Abstract
Adeno-associated virus (AAV) has emerged as a pivotal tool in neuroscience research, owing to its remarkable versatility and efficiency in delivering genetic material to diverse cell types within the nervous system. This mini review aims to underscore the advanced applications of AAV vectors in neuroscience and their profound potential to revolutionize our understanding of brain function and therapeutic interventions for neurological disorders. By providing a concise overview of the latest developments and strategies employing AAV vectors, this review illuminates the transformative role of AAV technology in unraveling the complexities of neural circuits and paving the way for innovative treatments. Through elucidating the multifaceted capabilities of AAV-mediated gene delivery, this review underscores its pivotal role as a cornerstone in contemporary neuroscience research, promising remarkable insights into the intricacies of brain biology and offering new avenues for therapeutic intervention.
Keywords: Adeno-associated virus, Viral vectors, Neuroscience
AAV was initially discovered as a contaminant of adenovirus stocks in the 1960s. Today, AAVs are currently among the most frequently used viral vectors for gene delivery to the brain due to the lack of cytotoxicity and the long-term expression in neurons [1], moreover they are an attractive candidate for creating viral vectors for gene therapy due to their ability to infect humans [2]. Remarkably, AAV causes a very mild immune response, making it a great candidate for gene therapy with minimal risk of adverse effects. AAV has a linear single strand DNA genome of approximately 4.7 kb in length. Lacking its own polymerase, the AAV genome relies on cellular polymerases for replication. Although AAV wide distribution, its virtually non-existent pathogenicity can be attributed to its inability to replicate on its own. Instead, they necessitate a co-infecting virus capable of replication to initiate a productive infection. In its genome are present only replication and capsid genes for viral replication regulation and capsid structure [3]. During infection cycle AAV enters the cell through receptor-mediated endocytosis and it is transported to the nucleus via clathrin coated vesicle. Once within the nucleus the virion capsid is lost and the viral genome released, where it can either integrate into the host cell genome (in the absence of a co-infected virus) or proceed to lytic cycle (in presence of co-infected virus) [4].
AAV-mediated gene delivery
Unlike conventional tracers, neurotropic viruses carry information encoded by nucleic acids, they can be genetically engineered for specific desired properties and can be used to introduce foreign genes that can be used to easily detect their presence or, more powerfully, to directly monitor or manipulate neuronal function. Viral-mediated approach enabled space- (targeting a specific brain region) and time-specific approaches (targeting any stage of animal’s life). AAVs are used in neuroscience to efficiently get genetic material into a cell, and even deliver this genetic material to specific cells in an organism.
Depending on experiment aims, researchers can choose the delivery modality of viral particles, either by direct injection into specific anatomic brain regions [5, 6] or via a widespread route [7, 8] for a global viral expression (Table 1). Stereotaxic injection enables infection of a restricted and precise brain area, thereby reducing potential off-target effects, while intravenous and retro-orbital injections (both employing serotypes permeable to the blood-brain barrier) allow uniform, noninvasive gene delivery to the central and peripheral nervous systems [9].
Table 1.
Viral vectors administration routes: local expression is reached by stereotaxic injection in a precise brain area and global transfection through intravenous, retro-orbital or cerebral spinal fluid injection
| LOCAL | GLOBAL | |
|---|---|---|
| ADVANTAGES |
• Restricted expression to injection site • Time-specific administration at desired developmental stage • Reduced off-target activity respect to global administration • High transgene expression level • Small viral titer and volume |
• CNS- or PNS-wide transduction • Uniform viral expression • Sparse or anatomically distributed cell types labeling can be achieved • Permeable to blood-brain barrier • Minimally invasive or non-invasive approach • No need of stereotaxic apparatus or surgical expertise |
| DISADVANTAGES |
• Invasive surgery • Need of stereotaxic apparatus • Tissue damage of the injected area • Expression gradient from injection site • Difficulties in reaching deep brain area |
• High volume and viral titer are required • Possible off-target effects • Non-selective approach • Limited by animal body weight |
In recent years, significant advancements in neurosurgical techniques have paralleled the progress of AAV-based gene therapy, profoundly impacting the field of neuroscience (reviewed by [10, 11]). Techniques such as convection-enhanced delivery and intrathecal administration have revolutionized the precise targeting and distribution of AAV vectors within the central nervous system [12, 13]. These methods enable widespread transduction of target cells, offering promising therapeutic avenues for conditions like epilepsy [14], neurotransmitter-related disorders [15] and other neurological disorders (reviewed by [16–18]). Additionally, novel vector infusion methods, including focused ultrasound-mediated delivery, enhance the specificity of AAV vector targeting within the CNS, minimizing off-target effects [19, 20]. Ongoing clinical trials and studies have showcased the transformative potential of these advanced neurosurgical approaches in treating a myriad of neurological disorders. These advancements represent a synergistic relationship between AAV vectors and neurosurgical techniques, thereby highlighting their pivotal role in shaping the future of neuroscience research and therapy.
AAV for controlling brain activity
With the development of cell-specific promoters and enhancers, AAV vectors can target specific cell types within complex brain circuits, allowing researchers to dissect neural circuits with unprecedented precision. AAV vectors boast remarkable specificity, enabling researchers to target distinct cell types and neural circuits with pinpoint accuracy. This precision revolutionizes our understanding of brain function by allowing manipulation at the cellular and circuit levels, unlocking new insights into neural connectivity and correlation to behavior.
The utilization of AAV-mediated delivery for optogenetic and chemogenetic tools offers precise manipulation of neuronal activity, facilitating the exploration of neural circuit dynamics and behavior in both healthy and pathological conditions. These genetic strategies involve the delivery of photoactivatable ion channels named opsins for optogenetics (reviewed by [21–23]) or ligand-gated G-coupled ion channels named designer receptors activated by designer drugs (DREADDs) for chemogenetics (reviewed by [24–26]). In optogenetic applications, the expression of channelrhodopsins (ChR) or halorhodopsins (HR) permits cellular access via light stimulation, enabling the activation or inhibition of neuronal activity within target cells [27, 28]. Conversely, in chemogenetics, the expression of engineered Gq- or Gi-coupled human muscarinic acetylcholine receptors (hM3Dq or hM4Di) enables activation or inhibition via the administration of a specific drug (clozapine N-oxide, CNO) [29]. However, combining these two systems is hindered by the shared drug activation mechanism. To address this limitation, Vardy et al. developed a novel Gi DREADD based on the human κ-opioid receptor, known as KORD, which is activated by a distinct ligand, Salvinorin B (SALB), thus allowing for the implementation of multi-application chemogenetic systems [30]. These technologies allow mapping and dissecting brain circuit in a time- and cell-type specific manner through spatiotemporal expression of chemo- and opto-genetic reporters, which may also be employed in combination due to their different induction methods (light stimulation for optogenetics and drug administration for chemogenetics) [31].
With the advent of optogenetics and chemogenetics, AAV-mediated delivery of light-sensitive and ligand-responsive proteins empowers researchers to modulate neuronal activity with exquisite temporal and spatial resolution. Cre-dependent viral vectors allow efficient gene activation in region- and time-specific manner by delivering the viral vectors to a restricted area at the desired developmental stage. This remarkable control sheds light on the dynamics of neural circuits, offering profound insights into brain function and dysfunction.
AAV for neuronal tracing
AAV vectors offer high transduction efficiency and long-lasting gene expression in neurons and glial cells, making them invaluable tools for studying gene function and manipulating neural circuits.
Overall, viral tracers can be classified into two categories based on their ability to traverse synapses: non-transsynaptic and transsynaptic. Non-transsynaptic viruses are confined to the neurons they infect and cannot extend across synapses to neighboring neurons. In contrast, transsynaptic viruses have the capacity to cross synapses and propagate to interconnected neurons. Both categories include viruses that are transported either anterogradely or retrogradely along axons. Extensive reviews have provided detailed insights into the diverse properties and applications of these viruses [32, 33]. Notably, among AAV variants, only AAV1 and AAV9 exhibit transsynaptic spreading [34, 35], while others do not.
The direction of spread of each virus type which is essential to identify the neuronal connection to the injection site, especially in tracing experiments. It is fundamental to know the region where the virus is injected, corresponding to the start of infection and the direction of the viral spread and the area of expression, essential to identify and map the neural circuit. For this reason, viral vectors are divided into two categories depending on the directionality of viral transport into the cell respect to the injection site: retrograde and anterograde tracers. Retrograde tracers enter at the level of presynaptic terminal and move retrogradely to cell body of the infected neurons by axonal transport, thus allowing the identification of projecting neurons; vice versa anterograde tracers infect the cell body and are transported to axonal processes, thus allowing the visualization of neurons and their targets [36] (Fig. 1). Most AAV serotypes exhibit anterograde transport properties, as exemplified by one of the latest tools, AAV13, with rigorous anterograde labeling [37]. Conversely, AAV2-retro [38], AAV9-retro [39], AAV-DJ/9 [40] and AAV11 [41] are retrogradely transported.
Fig. 1.
Anterograde and retrograde labeling by viral tracers. Tracers are categorized as anterograde or retrograde based on their direction of travel within neurons, either from or towards the cell body respectively. Anterograde tracers (marked in red) allow the visualization of neurons that receive information from the injection site, since they are absorbed by neuronal cell bodies at the injection site and then travel along the axon to reach terminal processes. Conversely, retrograde tracers (marked in blue) allow to visualize neurons that send projections to the injection site, since they are absorbed by terminals and transported back to the cell body
AAV engineering and capsid optimization
Numerous strains of AAV have been discovered in nature, categorized into various serotypes based on the unique antigenicity of their capsid proteins. These distinct serotypes exhibit varying tissue tropism, dictating their specificity for infecting different tissues (Table 2). By utilizing different capsid proteins during packaging, AAV vectors can adopt diverse serotypes. Active research is focused on the identification of capsid variants having a superior efficacy profile [42].
Table 2.
Different AAV serotypes for tissue specificity
| TISSUE | AAV SEROTYPES |
|---|---|
| Brain | AAV1, AAV2, AAV5, AAV8, AAV9, AAV11, AAV13, AAV-PHP.eB, AAV2-retro, AAV9-retro AAV-DJ/9 |
| Retina | AAV1, AAV2, AAV4, AAV5, AAV8, AAV9 |
| Spinal nerves | AAV2-retro, AAV-PHP.S |
To improve and refine AAV tropisms, extensive research has been conducted on the mutational tolerance of AAV capsids. Permissive sites for deliberate or random amino acid substitutions and insertions have been identified through various studies [43, 44]. Additionally, methods for constructing serotype-shuffled chimeric capsids have been developed (for detailed information see the following articles [45–47]).
Numerous engineered capsids with distinct tropisms encompass variants that exhibit: (i) confined or broad transduction within the central nervous system (CNS) or peripheral nervous system (PNS); (ii) cell-type specificity; or (iii) retrograde transport in neurons.
-
i)
Stereotaxic injection facilitates infection within a confined and specific region of the brain, minimizing the risk of off-target effects. In contrast, intravenous and retro-orbital injections, utilizing serotypes capable of crossing the blood-brain barrier, offer consistent and non-invasive gene delivery to both the central and peripheral nervous systems [9]. Moreover, numerous serotypes exhibit broad neuronal and glial transduction within the brain following direct intraparenchymal injection [48].
-
ii)
Cell-type specific infection can be accomplished through the integration of AAV delivery with recombinase systems or gene expression driven by tissue-specific promoters. Cre-dependent viral vectors enable precise gene activation in both spatial and temporal manners by directing the viral vectors to a delimited area during the desired developmental stage [5, 8]. The collaboration between viral vectors and the Cre/lox system has vastly expanded their utility, capitalizing on the adaptability of recombinase-based technology and the rapidity and adaptability of viral injections. AAV can carry the gene of interest under the control of a constitutive promoter like CMV or EF1a promoter [49], or it can be selectively expressed in a more defined subset of cells using a tissue-specific promoter to explore gene profiles and functions within various subpopulations [50, 51].
-
iii)
Targeting the soma of projection neurons becomes crucial for selectively manipulating neurons that terminate at specific sites. Achieving this specificity involves delivering an AAV engineered for retrograde transduction, such as AAV2-retro [38], AAV9-retro [39], AAV-DJ/9 [40], and AAV11 [41] to the projection sites. Consequently, only the cell bodies of neurons terminating in that area will be effectively targeted.
Ongoing efforts to engineer AAV capsids for enhanced tropism, transduction efficiency, and reduced immunogenicity are expanding the utility of AAV vectors in neuroscience research and gene therapy.
Limitations
In this mini-review, we’ve explored the advanced applications of adeno-associated virus (AAV) vectors in neuroscience. While AAV vectors offer several advantages, including long-term gene expression, and low immunogenicity, it is important to acknowledge certain limitations, particularly regarding transduction efficiency. Despite their widespread use and efficacy, AAV vectors may not achieve the same level of transduction efficiency as some other viral vectors. For instance, comparative studies have shown that AAV vectors typically require higher multiplicity of infection (MOI) compared to certain other viral vectors, such as lentiviral vectors, to achieve similar levels of transduction [52, 53]. Several factors contribute to the limitations of AAV transduction efficiency. These include the natural tropism of AAV serotypes [54, 55], which may not match the target cell type, as well as the presence of pre-existing neutralizing antibodies in the host that can inhibit vector entry [56–58]. Additionally, the relatively small packaging capacity of AAV vectors restricts the size of the transgene that can be delivered, limiting their utility for certain applications requiring larger genetic payloads. Researchers should carefully consider these limitations when choosing vector systems. While AAV’s safety and targeted delivery are advantageous, vectors like herpes simplex virus (HSV) or lentivirus may offer higher transduction efficiency due to their unique characteristics. While AAV remains a valuable tool, its limitations in transduction efficiency must be taken into account.
Conclusions
In conclusion, AAV vectors have revolutionized neuroscience research by enabling precise manipulation of neural circuits and offering promising therapeutic strategies for treating neurological disorders. Continued advancements in vector design and delivery techniques will further enhance the utility and efficacy of AAV-based approaches in unraveling the complexities of the brain and developing novel treatments for neurological diseases.
Author contributions
Antea Minetti contributed to the study conception, design and writing.
Funding
No funds, grants, or other support was received.
Open access funding provided by Università di Pisa within the CRUI-CARE Agreement.
Data availability
No datasets were generated or analysed during the current study.
Declarations
Ethical approval
Not applicable.
Consent for publication
Not applicable.
Informed consent
Not applicable.
Competing interests
The author declares she has no financial interests.
Footnotes
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Change history
6/5/2024
A Correction to this paper has been published: 10.1007/s11033-024-09671-7
References
- 1.Bulcha JT, Wang Y, Ma H, Tai PWL, Gao G (2021) Viral vector platforms within the gene therapy landscape. Signal Transduct Target Ther 6(1):1–24. 10.1038/s41392-021-00487-6 [DOI] [PMC free article] [PubMed]
- 2.Lundstrom K (2023) Viral vectors in gene therapy: where do we stand in 2023? Viruses 15(3):1–35. 10.3390/v15030698 [DOI] [PMC free article] [PubMed]
- 3.Zengel J, Carette JE (2020) Structural and cellular biology of adeno-associated virus attachment and entry. Adv Virus Res 106:39–84. 10.1016/bs.aivir.2020.01.002. [DOI] [PubMed]
- 4.Wang D, Tai PWL, Gao G (2019) Adeno-associated virus vector as a platform for gene therapy delivery. Nat Rev Drug Discov 18(5):358–378. 10.1038/s41573-019-0012-9 [DOI] [PMC free article] [PubMed]
- 5.Ravindra Kumar S et al (2020) Multiplexed cre-dependent selection yields systemic AAVs for targeting distinct brain cell types. Nat Methods 17(5):541–550. 10.1038/s41592-020-0799-7 [DOI] [PMC free article] [PubMed]
- 6.Rocchi F et al (2022) Increased fMRI connectivity upon chemogenetic inhibition of the mouse prefrontal cortex. Nat Commun 13(1056). 10.1038/s41467-022-28591-3 [DOI] [PMC free article] [PubMed]
- 7.Chan KY et al (2017) Engineered AAVs for efficient noninvasive gene delivery to the central and peripheral nervous systems. Nat Neurosci 20(8):1172–1179. 10.1038/nn.4593 [DOI] [PMC free article] [PubMed]
- 8.Deverman BE et al (2016) Cre-dependent selection yields AAV variants for widespread gene transfer to the adult brain. Nat Biotechnol 34(2):204–209. 10.1038/nbt.3440 [DOI] [PMC free article] [PubMed]
- 9.Li C, Samulski RJ (2020) Engineering adeno-associated virus vectors for gene therapy. Nat Rev Genet 21(4):255–272. 10.1038/s41576-019-0205-4 [DOI] [PubMed]
- 10.Mendell JR et al (2021) Current clinical applications of in vivo gene therapy with AAVs. Mol Ther 29(2):464–488. 10.1016/j.ymthe.2020.12.007 [DOI] [PMC free article] [PubMed]
- 11.Hudry E, Vandenberghe LH (2019) Therapeutic AAV gene transfer to the nervous system: a clinical reality. Neuron 101(5):839–862. 10.1016/j.neuron.2019.02.017 [DOI] [PMC free article] [PubMed]
- 12.Naidoo J, Fiandaca M, Lonser RR, Bankiewicz K (2019) Convection-enhanced drug delivery in the central nervous system. Nervous system drug delivery: principles and practice. pp 335–350. 10.1016/B978-0-12-813997-4.00016-5
- 13.Ajeeb R, Clegg JR (2023) Intrathecal delivery of macromolecules: clinical status and emerging technologies. Adv Drug Deliv Rev 199:114949. 10.1016/j.addr.2023.114949 [DOI] [PubMed]
- 14.Bettegazzi B, Cattaneo S, Simonato M, Zucchini S, Soukupova M (2024) Viral vector-based gene therapy for epilepsy: what does the future hold? Mol Diagn Ther 28(1):5–13. 10.1007/s40291-023-00687-6 [DOI] [PMC free article] [PubMed]
- 15.Chu WS, Ng J, Waddington SN, Kurian MA (2024) Gene therapy for neurotransmitter-related disorders. J Inherit Metab Dis 47(1):176–191. 10.1002/jimd.12697 [DOI] [PMC free article] [PubMed]
- 16.Hitti FL, Gonzalez-Alegre P, Lucas TH (2019) Gene therapy for neurologic disease: a neurosurgical review. World Neurosurg 121:261–273. 10.1016/j.wneu.2018.09.097 [DOI] [PubMed]
- 17.Deverman BE, Ravina BM, Bankiewicz KS, Paul SM, Sah DWY (2018) Gene therapy for neurological disorders: progress and prospects. Nat Rev Drug Discov 17(9):641–659. 10.1038/nrd.2018.110 [DOI] [PubMed]
- 18.Lonser RR, Akhter AS, Zabek M, Elder JB, Bankiewicz KS (2021) Direct convective delivery of adeno-associated virus gene therapy for treatment of neurological disorders. J Neurosurg 134(6):1751–1763. 10.3171/2020.4.JNS20701 [DOI] [PubMed]
- 19.Kofoed RH et al (2022) Efficacy of gene delivery to the brain using AAV and ultrasound depends on serotypes and brain areas. J Controlled Release 351:667–680. 10.1016/j.jconrel.2022.09.048 [DOI] [PubMed]
- 20.Ye D et al (2022) Incisionless targeted adeno-associated viral vector delivery to the brain by focused ultrasound-mediated intranasal administration. EBioMedicine 84(104277):1–14. 10.1016/j.ebiom.2022.104277 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Takao T, Yamada D, Takarada T (2022) Mouse model for optogenetic genome engineering. Acta Med Okayama 76(1):1–5. 10.18926/AMO/63202 [DOI] [PubMed] [Google Scholar]
- 22.Chen W et al (2022) The roles of optogenetics and technology in neurobiology: a review. Front Aging Neurosci 14. 10.3389/fnagi.2022.867863 [DOI] [PMC free article] [PubMed]
- 23.Altahini S, Arnoux I, Stroh A (2024) Optogenetics 2.0: challenges and solutions towards a quantitative probing of neural circuits. Biol Che 405(1):43–54. 10.1515/hsz-2023-0194 [DOI] [PubMed]
- 24.Atasoy D, Sternson SM (2018) Chemogenetic tools for causal cellular and neuronal biology. Physiol Rev 98(1):391–418. 10.1152/physrev.00009.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Vlasov K, Van Dort CJ, Solt K (2018) Optogenetics and Chemogenetics, vol 603, 1st edn. Elsevier Inc. 10.1016/bs.mie.2018.01.022 [DOI] [PMC free article] [PubMed]
- 26.Campbell EJ, Marchant NJ (2018) The use of chemogenetics in behavioural neuroscience: receptor variants, targeting approaches and caveats. Br J Pharmacol 175(7):994–1003. 10.1111/bph.14146 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Deisseroth K, Hegemann P (2017) The form and function of channelrhodopsin. Science 357(6356):1–9. 10.1126/science.aan5544 [DOI] [PMC free article] [PubMed]
- 28.Govorunova EG, Sineshchekov OA, Spudich JL (2022) Emerging diversity of channelrhodopsins and their structure-function relationships. Front Cell Neurosci 15. Frontiers Media S.A. 10.3389/fncel.2021.800313 [DOI] [PMC free article] [PubMed]
- 29.Zhu H, Roth BL (2015) DREADD: a chemogenetic GPCR signaling platform. Int J Neuropsychopharmacol 18(1). 10.1093/ijnp/pyu007 [DOI] [PMC free article] [PubMed]
- 30.Vardy E et al (2015) A new DREADD facilitates the multiplexed chemogenetic interrogation of behavior. Neuron 86(4):936–946. 10.1016/j.neuron.2015.03.065 [DOI] [PMC free article] [PubMed]
- 31.Berglund K, Tung JK, Higashikubo B, Gross RE, Moore CI, Hochgeschwender U (2016) Combined optogenetic and chemogenetic control of neurons. Methods Mol Biol 1408:207–225. 10.1007/978-1-4939-3512-3_14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Cong W, Shi Y, Qi Y, Wu J, Gong L, He M (2020) Viral approaches to study the mammalian brain: lineage tracing, circuit dissection and therapeutic applications. J Neurosci Methods 335(108629):1–11. 10.1016/j.jneumeth.2020.108629 [DOI] [PubMed]
- 33.Xu X et al (2020) Viral vectors for neural circuit mapping and recent advances in trans-synaptic anterograde tracers. Neuron 107(6):1029–1047. 10.1016/j.neuron.2020.07.010 [DOI] [PMC free article] [PubMed]
- 34.Zingg B, Dong HW, Tao HW, Zhang LI (2022) Application of AAV1 for anterograde transsynaptic circuit mapping and input-dependent neuronal cataloging. Curr Protoc 2(1). 10.1002/cpz1.339 [DOI] [PMC free article] [PubMed]
- 35.Zingg B, Peng B, Huang J, Tao HW, Zhang LI (2020) Synaptic specificity and application of anterograde transsynaptic AAV for probing neural circuitry. J Neurosci 40(16):3250–3267. 10.1523/JNEUROSCI.2158-19.2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Zingg B et al (2017) AAV-mediated anterograde transsynaptic tagging: mapping corticocollicular input-defined neural pathways for defense behaviors. Neuron 93(1):33–47. 10.1016/j.neuron.2016.11.045 [DOI] [PMC free article] [PubMed]
- 37.Han Z et al (2022) AAV13 enables precise targeting of local neural populations. Int J Mol Sci 23(12806):1–13. 10.3390/ijms232112806 [DOI] [PMC free article] [PubMed]
- 38.Tervo DGR et al (2016) A designer AAV variant permits efficient retrograde access to projection neurons. Neuron 92(2):372–382. 10.1016/j.neuron.2016.09.021 [DOI] [PMC free article] [PubMed]
- 39.Lin K et al (2020) AAV9-Retro mediates efficient transduction with axon terminal absorption and blood–brain barrier transportation. Mol Brain 13(138):1–12. 10.1186/s13041-020-00679-1 [DOI] [PMC free article] [PubMed]
- 40.Düring DN et al (2020) Fast retrograde access to projection neuron circuits underlying vocal learning in songbirds. Cell Rep 33(6). 10.1016/j.celrep.2020.108364 [DOI] [PMC free article] [PubMed]
- 41.Han Z et al (2023) AAV11 enables efficient retrograde targeting of projection neurons and enhances astrocyte-directed transduction. Nat Commun 14(3792):1–13. 10.1038/s41467-023-39554-7 [DOI] [PMC free article] [PubMed]
- 42.Weinmann J, Grimm D (2017) Next-generation AAV vectors for clinical use: an ever-accelerating race. Virus Genes 53(5):707–713. 10.1007/s11262-017-1502-7 [DOI] [PubMed]
- 43.Adachi K, Enoki T, Kawano Y, Veraz M, Nakai H (2014) Drawing a high-resolution functional map of adeno-associated virus capsid by massively parallel sequencing. Nat Commun 5(3075). 10.1038/ncomms4075 [DOI] [PMC free article] [PubMed]
- 44.Ogden PJ, Kelsic ED, Sinai S, Church GM (2019) Comprehensive AAV capsid fitness landscape reveals a viral gene and enables machine-guided design. Science 366:1139–1143. 10.1126/science.aaw2900 [DOI] [PMC free article] [PubMed]
- 45.Viney L et al (2021) Adeno-associated virus (AAV) capsid chimeras with enhanced infectivity reveal a core element in the AAV genome critical for both cell transduction and Capsid Assembly. J Virol 95(7). 10.1128/jvi.02023-20 [DOI] [PMC free article] [PubMed]
- 46.Challis RC et al (2022) Annual Review of Neuroscience Adeno-Associated Virus Toolkit to target diverse brain cells. Annu Rev Neurosci 45:447–469. 10.1146/annurev-neuro-111020 [DOI] [PubMed] [Google Scholar]
- 47.Lee EJ, Guenther CM, Suh J (2018) Adeno-associated virus (AAV) vectors: Rational design strategies for capsid engineering. Curr Opin Biomed Eng 7:58–63. 10.1016/j.cobme.2018.09.004 [DOI] [PMC free article] [PubMed]
- 48.Watakabe A, Sadakane O, Hata K, Ohtsuka M, Takaji M, Yamamori T (2017) Application of viral vectors to the study of neural connectivities and neural circuits in the marmoset brain. Dev Neurobiol 77(3):354–372. 10.1002/dneu.22459 [DOI] [PMC free article] [PubMed]
- 49.Fenno LE et al (2014) Targeting cells with single vectors using multiple-feature boolean logic. Nat Methods 11(7):763–772. 10.1038/nmeth.2996 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Koh W, Park YM, Lee SE, Lee CJ (2017) AAV-mediated astrocyte-specific gene expression under human ALDH1L1 promoter in mouse thalamus. Exp Neurobiol 26(6):350–361. 10.5607/en.2017.26.6.350 [DOI] [PMC free article] [PubMed]
- 51.Le N, Appel H, Pannullo N, Hoang T, Blackshaw S (2022) Ectopic insert-dependent neuronal expression of GFAP promoter-driven AAV constructs in adult mouse retina. Front Cell Dev Biol 1(1). 10.3389/fcell.2022.914386 [DOI] [PMC free article] [PubMed]
- 52.Colella P, Ronzitti G, Mingozzi F (2018) Emerging issues in AAV-Mediated in vivo gene therapy. Mol Ther Methods Clin Dev 8:87–104. 10.1016/j.omtm.2017.11.007 [DOI] [PMC free article] [PubMed]
- 53.Bulcha JT, Wang Y, Ma H, Tai PWL, Gao G (2021) Viral vector platforms within the gene therapy landscape. Signal Transduction and Targeted Therapy 6(1). 10.1038/s41392-021-00487-6 [DOI] [PMC free article] [PubMed]
- 54.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):1–16. 10.1371/journal.pone.0076310 [DOI] [PMC free article] [PubMed]
- 55.Yang OJ et al (2023) Evaluating the transduction efficiency of systemically delivered AAV vectors in the rat nervous system. Front Neurosci 17:1–13. 10.3389/fnins.2023.1001007 [DOI] [PMC free article] [PubMed]
- 56.Mingozzi F, High KA (2017) Overcoming the host Immune Response to Adeno-Associated Virus Gene Delivery vectors: the race between Clearance, Tolerance, neutralization, and escape. Annu Rev Virol 4:511–534. 10.1146/annurev-virology [DOI] [PubMed] [Google Scholar]
- 57.Rossi A et al (2019) Vector uncoating limits adeno-associated viral vector-mediated transduction of human dendritic cells and vector immunogenicity. Sci Rep 9(1):1–14. 10.1038/s41598-019-40071-1 [DOI] [PMC free article] [PubMed]
- 58.Zengel J et al (2023) Hardwiring tissue-specific AAV transduction in mice through engineered receptor expression. Nat Methods 20(7):1070–1081. 10.1038/s41592-023-01896-x [DOI] [PMC free article] [PubMed]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No datasets were generated or analysed during the current study.

