Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Aug 1.
Published in final edited form as: Curr Opin Neurobiol. 2020 Apr 25;63:67–76. doi: 10.1016/j.conb.2020.03.011

Viral vectors for neuronal cell type-specific visualization and manipulations

Yuanyuan Liu 1, Shane Hegarty 2,3, Carla Winter 2,4, Fan Wang 5, Zhigang He 2,3
PMCID: PMC7484153  NIHMSID: NIHMS1603336  PMID: 32344323

Abstract

Characterizing neuronal cell types demands efficient strategies for specific labeling and manipulation of individual subtypes to dissect their connectivity and functions. Recombinant viral technology offers a powerful toolbox for targeted transgene expression in specific neuronal populations. In order to achieve cell type-specific targeting, exciting progress has been made to: alter viral tropisms, design rational delivery strategies, and drive selective expression patterns with engineered DNA sequences in viral genomes. For the latter case, emerging single-cell genomic analyses provide rich databases. In this review, we will summarize current status, and point out challenges, of using viral vectors for neuronal cell type-specific visualization and manipulations. With concerted efforts, progress will continue to be made towards developing viral vectors for the vast array of neuronal subtypes in the mammalian nervous system.

Introduction

The ability to examine and manipulate the function of specific brain nuclei and neuronal types is critical for understanding how brain circuitry works. This is evident from the widespread applications of transgenic mouse driver lines, which enable specific transgene expression in a few selected cell types or brain regions [1]. In light of the complex nature of the nervous system, more versatile tools of this kind would be extremely desirable. This is an even more pressing issue with current advances in single cell analysis methods, such as single-cell RNA sequencing (scRNA-seq), that are characterizing ever-growing numbers of neuronal cell types in an increasing number of mammalian species (including mice and humans). In principle, if a neuronal type is uniquely defined by the combinatorial expression of a set of genes, one could target such neurons specifically using sophisticated genetics. For example, multiple transgenic animals can be generated in which each marker gene is used to drive either a different DNA recombinase (e.g. Cre, Flp, Dre), or a reporter line, so that a desired transgene (e.g. XFP, opsins, activity indicators) is only expressed when all recombinases and the reporter are expressed in the same neuronal cells. While the advancement of CRISPR technology has made it possible to generate gene-targeted animals efficiently, crossing these different lines to achieve cell-type specific expression is still a lengthy and expensive process.

On the other hand, recombinant viral vectors with cross-species targeting and minimal cytotoxic effects offer unprecedented delivery systems to selectively express different desired transgenes in specific neuronal populations. In addition to the natural tropisms of individual viral types [2,3], their transduction specificity can, in principle, be refined by altering their capsid or envelope proteins (to dictate which cells to bind and enter) and/or the promoter/enhancer in the viral genome (to determine whether or when to express the target gene). Combining these techniques to alter viral tropism with various delivery methods would allow recombinant viral vectors to serve as a collection of region-selective, neuron type-specific, and activity-dependent tools that can be readily used in multiple species. Here we review four main strategies for, and recent progresses toward, designing and optimizing viral vectors for neuronal type-specific expression, namely: (1) axonal projection-dependent; (2) viral serotype/tropism-based; (3) gene regulatory element (promoter/enhancer/microRNA)-driven; and (4) delivery-controlled strategies for targeting of distinct neuronal cell types (Figure 1).

Figure 1: Strategies for neuronal cell type-specific targeting with viral vectors.

Figure 1:

A) Axonal projection-dependent retrograde labeling: viruses with axonal tropism (rabies, HiRet lentivirus, CAV2, and retroAAV) mediate retrograde targeting for neurons of interest in both the brain and spinal cord. B) Viral serotype/capsid determines tissue, and in some cases cell-type specific, tropism. Here, AAV color indicates various capsids that correlate with the specific tissue or cell-type that the AAV is able to infect. Specifically: PHP.eB (red) is preferentially expressed in the CNS, PHP.S (green) in the PNS, AAV2 (purple) in RGCs and amacrine cells, ShH10 (yellow) in muller glia, and AAV2.7m8 (blue) in photoreceptors. Grey indicates cells that are not targeted. C) Sequencing and -omics approaches can be harnessed to identify novel gene regulatory elements (e.g. promoters, enhancers, microRNA) to label specific neuronal and non-neuronal subtypes. D) Finally, targeting of distinct neuronal cell types can be aided by unique delivery methods (stereotaxic, intrathecal, tail-vein, intravitreal, and sub-retinal). [Portions of this figure used and/or altered artwork from https://smart.servier.com/; Purkinje neuron from Ramón y Cajal].

Viral vectors for axonal projection-dependent targeting of specific neurons

Neurons residing within one brain region expressing similar genetic markers can have distinct projection targets and can thereby be classified based on their efferent axonal innervation patterns [46]. Thus, one can use recombinant viruses capable of infecting neurons from their axon terminals to target a more specific cell type within a larger neuronal population through retrograde viral infection (Figure 1A). Many types of viral vectors have axonal tropism (Table 1), and four different types of retrograde viral vectors have been developed to this end.

Table 1.

Summary of features of recombinant viruses commonly used in neuroscience

Family Genome Neurotropism Neuronal transport Features Limitations
Direction Cross-synapse
Adeno-associated virus (AAV) Single strand DNA Engineered capsid: e.g. PhP.eB (CNS), PhP.S (PNS) Enhancer/promotor specific Anterograde; Engineered for retrograde: rAAV2-retro No Exception: AAV1or 9-Cre, Anterograde Do not integrate; High and stable expression; Selective neurotropism; Capable of recombinase-based modification Low packaging capacity
Lentivirus Single strand (+) RNA No pseudotyping: immune cells Pseudotyping (VSV-G): neuron and glia Anterograde; Engineered for retrograde: FuG-B, C & E No Viral genome integrates into host genome; Selective targeting by pseudotyping (EnV-TVA); Capable of recombinase-based modification Expression level low
Rabies virus Single strand (−) RNA Neurotrophic Retrograde Retro-Exception: Sensory neurons, Anterograde Engineered for non-crossing retrograde (RV-dG) or mono-synaptic (RV-dG with G) transmission; Selective targeting by pseudotyping (EnV-TVA) Highly cyto-toxic; Incompatible with Cre/Flp
α-herpes viruses: Pseudorabies virus (PRV) and HSV1 Double strand DNA Neurotrophic Bi-directional Strain dependent: PRV: Retrograde HSV1: Retro-/Anterograde Poly-synaptic, bi-directional transmission; Engineered for mono-synaptic transmission (HSV: H129-dTK) Difficult for genome engineering; Highly cyto-toxic

Recombinant rabies viruses:

It has long been known that rabies viruses infect mammalian neurons from axon terminals [7]. However, the use of rabies viruses for manipulating or labeling specific neuronal populations is limited because they are neurotoxic and uncontrollably propagate between hierarchically-connected neurons. Within the five genes of rabies viral genome, only G encodes the glycoprotein to mediate retrograde, trans-synaptic viral transmission. Thus, when the rabies envelope glycoprotein (RG) was replaced by fluorescent proteins (e.g. GFP), the recombinant rabies virus is used for non-transsynaptic retrograde neuronal labeling [8]. To further reduce its neurotoxicity, Wickersham’s group developed a double deletion (L and G proteins) recombinant rabies virus [9]. This new version enables long-term anatomical or physiological study of retrogradely-targeted projection neurons.

Retrograde lentiviruses:

Considering the aforementioned limitations of the rabies viruses, a promising alternative candidate is the notably less toxic lentivirus genus. Lentiviruses are single stranded RNA viruses that stably integrate into the host genome of both pre- and post-mitotic cells [2]. In light of previous findings that RG pseudotyped equine infectious anemia virus (EIAV) is capable of retrograde tracing [10], Kobayashi’s group decided to pseudotype lentiviruses using fusion proteins incorporating RG domains. They discovered that packaging lentiviruses with a fusion envelope composed of the extracellular domain of RG and the transmembrane and intracellular domain of the vesicular stomatitis virus (VSV) glycoprotein resulted in retrograde lentiviral vectors (called HiRet-LV, also called RG-LV) that were highly effective in infecting axon terminals of long-distance projection neurons in in the brain and spinal cord of both rodents and non-human primates (NHPs) [1113]. Retrograde lentiviruses have since been successfully used by multiple labs for axonal projection-dependent labeling and manipulation of neuronal cell types [1417].

CAV-2:

Canine adenovirus type 2 (CAV-2) displays potent retrograde axonal transport ability, through its selective interaction with the coxsackievirus and adenovirus receptor (CAR) at axon terminals [18]. CAV-2 can drive stable, long-term transgene expression with minimal cytotoxicity and has been effectively used as a retrograde vector to study many circuits [19,20]. However, CAV-2 has limited tropism since many neurons do not express the CAR receptor. This can be overcome by overexpression of CAR in candidate projection neurons [21].

rAAV2-retro:

Using in vivo screening for adeno-associated virus (AAV) capsids that would enable the packaged AAVs to be efficiently taken up by axonal terminals, recombinant AAV2-retro (rAAV2-retro) capsid was discovered to exhibit high efficiency in retrograde labeling of long-projecting neurons such as corticospinal neurons [22]. rAAV2-retro vector has broad, but not pan-neuronal, tropism for retrograde infection and can drive high-level gene expression like other AAVs. Thus, it is a very versatile retrograde vector, but which needs its tropism for desired neurons tested before use.

Applications of retrograde vectors:

The development of retrograde viral tools enables virus-based intersectional approaches to precisely manipulate target projection neurons that are embedded in a mixture of different neuronal populations. To achieve this, the retrograde viral vectors encoding recombinases (e.g. Cre or Flp) are often injected at specific sites where the axons of the target projection neurons innervate. These viral particles are selectively absorbed by axon terminals and then undergo retrograde gene transfer. Next, AAVs encoding Cre- or Flp-dependent genes (e.g. XFP, DTR, ChR2 or GCamp6) are injected into areas where the somas of the targeted projection neurons are located (Figure 2). Such a multi-step, axonal projection-dependent intersectional approach has been successfully applied in sophisticated neuronal circuit mapping and functional characterization in both rodent and NHP models [4,16,17,2325]. More recently, combining this approach with brain clearing and 3D-imaging reconstruction, the Janelia MouseLight project has reconstructed the entire cell bodies and neurites of >1000 cortical projection neurons in the mouse brain, revealing previously unknown subtypes of cortical projection neurons [26] (MouseLight Neuron Browser: http://ml-neuronbrowser.janelia.org).

Figure 2: Viral-based intersectional targeting approach.

Figure 2:

A) Schematic drawing of viral-based intersectional targeting approach. B) Images of a coronal brain section showing corticospinal neurons targeted by an engineered lenti-viral vector (HiRet-GFP, green), a traditional neuronal tracer (BDA, red) and their merge. The cartoon drawing demonstrated that almost all corticospinal neurons randomly labeled by BDA are co-labeled with HiRet-GFP and thus indicated the high efficiency of viral mediated retrograde labeling. In addition, compared to BDA, viral based retrograde labeling allows further functional observation and manipulations of targeted neurons.

AAV serotype/capsids-based cell type-specific neuronal targeting

Recombinant AAVs (rAAVs) are comprised of a single-stranded DNA genome with a capsid outer shell and are unable to integrate into host genomes, unlike naturally occurring ones that indeed integrate their genome into a preferred locus of the host genome [27]. Because of minimal cytotoxic effects and different tropisms associated with individual serotypes, AAV vectors have been increasingly used for mapping brain connectivity and for monitoring and manipulating neuronal functions in many vertebrate species, including mouse, rat, songbird, and NHPs. In addition, AAV vectors are also used clinically for gene therapy [28]. In most cases, the serotypes of rAAVs are determined by variations in their capsid proteins [29], with AAV1-9 being the most commonly used. Most of these serotypes are capable of infecting many neuronal cells, although with various efficacies in targeting different neuronal and glial cell types [3032]. For example, intravitreally-injected AAV2 is highly effective to generally target retinal ganglion cells [33,34].

The existence of many serotypes raises the possibility of altering/screening capsid proteins to achieve cell type-specific infection, and has inspired many such efforts. In fact, the above mentioned rAAV-retro capsid was developed based on similar ideas. Briefly, directed evolution, which involves mutagenesis of the Cap gene followed by iterative rounds of phenotypic screening, has been widely used to identify a desired viral capsid. For example, most natural AAVs have limited ability to cross the blood brain barrier (BBB). By using a Cre-dependent selection system, Gradinaru’s group discovered novel capsids named PHP.B that can cross the BBB to infect the entire nervous system [35]. In a follow-up study, they further developed engineered capsids named PHP.eB and PHP.S that preferentially target CNS or PNS, respectively, through systemic administration [36]. Using similar approaches, an AAV2 variant named 7m8 and an AAV6 variant named ShH10 have been identified for enhanced selectivity for photoreceptors and Muller glia in the murine retina, respectively [37,38] (Figure 1B). Another strategy adopts a reverse principle to identify ancestral nodes or putative parents of currently available Cap proteins. Using this “back to future” approach, Anc80L65 was discovered and has showed strong potency for gene therapy of the inner ear [39,40]. We envision more variants of capsid proteins for targeting distinct neuronal cell types will be developed and optimized in the near future.

Identifying promoters/enhancers and other regulatory elements that can drive cell type-specific expression

Since neuronal cell types (or non-neuronal cell types in the nervous system) are determined by expression of marker gene(s), an approach to specifically label or manipulate distinct populations is to identify unique gene regulatory elements (promoters/enhancers) that fit into the packaging capacity of AAVs (4.7kb) to drive cell type-specific gene expression. Previous studies have identified a number of promoters useful for this purpose, such as human synapsin 1 (hSyn1) promoter for (pan-)neuronal specific expression [41], GFAP promoter for astrocytes [42], MBP for oligodendrocytes [43], tyrosine hydroxylase for catecholaminergic cells [36], L7/PCP2 for Purkinje cells [44], and FEV for serotonergic cells [45] (Figure 1C). In addition, since activity-dependent genetic methods are fruitful in tagging and manipulating functionally-activated ensembles of neurons [4649], several studies have attempted to develop viral vectors using enhancers or promoters of immediate early genes, such as Fos or Arc [50] [51] [52] [53]. For example, an AAV vector in which the Fos promoter drives the expression of tTA was used to label different amygdala neurons activated by either aversive or appetitive stimuli [50], while AAVs containing the Arc promoter driving CreERT2 were used to label activated prefrontal cortex neurons [51]. The main caveat for AAVs using activity-dependent promoter/enhancers is a background basal expression which is independent of neuronal activity, and could therefore result in random labeling of non-functionally relevant neurons. To address this, Sorenson et al. developed AAVs with a Robust Activity Marking (RAM) system that consists of a synthetic neuronal activity-dependent promoter, which has very low expression in basal conditions, strong induction by neural activity and is under the temporal control of a modified Tet-Off system [54].

Suffice it to say, with the ever-increasing numbers of neuronal cell types being identified in the mammalian brain through the expansion of the single-cell RNAseq (scRNA-seq) technology, more enhancers/promoters that can drive specific expression using AAVs are urgently needed. Owing to recent advancement in scRNA-seq, Assay for Transposase-Accessible Chromatin using sequencing (ATAC-Seq) and other “-omic” methods, important progress has been made toward this end. Below we briefly review these methods. One method to identify enhancers is to search for evolutionarily-conserved DNA regulatory elements. By comparative genomic analysis, Visel and Pennacchino identified several hundred ultra-conserved non-coding elements in the human genome [55,56]. When tested in early embryos using transient transgenesis, many elements drove gene expression reproducibly in restricted regions of the embryonic brain [57]. Follow-up studies suggested many of these regulatory elements could act as enhancers for specific brain regions [58], and restrict expression to particular neuronal subtypes such as using the Dlx5/6 enhancer for GABAergic neurons [59]. However, many of the ultra-conserved enhancers are developmental enhancers, and fail to act in the adult brain (P. Williams and Z. He, unpublished data).

Second, mapping the open chromatin regions with advanced technologies has led to the identification of cell type-restricted gene regulatory elements (GREs) or enhancers. For example, Mich et al. used ATAC-seq to discover specific enhancer elements for diverse neural types within human cortex, which were then cloned into AAV vectors and tested in the mouse brain. Several enhancers were shown to enable labeling and perturbation of specific cortical neuronal subtypes [60] Furthermore, using the principles of massively parallel reporter assays (MPRA) with scRNA-seq, Hrvatin et al. designed a Paralleled Enhancer Single Cell Assay (PESCA) to identify and functionally assess the specificity of hundreds of GREs across the full complement of cell types present in the brain [61]. They identified GREs that confine AAV expression to somatostatin (SST)-expressing interneurons, which can be used to selectively modulate their neuronal activity [61]. We envision ATAC-seq and PESCA related approaches will generate a large database of neuronal subtype-specific enhancers.

Combining these different GREs may further optimize neuron type-specific AAV vectors. With this in mind, Jüttner et al. created a library of 230 AAVs, each with a different synthetic promoter designed based on identified “transcriptional code” for adult retinal cell types [62], and found that a number of these AAVs specifically target expression to neuronal and glial cell types in mouse and NHP retina in vivo and in the human retina in vitro [63]. Using logical combinations of different AAVs containing different synthetic promoters in an intersectional manner, they further achieved specific targeting of retinal cell types for manipulation and recording of neuronal function [63]. Thus, combinations of AAVs containing different GREs used in an intersectional manner will further improve the specificity of viral vectors for targeting desired neuronal types.

Third, promoters and enhancers may have sequences with the length beyond the packaging capacity of AAVs, but the recognition sites of microRNA (miRNA) are small (about 22 nucleotides) and can be easily packaged into viral vectors. Using the ability of miRNAs in restricting gene expression in certain neuronal types, AAVs with tandem copies of short miRNA-target sequences (miRNA-TSs) in their 3’ untranslated region of the genome have been characterized [64]. Recently, Keaveney et al. developed a miRNA-guided neuron tag (“mAGNET”) to restrict transgene expression to inhibitory (GABA+) neurons in the mouse neocortex (GABA mAGNET) [65]. In light of highly enriched expression of miRNAs in the mammalian brain, microRNA-based methods have huge potential. Mining the single-cell miRNA-seq database should facilitate the identification of new cell type-specific miRNAs.

Delivery-method controlled cell type-restricted targeting

In the nervous system, neuronal cells are defined by both their function and anatomical location. Thus, the method used to deliver viral vectors is another important consideration when attempting to restrict viral expression to precise neuronal cell types. The three most common virus delivery methods to target neurons are stereotaxic, intrathecal, and tail vein injection (Figure 1D), each with their own advantages and limitations, and which should be considered in parallel with the tropism of the particular viral vector of choice. For example, tail vein injection offers an accessible delivery method, but most viruses cannot cross the BBB. As discussed above, the newly developed AAV-PHP.B vectors are able to cross BBB in rodents, and are thus suitable for systemic injection to target CNS neurons [35]. Stereotaxic injection is most commonly used for targeting cells in different parts of the brain or spinal cord; however, it is a more invasive route of administration. In contrast, intrathecal injection is less invasive, but preferentially targets the dorsal root ganglia (DRGs) and spinal dorsal horn, with a decreasing expression gradient from lumbar to cervical spinal cord segments [66]. Such physical limitations to viral vector spread or entry can be addressed through, for example, adjusting body positioning post viral delivery [67]. More recently, bilateral intravenous injection of relatively large volumes of AAV9 into the transverse sinuses of early postnatal animals has been employed as a simpler way to achieve widespread and robust infection of the mouse brain [68].

In addition to this, different viruses display distinct infection profiles even when the same delivery method is chosen. For example, AAV2 has a more restricted, localized expression profile than other AAV serotypes when stereotaxically-injected into the CNS [30]. It is important to note that structural constraints can create boundaries for viral diffusion. This is nicely illustrated by distinct efficacies of different injection protocols in targeting different populations of retinal cells [69]. For example, intravitreal injection of AAVs is optimal for targeting inner retinal neurons/Müller glia, while subretinal AAV delivery is optimal for targeting the Retinal Pigment Epithelium (RPE) /photoreceptors [3]. Another example takes advantage of a transiently-compromised BBB after spinal cord injury. In this case, tail vein injection of AAV9 results in peri-lesion neuronal targeting [70]. Thus, the characteristic features and limitations of different delivery methods can be exploited to restrict infection of specific viral vectors to particular neuronal cell types and CNS regions of interest.

Other notes on using viral vectors to target specific cell types

While most efforts have aimed to achieve restricted and high-level gene expression with rAAV vectors, little attention has been paid to the toxicity associated with these AAV vectors, a crucial issue for their applications in human gene therapy [69,71]. Non-selective targeting of cell types represents a key issue. Based on the studies showing that AAV9 is able to transduce spinal motor neurons when administered intravenously at high doses [72,73], this approach was approved for expressing non-mutated survival of motor neuron (SMN) protein to treat infants with spinal muscular atrophy (SMA) [74]. However, recent studies have shown that high doses of AAV9 administration trigger sensory side-effects, with mononuclear cell inflammation in the dorsal root ganglion and cell death [75]. Based on such concerns, some of SMA clinical studies were halted (https://www.statnews.com/2019/10/30/novartis-zolgensma-gene-therapy-study-halted-by-fda-on-animal-safety-concerns/). Moreover, a recent clinical trial of an AAV9-mediated gene therapy for Duchenne muscular dystrophy was halted due to kidney injury and decreases in red blood cell counts in patients after intravenous viral infusion (https://www.the-scientist.com/news-opinion/trial-of-gene-therapy-for-duchenne-muscular-dystrophy-put-on-hold-66711). Similarly, by testing multiple AAV genome structures and capsid types using subretinal injection in mice, Xiong et al. found a strong correlation between cis-regulatory sequences and toxicity [76] . Interestingly, AAVs with any one of three broadly active promoters, or an RPE-specific promoter, were toxic, while AAVs with four different photoreceptor-specific promoters were not toxic at the highest doses tested. With increased efforts in using AAVs to express specific proteins to manipulate neuronal functions, distinguishing the true effects versus the noise/inflammation/toxicity associated with viral vectors should be considered with greater caution. Thus, refining these viral vectors will facilitate the development of new gene therapy protocols.

Finally, the optimizations of viruses for neuronal targeting discussed herein have limitations in their translational potential due to diversity in cross-species tropism of viruses. Indeed, the retinal cell types targeted by specific synthetic promoter AAVs varied widely between mouse and NHP/human retinas [63]. Moreover, the PHP.B virus discussed above, which crosses BBB and remarkably transduces CNS following intravenous infusion in mice [35], was not found to do the same in NHPs [77]. Therefore, in these cases, optimizing cell-type targeting in mice yields vectors that are unlikely to optimally target the same cell type in NHPs/humans. On the other hand, the ability of an AAV to target a neural cell group in NHPs is a good predictor for targeting the same cell group in humans [63], suggesting that phylogenetically closer species have more correlated responses to virus infection.

Concluding remarks

Developments in the past decade have shown that recombinant viral tools have started to transform our ability to map, observe and manipulate neuronal circuits in experimental models, and to design gene therapy strategies in humans. While this review has been mainly focused on targeting specific neuronal types, recombinant viral vectors have also been used for monosynaptic tracing of neuronal connections [1,78]. In brief, the vectors for retrograde trans-synaptic tracing include the SADB19-base vectors [79], and a less toxic CVS-N2c strain [80]. Recent studies also led to the development of anterograde trans-synaptic tracing vectors, such as the recombinant HSV-129 (Table 1) [81,82], AAV1 or AAV9-Cre [83], and VSV [84]. Furthermore, several studies have combined cell type-specific viral vectors with such “trans-synaptic” or “trans-neuronal vectors” to dissect input-output circuits [24].

In conclusion, it is an exciting time in neuroscience as we continue to make progress into uncovering the diversity of neuronal cell types. Using the methods (projection-based, serotype-restricted, GRE-dependent, and delivery-controlled) described here, their combinations, and other yet to be discovered methods to generate viral vectors, in the not too distant future it will be possible to target identified neuronal subtypes with even greater precision. This will allow us to determine the connectivity and functions of various types of neurons in the mammalian brain, and use such knowledge to develop therapies for brain disorders.

Acknowledgment

This work is supported by NIH grants U01MH109107, R01NS109947, and P30EY012196.

References:

* of special interest:

** of outstanding interest:

  • 1.Luo L, Callaway EM, Svoboda K: Genetic Dissection of Neural Circuits: A Decade of Progress. Neuron 2018, 98:256–281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Nassi JJ, Cepko CL, Born RT, Beier KT: Neuroanatomy goes viral! Front Neuroanat 2015, 9:80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Schon C, Biel M, Michalakis S: Retinal gene delivery by adeno-associated virus (AAV) vectors: Strategies and applications. Eur J Pharm Biopharm 2015, 95:343–352. [DOI] [PubMed] [Google Scholar]
  • 4.Beier KT, Gao XJ, Xie S, DeLoach KE, Malenka RC, Luo L: Topological Organization of Ventral Tegmental Area Connectivity Revealed by Viral-Genetic Dissection of Input-Output Relations. Cell Rep 2019, 26:159–167. e156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Ren J, Friedmann D, Xiong J, Liu CD, Ferguson BR, Weerakkody T, DeLoach KE, Ran C, Pun A, Sun Y, et al. : Anatomically Defined and Functionally Distinct Dorsal Raphe Serotonin Sub-systems. Cell 2018, 175:472–487. e420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Stachniak TJ, Ghosh A, Sternson SM: Chemogenetic synaptic silencing of neural circuits localizes a hypothalamus-->midbrain pathway for feeding behavior. Neuron 2014, 82:797–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Kelly RM, Strick PL: Rabies as a transneuronal tracer of circuits in the central nervous system. J Neurosci Methods 2000, 103:63–71. [DOI] [PubMed] [Google Scholar]
  • 8.Wickersham IR, Finke S, Conzelmann KK, Callaway EM: Retrograde neuronal tracing with a deletion-mutant rabies virus. Nat Methods 2007, 4:47–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Chatterjee S, Sullivan HA, MacLennan BJ, Xu R, Hou Y, Lavin TK, Lea NE, Michalski JE, Babcock KR, Dietrich S, et al. : Nontoxic, double-deletion-mutant rabies viral vectors for retrograde targeting of projection neurons. Nat Neurosci 2018, 21:638–646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Mazarakis ND, Azzouz M, Rohll JB, Ellard FM, Wilkes FJ, Olsen AL, Carter EE, Barber RD, Baban DF, Kingsman SM, et al. : Rabies virus glycoprotein pseudotyping of lentiviral vectors enables retrograde axonal transport and access to the nervous system after peripheral delivery. Hum Mol Genet 2001, 10:2109–2121. [DOI] [PubMed] [Google Scholar]
  • 11.Kato S, Kobayashi K, Inoue K, Kuramochi M, Okada T, Yaginuma H, Morimoto K, Shimada T, Takada M, Kobayashi K: A lentiviral strategy for highly efficient retrograde gene transfer by pseudotyping with fusion envelope glycoprotein. Hum Gene Ther 2011, 22:197–206. [DOI] [PubMed] [Google Scholar]
  • 12.Kobayashi K, Inoue KI, Tanabe S, Kato S, Takada M, Kobayashi K: Pseudotyped Lentiviral Vectors for Retrograde Gene Delivery into Target Brain Regions. Front Neuroanat 2017, 11:65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Kato S, Sugawara M, Kobayashi K, Kimura K, Inoue KI, Takada M, Kobayashi K: Enhancement of the transduction efficiency of a lentiviral vector for neuron-specific retrograde gene delivery through the point mutation of fusion glycoprotein type E. J Neurosci Methods 2019, 311:147–155. [DOI] [PubMed] [Google Scholar]
  • 14.Nelson A, Schneider DM, Takatoh J, Sakurai K, Wang F, Mooney R: A circuit for motor cortical modulation of auditory cortical activity. J Neurosci 2013, 33:14342–14353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Et Stanek, Rodriguez E, Zhao S, Han BX, Wang F: Supratrigeminal Bilaterally Projecting Neurons Maintain Basal Tone and Enable Bilateral Phasic Activation of Jaw-Closing Muscles. J Neurosci 2016, 36:7663–7675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Wang X, Liu Y, Li X, Zhang Z, Yang H, Zhang Y, Williams PR, Alwahab NSA, Kapur K, Yu B, et al. : Deconstruction of Corticospinal Circuits for Goal-Directed Motor Skills. Cell 2017, 171:440–455. e414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Liu Y, Latremoliere A, Li X, Zhang Z, Chen M, Wang X, Fang C, Zhu J, Alexandre C, Gao Z, et al. : Touch and tactile neuropathic pain sensitivity are set by corticospinal projections. Nature 2018, 561:547–550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Salinas S, Schiavo G, Kremer EJ: A hitchhiker’s guide to the nervous system: the complex journey of viruses and toxins. Nat Rev Microbiol 2010, 8:645–655. [DOI] [PubMed] [Google Scholar]
  • 19.Junyent F, Kremer EJ: CAV-2--why a canine virus is a neurobiologist’s best friend. Curr Opin Pharmacol 2015, 24:86–93. [DOI] [PubMed] [Google Scholar]
  • 20.Del Rio D, Beucher B, Lavigne M, Wehbi A, Gonzalez Dopeso-Reyes I, Saggio I, Kremer EJ: CAV-2 Vector Development and Gene Transfer in the Central and Peripheral Nervous Systems. Front Mol Neurosci 2019, 12:71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Li SJ, Vaughan A, Sturgill JF, Kepecs A: A Viral Receptor Complementation Strategy to Overcome CAV-2 Tropism for Efficient Retrograde Targeting of Neurons. Neuron 2018, 98:905–917. e905. [DOI] [PubMed] [Google Scholar]
  • 22.Tervo DG, Hwang BY, Viswanathan S, Gaj T, Lavzin M, Ritola KD, Lindo S, Michael S, Kuleshova E, Ojala D, et al. : A Designer AAV Variant Permits Efficient Retrograde Access to Projection Neurons. Neuron 2016, 92:372–382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Beier KT, Steinberg EE, DeLoach KE, Xie S, Miyamichi K, Schwarz L, Gao XJ, Kremer EJ, Malenka RC, Luo L: Circuit Architecture of VTA Dopamine Neurons Revealed by Systematic Input-Output Mapping. Cell 2015, 162:622–634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Schwarz LA, Miyamichi K, Gao XJ, Beier KT, Weissbourd B, DeLoach KE, Ren J, Ibanes S, Malenka RC, Kremer EJ, et al. : Viral-genetic tracing of the input-output organization of a central noradrenaline circuit. Nature 2015, 524:88–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Kinoshita M, Matsui R, Kato S, Hasegawa T, Kasahara H, Isa K, Watakabe A, Yamamori T, Nishimura Y, Alstermark B, et al. : Genetic dissection of the circuit for hand dexterity in primates. Nature 2012, 487:235–238. [DOI] [PubMed] [Google Scholar]
  • 26.Winnubst J, Bas E, Ferreira TA, Wu Z, Economo MN, Edson P, Arthur BJ, Bruns C, Rokicki K, Schauder D, et al. : Reconstruction of 1,000 Projection Neurons Reveals New Cell Types and Organization of Long-Range Connectivity in the Mouse Brain. Cell 2019, 179:268–281. e213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.McCarty DM, Young SM Jr, Samulski RJ: Integration of adeno-associated virus (AAV) and recombinant AAV vectors. Annu Rev Genet 2004, 38:819–845. [DOI] [PubMed] [Google Scholar]
  • 28.High KA, Roncarolo MG: Gene Therapy. N Engl J Med 2019, 381:455–464. [DOI] [PubMed] [Google Scholar]
  • 29.Chen SH, Haam J, Walker M, Scappini E, Naughton J, Martin NP: Recombinant Viral Vectors as Neuroscience Tools. Curr Protoc Neurosci 2019, 87:e67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Taymans JM, Vandenberghe LH, Haute CV, Thiry I, Deroose CM, Mortelmans L, Wilson JM, Debyser Z, Baekelandt V: Comparative analysis of adeno-associated viral vector serotypes 1, 2, 5, 7, and 8 in mouse brain. Hum Gene Ther 2007, 18:195–206. [DOI] [PubMed] [Google Scholar]
  • 31.Howard DB, Powers K, Wang Y, Harvey BK: Tropism and toxicity of adeno-associated viral vector serotypes 1, 2, 5, 6, 7, 8, and 9 in rat neurons and glia in vitro. Virology 2008, 372:24–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Srivastava A: In vivo tissue-tropism of adeno-associated viral vectors. Curr Opin Virol 2016, 21:75–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Park KK, Liu K, Hu Y, Smith PD, Wang C, Cai B, Xu B, Connolly L, Kramvis I, Sahin M, et al. : Promoting axon regeneration in the adult CNS by modulation of the PTEN/mTOR pathway. Science 2008, 322:963–966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Petrs-Silva H, Dinculescu A, Li Q, Deng WT, Pang JJ, Min SH, Chiodo V, Neeley AW, Govindasamy L, Bennett A, et al. : Novel properties of tyrosine-mutant AAV2 vectors in the mouse retina. Mol Ther 2011, 19:293–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Deverman BE, Pravdo PL, Simpson BP, Kumar SR, Chan KY, Banerjee A, Wu WL, Yang B, Huber N, Pasca SP, et al. : Cre-dependent selection yields AAV variants for widespread gene transfer to the adult brain. Nat Biotechnol 2016, 34:204–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Chan KY, Jang MJ, Yoo BB, Greenbaum A, Ravi N, Wu WL, Sanchez-Guardado L, Lois C, Mazmanian SK, Deverman BE, et al. : Engineered AAVs for efficient noninvasive gene delivery to the central and peripheral nervous systems. Nat Neurosci 2017, 20:1172–1179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Dalkara D, Byrne LC, Klimczak RR, Visel M, Yin L, Merigan WH, Flannery JG, Schaffer DV: In vivo-directed evolution of a new adeno-associated virus for therapeutic outer retinal gene delivery from the vitreous. Sci Transl Med 2013, 5:189ra176. [DOI] [PubMed] [Google Scholar]
  • 38.Koerber JT, Klimczak R, Jang JH, Dalkara D, Flannery JG, Schaffer DV: Molecular evolution of adeno-associated virus for enhanced glial gene delivery. Mol Ther 2009, 17:2088–2095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Zinn E, Pacouret S, Khaychuk V, Turunen HT, Carvalho LS, Andres-Mateos E, Shah S, Shelke R, Maurer AC, Plovie E, et al. : In Silico Reconstruction of the Viral Evolutionary Lineage Yields a Potent Gene Therapy Vector. Cell Rep 2015, 12:1056–1068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Landegger LD, Pan B, Askew C, Wassmer SJ, Gluck SD, Galvin A, Taylor R, Forge A, Stankovic KM, Holt JR, et al. : A synthetic AAV vector enables safe and efficient gene transfer to the mammalian inner ear. Nat Biotechnol 2017, 35:280–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Kugler S, Kilic E, Bahr M: 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 Ther 2003, 10:337–347. [DOI] [PubMed] [Google Scholar]
  • 42.Lee Y, Messing A, Su M, Brenner M: GFAP promoter elements required for region-specific and astrocyte-specific expression. Glia 2008, 56:481–493. [DOI] [PubMed] [Google Scholar]
  • 43.von Jonquieres G, Frohlich D, Klugmann CB, Wen X, Harasta AE, Ramkumar R, Spencer ZH, Housley GD, Klugmann M: Recombinant Human Myelin-Associated Glycoprotein Promoter Drives Selective AAV-Mediated Transgene Expression in Oligodendrocytes. Front Mol Neurosci 2016, 9:13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.El-Shamayleh Y, Kojima Y, Soetedjo R, Horwitz GD: Selective Optogenetic Control of Purkinje Cells in Monkey Cerebellum. Neuron 2017, 95:51–62. e54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.de Leeuw CN, Korecki AJ, Berry GE, Hickmott JW, Lam SL, Lengyell TC, Bonaguro RJ, Borretta LJ, Chopra V, Chou AY, et al. : rAAV-compatible MiniPromoters for restricted expression in the brain and eye. Mol Brain 2016, 9:52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Barth AL, Gerkin RC, Dean KL: Alteration of neuronal firing properties after in vivo experience in a FosGFP transgenic mouse. J Neurosci 2004, 24:6466–6475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Liu X, Ramirez S, Pang PT, Puryear CB, Govindarajan A, Deisseroth K, Tonegawa S: Optogenetic stimulation of a hippocampal engram activates fear memory recall. Nature 2012, 484:381–385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Allen WE, DeNardo LA, Chen MZ, Liu CD, Loh KM, Fenno LE, Ramakrishnan C, Deisseroth K, Luo L: Thirst-associated preoptic neurons encode an aversive motivational drive. Science 2017, 357:1149–1155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Sakurai K, Zhao S, Takatoh J, Rodriguez E, Lu J, Leavitt AD, Fu M, Han BX, Wang F: Capturing and Manipulating Activated Neuronal Ensembles with CANE Delineates a Hypothalamic Social-Fear Circuit. Neuron 2016, 92:739–753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kim J, Pignatelli M, Xu S, Itohara S, Tonegawa S: Antagonistic negative and positive neurons of the basolateral amygdala. Nat Neurosci 2016, 19:1636–1646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ye L, Allen WE, Thompson KR, Tian Q, Hsueh B, Ramakrishnan C, Wang AC, Jennings JH, Adhikari A, Halpern CH, et al. : Wiring and Molecular Features of Prefrontal Ensembles Representing Distinct Experiences. Cell 2016, 165:1776–1788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kawashima T, Okuno H, Nonaka M, Adachi-Morishima A, Kyo N, Okamura M, Takemoto-Kimura S, Worley PF, Bito H: Synaptic activity-responsive element in the Arc/Arg3.1 promoter essential for synapse-to-nucleus signaling in activated neurons. Proc Natl Acad Sci U S A 2009, 106:316–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kawashima T, Kitamura K, Suzuki K, Nonaka M, Kamijo S, Takemoto-Kimura S, Kano M, Okuno H, Ohki K, Bito H: Functional labeling of neurons and their projections using the synthetic activity-dependent promoter E-SARE. Nat Methods 2013, 10:889–895. [DOI] [PubMed] [Google Scholar]
  • 54.Sorensen AT, Cooper YA, Baratta MV, Weng FJ, Zhang Y, Ramamoorthi K, Fropf R, LaVerriere E, Xue J, Young A, et al. : A robust activity marking system for exploring active neuronal ensembles. Elife 2016, 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Pennacchio LA, Ahituv N, Moses AM, Prabhakar S, Nobrega MA, Shoukry M, Minovitsky S, Dubchak I, Holt A, Lewis KD, et al. : In vivo enhancer analysis of human conserved non-coding sequences. Nature 2006, 444:499–502. [DOI] [PubMed] [Google Scholar]
  • 56.Visel A, Prabhakar S, Akiyama JA, Shoukry M, Lewis KD, Holt A, Plajzer-Frick I, Afzal V, Rubin EM, Pennacchio LA: Ultraconservation identifies a small subset of extremely constrained developmental enhancers. Nat Genet 2008, 40:158–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Pattabiraman K, Golonzhka O, Lindtner S, Nord AS, Taher L, Hoch R, Silberberg SN, Zhang D, Chen B, Zeng H, et al. : Transcriptional regulation of enhancers active in protodomains of the developing cerebral cortex. Neuron 2014, 82:989–1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Dickel DE, Ypsilanti AR, Pla R, Zhu Y, Barozzi I, Mannion BJ, Khin YS, Fukuda-Yuzawa Y, Plajzer-Frick I, Pickle CS, et al. : Ultraconserved Enhancers Are Required for Normal Development. Cell 2018, 172:491–499. e415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Dimidschstein J, Chen Q, Tremblay R, Rogers SL, Saldi GA, Guo L, Xu Q, Liu R, Lu C, Chu J, et al. : A viral strategy for targeting and manipulating interneurons across vertebrate species. Nat Neurosci 2016, 19:1743–1749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Mich JK, Hess EE, Graybuck LT, Somasundaram S, Miller JA, Ding Y, Shapovalova NV, Fong O, Yao S, Mortrud M, et al. : Epigenetic landscape and AAV targeting of human neocortical cell classes. bioRxiv 2019:555318. [Google Scholar]
  • 61.Hrvatin S, Tzeng CP, Nagy MA, Stroud H, Koutsioumpa C, Wilcox OF, Assad EG, Green J, Harvey CD, Griffith EC, et al. : A scalable platform for the development of cell-type-specific viral drivers. Elife 2019, 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Siegert S, Cabuy E, Scherf BG, Kohler H, Panda S, Le YZ, Fehling HJ, Gaidatzis D, Stadler MB, Roska B: Transcriptional code and disease map for adult retinal cell types. Nat Neurosci 2012, 15:487–495, s481–482. [DOI] [PubMed] [Google Scholar]
  • 63.Juttner J, Szabo A, Gross-Scherf B, Morikawa RK, Rompani SB, Hantz P, Szikra T, Esposti F, Cowan CS, Bharioke A, et al. : Targeting neuronal and glial cell types with synthetic promoter AAVs in mice, non-human primates and humans. Nat Neurosci 2019, 22:1345–1356. [DOI] [PubMed] [Google Scholar]
  • 64.Sayeg MK, Weinberg BH, Cha SS, Goodloe M, Wong WW, Han X: Rationally Designed MicroRNA-Based Genetic Classifiers Target Specific Neurons in the Brain. ACS Synth Biol 2015, 4:788–795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Keaveney MK, Tseng HA, Ta TL, Gritton HJ, Man HY, Han X: A MicroRNA-Based Gene-Targeting Tool for Virally Labeling Interneurons in the Rodent Cortex. Cell Rep 2018, 24:294–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Vulchanova L, Schuster DJ, Belur LR, Riedl MS, Podetz-Pedersen KM, Kitto KF, Wilcox GL, McIvor RS, Fairbanks CA: Differential adeno-associated virus mediated gene transfer to sensory neurons following intrathecal delivery by direct lumbar puncture. Mol Pain 2010, 6:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Castle MJ, Cheng Y, Asokan A, Tuszynski MH: Physical positioning markedly enhances brain transduction after intrathecal AAV9 infusion. Sci Adv 2018, 4:eaau9859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Barson D, Hamodi AS, Shen X, Lur G, Constable RT, Cardin JA, Crair MC, Higley MJ: Simultaneous mesoscopic and two-photon imaging of neuronal activity in cortical circuits. Nat Methods 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Roska B, Sahel JA: Restoring vision. Nature 2018, 557:359–367. [DOI] [PubMed] [Google Scholar]
  • 70.Chen B, Li Y, Yu B, Zhang Z, Brommer B, Williams PR, Liu Y, Hegarty SV, Zhou S, Zhu J, et al. : Reactivation of Dormant Relay Pathways in Injured Spinal Cord by KCC2 Manipulations. Cell 2018, 174:1599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Trapani I, Auricchio A: Seeing the Light after 25 Years of Retinal Gene Therapy. Trends Mol Med 2018, 24:669–681. [DOI] [PubMed] [Google Scholar]
  • 72.Foust KD, Wang X, McGovern VL, Braun L, Bevan AK, Haidet AM, Le TT, Morales PR, Rich MM, Burghes AH, et al. : Rescue of the spinal muscular atrophy phenotype in a mouse model by early postnatal delivery of SMN. Nat Biotechnol 2010, 28:271–274. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 73.Valori CF, Ning K, Wyles M, Mead RJ, Grierson AJ, Shaw PJ, Azzouz M: Systemic delivery of scAAV9 expressing SMN prolongs survival in a model of spinal muscular atrophy. Sci Transl Med 2010, 2:35ra42. [DOI] [PubMed] [Google Scholar]
  • 74.Mendell JR, Al-Zaidy S, Shell R, Arnold WD, Rodino-Klapac LR, Prior TW, Lowes L, Alfano L, Berry K, Church K, et al. : Single-Dose Gene-Replacement Therapy for Spinal Muscular Atrophy. N Engl J Med 2017, 377:1713–1722. [DOI] [PubMed] [Google Scholar]
  • 75.Hinderer C, Katz N, Buza EL, Dyer C, Goode T, Bell P, Richman LK, Wilson JM: Severe Toxicity in Nonhuman Primates and Piglets Following High-Dose Intravenous Administration of an Adeno-Associated Virus Vector Expressing Human SMN. Hum Gene Ther 2018, 29:285–298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Xiong W, Wu DM, Xue Y, Wang SK, Chung MJ, Ji X, Rana P, Zhao SR, Mai S, Cepko CL: AAV cis-regulatory sequences are correlated with ocular toxicity. Proc Natl Acad Sci U S A 2019, 116:5785–5794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Hordeaux J, Wang Q, Katz N, Buza EL, Bell P, Wilson JM: The Neurotropic Properties of AAV-PHP.B Are Limited to C57BL/6J Mice. Mol Ther 2018, 26:664–668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Callaway EM, Luo L: Monosynaptic Circuit Tracing with Glycoprotein-Deleted Rabies Viruses. J Neurosci 2015, 35:8979–8985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Wickersham IR, Lyon DC, Barnard RJ, Mori T, Finke S, Conzelmann KK, Young JA, Callaway EM: Monosynaptic restriction of transsynaptic tracing from single, genetically targeted neurons. Neuron 2007, 53:639–647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Reardon TR, Murray AJ, Turi GF, Wirblich C, Croce KR, Schnell MJ, Jessell TM, Losonczy A: Rabies Virus CVS-N2c(DeltaG) Strain Enhances Retrograde Synaptic Transfer and Neuronal Viability. Neuron 2016, 89:711–724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Zeng WB, Jiang HF, Gang YD, Song YG, Shen ZZ, Yang H, Dong X, Tian YL, Ni RJ, Liu Y, et al. : Anterograde monosynaptic transneuronal tracers derived from herpes simplex virus 1 strain H129. Mol Neurodegener 2017, 12:38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Wojaczynski GJ, Engel EA, Steren KE, Enquist LW, Patrick Card J: The neuroinvasive profiles of H129 (herpes simplex virus type 1) recombinants with putative anterograde-only transneuronal spread properties. Brain Struct Funct 2015, 220:1395–1420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Zingg B, Chou XL, Zhang ZG, Mesik L, Liang F, Tao HW, Zhang LI: AAV-Mediated Anterograde Transsynaptic Tagging: Mapping Corticocollicular Input-Defined Neural Pathways for Defense Behaviors. Neuron 2017, 93:33–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Mundell NA, Beier KT, Pan YA, Lapan SW, Goz Ayturk D, Berezovskii VK, Wark AR, Drokhlyansky E, Bielecki J, Born RT, et al. : Vesicular stomatitis virus enables gene transfer and transsynaptic tracing in a wide range of organisms. J Comp Neurol 2015, 523:1639–1663. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES