Adeno-Associated Viral-Mediated Gene Transfer
Gaetano Romano (gromano at temple dot edu)
Department of Biology, College of Science and Technology, Temple University, 1900 North 12th Street, Philadelphia, PA 19122, U.S.A.
DOI
//dx.doi.org/10.13070/mm.en.9.2796
Date
last modified : 2023-03-25; original version : 2019-07-16
Cite as
MATER METHODS 2019;9:2796
Abstract

An overview on adeno-associated viral (AAV) vectors

Introduction

AAV is a non-enveloped small single-stranded DNA virus, its genome is 4.7 kb long, belongs to the genera of the parvoviridae and has a life cycle of two phases: latent infection and lytic phase [1]. Interestingly, AAV is non-pathogenic in humans [2]. The AAV reproductive and lytic phases in mammalian cells require a helper virus, such as adenovirus, herpesvirus, or vaccinia virus [1]. After viral entry, human AAV integrates its genome into q arm of chromosome 19, between q13.3 and qter, which is also termed AAVS1 [3]. The AAV genome integration into the AAVS1 is safe and does not cause insertional mutagenesis events that may lead to carcinogenesis [2, 4], although such optimism is under scrutiny [5, 6].

Adeno-Associated Viral-Mediated Gene Transfer figure 1
Figure 1. A schematic representation of wild-type AAV and recombinant AAV-derived vector system.

AAV can also infect non-dividing cells and has a wide range of cell tropism [2]. The AAV genomic organization is relatively simple. Two inverted terminal repeats (ITRs) are situated at the 5’ and 3’end of the viral genome, which contains only two genes: Rep and Cap (Fig. 1). The ITRs contain the viral promoter and the signals that are responsible of the integration of the viral genome into the cellular chromosome [2]. Rep gene encodes four structural proteins termed Rep 78, Rep 68, Rep 52 and Rep 40, whereas Cap encodes three structural protein termed VP1, VP2 and VP3 [2]. Rep 68 and Rep 78 mediate the AAV genome integration into the safe AAVS1 site of the cellular chromosome [2]. Ogden PJ et al systematically evaluated the fitness landscape of mutant AAV2 Cap gene; AAV2 forms the first U.S. FDA-approved gene therapy [7]. Other experiments also used AAV2 [8]. More AAV-based gene therapies have been approved by FDA [9]. Chan KY et al optimized AAVs for specific gene delivery to central (AAV-PHP.eB) and peripheral (AAV-PHP.S) nervous systems [10]. Y Lu et al introduced OCT4, SOX2 and KLF4 as a polycistron in an AAV9 vector to regenerate retinal ganglion cells in mice [11]. Rauch JN et al infected mice to deliver shRNA against LRP1 with AAV-PHP.eB [12]. J Hordeaux et al integrated an microRNA 183 target sequence in AAV vectors to reduce the expression of transgenes in dorsal root ganglion, and thus reduce the dorsal root ganglion toxicity in AAV-based gene therapies [13].

Production of AAV-derived Vector Systems

The interest in AAV-mediated gene transfer is motivated by two major factors: the non-pathogenicity of the wild-type virus in humans and its ability to infect non-dividing cells [1, 2]. The engineering of AAV-derived vector systems only requires the removal of the Rep and Cap genes from the shuttle vector, which contains the two ITRs, an internal exogenous enhancer/promoter that drives the expression of the transgene and a poly A tail (Fig. 1). Rep and Cap genes are placed in a separate plasmid, which has to be co-transfected with the shuttle vector into packaging cells [2]. Following the two-plasmid co-transfection into packaging cells, an adenovirus infection is required to induce AAV lytic phase, which is induced by the adenoviral early E1 and E4 genes [2].

Main propertiesDrawbacks and potential adverse effects in therapy
  • AAV vectors can also transduce non-dividing cells.
  • High titers (1010 tu/ml).
  • Broad cell tropism.
  • Integration of AAV vector into the cell genome allows for a stable transgene expression.
  • Wild-type AAV is not pathogenic in humans.
  • Helper viruses are no longer needed for the production of AAV vectors.
  • AAV vector particles can be easily isolated with columns, instead of using CsCl.
  • Small genome (5 kb).
  • Various AAV serotypes are available for in vivo gene transfer applications (avoidance of humoral immune responses).
  • Limited capacity to accommodate transgenes (the maximum is roughly 4 kb), which can be remedied through dual AAVs with split inteins [14].
  • Insertional mutagenesis.
  • Inefficient for the transduction of stem cells.
  • Humoral immune responses neutralize AAV vector particles in vivo.
Table 1. Advantages and drawbacks of AAV-mediated gene transfer. (Abbreviation: tu/ml = transducing units per milliliter).

More recent protocols do not require the use of a helper virus to generate AAV-derived vector particles. These protocols utilize a plasmid encoding for the necessary helper virus factors, which is co-transfected into packaging cells, along with the shuttle vector and the plasmid that containing the Rep and Cap genes [15]. The possibility of using a three-plasmid co-transfection without the need of a helper virus has improved the clinical grade applications of AAV-derived vectors.

AAV vectors are commonly used in experiments involving brains or neurons. S Espinoza et al designed and injected into mouse brains an AAV9 vector expressing SINEUP RNA for GDNF expression [16]. Zhang X et al injected AAV2/9-CAG-DIO-taCasp3-TEVp, AAV2/9-hsyn-DIO-GCaMP6m, AAV9-EF1α-DIO-hM3D(Gq)-mCherry, AAV9-EF1α-DIO-hM4D(Gi)-mCherry, AAV9-EF1α-DIO-eYFP and AAV2/9-EF1α-DIO-ChR2-mCherry into the central nucleus of the amygdala and the paraventricular nucleus in mice to study the neuronal control of humoral immune responses in spleen [17]. Moya IM et al injected AAV8.TBG.PI.Cre.rBG from UPenn Core into mice to express Cre recombinase in hepatocytes [18]. Siciliano CA et al injected AAV5-CaMKIIα-eNpHR3.0-eYFP or AAV5-DIO-ChR2-eYFP, and their controls AAV5-CaMKIIα-eYFP and AAV5-DIO-eYFP into mouse medial prefrontal cortex to conduct optogenetic experiments [19]. Patzke C et al generated conditional synapsin-1 knockout human ES H1 cells with adeno-associated viruses through cre-recombination [20]. Marshel JH et al transduced neurons with an adeno-associated viral vector carrying enhanced YFP and optogenetic gene ChRmine along with the trafficking sequence, ER export signal, and the CaMKIIalpha promoter [21]. Szőnyi A et al injected into mouse brains AAV2/1-EF1a-DIO-GCaMP6f, AAV2/5-EF1αDIO-eYFP, AAV2/5-EF1α-DIO-mCherry, AAV2/5-CAG-FLEX-ArchT-GFP, AAV2/5-EF1α-DIO-hChR2(H134R)-eYFP for anterograde tracing and optogenetic experiments to study the role of brainstem nucleus incertus GABAergic cells in contextual memory formation [22]. Marvin JS et al transfected HEK293 cells with the adeno-associated virus (AAV) plasmid coding for the glutamate sensor iGluSnFR, helper plasmids encoding rep and cap genes (pRV1 and pH21), and adenoviral helper pFΔ6 from Stratagene using the calcium phosphate transfection method, and purified the virus particles from cell lysate by HiTrap heparin HP columns from GE Healthcare [23]. Zott B et al injected these kinds of GluSnFr viral constructs into mouse hippocampal CA1 regions to study the effect of beta-amyloid peptides on glutamate levels in brain neurons in vivo [24]. AAV2/5 serotype is thought to infect astrocytes preferentially in mouse brains [25]. M Aubert et al evaluated the infectiveness of four serotypes: AAV1, AAV8, AAV-PHP.S, and AAV-Rh10 among neurons of the peripheral nerve system [26].

It is of interest to note that multiplicity of Infection/MOI or AAV titer differs up to an order when determined by two different appraches: ddPCR and qPCR [27].

Organization Sample reference
AddGeneAAV9-Syn-GCaMP6 [28] ; pAAV-CBA-DIO-hM4Di-mCherry, AAV-CMV-LOX-STOP-LOX-mG-CaMP3.0 [29] ; AAV2/8-hSyn-DIOmCherry, AAV2/8-hSyn-DIO-hM3D(Gq)-mCherry [30] ; AAV9-CAG-DIO-tdTomato-WPRE-bGH, AAV9-EF1-DIO-hChR2(H134R)-mCherry-WPRE-hGH [31]
UNC Vector CoreAAV2/5-EF1α-DIO-eYFP, AAV2/5-EF1α-DIO-mCherry, AAV2/5-EF1α-DIO-hChR2(H134R)-eYFP; AAV2/5-CAG-FLEX-ArchT-GFP [30] ; AAV2/5-EF1αDIO-eYFP, AAV2/5-EF1α-DIO-mCherry, AAV2/5-CAG-FLEX-ArchT-GFP [22] ; AAV1-EF1a-FLEX-TVAmCherry [32], AAV5-DIO-ChR2-EYFP [33]
Salk GT3 CoreAAV2/8-hSyn-FLEX-TVA-p2A-eGFP-p2AoG [30]
Stanford Viral CoreAAV8-EF1a-fDIO-GCAMP6m-WPRE-SV40 [31]
Boston Children Viral CoreAAV9-hSyn-DIO-hM3D(Gq)-mCherry, AAV9-CAG-DIO-ChR2(H134R)-mCherry-WPRE-hGH [31]
Janelia Viral CoreAAV9-CAG-DIO-ChR2(H134R)-mCherry-WPRE-hGH [31] ; AAV1-G(N2C)-mKate [32]
TIGEM AAV Vector CorepAAV2.1 or pZac with inteins [34]
UPenn Vector CorepAAV2/9, pAAV2/hu68 [13], AAV8.TBG.PI.Cre.rBG [18], AAV1.hSyn.Cre.WPRE.hGH [24], AAV2/5-EF1α-DIO-hChR2(H134R)-eYFP [22]
Vector BiolabsAAV9-tMCK-eGFP-WPRE etc [35]
Canadian Neurophotonics Platform Viral Vector Core FacilityAAV2/9-CAG-DIO-chABC etc [36]
Table 2. List of companies and organizations that provide the components for the production of AAV-derived vectors.

The advantages and drawbacks of AAV-mediated gene transfer are reported in Table 1. Two main shortcomings of AAV-derived vectors are related to the limited capacity to accommodate transgenes, which is in the range of 4 kb, and insertional mutagenesis [2]. Dual AAVs with split inteins can remedy the limited capacity issue [14]. Unfortunately, AAV-derived vectors integrate their genomes randomly into cellular chromosomes, in contrast to wild-type AAV [4]. Some studies reported that AAV-mediated gene transfer induced hepatocellular carcinomas and angiosarcomas in mice [4]. Worryingly, a number of studies showed that the random integration of AAV-derived vectors occurs preferentially at actively transcribed genes of the target cell genome, which, in turn, may lead to critical DNA damage and/or silencing of tumor suppressor genes [4].

A list of companies and organization that provide the components for the production of AAV-derived vectors is reported in Table 2. Some of them also publish standardized protocols such as Salk GT3 Core [37].

Titers and Purity of AAV-Derived Vector Particles
Company Type of assay Reference
Addgene qPCR
Next generation sequencing (NGS)
Azura Genomics qPCR
Bio-Rad ddPCR
NGS
Biotium Fluorescence-mediated viral nucleic acid detection
Cell Biolabs
Fluorescence-mediated viral nucleic acid detection QuickTiter AAV Quantitation Kit [25]
GeneCopoeia AAV-encoded GFP
NGS
Gyrolab ELISA
Lumiprobe Fluorescence-mediated viral nucleic acid detection (PicoGreen)
Malvern Panalytical Dynamic light scattering
Precision Medicine ddPCR
Progen ELISA
Takara / Clontech qPCR AAVpro Titration Kit [14, 38]
Vector Biolabs AAV-encoded GFP
Vigene Biosciences AAV-encoded GFP
Wyatt Technology Dynamic light scattering
Table 1. List of companies that provide assays measuring the titers of AAV-derived vectors.

The quantification of AAV-derived vector particles can help control and optimize the efficiency of AAV-mediated gene transfer into mammalian cells [15]. Several systems are used to assess the titers of AAV vector stocks, such as dot-blot hybridization [39], Southern blot [40], UV-based spectrophotometry [41], enzyme-linked immunosorbent assay (ELISA) [42], PicoGreen-based fluorimetry [43], quantitative real-time polymerase chain reaction (qPCR) [44] and droplet digital PCR (ddPCR) [45]. UV-based spectrophotometry measures the UV absorbance of denatured AAV-derived vector particles in solution [41]. The fluorescent dye PicoGreen stains the encapsidated AAV transfer vectors genome [43]. Droplet digital PCR (ddPCR) does not require a standard curve, as it can quantify directly and with high precision specific types of DNA copies [45]. Dynamic light scattering can assess the size of AAV-based vector particles and determine if some particles have formed aggregates. Next-generation sequencing (NGS) can examine the possible mis-incorporation of genetic elements inside AAV-derived vector particles [46, 47]. A number of assays are commercially available and are listed in Table 3.

Some companies provide purified AAV-derived vector particles encoding green fluorescent protein (GFP), which can be utilized to assess the efficiency of AAV-mediated gene transfer in a variety of mammalian cell types (Table 3). GFP is encoded in different serotypes of AAV-based vectors, in order to compare their gene transduction efficiency in particular cell types.

Update (March 2023).

Studies are currently underway for the full characterization of in vivo transduction properties of various subtypes of AAV-based vector systems. This may require the application of spatial transcriptomics, which is an emerging technology that allows for the readout of thousands of mRNA sequences in tissue sections, without altering the overall context of the three-dimensional structure of the analyzed tissues [48-61]. Indeed, spatial transcriptomics can also be utilized for the three-dimensional analyses of viral vectors-encoded transgenes among various types of transduced cells and/or tissues, with a particular emphasis on the central nervous system [62-65].

A recent report has improved and characterized a promising ultrasensitive system, which was based on sequential fluorescence in situ hybridization (USeqFISH), to determine the three-dimensional transcriptomic profiling of endogenous and viral vector-encoded mRNA with a short barcode [66]. The study was conducted in the following animal models: mouse, marmoset and rhesus macaque. Single doses of AAV-derived vector particles were injected into the central nervous systems of the animals. A pool of six serotypes AAV serotypes was used in the experiments. The AAV-derived vectors pool included a new variant, termed AAV-PHP.AX, which has shown a particular efficiency in transducing neurons and astrocytes in the in vivo system [66-68]. The AAV-encoded mRNAs are typically abundant in transduced cell populations and may vary from 10 to 100 transcripts per cell [69]. The copy numbers of AAV-encoded transcripts are comparable to the expression levels of endogenous cellular genes. Usually, the spatial transcriptomics modalities involve dozens of probes per mRNA target to ensure an adequate specificity and signal intensity, which may require a rather long barcode ranging from 0.5 to 1 kilobase. The length of the barcode may pose an obstacle for the detection of AAV-encoded mRNA, due to the limited packaging capacity of AAV vector particles, which is lower than 4.7 kb. With the intent to reduce the length of the probes for the specific detection of the AAV-encoded mRNA barcode, USeqFISH combines signal amplification with sequential labeling to produce brighter and more sensitive mRNA-associated signals than conventional amplification systems for spatial transcriptomic analysis. The signal amplification can be based either on rolling-circle amplification (RCA) [70], or hybridization chain reaction (HCR) [71]. The increment of the signal sensitivity results in the use of just four, or even less, labelled oligonucleotide probes for the detection of each target gene, along with cell type-specific markers, in different regions of the mammalian brain [59, 60, 72]. Signal amplification systems combined with sequential labeling with shorter mRNA barcodes may allow for the simultaneous readout of approximately 50 genes in brain tissue sections.

References
  1. Blacklow N, Hoggan M, Kapikian A, Austin J, Rowe W. Epidemiology of adenovirus-associated virus infection in a nursery population. Am J Epidemiol. 1968;88:368-78 pubmed
  2. Romano G, Michell P, Pacilio C, Giordano A. Latest developments in gene transfer technology: achievements, perspectives, and controversies over therapeutic applications. Stem Cells. 2000;18:19-39 pubmed
  3. Romano G. Gene transfer in experimental medicine. Drug News Perspect. 2003;16:267-76 pubmed
  4. Romano G, Marino I, Pentimalli F, Adamo V, Giordano A. Insertional mutagenesis and development of malignancies induced by integrating gene delivery systems: implications for the design of safer gene-based interventions in patients. Drug News Perspect. 2009;22:185-96 pubmed publisher
  5. Kaiser J. How safe is a popular gene therapy vector?. Science. 2020;367:131 pubmed publisher
  6. Nguyen G, Everett J, Kafle S, Roche A, Raymond H, Leiby J, et al. A long-term study of AAV gene therapy in dogs with hemophilia A identifies clonal expansions of transduced liver cells. Nat Biotechnol. 2020;: pubmed publisher
  7. Ogden P, Kelsic E, Sinai S, Church G. Comprehensive AAV capsid fitness landscape reveals a viral gene and enables machine-guided design. Science. 2019;366:1139-1143 pubmed publisher
  8. Tsai N, Wang F, Toma K, Yin C, Takatoh J, Pai E, et al. Trans-Seq maps a selective mammalian retinotectal synapse instructed by Nephronectin. Nat Neurosci. 2022;25:659-674 pubmed publisher
  9. FDA Approves First Gene Therapy for the Treatment of High-Risk, Non-Muscle-Invasive Bladder Cancer. Available from: www.fda.gov/news-events/press-announcements/fda-approves-first-gene-therapy-treatment-high-risk-non-muscle-invasive-bladder-cancer
  10. Chan K, Jang M, Yoo B, Greenbaum A, Ravi N, Wu W, et al. Engineered AAVs for efficient noninvasive gene delivery to the central and peripheral nervous systems. Nat Neurosci. 2017;20:1172-1179 pubmed publisher
  11. Lu Y, Brommer B, Tian X, Krishnan A, Meer M, Wang C, et al. Reprogramming to recover youthful epigenetic information and restore vision. Nature. 2020;588:124-129 pubmed publisher
  12. Rauch J, Luna G, Guzman E, Audouard M, Challis C, Sibih Y, et al. LRP1 is a master regulator of tau uptake and spread. Nature. 2020;580:381-385 pubmed publisher
  13. Hordeaux J, Buza E, Jeffrey B, Song C, Jahan T, Yuan Y, et al. MicroRNA-mediated inhibition of transgene expression reduces dorsal root ganglion toxicity by AAV vectors in primates. Sci Transl Med. 2020;12: pubmed publisher
  14. Levy J, Yeh W, Pendse N, Davis J, Hennessey E, Butcher R, et al. Cytosine and adenine base editing of the brain, liver, retina, heart and skeletal muscle of mice via adeno-associated viruses. Nat Biomed Eng. 2020;4:97-110 pubmed publisher
  15. Romano G. Current development of adeno-associated viral vectors. Drug News Perspect. 2005;18:311-6 pubmed
  16. Espinoza S, Scarpato M, Damiani D, Managò F, Mereu M, Contestabile A, et al. SINEUP Non-coding RNA Targeting GDNF Rescues Motor Deficits and Neurodegeneration in a Mouse Model of Parkinson's Disease. Mol Ther. 2019;: pubmed publisher
  17. Zhang X, Lei B, Yuan Y, Zhang L, Hu L, Jin S, et al. Brain control of humoral immune responses amenable to behavioural modulation. Nature. 2020;581:204-208 pubmed publisher
  18. Moya I, Castaldo S, Van den Mooter L, Soheily S, Sansores Garcia L, Jacobs J, et al. Peritumoral activation of the Hippo pathway effectors YAP and TAZ suppresses liver cancer in mice. Science. 2019;366:1029-1034 pubmed publisher
  19. Siciliano C, Noamany H, Chang C, Brown A, Chen X, Leible D, et al. A cortical-brainstem circuit predicts and governs compulsive alcohol drinking. Science. 2019;366:1008-1012 pubmed publisher
  20. Patzke C, Brockmann M, Dai J, Gan K, Grauel M, Fenske P, et al. Neuromodulator Signaling Bidirectionally Controls Vesicle Numbers in Human Synapses. Cell. 2019;179:498-513.e22 pubmed publisher
  21. Marshel J, Kim Y, Machado T, Quirin S, Benson B, Kadmon J, et al. Cortical layer-specific critical dynamics triggering perception. Science. 2019;365: pubmed publisher
  22. Szonyi A, Sos K, Nyilas R, Schlingloff D, Domonkos A, Takács V, et al. Brainstem nucleus incertus controls contextual memory formation. Science. 2019;364: pubmed publisher
  23. Marvin J, Scholl B, Wilson D, Podgorski K, Kazemipour A, Müller J, et al. Stability, affinity, and chromatic variants of the glutamate sensor iGluSnFR. Nat Methods. 2018;15:936-939 pubmed publisher
  24. Zott B, Simon M, Hong W, Unger F, Chen Engerer H, Frosch M, et al. A vicious cycle of β amyloid-dependent neuronal hyperactivation. Science. 2019;365:559-565 pubmed publisher
  25. Wu Z, Parry M, Hou X, Liu M, Wang H, Cain R, et al. Gene therapy conversion of striatal astrocytes into GABAergic neurons in mouse models of Huntington's disease. Nat Commun. 2020;11:1105 pubmed publisher
  26. Aubert M, Strongin D, Roychoudhury P, Loprieno M, Haick A, Klouser L, et al. Gene editing and elimination of latent herpes simplex virus in vivo. Nat Commun. 2020;11:4148 pubmed publisher
  27. Vaidyanathan S, Salahudeen A, Sellers Z, Bravo D, Choi S, Batish A, et al. High-Efficiency, Selection-free Gene Repair in Airway Stem Cells from Cystic Fibrosis Patients Rescues CFTR Function in Differentiated Epithelia. Cell Stem Cell. 2020;26:161-171.e4 pubmed publisher
  28. Badimon A, Strasburger H, Ayata P, Chen X, Nair A, Ikegami A, et al. Negative feedback control of neuronal activity by microglia. Nature. 2020;: pubmed publisher
  29. Qian H, Kang X, Hu J, Zhang D, Liang Z, Meng F, et al. Reversing a model of Parkinson's disease with in situ converted nigral neurons. Nature. 2020;582:550-556 pubmed publisher
  30. Szonyi A, Zichó K, Barth A, Gönczi R, Schlingloff D, Török B, et al. Median raphe controls acquisition of negative experience in the mouse. Science. 2019;366: pubmed publisher
  31. Bai L, Mesgarzadeh S, Ramesh K, Huey E, Liu Y, Gray L, et al. Genetic Identification of Vagal Sensory Neurons That Control Feeding. Cell. 2019;179:1129-1143.e23 pubmed publisher
  32. Zhang J, Jin H, Zhang W, Ding C, O Keeffe S, Ye M, et al. Sour Sensing from the Tongue to the Brain. Cell. 2019;179:392-402.e15 pubmed publisher
  33. Adaikkan C, Middleton S, Marco A, Pao P, Mathys H, Kim D, et al. Gamma Entrainment Binds Higher-Order Brain Regions and Offers Neuroprotection. Neuron. 2019;102:929-943.e8 pubmed publisher
  34. Tornabene P, Trapani I, Minopoli R, Centrulo M, Lupo M, de Simone S, et al. Intein-mediated protein trans-splicing expands adeno-associated virus transfer capacity in the retina. Sci Transl Med. 2019;11: pubmed publisher
  35. Palla A, Ravichandran M, Wang Y, Alexandrova L, Yang A, Kraft P, et al. Inhibition of prostaglandin-degrading enzyme 15-PGDH rejuvenates aged muscle mass and strength. Science. 2020;: pubmed publisher
  36. Tansley S, Gu N, Guzmán A, Cai W, Wong C, Lister K, et al. Microglia-mediated degradation of perineuronal nets promotes pain. Science. 2022;:eabl6773 pubmed publisher
  37. Kataoka N, Shima Y, Nakajima K, Nakamura K. A central master driver of psychosocial stress responses in the rat. Science. 2020;367:1105-1112 pubmed publisher
  38. Choi C, Park J, Kim H, Chang K, Park J, MIN K. DSCR1 upregulation enhances dural meningeal lymphatic drainage to attenuate amyloid pathology of Alzheimer's disease. J Pathol. 2021;255:296-310 pubmed publisher
  39. Samulski R, Chang L, Shenk T. Helper-free stocks of recombinant adeno-associated viruses: normal integration does not require viral gene expression. J Virol. 1989;63:3822-8 pubmed
  40. McCarty D, Monahan P, Samulski R. Self-complementary recombinant adeno-associated virus (scAAV) vectors promote efficient transduction independently of DNA synthesis. Gene Ther. 2001;8:1248-54 pubmed
  41. Sommer J, Smith P, Parthasarathy S, Isaacs J, Vijay S, Kieran J, et al. Quantification of adeno-associated virus particles and empty capsids by optical density measurement. Mol Ther. 2003;7:122-8 pubmed
  42. Sondhi D, Peterson D, Giannaris E, Sanders C, Mendez B, De B, et al. AAV2-mediated CLN2 gene transfer to rodent and non-human primate brain results in long-term TPP-I expression compatible with therapy for LINCL. Gene Ther. 2005;12:1618-32 pubmed
  43. Piedra J, Ontiveros M, Miravet S, Penalva C, Monfar M, Chillon M. Development of a rapid, robust, and universal picogreen-based method to titer adeno-associated vectors. Hum Gene Ther Methods. 2015;26:35-42 pubmed publisher
  44. D Costa S, Blouin V, Broucque F, Penaud Budloo M, Francois A, Perez I, et al. Practical utilization of recombinant AAV vector reference standards: focus on vector genomes titration by free ITR qPCR. Mol Ther Methods Clin Dev. 2016;5:16019 pubmed publisher
  45. Lock M, Alvira M, Chen S, Wilson J. Absolute determination of single-stranded and self-complementary adeno-associated viral vector genome titers by droplet digital PCR. Hum Gene Ther Methods. 2014;25:115-25 pubmed publisher
  46. Saveliev A, Liu J, Li M, Hirata L, Latshaw C, Zhang J, et al. Accurate and Rapid Sequence Analysis of Adeno-Associated Virus Plasmids by Illumina Next-Generation Sequencing. Hum Gene Ther Methods. 2018;: pubmed publisher
  47. Stutika C, Gogol Döring A, Botschen L, Mietzsch M, Weger S, Feldkamp M, et al. A Comprehensive RNA Sequencing Analysis of the Adeno-Associated Virus (AAV) Type 2 Transcriptome Reveals Novel AAV Transcripts, Splice Variants, and Derived Proteins. J Virol. 2016;90:1278-89 pubmed publisher
  48. Ståhl P, Salmén F, Vicković S, Lundmark A, Navarro J, Magnusson J, et al. Visualization and analysis of gene expression in tissue sections by spatial transcriptomics. Science. 2016;353:78-82 pubmed publisher
  49. Eng C, Lawson M, Zhu Q, Dries R, Koulena N, Takei Y, et al. Transcriptome-scale super-resolved imaging in tissues by RNA seqFISH. Nature. 2019;568:235-239 pubmed publisher
  50. Chen K, Boettiger A, Moffitt J, Wang S, Zhuang X. RNA imaging. Spatially resolved, highly multiplexed RNA profiling in single cells. Science. 2015;348:aaa6090 pubmed publisher
  51. Rodriques S, Stickels R, Goeva A, Martin C, Murray E, Vanderburg C, et al. Slide-seq: A scalable technology for measuring genome-wide expression at high spatial resolution. Science. 2019;363:1463-1467 pubmed publisher
  52. Lee J, Daugharthy E, Scheiman J, Kalhor R, Yang J, Ferrante T, et al. Highly multiplexed subcellular RNA sequencing in situ. Science. 2014;343:1360-3 pubmed publisher
  53. Cho C, Xi J, Si Y, Park S, Hsu J, Kim M, et al. Microscopic examination of spatial transcriptome using Seq-Scope. Cell. 2021;184:3559-3572.e22 pubmed publisher
  54. Ortiz C, Carl xe9 n M, Meletis K. Spatial Transcriptomics: Molecular Maps of the Mammalian Brain. Annu Rev Neurosci. 2021;44:547-562 pubmed publisher
  55. Rao A, Barkley D, França G, Yanai I. Exploring tissue architecture using spatial transcriptomics. Nature. 2021;596:211-220 pubmed publisher
  56. Wang X, Allen W, Wright M, Sylwestrak E, Samusik N, Vesuna S, et al. Three-dimensional intact-tissue sequencing of single-cell transcriptional states. Science. 2018;361: pubmed publisher
  57. Chen F, Wassie A, Cote A, Sinha A, Alon S, Asano S, et al. Nanoscale imaging of RNA with expansion microscopy. Nat Methods. 2016;13:679-84 pubmed publisher
  58. Alon S, Goodwin D, Sinha A, Wassie A, Chen F, Daugharthy E, et al. Expansion sequencing: Spatially precise in situ transcriptomics in intact biological systems. Science. 2021;371: pubmed publisher
  59. Codeluppi S, Borm L, Zeisel A, La Manno G, van Lunteren J, Svensson C, et al. Spatial organization of the somatosensory cortex revealed by osmFISH. Nat Methods. 2018;15:932-935 pubmed publisher
  60. Wang Y, Eddison M, Fleishman G, Weigert M, Xu S, Wang T, et al. EASI-FISH for thick tissue defines lateral hypothalamus spatio-molecular organization. Cell. 2021;184:6361-6377.e24 pubmed publisher
  61. Gyllborg D, Langseth C, Qian X, Choi E, Salas S, Hilscher M, et al. Hybridization-based in situ sequencing (HybISS) for spatially resolved transcriptomics in human and mouse brain tissue. Nucleic Acids Res. 2020;48:e112 pubmed publisher
  62. Nectow A, Nestler E. Viral tools for neuroscience. Nat Rev Neurosci. 2020;21:669-681 pubmed publisher
  63. Bedbrook C, Deverman B, Gradinaru V. Viral Strategies for Targeting the Central and Peripheral Nervous Systems. Annu Rev Neurosci. 2018;41:323-348 pubmed publisher
  64. Zhu D, Schieferecke A, Lopez P, Schaffer D. Adeno-Associated Virus Vector for Central Nervous System Gene Therapy. Trends Mol Med. 2021;27:524-537 pubmed publisher
  65. Wang D, Tai P, Gao G. Adeno-associated virus vector as a platform for gene therapy delivery. Nat Rev Drug Discov. 2019;18:358-378 pubmed publisher
  66. Jang M, Coughlin G, Jackson C, Chen X, Chuapoco M, Vendemiatti J, et al. Spatial transcriptomics for profiling the tropism of viral vectors in tissues. Nat Biotechnol. 2023;: pubmed publisher
  67. Ravindra Kumar S, Miles T, Chen X, Brown D, Dobreva T, Huang Q, et al. Multiplexed Cre-dependent selection yields systemic AAVs for targeting distinct brain cell types. Nat Methods. 2020;17:541-550 pubmed publisher
  68. Deverman B, Pravdo P, Simpson B, Kumar S, Chan K, Banerjee A, et al. Cre-dependent selection yields AAV variants for widespread gene transfer to the adult brain. Nat Biotechnol. 2016;34:204-9 pubmed publisher
  69. Brown D, Altermatt M, Dobreva T, Chen S, WANG A, Thomson M, et al. Deep Parallel Characterization of AAV Tropism and AAV-Mediated Transcriptional Changes via Single-Cell RNA Sequencing. Front Immunol. 2021;12:730825 pubmed publisher
  70. Larsson C, Grundberg I, Söderberg O, Nilsson M. In situ detection and genotyping of individual mRNA molecules. Nat Methods. 2010;7:395-7 pubmed publisher
  71. Choi H, Schwarzkopf M, Fornace M, Acharya A, Artavanis G, Stegmaier J, et al. Third-generation in situ hybridization chain reaction: multiplexed, quantitative, sensitive, versatile, robust. Development. 2018;145: pubmed publisher
  72. Sun Y, Chen X, Fischer S, Lu S, Zhan H, Gillis J, et al. Integrating barcoded neuroanatomy with spatial transcriptional profiling enables identification of gene correlates of projections. Nat Neurosci. 2021;24:873-885 pubmed publisher
ISSN : 2329-5139