20):10262-10269.
    [3]
    Aschauer DF, Kreuz S, Rumpel S. 2013. Analysis of transduction efficiency, tropism and axonal transport of AAV serotypes 1, 2, 5, 6, 8 and 9 in the mouse brain. PLoS One, 8(9):e76310.
    [4]
    Bartlett JS, Samulski RJ, McCown TJ. 1998. Selective and rapid uptake of adeno-associated virus type 2 in brain. Human Gene Therapy, 9(8):1181-1186.
    [5]
    Bennett J. 2003. Immune response following intraocular delivery of recombinant viral vectors. Gene Therapy, 10(11):977-982.
    [6]
    Bish LT, Morine K, Sleeper MM, Sanmiguel J, Wu D, Gao GP, Wilson JM, Sweeney HL. 2008. Adeno-associated virus (AAV) serotype 9 provides global cardiac gene transfer superior to AAV1, AAV6, AAV7, and AAV8 in the mouse and rat. Human Gene Therapy, 19(12):1359-1368.
    [7]
    Burger C, Gorbatyuk OS, Velardo MJ, Peden CS, Williams P, Zolotukhin S, Reier PJ, Mandel RJ, Muzyczka N. 2004. Recombinant AAV viral vectors pseudotyped with viral capsids from serotypes 1, 2, and 5 display differential efficiency and cell tropism after delivery to different regions of the central nervous system. Molecular Therapy, 10(2):302-317.
    [8]
    Cearley CN, Wolfe JH. 2006. Transduction characteristics of adeno-associated virus vectors expressing cap serotypes 7, 8, 9, and Rh10 in the mouse brain. Molecular Therapy, 13(3):528-537.
    [9]
    Carter P & Samulski R. 2000. Adeno-associated viral vectors as gene delivery vehicles. International journal of molecular medicine, 6(1), 17-44.
    [10]
    Cockrell AS, Kafri T. 2007. Gene delivery by lentivirus vectors. Molecular Biotechnology, 36(3):184-204.
    [11]
    Costantini LC, Bakowska JC, Breakefield XO, Isacson O. 2000. Gene therapy in the CNS. Gene Therapy, 7(2):93-109.
    [12]
    Cronin J, Zhang XY, Reiser J. 2005. Altering the tropism of lentiviral vectors through pseudotyping. Current Gene Therapy, 5(4):387-398.
    [13]
    Davidoff AM, Gray JT, Ng CY, Zhang YB, Zhou JF, Spence Y, Bakar Y, Nathwani AC. 2005. Comparison of the ability of adeno-associated viral vectors pseudotyped with serotype 2, 5, and 8 capsid proteins to mediate efficient transduction of the liver in murine and nonhuman primate models. Molecular Therapy, 11(6):875-888.
    [14]
    Davidson BL, Stein CS, Heth JA, Martins I, Kotin RM, Derksen TA, Zabner J, Ghodsi A, Chiorini JA. 2000. Recombinant adeno-associated virus type 2, 4, and 5 vectors:transduction of variant cell types and regions in the mammalian central nervous system. Proceedings of the National Academy of Sciences of the United States of America, 97(7):3428-3432.
    [15]
    Davidson BL, Breakefield XO. 2003. Viral vectors for gene delivery to the nervous system. Nature Reviews Neuroscience, 4(5):353-364.
    [16]
    Dittgen T, Nimmerjahn A, Komai S, Licznerski P, Waters J, Margrie TW, Helmchen F, Denk W, Brecht M, Osten P. 2004. Lentivirus-based genetic manipulations of cortical neurons and their optical and electrophysiological monitoring in vivo. Proceedings of the National Academy of Sciences of the United States of America, 101(52):18206-18211.
    [17]
    Dodiya HB, Bjorklund T, Stansell J III, Mandel RJ, Kirik D, Kordower JH. 2010. Differential transduction following basal ganglia administration of distinct pseudotyped AAV capsid serotypes in nonhuman primates. Molecular Therapy, 18(3):579-587.
    [18]
    Elder GA, Gama Sosa MA, De Gasperi R. 2010. Transgenic mouse models of Alzheimer's disease. Mount Sinai Journal of Medicine:A Journal of Translational and Personalized Medicine, 77(1):69-81.
    [19]
    Fenno L, Yizhar O, Deisseroth K. 2011. The development and application of optogenetics. Annual Review of Neuroscience, 34(1):389-412.
    [20]
    Gao GP, Alvira MR, Wang L, Calcedo R, Johnston J, Wilson JM. 2002. Novel adeno-associated viruses from rhesus monkeys as vectors for human gene therapy. Proceedings of the National Academy of Sciences of the United States of America, 99(18):11854-11859.
    [21]
    Gerits A, Vancraeyenest P, Vreysen S, Laramée ME, Michiels A, Gijsbers R, Van den Haute C, Moons L, Debyser Z, Baekelandt V, Arckens L, Vanduffel W. 2015. Serotype-dependent transduction efficiencies of recombinant adeno-associated viral vectors in monkey neocortex. Neurophotonics, 2(3):031209.
    [22]
    Gruh I, Wunderlich S, Winkler M, Schwanke K, Heinke J, Blömer U, Ruhparwar A, Rohde B, Li RK, Haverich A. 2008. Human CMV immediate-early enhancer:a useful tool to enhance cell-type-specific expression from lentiviral vectors. The Journal of Gene Medicine, 10(1):21-32.
    [23]
    Hioki H, Kameda H, Nakamura H, Okunomiya T, Ohira K, Nakamura K, Kuroda M, Furuta T, Kaneko T. 2007. Efficient gene transduction of neurons by lentivirus with enhanced neuron-specific promoters. Gene Therapy, 14(11):872-882.
    [24]
    Jennings CG, Landman R, Zhou Y, Sharma J, Hyman J, Movshon JA, Qiu ZL, Roberts AC, Roe AW, Wang XQ, Zhou HH, Wang LP, Zhang F, Desimone R, Feng GP. 2016. Opportunities and challenges in modeling human brain disorders in transgenic primates. Nature Neuroscience, 19(9):1123-1130.
    [25]
    Kaplitt MG, Leone P, Samulski RJ, Xiao X, Pfaff DW, O'Malley KL, During MJ. 1994. Long-term gene expression and phenotypic correction using adeno-associated virus vectors in the mammalian brain. Nature Genetics, 8(2):148-154.
    [26]
    Kirik D, Rosenblad C, Burger C, Lundberg C, Johansen TE, Muzyczka N, Mandel RJ, Björklund A. 2002. Parkinson-like neurodegeneration induced by targeted overexpression of α-synuclein in the nigrostriatal system. The Journal of Neuroscience, 22(7):2780-2791.
    [27]
    Kügler S, Kilic E, Bähr M. 2003. Human synapsin 1 gene promoter confers highly neuron-specific long-term transgene expression from an adenoviral vector in the adult rat brain depending on the transduced area. Gene Therapy, 10(4):337-347.
    [28]
    Lai ZN, Brady RO. 2002. Gene transfer into the central nervous system in vivo using a recombinant lentivirus vector. Journal of Neuroscience Research, 67(3):363-371.
    [29]
    Lawlor PA, Bland RJ, Mouravlev A, Young D, During MJ. 2009. Efficient gene delivery and selective transduction of glial cells in the mammalian brain by AAV serotypes isolated from nonhuman primates. Molecular Therapy, 17(10):1692-1702.
    [30]
    Lewis P, Hensel M, Emerman M. 1992. Human immunodeficiency virus infection of cells arrested in the cell cycle. The EMBO Journal, 11(8):3053-3058.
    [31]
    Liu Z, Li X, Zhang JT, Cai YJ, Cheng TL, Cheng C, Wang Y, Zhang CC, Nie YH, Chen ZF, Bian WJ, Zhang L, Xiao JQ, Lu B, Zhang YF, Zhang XD, Sang X, Wu JJ, Xu X, Xiong ZQ, Zhang F, Yu X, Gong N, Zhou WH, Sun Q, Qiu ZL. 2016. Autism-like behaviours and germline transmission in transgenic monkeys overexpressing MeCP2. Nature, 530(7588):98-102.
    [32]
    Markakis EA, Vives KP, Bober J, Leichtle S, Leranth C, Beecham J, Elsworth JD, Roth RH, Samulski RJ, Redmond Jr DE. 2010. Comparative transduction efficiency of AAV vector serotypes 1-6 in the substantia nigra and striatum of the primate brain. Molecular Therapy, 18(3):588-593.
    [33]
    Mayford M, Baranes D, Podsypanina K, Kandel ER. 1996. The 3'-untranslated region of CaMKIIα is a cis-acting signal for the localization and translation of mRNA in dendrites. Proceedings of the National Academy of Sciences of the United States of America, 93(23):13250-13255.
    [34]
    Miller DG, Adam MA, Miller AD. 1990. Gene transfer by retrovirus vectors occurs only in cells that are actively replicating at the time of infection. Molecular and Cellular Biology, 10(8):4239-4242.
    [35]
    Mingozzi F, High KA. 2013. Immune responses to AAV vectors:overcoming barriers to successful Gene Therapy. Blood, 122(1):23-36.
    [36]
    Miyoshi H, Blömer U, Takahashi M, Gage FH, Verma IM. 1998. Development of a self-inactivating lentivirus vector. Journal of Virology, 72(10):8150-8157.
    [37]
    Naldini L, Blömer U, Gallay P, Ory D, Mulligan R, Gage FH, Verma IM, Trono D. 1996. In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector. Science, 272(5259):263-267.
    [38]
    Nayak S, Herzog RW. 2010. Progress and prospects:immune responses to viral vectors. Gene Therapy, 17(3):295-304.
    [39]
    Niu YY, Shen B, Cui YQ, Chen YC, Wang JY, Wang L, Kang Y, Zhao XY, Si W, Li W, Xiang AP, Zhou JK, Guo XJ, Bi Y, Si CY, Hu B, Dong GY, Wang H, Zhou ZM, Li TQ, Tan T, Pu XQ, Wang F, Ji SH, Zhou Q, Huang XX, Ji WZ, Sha JH. 2014. Generation of gene-modified cynomolgus monkey via Cas9/RNA-mediated gene targeting in one-cell embryos. Cell, 156(4):836-843.
    [40]
    Niu YY, Guo XY, Chen YC, Wang CE, Gao JQ, Yang WL, Kang Y, Si W, Wang H, Yang SH, Li S, Ji W, Li XJ. 2015. Early Parkinson's disease symptoms in α-synuclein transgenic monkeys. Human Molecular Genetics, 24(8):2308-2317.
    [41]
    Okano H, Hikishima K, Iriki A, Sasaki E. 2012. The common marmoset as a novel animal model system for biomedical and neuroscience research applications. Seminars in Fetal and Neonatal Medicine, 17(6):336-340.
    [42]
    Orth M, Tabrizi SJ. 2003. Models of Parkinson's disease. Movement Disorders, 18(7):729-737.
    [43]
    Paxinos G, Huang XF, Toga AW. 2000. The Rhesus Monkey Brain in Stereotaxic Coordinates. San Diego:Academic Press.
    [44]
    Rabinowitz JE, Rolling F, Li CW, Conrath H, Xiao WD, Xiao X, Samulski RJ. 2002. Cross-packaging of a single adeno-associated virus (AAV) type 2 vector genome into multiple AAV serotypes enables transduction with broad specificity. Journal of Virology, 76(2):791-801.
    [45]
    Ran FA, Hsu PD, Lin CY, Gootenberg JS, Konermann S, Trevino AE, Scott DA, Inoue A, Matoba S, Zhang Y, Zhang F. 2013. Double nicking by RNA-guided CRISPR Cas9 for enhanced genome editing specificity. Cell, 154(6):1380-1389.
    [46]
    Ridet JL, Bensadoun JC, Déglon N, Aebischer P, Zurn AD. 2006. Lentivirus-mediated expression of glutathione peroxidase:neuroprotection in murine models of Parkinson's disease. Neurobiology of Disease, 21(1):29-34.
    [47]
    Samaranch L, Salegio EA, San Sebastian W, Kells AP, Foust KD, Bringas JR, Lamarre C, Forsayeth J, Kaspar BK, Bankiewicz KS. 2012. Adeno-associated virus serotype 9 transduction in the central nervous system of nonhuman primates. Human Gene Therapy, 23(4):382-389.
    [48]
    Scheyltjens I, Laramée ME, Van den Haute C, Gijsbers R, Debyser Z, Baekelandt V, Vreysen S, Arckens L. 2015. Evaluation of the expression pattern of rAAV2/1, 2/5, 2/7, 2/8, and 2/9 serotypes with different promoters in the mouse visual cortex. Journal of Comparative Neurology, 523(14):2019-2042.
    [49]
    Schmidt M, Voutetakis A, Afione S, Zheng CY, Mandikian D, Chiorini JA. 2008. Adeno-associated virus type 12 (AAV12):a novel AAV serotype with sialic acid- and heparan sulfate proteoglycan-independent transduction activity. Journal of Virology, 82(3):1399-1406.
    [50]
    Schorpp M, Jäger R, Schellander K, Schenkel J, Wagner EF, Weiher H, Angel P. 1996. The human ubiquitin C promoter directs high ubiquitous expression of transgenes in mice. Nucleic Acids Research, 24(9):1787-1788.
    [51]
    Shultz S, Opie C, Atkinson QD. 2011. Stepwise evolution of stable sociality in primates. Nature, 479(7372):219-222.
    [52]
    Taymans JM, Vandenberghe LH, Van Den Haute C, Thiry I, Deroose CM, Mortelmans L, Wilson JM, Debyser Z, Baekelandt V. 2007. Comparative analysis of adeno-associated viral vector serotypes 1, 2, 5, 7, and 8 in mouse brain. Human Gene Therapy, 18(3):195-206.
    [53]
    Towne C, Schneider BL, Kieran D, Redmond Jr DE, Aebischer P. 2010. Efficient transduction of non-human primate motor neurons after intramuscular delivery of recombinant AAV serotype 6. Gene Therapy, 17(1):141-146.
    [54]
    Watakabe A, Ohtsuka M, Kinoshita M, Takaji M, Isa K, Mizukami H, Ozawa K, Isa T, Yamamori T. 2015. Comparative analyses of adeno-associated viral vector serotypes 1, 2, 5, 8 and 9 in marmoset, mouse and macaque cerebral cortex. Neuroscience Research, 93:144-157.
    [55]
    Weinberg MS, Samulski RJ, McCown TJ. 2013. Adeno-associated virus (AAV) Gene Therapy for neurological disease. Neuropharmacology, 69:82-88.
    [56]
    Wilhelm F, Winkler U, Morawski M, Jäger C, Reinecke L, Rossner MJ, Hirrlinger PG, Hirrlinger J. 2011. The human ubiquitin C promoter drives selective expression in principal neurons in the brain of a transgenic mouse line. Neurochemistry International, 59(6):976-980.
    [57]
    Wiznerowicz M, Trono D. 2005. Harnessing HIV for therapy, basic research and biotechnology. Trends in Biotechnology, 23(1):42-47.
    [58]
    Wong LF, Goodhead L, Prat C, Mitrophanous KA, Kingsman SM, Mazarakis ND. 2006. Lentivirus-mediated gene transfer to the central nervous system:therapeutic and research applications. Human Gene Therapy, 17(1):1-9.
    [59]
    Xiao WD, Chirmule N, Berta SC, McCullough B, Gao GP, Wilson JM. 1999. Gene therapy vectors based on adeno-associated virus type 1. Journal of Virology, 73(5):3994-4003.
    [60]
    Xiao X. 2003. Virus-based vectors for gene expression in mammalian cells:adeno-associated virus. New Comprehensive Biochemistry, 38:93-108.
    [61]
    Yang MF, Miao JY, Rizak J, Zhai RW, Wang ZB, Huma T, Li T, Zheng N, Wu SH, Zheng YW, Fan XN, Yang JZ, Wang JH, Yang SC, Ma YY, Lü LB, He RQ, Hu XT. 2014. Alzheimer's disease and methanol toxicity (part 2):lessons from four rhesus macaques (Macaca mulatta) chronically fed methanol. Journal of Alzheimer's Disease, 41(4):1131-1147.
    [62]
    Yang R, Wang QJ, Min LQ, Sui RB, Li J, Liu XW. 2013. Monosialoanglioside improves memory deficits and relieves oxidative stress in the hippocampus of rat model of Alzheimer's disease. Neurological Sciences, 34(8):1447-1451.
    [63]
    Zhang B. 2017. Consequences of early adverse rearing experience(EARE) on development:insights from non-human primate studies Zoological Research, 38(1):7-35.
    [64]
    Zincarelli C, Soltys S, Rengo G, Rabinowitz JE. 2008. Analysis of AAV serotypes 1-9 mediated gene expression and tropism in mice after systemic injection. Molecular Therapy, 16(6):1073-1080.
    [65]
    Zufferey R, Nagy D, Mandel RJ, Naldini L, Trono D. 1997. Multiply attenuated lentiviral vector achieves efficient gene delivery in vivo. Nature Biotechnology, 15(9):871-875.
  • 加载中

Catalog

    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    Article Metrics

    Article views (2140) PDF downloads(1512) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return