Next Article in Journal
Characterization of the Mitochondrial Genome of a Wheat AL-Type Male Sterility Line and the Candidate CMS Gene
Next Article in Special Issue
Parkinson’s Disease: A Prionopathy?
Previous Article in Journal
Molecular Mechanisms of Drug Resistance in Glioblastoma
Previous Article in Special Issue
Proteolytic α-Synuclein Cleavage in Health and Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Adeno-Associated Viral Vectors as Versatile Tools for Parkinson’s Research, Both for Disease Modeling Purposes and for Therapeutic Uses

by
Ana Fajardo-Serrano
1,2,3,*,
Alberto J. Rico
1,2,3,
Elvira Roda
1,2,3,
Adriana Honrubia
1,2,3,
Sandra Arrieta
1,2,3,
Goiaz Ariznabarreta
1,2,3,
Julia Chocarro
1,2,3,
Elena Lorenzo-Ramos
1,2,3,
Alvaro Pejenaute
1,2,3,
Alfonso Vázquez
3,4 and
José Luis Lanciego
1,2,3,*
1
Centro de Investigación Médica Aplicada (CIMA), Department of Neurosciences, Universidad de Navarra, 31008 Pamplona, Spain
2
Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 28031 Madrid, Spain
3
Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
4
Department of Neurosurgery, Servicio Navarro de Salud, Complejo Hospitalario de Navarra, 31008 Pamplona, Spain
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(12), 6389; https://doi.org/10.3390/ijms22126389
Submission received: 24 May 2021 / Revised: 9 June 2021 / Accepted: 11 June 2021 / Published: 15 June 2021
(This article belongs to the Special Issue Neurobiology of Parkinson’s Disease)

Abstract

:
It is without any doubt that precision medicine therapeutic strategies targeting neurodegenerative disorders are currently witnessing the spectacular rise of newly designed approaches based on the use of viral vectors as Trojan horses for the controlled release of a given genetic payload. Among the different types of viral vectors, adeno-associated viruses (AAVs) rank as the ones most commonly used for the purposes of either disease modeling or for therapeutic strategies. Here, we reviewed the current literature dealing with the use of AAVs within the field of Parkinson’s disease with the aim to provide neuroscientists with the advice and background required when facing a choice on which AAV might be best suited for addressing a given experimental challenge. Accordingly, here we will be summarizing some insights on different AAV serotypes, and which would be the most appropriate AAV delivery route. Next, the use of AAVs for modeling synucleinopathies is highlighted, providing potential readers with a landscape view of ongoing pre-clinical and clinical initiatives pushing forward AAV-based therapeutic approaches for Parkinson’s disease and related synucleinopathies.

1. AAV Landscapes

Adeno-associated viral vectors (AAVs) are members of the Parvoviridae family and Dependoparvovirus genus. AAVs are non-pathogenic and replication-defective viruses, requiring a co-infection with adenovirus or herpes simplex virus to complete the replication cycle. In the absence of a helper virus, AAVs can produce a latent infection in which the viral genome persists, integrated in infected cells [1]. AAVs were first identified by electron microscopy as contaminants of adenoviral stocks produced in in vitro preparations [2]. AAVs are characterized by their non-enveloped icosahedral capsid carrying a 4.7 kilobase single-stranded linear DNA genome packaging that contains two open-reading frames encoding four non-structural proteins (Rep40, 52, 68, and 78), the capsid (Cap) gene encoding for three structural proteins (VP1, VP2, and VP3), and a small viral co-factor required for assembly known as the assembly-activating protein. The replication gene (Rep) encodes four proteins that are essential for replication, packaging, and transcriptional regulation, and it is the key component in establishing the site-specific integration. The genome is flanked by two 145 nucleotides inverted terminal repeats (ITRs) that fold into T-shaped hairpins, which are the only essential sequences required in cis for the replication and packaging of the AAV genome [3]. Although the viral capsid structure is preserved, different capsid surface domains and slight variations in the amino acid composition result in different AAV serotypes, with up to 12 natural serotypes (from AAV1 to AAV12) and more than 100 variants being described to date [4]. A preferential CNS tropism has been reported for AAV1, AAV2, AAV4, AAV5, AAV7, AAV8, AAV9, and AAVrh10 [5], where AAV9 and AAVrh10 ranked as the serotypes showing the highest CNS tropism (Tanguy et al., 2015; Karim Bey et al., 2020).
AAV vectors containing the Rep sequence have a preferential integration into the AAVS1 site (only present in humans and NHP) of Ch19, whereas most Rep-independent recombinant AAVs vectors will not include Rep and will not integrate specifically. AAVs are promising for treating a number of neurological disorders [6]. However, random integration may result in mutagenesis potential and oncogenesis [7]. Accordingly, for the production of a given recombinant AAV, the Rep and Cap genes of the wild-type AAV are replaced by the transgene of interest and the regulatory sequences required for the promoter and the poly A sequence [8,9,10]. It is worth noting that AAVs allow transduction of both dividing and non-dividing cells, affording a stable, long-term gene expression, maintained up to 8 years in non-human primates (NHPs) [11] and up to 10 years in humans [12]. The ability of AAVs for transducing post-mitotic cells such as neurons is an important characteristic when dealing with AAV use in neurodegenerative disorders, as these viral vectors currently represent the safest choice for gene therapy. Therapeutic approaches other than AAVs, such as small molecules, nanocarriers, and exosomes (among others) are currently under development, collectively also representing appealing choices that still remain in early developmental stages [13,14,15]. Nevertheless, the AAV field is currently in the spotlight, already entering the clinical field, and based on robust preclinical evidence. Indeed, AAV-based therapies afford a stable, long-term expression for a given gene, with a disease-modifying potential after one single AAV delivery.
There are two main approaches for gene transfer into the CNS, the in vivo and ex vivo strategies. In the ex vivo approach, neurons are genetically engineered to produce the therapeutic protein and then transplanted into the host. The in vivo approach involves the direct insertion of genetic material into the CNS, mainly through the injection of viruses containing the gene of interest [16]. For the purposes of this review, we will remain focused only on the use of AAVs for the in vivo approach.
When designing any given AAV-based experiment in the CNS field, a number of important items regarding the AAV structure itself need to be taken into consideration, namely the selection of the best suited AAV serotype together with the choice of the most convenient promoter (e.g., the sequence driving transgene expression). That said, it is also known that different AAV serotypes exhibit preferred cellular-specific tropism; therefore, the AAV serotype choice can further be tailored when targeting a given brain territory and indeed important differences can be observed between AAV deliveries in adult vs. neonatal specimens [17].
Promoters can broadly be categorized as either ubiquitous or cell-specific, i.e., ubiquitous promoters drive transgene transduction both in neurons and glial cells, whereas a broad portfolio exists for specific expression in neurons vs. different types of glial cells, as well as within a number of neuronal phenotypes [18]. Regarding ubiquitous promoters for CNS delivery, CAG, CBA, GUSB, and EF1α are the ones most commonly used [18]. CAG is a strong synthetic promoter frequently used in mammalian expression vectors composed by the cytomegalovirus early enhancer element (C), the promoter of the chicken beta-actin gene (A), and the splice acceptor of the rabbit beta-globin gene (G). CBA is the chicken beta-actin promoter; GUSB is the promoter of beta-glucuronidase. Finally, EF1α is a constitutive promoter of human elongation factor-1 alpha.
Although using an efficient promoter is obviously a pre-requisite for any successful gene therapy approach, it is worth noting that different promoters may have different potencies when driving transgene expression. Indeed, when considering clinical translation, the choice of a strong promoter might not be viewed as the best strategy, bearing in mind that undesired side effects such as toxicity phenomena driven by unwanted excessive transduction of the encoded gene may appear. In other words, within the pre-clinical stage of development, one should ensure appropriate selection of the weakest promoter driving sufficient transgene expression.
Moreover, interactions between AAV capsids and promoters driving cell type transduction regardless of virus permissivity have been recently reported in both rodent and NHPs, and these interactions include widely-used promoters such as CBA and JetI [19,20].
In summary, a balanced choice between the AAV serotype and promoter is required for a successful outcome of any gene therapy-based experiment. Moreover, and considering the limited cloning capacity of AAVs, the use of small-sized promoters is a convenient choice for leaving enough cargo space to accommodate large genes of interest, if needed [21,22].

2. Routes for AAV Delivery

Once a final choice is made on the selected AAV serotype and promoter, the next decision to be taken is which would be the most appropriate delivery route maximizing viral transduction within the desired area. It is worth stressing the fact that the choice of delivery route represents a critical issue in terms of safety and efficacy of the desired therapeutics. Besides delivery routes targeting neurosensory organs such as the eye or the cochlea, the most frequently used approaches for CNS applications can be broadly categorized into: (i) intraparenchymal, (ii) intra-CSF (intrathecal or lumbar administration, intracisternal and intracerebroventricular), and (iii) intravenous (Hudry and Vandenberghe, 2019). Moreover, the choice of a given delivery route is also often disease-tailored or even customized by disease stages of progression (Table 1).
The most commonly used routes for AAV deliveries within the context of Parkinson disease are the intraparenchymal, intra-CSF (lumbar, intracisternal, or intracerebroventricular), and intravenous. Other options less commonly used are either subpial [106,107] or intranasal [108]. The intranasal route of administration has often been considered for experimental therapies in the field of Parkinson’s disease (PD), other than AAV deliveries [13,14]. See Figure 1.

2.1. Intraparenchymal Deliveries

Intraparenchymal AAV delivery stands on stereotaxic surgery, a procedure where a needle or cannula is inserted directly in the desired target area, as defined with three coordinates (e.g., rostrocaudal, mediolateral, and dorsoventral coordinates). By delivering the viral vector this way, a high transduction efficiency is expected; therefore, the intraparenchymal delivery is the choice most frequently used in both pre-clinical and clinical studies (see Table 1 and Table 2) [109,110,111]. This route of administration offers a number of advantages, such as (i) maximizing viral concentrations into a specific brain area, (ii) no need for bypassing the blood brain barrier (BBB), (iii) broad cellular transduction expected even when infusing small volumes of viral particles, (iv) reduced immune response, and (v) very limited presence, if any, of off-target effects, i.e., transgene expression in peripheral body organs other than the brain. Reduced budgetary constraints also represent an additional advantage when compared to either intra-CSF or intravenous deliveries, where much larger volumes and higher viral titrations are required. Not surprisingly, the intraparenchymal delivery route has been appointed as the most popular choice for most of the currently ongoing clinical trials dealing with CNS neurodegenerative disorders, in particular for brain disorders where the initial pathological insult is limited to specific brain areas such as the substantia nigra in Parkinson’s disease (Table 2). When considering intraparenchymal AAV delivery, transduction levels in the brain are dependent on the (i) injected volume, (ii) rate of injection, (iii) viral titration, and (iv) affinity of a given AAV serotype for cell receptors at the injection site. For intraparenchymal deliveries targeting broad brain areas like the caudate-putamen nuclei, where maximizing the transduction area within the target structure is desired, the use of injection strategies such as pressurized convection-enhanced delivery (CED) [111,112,113] likely represents the most convenient choice.
It is also worth noting that the gene therapy field is a quickly changing scenario, with new arrivals being incorporated at a breath-taking speed, in particular those related to the new viral capsid variants designed for the retrograde spread of the AAV particles through brain circuits, ultimately transducing neurons from distant territories innervating the injection site. Although a number of AAV serotypes exhibit some inherent degree of retrograde spread, AAV6 and AAV9 in particular [61,128], newly-designed AAV capsid variants such as AAV2-retro [129], AAV-TT [82], AAV2-HBKO [83], and “barcoded” AAV-MNM004 and AAV-MNM008 [84] enable widespread transgene expression through retrograde transport. AAV2-retro and AAV2-HBKO have been successfully tested in both rodents and non-human primates [50,81,83], whereas AAV-TT, AAV-MNM004, and AAV-MNM008 have been so far only tested in rodents [82,84]. These AAV capsid variants share the option of transducing multiple brain areas with just one AAV deposit; therefore, they are very much appealing for the development of gene therapy treatments for neurodegenerative disorders characterized by the widespread aggregation of misfolded proteins. A similar rationale also applies when using retrogradely spreading AAVs for the purpose of modeling disseminated neurodegenerative proteinopathies in experimental animals. In this regard, the choice of the area best suited for retro-AAV delivery is dependent on the neurodegenerative disorder to be modeled.

2.2. Intra-CSF Deliveries

When considering AAV deliveries within CSF spaces, the distribution of viral particles will depend on the CSF circulation patterns, thereby raising some concerns about the inter-subject variability of intra-CSF deliveries. CSF is produced by choroid plexuses through brain ventricles, circulating through the ventricular system and finally reaching the subarachnoid space from where it is drained to the peripheral blood circulation [130] (Figure 2). CSF is produced continuously, being fully replaced up to five times per day.
Intra-CSF AAV deliveries collectively represent another feasible way for viral vector administration. This administration route bypasses the BBB, and it is less invasive than the intraparenchymal delivery (e.g., for intra-CSF deliveries other than the lateral ventricles, there is no need for stereotaxic surgery). Furthermore, when compared to intravenous administration, intra-CSF deliveries are expected to induce a lower immune response in peripheral organs. It can be performed either through lumbar puncture (e.g., intrathecal), the cisterna magna, or into the lateral cerebral ventricles [131]. Both AAV2.5 and AAV9 serotypes are able to achieve cellular transduction in the brain and spinal cord parenchyma following an intrathecal injection [86], and the same holds true for AAV-PHP.B [99] and AAVrh10 [96]. Although this delivery route has its own inherent advantages, vector dilution and the limited penetration/transduction of deep brain structures (e.g., poor diffusion from ventricles and subarachnoid space) are both limiting factors that need to be properly balanced before pushing forward intra-CSF deliveries for the purposes of animal modeling and/or disease interventions with a gene therapy therapeutic candidate. Intrathecal access to the subarachnoid CSF is achieved in routine clinical practice. Although less commonly used in daily clinical practice, access to the cisterna magna though an intracisternal approach instead of lumbar puncture can be 10 to 100-fold more efficient for brain transduction [95], despite a potential risk of damaging the medulla oblongata. In studies conducted in dogs, where intracerebroventricular and intracisternal delivery routes were compared, a similar transduction efficacy has been observed; however, encephalitis was reported as an unwanted event in animals after intracerebroventricular administration [131].

2.3. Intravenous Deliveries

The case of intravenous AAV administration is viewed as a relatively simple procedure, and much less invasive; however, it is also less effective for brain transduction as a result of the limited BBB passage for most AAV serotypes. Furthermore, when AAVs are delivered this way, a number of unwanted issues need to be taken into consideration, such as off-target effects, associated immune response, and potential toxicity phenomena. Although it is not an intrinsic feature of AAVs, some viral vector serotypes such as AAVrh10 [132] and AAV9 (the latter to a smaller extent, see [94,133,134]) can cross the BBB. Within the field of Parkinson’s disease, it seems that the limited penetration of the BBB by any given AAV serotype would result in a non-relevant therapeutic concentration ultimately reaching deep brain structures like the basal ganglia. In an attempt to overcome these limitations, an AAV9 variant known as AAV-PHP.B was introduced in 2016 [135], and was designed for more efficient BBB bypassing. It has been reported elsewhere that the improved penetration of the BBB by AAV-PHP.B is limited to the C57BL/6J mice strain [136], and the same likely applies to non-human primates [98]. Moreover, it is worth noting that BBB bypassing is more effective in neonatal rodent brains than in adults [137,138]. More recently, an improved version named AAV-PHP.eB has been released, showing a more efficient neuronal transduction in rats after intravenous administration [104,139]. At present, consensus exists on improved brain transduction of AAV-PHP.B variants compared to native AAV9 when injected into the cisterna magna in rodents [92]; however, the use of AAV-PHP.B in non-human primates is still a matter of debate. Moreover, the expression of the lymphocyte antigen-6 receptor (Ly6a) likely is the determining factor for the differential efficacy of AAV-PHP.B in transducing the CNS [140] across different mouse strains, as recently reported elsewhere [141]. Ly6a is almost exclusively expressed in brain endothelial cells, except for a minor expression in astrocytes and microglia, thus facilitating the BBB passage [140]. Interestingly, the presence of the Ly6 locus in different mice strains is required for ensuring the transduction ability of some AAV serotypes [142]. Indeed, the lack of the Ly6a gene in NHPs likely explains the suboptimal performance of AAV-PHP.B in these animals [140,142,143].

2.4. Other Delivery Routes

Although technically very demanding, a subpial delivery route represents another feasible choice at hand. Subpial deliveries at cervical and/or lumbar spinal cord levels have been carried out in mice, pigs, and NHPs, showing efficient transduction of the spinal cord motor neurons [107]. When compared to the intrathecal administration of AAVs, the subpial approach seems to result in superior transgene expression. Bearing in mind that the length of the spinal cord in adult pigs of 35–40 kg of body weight is roughly similar to that of humans, a similar transduction would be expected [106].
Finally, the intranasal delivery route can also be considered as an alternative, less-invasive option. From the available AAV serotypes, AAV8 and AAV9 show the highest tropism for efficiently transducing olfactory sensory neurons [108,144]. In this regard, the intranasal approach has been suggested as an easy way for gaining direct CNS access, with an appealing potential for the treatment of neurodegenerative disorders [137].
When considering delivery routes all together, beyond the pros and cons stated above, two additional items merit extra attention. On the one hand, when compared to intraparenchymal deliveries, it is without doubt that intra-CSF and intravenous administrations require higher AAV titrations and volumes, something that can be viewed as an additional disadvantage particularly related to dose-escalation productions for clinical use. On the other hand, the presence of pre-existing neutralizing antibodies also represents a matter of concern, particularly regarding delivery routes other than intraparenchymal.

3. AAVs for Animal Modeling of Parkinson’s Disease

Although neurotoxin-based mammalian animal models of PD (6-OHDA and MPTP) have been instrumental in setting up the basis of our current understanding of basal ganglia function and dysfunction [145,146,147], these symptomatic models have failed to recapitulate the known neuropathology of this neurodegenerative disorder, where dopaminergic neurons die upon the progressive intracytoplasmic aggregation of misfolded alpha-synuclein (α-syn). Accordingly, the field is quickly moving forward towards the implementation of animal models of PD-like synucleinopathies in an attempt to lay the groundwork needed for the design and testing disease-modifying therapies [148].

3.1. Models Based on AAV-Mediated α-syn Overexpression

The discovery that α-syn is the major component of Lewy bodies [149] drastically changed the field of animal modeling in PD. Initial attempts were based on the development of several different murine transgenic mice lines overexpressing different forms of either mutated or wild-type α-syn. Although these transgenic mice lines are appealing choices for testing new therapeutic candidates targeting α-syn aggregation, in most cases these transgenic models lacked the appropriate phenotype and, indeed, neuronal loss in the substantia nigra pars compacta (SNpc) is often weak or even absent [150].
While transgenic mice have driven significant advances in the understanding of synucleinopathies, the use of viral vectors coding for α-syn have quickly become more popular in the field. These models stand on the use of viral vectors directly injected into the SNpc for transducing dopaminergic neurons with α-syn. From the available arsenal of viral vectors, both lentiviruses and AAVs have been the most often used choices [32,151]. The lentivirus-mediated transfer of α-syn resulted in dopaminergic (DA) neuronal loss in the SNpc after 6 weeks post-delivery [152]. Moreover, the use of AAVs overexpressing human A53T α-synuclein in rats resulted in the strong transduction of α-syn within TH+ neurons in the SNpc (above 90% of cells expressing α-syn), further inducing a variable degree of DA cell loss with different follow-up times [65,153,154,155]. Both AAV2 and AAV5 serotypes coding for α-syn have also been used in marmosets, achieving a well-established neuropathology together with a consistent DA cell loss ranging from 30 to 80% [146,151,153]. Quite a similar approach has been successfully tested in Macaca fascilularis, resulting in an up to 50% loss of TH+ neurons after a survival time of 17 weeks [156]. More recently, and by taking advantage of an AAV9 coding for A53T mutated α-syn, our group has reported a 39% loss of DA neurons in Macaca fascicularis 12 weeks post injection into the SNpc [157]. Although different choices made regarding AAV serotypes, promoters chosen, and ranges of survival times may account for the reported data dealing with the percentages reported above for α-syn induced DA cell death, it seems clear that the AAV-mediated enhancement of α-syn currently is in the spotlight in terms of development and validation of a new generation of animal models of synucleinopathy.
When using AAV coding for α-syn for the purposes of PD modeling, there are a number of issues that need to be taken into consideration. For instance, it is well known that marmosets harbor a naturally occurring A53T mutation in the SNCA gene, which could generate protection against A53T overexpression [72,148,158]. Next, although the A53T mutation is the one most broadly used, AAVs carrying mutations other than A53T, such as A30P and E36K, have also been successfully used in mice and marmosets [151,152]. Finally, it has been recently reported that the introduction of additional elements such as the woodchuck hepatitis virus post-transcriptional regulatory element (WPRE) enabled a greater nigrostriatal degeneration using AAV9 in mice [73] when compared with AAVs lacking WPRE.

3.2. Models Based on AAV-Mediated Neuromelanin Overexpression

Although animal models based on the AAV-mediated expression of α-syn collectively represent a major step forward in the field of PD and related synucleinopathies, it should be noted that these models share a common caveat, i.e., the obtained AAV-mediated expression of α-syn is well above what occurs naturally in physiological terms. In other words, the α-syn expression obtained this way is too high; therefore, although the loss of DA neurons is more progressive than neurotoxin-based models, it is quicker than desired, and DA neurons die too early from excessive α-syn production, i.e., before having a chance of aggregating α-syn in the form of Lewy bodies, as occurs in the human pathological conditions.
That said, it is worth recognizing that PD is a degenerative disorder only observed in humans, and animal species other than humans are not suffering from this disease. When thinking about what drives this specific susceptibility, the neuromelanin-related pigmentation of the SNpc in humans (the same applies to the locus cerouleus) is something unique, not shared with any other animal species; therefore, it can be hypothesized that neuromelanin accumulation may account for DA cell vulnerability. Indeed, neuromelanin aggregation in humans is a time-dependent phenomenon, something that needs to be taken into consideration bearing in mind that advanced age is the main known risk for PD. Only aged non-human primates exhibited some degree of pigmentation in the SNpc and locus cerouleus, although to an extent not comparable to that of the human SNpc. Such a physiological, time-dependent aggregation of neuromelanin in NHPs is preferentially observed in DA neurons located in the lateral and caudal regions of the SNpc, where the most vulnerable DA neurons are located (unpublished data).
While to what extent neuromelanin aggregation can be blamed for DA cell vulnerability is still an unresolved debate, a direct relationship between Lewy bodies and neuromelanin aggregates has been reported elsewhere [159]. Although this rationale (together with more evidence available in the literature remaining outside the focus of this review) can be taken as a smoking gun linking neuromelanin aggregation with PD pathophysiology, the most commonly used laboratory experimental animals lack neuromelanin, so such a potential link has been neglected so far in the field.
This scenario has completely changed very recently upon the design and validation of an AAV-based rodent model of PD characterized by neuromelanin aggregation [160]. These researchers have injected an AAV coding for human tyrosinase (the rate-limiting enzyme for neuromelanin) into the SNpc in rats. When injected this way, an age-dependent aggregation of neuromelanin is observed in the rat SNpc up to similar levels than those observed in elderly humans, together with a time-dependent PD-like symptomatic phenotype, nigrostriatal degeneration, and the presence of Lewy body-like aggregates. Although the mechanisms sustaining a link between neuromelanin production and α-syn accumulation are still poorly understood, a major advantage of this model when compared to AAV-mediated models of synucleinopathy is represented by the fact that the observed α-syn aggregation is physiological and driven by neuromelanin expression rather than directly induced by the injected AAV itself. In summary, this new model currently holds great promise and likely represents the animal model so far best mimicking the known pathophysiology of PD.

3.3. Other AAV-Based Models

Although models based on AAV-mediated α-syn overexpression are by far the ones most commonly used at present, a number of alternatives have been made available, including models related to leucine-rich repeated kinase 2 (LRRK2), and PTEN-induced kinase 1 (PINK1).
LRRK2 mutations are associated with autosomal dominant inheritance in familiar PD [161]. The generation of viral vector-mediated LRRK2 PD models using either adenoviruses or lentiviruses in rodents has been reported elsewhere [162,163]. LRRK2-based models failed to recapitulate several aspects of PD neuropathology, such as Lewy bodies and α-syn aggregates.
PTEN-induced kinase 1 (PINK1) is a serine/threonine-protein kinase. Knocking-down PINK1 with AAVs did not generated a significant dopaminergic cell death with a follow-up of up to 3 weeks in rodents [164]. Further research efforts will be needed to properly validate animal models based on the AAV-mediated administration of PINK1 interference mRNAs.

4. Use of AAV-Mediated Gene Transfer for Therapeutic Purposes

The use of AAVs for the treatment of CNS disorders exemplifies the translation of pre-clinical evidence towards clinical trials, beginning with pioneer experiences [8,165] up to a growing list of clinical trials (Table 2). Among the different AAV serotypes available, AAV2 and AAV9 ranked as the most commonly used within the context of PD (Table 2).
When considering PD under a simplistic view as a basal ganglia-related disorder primarily affecting the nigrostriatal pathway, the most rational scenario implies an intraparenchymal delivery route administering a given AAV either into the SNpc or in the striatum [166,167]. Although the SNpc is located very deeply in the brain and thus represents a demanding surgical target, its relatively small size makes the SNpc an affordable area to be covered by a direct intraparenchymal injection of an AAV. On the other hand, targeting the striatum is a more feasible surgical approach and indeed by going this way larger volumes of AAVs can be accommodated. Not surprisingly, most of the ongoing clinical trials are based on focused AAV deliveries targeting either the striatum or the SNpc (Table 2).
Gene therapy ongoing clinical trials for PD can broadly be categorized into (i) dopamine-related, (ii) neurotrophic factors, (iii) neurotransmitters and neuromodulation, and (iv) disease-specific mutations. Dopamine-related approaches take advantage of AAV coding for dopa-decarboxylase (AADC), the enzyme in charge of converting levodopa into dopamine. Early-stage clinical trials with AAV-AADC showed good safety and efficacy profiles, directly related to the percentage of the putaminal area transduced with the therapeutic vector [114,115,168,169], paving the ground for an ongoing phase II trial. However, AAV-mediated restoration of dopamine levels cannot be viewed as a disease-modifying strategy for PD. Quite the opposite applies to a number of gene therapy initiatives based on the use of AAVs as carriers of genes coding for neurotrophic factors such as glial cell-line neurotrophic factor (GDNF), and its close relative known as neurturin (NRTN). Both GDNF and NRTN exert a dopaminotrophic effect and are therefore likely good candidates for gene therapies. By taking advantage of intraputaminal AAV2-GDNF deliveries, disease progression was stabilized with a follow-up of 18 months, together with an enhanced uptake of the radiotracer F-Dopa [170] despite a somewhat limited transduced area of the putamen. Obtained endpoints using AAV2-NRTN instead of AAV2-GDNF under a similar approach showed less conclusive results [126], likely due to NRTN-increased expression restricted to the injection sites [171].
A completely different rationale underlies an AAV-based initiative for tuning down the hyperexcitability of the subthalamic nucleus (STN) that typically accounts in PD. With the purpose of switching the functional activity of the STN from excitation to inhibition, using AAV coding for the enzyme glutamic acid decarboxylase (GAD), [172]. Obtained results showed the safety of this approach, together with clinical improvement (measured by UPDRS motor scores) after 12 months of follow-ups. However, a subsequent phase II randomized trial failed to demonstrate motor improvement [121].
The demonstration that heterozygous mutations in the GBA1 gene (coding for a lysosomal enzyme known as glucocerebrosidase; GCase) are the main genetic risk factors for PD [173] has opened strong interest for increasing GCase activity as a potential disease-modifying treatment. Promising results obtained in pre-clinical studies carried out in mice and in NHPs [105,157,174] have paved the way for clinical translation, and in this regard it is worth noting that there is an ongoing clinical trial with PD patients carrying heterozygous GBA1 mutations through the intracisternal delivery of AAV9-GBA1.

5. Conclusions

It is without a doubt that we are at the beginning of a promising journey where gene therapy-based approaches will progressively gain appeal and popularity. Regarding the use of AAVs for the purposes of animal modeling, it is clear that any animal model has its own strengths and shortcomings. However, by taking advantage of the impressive versatility of AAVs at present we are closer than ever when adequately mimicking the underlying pathophysiology that typically characterizes PD. Furthermore, when coming to translational initiatives for disease-modifying purposes, both earlier and ongoing attempts have succeeded in demonstrating safety; although, efficacy needs to be improved. In this regard, finding a proper balance between the underlying rationale for a given approach and the most appropriate route for AAV deliveries are the key items ultimately driving clinical success. Within this scenario, it should be stressed that brain function and dysfunction specifically stand on neuronal circuits (e.g., neuronal circuits provide the brain with its unique identity and operational principles when compared to any other body organ); therefore, researchers and clinicians going this way should ensure they have a strong neuroanatomical background. Finally, it is worth noting that this is a quickly changing scenario where the best is surely yet to come.

Author Contributions

Conceptualization, all authors; literature review, E.R., A.H., S.A., G.A., J.C., E.L.-R., A.P.; writing—original draft preparation, writing—review and editing, A.F.-S., A.J.R., A.V. and J.L.L.; supervision, A.F.-S., A.J.R. and J.L.L.; funding acquisition, J.L.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the ERC Advanced Grant, grant number 340527; CiberNed’s Intramural Program Grant, grant number PI2017/02; FEDER/Ministerio de Ciencia, Innovación y Universidades–Agencia estatal de Investigación, grant number BFU2017-82407-R and by the Department of Health of the Government of Navarra, grant numbers 046-2017_NAB7 and 011-1383-2019-000006 (PI031).

Institutional Review Board Statement

Submission has been approved by the Institutional Review Board Statement of the Centro de Investigación Médica Aplicada (CIMA), University of Navarra.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data reported here are available from authors upon reasonable request.

Acknowledgments

Cartoons included in Figure 1 and Figure 2 were taken and modified from Servier Medical Art (https://smart.servier.com/), last access 14 June 21, licensed under a Creative Commons Attribution 3.0 Unported License (https://creativecommons.org/licenses/by/3.0/), last access 14 June 2021.

Conflicts of Interest

J.L.L. is co-founder and paid consultant of Handl Therapeutics B.V. (Leuven, Belgium).

References

  1. Murlidharan, G.; Samulski, R.J.; Asokan, A. Biology of Adeno-Associated Viral Vectors in the Central Nervous System. Front. Mol. Neurosci. 2014, 7, 76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Atchison, R.W.; Casto, B.C.; Hammon, W.M. Adenovirus-Associated Defective Virus Particles. Science 1965, 149, 754–756. [Google Scholar] [CrossRef]
  3. Tseng, Y.-S.; Agbandje-McKenna, M. Mapping the AAV Capsid Host Antibody Response toward the Development of Second Generation Gene Delivery Vectors. Front. Immunol. 2014, 5, 9. [Google Scholar] [CrossRef] [Green Version]
  4. Salganik, M.; Hirsch, M.L.; Samulski, R.J. Adeno-Associated Virus as a Mammalian DNA Vector. Microbiol. Spectr. 2015, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Saraiva, J.; Nobre, R.J.; Pereira de Almeida, L. Gene Therapy for the CNS Using AAVs: The Impact of Systemic Delivery by AAV9. J. Control. Release 2016, 241, 94–109. [Google Scholar] [CrossRef] [PubMed]
  6. Haberman, R.P.; McCown, T.J.; Samulski, R.J. Inducible Long-Term Gene Expression in Brain with Adeno-Associated Virus Gene Transfer. Gene Ther. 1998, 5, 1604–1611. [Google Scholar] [CrossRef] [Green Version]
  7. McCarty, D.M.; Young, S.M.; Samulski, R.J. Integration of Adeno-Associated Virus (AAV) and Recombinant AAV Vectors. Annu. Rev. Genet. 2004, 38, 819–845. [Google Scholar] [CrossRef] [PubMed]
  8. Flotte, T.R.; Carter, B.J. Adeno-Associated Virus Vectors for Gene Therapy. Gene Ther. 1995, 2, 357–362. [Google Scholar]
  9. Carter, P.J.; Samulski, R.J. Adeno-Associated Viral Vectors as Gene Delivery Vehicles. Int. J. Mol. Med. 2000, 6, 17–27. [Google Scholar] [CrossRef]
  10. Gaj, T.; Sirk, S.J.; Shui, S.-L.; Liu, J. Genome-Editing Technologies: Principles and Applications. Cold Spring Harb. Perspect. Biol. 2016, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Hadaczek, P.; Eberling, J.L.; Pivirotto, P.; Bringas, J.; Forsayeth, J.; Bankiewicz, K.S. Eight Years of Clinical Improvement in MPTP-Lesioned Primates after Gene Therapy with AAV2-HAADC. Mol. Ther. 2010, 18, 1458–1461. [Google Scholar] [CrossRef] [PubMed]
  12. Leone, P.; Shera, D.; McPhee, S.W.J.; Francis, J.S.; Kolodny, E.H.; Bilaniuk, L.T.; Wang, D.-J.; Assadi, M.; Goldfarb, O.; Goldman, H.W.; et al. Long-Term Follow-up after Gene Therapy for Canavan Disease. Sci. Transl. Med. 2012, 4, 165ra163. [Google Scholar] [CrossRef] [Green Version]
  13. Aly, A.E.-E.; Waszczak, B.L. Intranasal Gene Delivery for Treating Parkinson’s Disease: Overcoming the Blood–Brain Barrier. Expert Opin. Drug Deliv. 2015, 12, 1923–1941. [Google Scholar] [CrossRef]
  14. Rehman, S.; Nabi, B.; Zafar, A.; Baboota, S.; Ali, J. Intranasal Delivery of Mucoadhesive Nanocarriers: A Viable Option for Parkinson’s Disease Treatment? Expert Opin. Drug Deliv. 2019, 16, 1355–1366. [Google Scholar] [CrossRef]
  15. Haney, M.J.; Klyachko, N.L.; Zhao, Y.; Gupta, R.; Plotnikova, E.G.; He, Z.; Patel, T.; Piroyan, A.; Sokolsky, M.; Kabanov, A.V.; et al. Exosomes as Drug Delivery Vehicles for Parkinson’s Disease Therapy. J. Control. Release 2015, 207, 18–30. [Google Scholar] [CrossRef] [Green Version]
  16. de Lima, M.C.P.; da Cruz, M.T.G.; Cardoso, A.L.C.; Simoes, S.; de Almeida, L.P. Liposomal and Viral Vectors for Gene Therapy of the Central Nervous System. Curr. Drug Targets CNS Neurol. Disord. 2005, 4, 453–465. [Google Scholar] [CrossRef]
  17. Gao, G.; Zhong, L.; Danos, O. Exploiting Natural Diversity of AAV for the Design of Vectors with Novel Properties. Methods Mol. Biol. 2011, 807, 93–118. [Google Scholar] [CrossRef] [PubMed]
  18. Haery, L.; Deverman, B.E.; Matho, K.S.; Cetin, A.; Woodard, K.; Cepko, C.; Guerin, K.I.; Rego, M.A.; Ersing, I.; Bachle, S.M.; et al. Adeno-Associated Virus Technologies and Methods for Targeted Neuronal Manipulation. Front. Neuroanat. 2019, 13, 93. [Google Scholar] [CrossRef] [Green Version]
  19. Powell, S.K.; Samulski, R.J.; McCown, T.J. AAV Capsid-Promoter Interactions Determine CNS Cell-Selective Gene Expression In Vivo. Mol. Ther. 2020, 28, 1373–1380. [Google Scholar] [CrossRef] [PubMed]
  20. Bohlen, M.O.; McCown, T.J.; Powell, S.K.; El-Nahal, H.G.; Daw, T.; Basso, M.A.; Sommer, M.A.; Samulski, R.J. Adeno-Associated Virus Capsid-Promoter Interactions in the Brain Translate from Rat to the Nonhuman Primate. Hum. Gene Ther. 2020, 31, 1155–1168. [Google Scholar] [CrossRef]
  21. Pignataro, D.; Sucunza, D.; Vanrell, L.; Lopez-Franco, E.; Dopeso-Reyes, I.G.; Vales, A.; Hommel, M.; Rico, A.J.; Lanciego, J.L.; Gonzalez-Aseguinolaza, G. Adeno-Associated Viral Vectors Serotype 8 for Cell-Specific Delivery of Therapeutic Genes in the Central Nervous System. Front. Neuroanat. 2017, 11, 2. [Google Scholar] [CrossRef] [Green Version]
  22. Pignataro, D.; Sucunza, D.; Rico, A.J.; Dopeso-Reyes, I.G.; Roda, E.; Rodríguez-Perez, A.I.; Labandeira-Garcia, J.L.; Broccoli, V.; Kato, S.; Kobayashi, K.; et al. Gene Therapy Approaches in the Non-Human Primate Model of Parkinson’s Disease. J. Neural. Transm. 2018, 125, 575–589. [Google Scholar] [CrossRef]
  23. Brady, M.L.; Raghavan, R.; Singh, D.; Anand, P.J.; Fleisher, A.S.; Mata, J.; Broaddus, W.C.; Olbricht, W.L. In Vivo Performance of a Microfabricated Catheter for Intraparenchymal Delivery. J. Neurosci. Methods 2014, 229, 76–83. [Google Scholar] [CrossRef] [PubMed]
  24. Burger, C.; Gorbatyuk, O.S.; Velardo, M.J.; Peden, C.S.; Williams, P.; Zolotukhin, S.; Reier, P.J.; Mandel, R.J.; Muzyczka, N. Recombinant AAV Viral Vectors Pseudotyped with Viral Capsids from Serotypes 1, 2, and 5 Display Differential Efficiency and Cell Tropism after Delivery to Different Regions of the Central Nervous System. Mol. Ther. 2004, 10, 302–317. [Google Scholar] [CrossRef]
  25. Joniec-Maciejak, I.; Ciesielska, A.; Wawer, A.; Sznejder-Pachołek, A.; Schwenkgrub, J.; Cudna, A.; Hadaczek, P.; Bankiewicz, K.S.; Członkowska, A.; Członkowski, A. The Influence of AAV2-Mediated Gene Transfer of Human IL-10 on Neurodegeneration and Immune Response in a Murine Model of Parkinson’s Disease. Pharmacol. Rep. 2014, 66, 660–669. [Google Scholar] [CrossRef]
  26. Theodore, S.; Cao, S.; McLean, P.J.; Standaert, D.G. Targeted Overexpression of Human Alpha-Synuclein Triggers Microglial Activation and an Adaptive Immune Response in a Mouse Model of Parkinson Disease. J. Neuropathol. Exp. Neurol. 2008, 67, 1149–1158. [Google Scholar] [CrossRef] [Green Version]
  27. Cao, S.; Theodore, S.; Standaert, D.G. Fcγ Receptors Are Required for NF-ΚB Signaling, Microglial Activation and Dopaminergic Neurodegeneration in an AAV-Synuclein Mouse Model of Parkinson’s Disease. Mol. Neurodegener. 2010, 5, 42. [Google Scholar] [CrossRef] [Green Version]
  28. Thome, A.D.; Standaert, D.G.; Harms, A.S. Fractalkine Signaling Regulates the Inflammatory Response in an α-Synuclein Model of Parkinson Disease. PLoS ONE 2015, 10, e0140566. [Google Scholar] [CrossRef] [Green Version]
  29. Du, Y.; Zhang, X.; Tao, Q.; Chen, S.; Le, W. Adeno-Associated Virus Type 2 Vector-Mediated Glial Cell Line-Derived Neurotrophic Factor Gene Transfer Induces Neuroprotection and Neuroregeneration in a Ubiquitin-Proteasome System Impairment Animal Model of Parkinson’s Disease. Neurodegener. Dis. 2013, 11, 113–128. [Google Scholar] [CrossRef]
  30. Steidinger, T.U.; Slone, S.R.; Ding, H.; Standaert, D.G.; Yacoubian, T.A. Angiogenin in Parkinson Disease Models: Role of Akt Phosphorylation and Evaluation of AAV-Mediated Angiogenin Expression in MPTP Treated Mice. PLoS ONE 2013, 8, e56092. [Google Scholar] [CrossRef]
  31. Hadaczek, P.; Wu, G.; Sharma, N.; Ciesielska, A.; Bankiewicz, K.; Davidow, A.L.; Lu, Z.-H.; Forsayeth, J.; Ledeen, R.W. GDNF Signaling Implemented by GM1 Ganglioside; Failure in Parkinson’s Disease and GM1-Deficient Murine Model. Exp. Neurol. 2015, 263, 177–189. [Google Scholar] [CrossRef]
  32. Klein, R.L.; Hamby, M.E.; Sonntag, C.F.; Millard, W.J.; King, M.A.; Meyer, E.M. Measurements of Vector-Derived Neurotrophic Factor and Green Fluorescent Protein Levels in the Brain. Methods 2002, 28, 286–292. [Google Scholar] [CrossRef]
  33. Bäck, S.; Peränen, J.; Galli, E.; Pulkkila, P.; Lonka-Nevalaita, L.; Tamminen, T.; Voutilainen, M.H.; Raasmaja, A.; Saarma, M.; Männistö, P.T.; et al. Gene Therapy with AAV2-CDNF Provides Functional Benefits in a Rat Model of Parkinson’s Disease. Brain Behav. 2013, 3, 75–88. [Google Scholar] [CrossRef]
  34. Ciesielska, A.; Mittermeyer, G.; Hadaczek, P.; Kells, A.P.; Forsayeth, J.; Bankiewicz, K.S. Anterograde Axonal Transport of AAV2-GDNF in Rat Basal Ganglia. Mol. Ther. 2011, 19, 922–927. [Google Scholar] [CrossRef] [Green Version]
  35. Ciesielska, A.; Sharma, N.; Beyer, J.; Forsayeth, J.; Bankiewicz, K. Carbidopa-Based Modulation of the Functional Effect of the AAV2-HAADC Gene Therapy in 6-OHDA Lesioned Rats. PLoS ONE 2015, 10, e0122708. [Google Scholar] [CrossRef]
  36. Moon, H.C.; Won, S.Y.; Kim, E.G.; Kim, H.K.; Cho, C.B.; Park, Y.S. Effect of Optogenetic Modulation on Entopeduncular Input Affects Thalamic Discharge and Behavior in an AAV2-α-Synuclein-Induced Hemiparkinson Rat Model. Neurosci. Lett. 2018, 662, 129–135. [Google Scholar] [CrossRef]
  37. Gasmi, M.; Herzog, C.D.; Brandon, E.P.; Cunningham, J.J.; Ramirez, G.A.; Ketchum, E.T.; Bartus, R.T. Striatal Delivery of Neurturin by CERE-120, an AAV2 Vector for the Treatment of Dopaminergic Neuron Degeneration in Parkinson’s Disease. Mol. Ther. 2007, 15, 62–68. [Google Scholar] [CrossRef] [PubMed]
  38. Gasmi, M.; Brandon, E.P.; Herzog, C.D.; Wilson, A.; Bishop, K.M.; Hofer, E.K.; Cunningham, J.J.; Printz, M.A.; Kordower, J.H.; Bartus, R.T. AAV2-Mediated Delivery of Human Neurturin to the Rat Nigrostriatal System: Long-Term Efficacy and Tolerability of CERE-120 for Parkinson’s Disease. NeuroBiol. Dis. 2007, 27, 67–76. [Google Scholar] [CrossRef]
  39. Johnston, L.C.; Su, X.; Maguire-Zeiss, K.; Horovitz, K.; Ankoudinova, I.; Guschin, D.; Hadaczek, P.; Federoff, H.J.; Bankiewicz, K.; Forsayeth, J. Human Interleukin-10 Gene Transfer Is Protective in a Rat Model of Parkinson’s Disease. Mol. Ther. 2008, 16, 1392–1399. [Google Scholar] [CrossRef] [PubMed]
  40. Janelidze, S.; Nordström, U.; Kügler, S.; Brundin, P. Pre-Existing Immunity to Adeno-Associated Virus (AAV)2 Limits Transgene Expression Following Intracerebral AAV2-Based Gene Delivery in a 6-Hydroxydopamine Model of Parkinson’s Disease. J. Gene Med. 2014, 16, 300–308. [Google Scholar] [CrossRef]
  41. Zheng, D.; Jiang, X.; Zhao, J.; Duan, D.; Zhao, H.; Xu, Q. Subthalamic HGAD65 Gene Therapy and Striatum TH Gene Transfer in a Parkinson’s Disease Rat Model. Neural. Plast. 2013, 2013, 263287. [Google Scholar] [CrossRef] [Green Version]
  42. Fitzsimons, H.L.; Riban, V.; Bland, R.J.; Wendelken, J.L.; Sapan, C.V.; During, M.J. Biodistribution and Safety Assessment of AAV2-GAD Following Intrasubthalamic Injection in the Rat. J. Gene Med. 2010, 12, 385–398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. De Miranda, B.R.; Rocha, E.M.; Castro, S.L.; Greenamyre, J.T. Protection from α-Synuclein Induced Dopaminergic Neurodegeneration by Overexpression of the Mitochondrial Import Receptor TOM20. NPJ Parkinsons Dis. 2020, 6, 38. [Google Scholar] [CrossRef]
  44. Ren, X.; Zhang, T.; Gong, X.; Hu, G.; Ding, W.; Wang, X. AAV2-Mediated Striatum Delivery of Human CDNF Prevents the Deterioration of Midbrain Dopamine Neurons in a 6-Hydroxydopamine Induced Parkinsonian Rat Model. Exp. Neurol. 2013, 248, 148–156. [Google Scholar] [CrossRef]
  45. Herzog, C.D.; Bishop, K.M.; Brown, L.; Wilson, A.; Kordower, J.H.; Bartus, R.T. Gene Transfer Provides a Practical Means for Safe, Long-Term, Targeted Delivery of Biologically Active Neurotrophic Factor Proteins for Neurodegenerative Diseases. Drug Deliv. Transl. Res. 2011, 1, 361–382. [Google Scholar] [CrossRef]
  46. Herzog, C.D.; Brown, L.; Kruegel, B.R.; Wilson, A.; Tansey, M.G.; Gage, F.H.; Johnson, E.M.; Bartus, R.T. Enhanced Neurotrophic Distribution, Cell Signaling and Neuroprotection Following Substantia Nigral versus Striatal Delivery of AAV2-NRTN (CERE-120). NeuroBiol. Dis. 2013, 58, 38–48. [Google Scholar] [CrossRef]
  47. Bartus, R.T.; Brown, L.; Wilson, A.; Kruegel, B.; Siffert, J.; Johnson, E.M.; Kordower, J.H.; Herzog, C.D. Properly Scaled and Targeted AAV2-NRTN (Neurturin) to the Substantia Nigra Is Safe, Effective and Causes No Weight Loss: Support for Nigral Targeting in Parkinson’s Disease. NeuroBiol. Dis. 2011, 44, 38–52. [Google Scholar] [CrossRef]
  48. Wang, C.Y.; Guo, H.Y.; Lim, T.M.; Ng, Y.K.; Neo, H.P.; Hwang, P.Y.K.; Yee, W.-C.; Wang, S. Improved Neuronal Transgene Expression from an AAV-2 Vector with a Hybrid CMV Enhancer/PDGF-Beta Promoter. J. Gene Med. 2005, 7, 945–955. [Google Scholar] [CrossRef] [PubMed]
  49. Bartus, R.T.; Herzog, C.D.; Chu, Y.; Wilson, A.; Brown, L.; Siffert, J.; Johnson, E.M.; Olanow, C.W.; Mufson, E.J.; Kordower, J.H. Bioactivity of AAV2-Neurturin Gene Therapy (CERE-120): Differences between Parkinson’s Disease and Nonhuman Primate Brains. Mov. Disord. 2011, 26, 27–36. [Google Scholar] [CrossRef]
  50. Weiss, A.R.; Liguore, W.A.; Domire, J.S.; Button, D.; McBride, J.L. Intra-Striatal AAV2.Retro Administration Leads to Extensive Retrograde Transport in the Rhesus Macaque Brain: Implications for Disease Modeling and Therapeutic Development. Sci. Rep. 2020, 10, 6970. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Richardson, R.M.; Kells, A.P.; Rosenbluth, K.H.; Salegio, E.A.; Fiandaca, M.S.; Larson, P.S.; Starr, P.A.; Martin, A.J.; Lonser, R.R.; Federoff, H.J.; et al. Interventional MRI-Guided Putaminal Delivery of AAV2-GDNF for a Planned Clinical Trial in Parkinson’s Disease. Mol. Ther. 2011, 19, 1048–1057. [Google Scholar] [CrossRef] [PubMed]
  52. Su, X.; Kells, A.P.; Huang, E.J.; Lee, H.S.; Hadaczek, P.; Beyer, J.; Bringas, J.; Pivirotto, P.; Penticuff, J.; Eberling, J.; et al. Safety Evaluation of AAV2-GDNF Gene Transfer into the Dopaminergic Nigrostriatal Pathway in Aged and Parkinsonian Rhesus Monkeys. Hum. Gene Ther. 2009, 20, 1627–1640. [Google Scholar] [CrossRef] [PubMed]
  53. Valles, F.; Fiandaca, M.S.; Eberling, J.L.; Starr, P.A.; Larson, P.S.; Christine, C.W.; Forsayeth, J.; Richardson, R.M.; Su, X.; Aminoff, M.J.; et al. Qualitative Imaging of Adeno-Associated Virus Serotype 2-Human Aromatic L-Amino Acid Decarboxylase Gene Therapy in a Phase I Study for the Treatment of Parkinson Disease. Neurosurgery 2010, 67, 1377–1385. [Google Scholar] [CrossRef] [PubMed]
  54. Yun, S.P.; Kim, D.; Kim, S.; Kim, S.; Karuppagounder, S.S.; Kwon, S.-H.; Lee, S.; Kam, T.-I.; Lee, S.; Ham, S.; et al. α-Synuclein Accumulation and GBA Deficiency Due to L444P GBA Mutation Contributes to MPTP-Induced Parkinsonism. Mol. Neurodegener. 2018, 13, 1. [Google Scholar] [CrossRef] [Green Version]
  55. Pajarillo, E.; Johnson, J.; Rizor, A.; Nyarko-Danquah, I.; Adinew, G.; Bornhorst, J.; Stiboller, M.; Schwerdtle, T.; Son, D.-S.; Aschner, M.; et al. Astrocyte-Specific Deletion of the Transcription Factor Yin Yang 1 in Murine Substantia Nigra Mitigates Manganese-Induced Dopaminergic Neurotoxicity. J. Biol. Chem. 2020, 295, 15662–15676. [Google Scholar] [CrossRef]
  56. Bordia, T.; Perez, X.A.; Heiss, J.; Zhang, D.; Quik, M. Optogenetic Activation of Striatal Cholinergic Interneurons Regulates L-Dopa-Induced Dyskinesias. NeuroBiol. Dis. 2016, 91, 47–58. [Google Scholar] [CrossRef] [Green Version]
  57. Chatterjee, D.; Bhatt, M.; Butler, D.; De Genst, E.; Dobson, C.M.; Messer, A.; Kordower, J.H. Proteasome-Targeted Nanobodies Alleviate Pathology and Functional Decline in an α-Synuclein-Based Parkinson’s Disease Model. NPJ Parkinsons Dis. 2018, 4, 25. [Google Scholar] [CrossRef] [Green Version]
  58. Gombash, S.E.; Manfredsson, F.P.; Kemp, C.J.; Kuhn, N.C.; Fleming, S.M.; Egan, A.E.; Grant, L.M.; Ciucci, M.R.; MacKeigan, J.P.; Sortwell, C.E. Morphological and Behavioral Impact of AAV2/5-Mediated Overexpression of Human Wildtype Alpha-Synuclein in the Rat Nigrostriatal System. PLoS ONE 2013, 8, e81426. [Google Scholar] [CrossRef] [Green Version]
  59. Wagner, L.M.; Nathwani, S.M.; Ten Eyck, P.P.; Aldridge, G.M. Local Cortical Overexpression of Human Wild-Type Alpha-Synuclein Leads to Increased Dendritic Spine Density in Mouse. Neurosci. Lett. 2020, 733, 135051. [Google Scholar] [CrossRef]
  60. Xilouri, M.; Brekk, O.R.; Landeck, N.; Pitychoutis, P.M.; Papasilekas, T.; Papadopoulou-Daifoti, Z.; Kirik, D.; Stefanis, L. Boosting Chaperone-Mediated Autophagy in Vivo Mitigates α-Synuclein-Induced Neurodegeneration. Brain 2013, 136, 2130–2146. [Google Scholar] [CrossRef] [Green Version]
  61. Löw, K.; Aebischer, P.; Schneider, B.L. Direct and Retrograde Transduction of Nigral Neurons with AAV6, 8, and 9 and Intraneuronal Persistence of Viral Particles. Hum. Gene Ther. 2013, 24, 613–629. [Google Scholar] [CrossRef] [Green Version]
  62. Kordower, J.H.; Dodiya, H.B.; Kordower, A.M.; Terpstra, B.; Paumier, K.; Madhavan, L.; Sortwell, C.; Steece-Collier, K.; Collier, T.J. Transfer of Host-Derived α Synuclein to Grafted Dopaminergic Neurons in Rat. NeuroBiol. Dis. 2011, 43, 552–557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Decressac, M.; Mattsson, B.; Lundblad, M.; Weikop, P.; Björklund, A. Progressive Neurodegenerative and Behavioural Changes Induced by AAV-Mediated Overexpression of α-Synuclein in Midbrain Dopamine Neurons. NeuroBiol. Dis. 2012, 45, 939–953. [Google Scholar] [CrossRef]
  64. Decressac, M.; Mattsson, B.; Weikop, P.; Lundblad, M.; Jakobsson, J.; Björklund, A. TFEB-Mediated Autophagy Rescues Midbrain Dopamine Neurons from α-Synuclein Toxicity. Proc. Natl. Acad. Sci. USA 2013, 110, E1817–E1826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Oliveras-Salvá, M.; Macchi, F.; Coessens, V.; Deleersnijder, A.; Gérard, M.; Van der Perren, A.; Van den Haute, C.; Baekelandt, V. Alpha-Synuclein-Induced Neurodegeneration Is Exacerbated in PINK1 Knockout Mice. NeuroBiol. Aging 2014, 35, 2625–2636. [Google Scholar] [CrossRef]
  66. Wang, L.; Wang, Z.; Xu, X.; Zhu, R.; Bi, J.; Liu, W.; Feng, X.; Wu, H.; Zhang, H.; Wu, J.; et al. Recombinant AAV8-Mediated Intrastriatal Gene Delivery of CDNF Protects Rats against Methamphetamine Neurotoxicity. Int. J. Med. Sci. 2017, 14, 340–347. [Google Scholar] [CrossRef] [Green Version]
  67. Wang, L.; Wang, Z.; Zhu, R.; Bi, J.; Feng, X.; Liu, W.; Wu, J.; Zhang, H.; Wu, H.; Kong, W.; et al. Therapeutic Efficacy of AAV8-Mediated Intrastriatal Delivery of Human Cerebral Dopamine Neurotrophic Factor in 6-OHDA-Induced Parkinsonian Rat Models with Different Disease Progression. PLoS ONE 2017, 12, e0179476. [Google Scholar] [CrossRef]
  68. McFarland, N.R.; Dimant, H.; Kibuuka, L.; Ebrahimi-Fakhari, D.; Desjardins, C.A.; Danzer, K.M.; Danzer, M.; Fan, Z.; Schwarzschild, M.A.; Hirst, W.; et al. Chronic Treatment with Novel Small Molecule Hsp90 Inhibitors Rescues Striatal Dopamine Levels but Not α-Synuclein-Induced Neuronal Cell Loss. PLoS ONE 2014, 9, e86048. [Google Scholar] [CrossRef] [PubMed]
  69. Wang, Q.-J.; Chen, A.-D.; Chen, H.-C.; Wang, D.-X.; Cai, Y.-T.; Yin, J.; Jing, Y.-H.; Gao, L.-P. Noncanonical Roles of Hα-Syn (A53T) in the Pathogenesis of Parkinson’s Disease: Synaptic Pathology and Neuronal Aging. Neural. Plast. 2020, 2020, 6283754. [Google Scholar] [CrossRef]
  70. Wang, D.; Zhong, L.; Li, M.; Li, J.; Tran, K.; Ren, L.; He, R.; Xie, J.; Moser, R.P.; Fraser, C.; et al. Adeno-Associated Virus Neutralizing Antibodies in Large Animals and Their Impact on Brain Intraparenchymal Gene Transfer. Mol. Ther. Methods Clin. Dev. 2018, 11, 65–72. [Google Scholar] [CrossRef] [Green Version]
  71. Hudry, E.; Andres-Mateos, E.; Lerner, E.P.; Volak, A.; Cohen, O.; Hyman, B.T.; Maguire, C.A.; Vandenberghe, L.H. Efficient Gene Transfer to the Central Nervous System by Single-Stranded Anc80L65. Mol. Ther. Methods Clin. Dev. 2018, 10, 197–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Bourdenx, M.; Dovero, S.; Engeln, M.; Bido, S.; Bastide, M.F.; Dutheil, N.; Vollenweider, I.; Baud, L.; Piron, C.; Grouthier, V.; et al. Lack of Additive Role of Ageing in Nigrostriatal Neurodegeneration Triggered by α-Synuclein Overexpression. Acta Neuropathol. Commun. 2015, 3, 46. [Google Scholar] [CrossRef] [Green Version]
  73. Sun, X.; Yu, X.; Zhang, L.; Zhao, W.; Wang, M.; Zhang, Y.; Li, X.; Gao, R.; Breger, L.S.; Dovero, S.; et al. Comparison of the Expression and Toxicity of AAV2/9 Carrying the Human A53T α-Synuclein Gene in Presence or Absence of WPRE. Heliyon 2021, 7, e06302. [Google Scholar] [CrossRef]
  74. Pabon, M.M.; Jernberg, J.N.; Morganti, J.; Contreras, J.; Hudson, C.E.; Klein, R.L.; Bickford, P.C. A Spirulina-Enhanced Diet Provides Neuroprotection in an α-Synuclein Model of Parkinson’s Disease. PLoS ONE 2012, 7, e45256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Yang, C.; Hao, F.; He, J.; Lu, T.; Klein, R.L.; Zhao, L.-R.; Duan, W.-M. Sequential Adeno-Associated Viral Vector Serotype 9-Green Fluorescent Protein Gene Transfer Causes Massive Inflammation and Intense Immune Response in Rat Striatum. Hum. Gene Ther. 2016, 27, 528–543. [Google Scholar] [CrossRef]
  76. Hao, F.; Yang, C.; Chen, S.-S.; Wang, Y.-Y.; Zhou, W.; Hao, Q.; Lu, T.; Hoffer, B.; Zhao, L.-R.; Duan, W.-M.; et al. Long-Term Protective Effects of AAV9-Mesencephalic Astrocyte-Derived Neurotrophic Factor Gene Transfer in Parkinsonian Rats. Exp. Neurol. 2017, 291, 120–133. [Google Scholar] [CrossRef]
  77. Subbarayan, M.S.; Hudson, C.; Moss, L.D.; Nash, K.R.; Bickford, P.C. T Cell Infiltration and Upregulation of MHCII in Microglia Leads to Accelerated Neuronal Loss in an α-Synuclein Rat Model of Parkinson’s Disease. J. Neuroinflammation 2020, 17, 242. [Google Scholar] [CrossRef] [PubMed]
  78. Yang, C.; Yang, W.-H.; Chen, S.-S.; Ma, B.-F.; Li, B.; Lu, T.; Qu, T.-Y.; Klein, R.L.; Zhao, L.-R.; Duan, W.-M. Pre-Immunization with an Intramuscular Injection of AAV9-Human Erythropoietin Vectors Reduces the Vector-Mediated Transduction Following Re-Administration in Rat Brain. PLoS ONE 2013, 8, e63876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Xue, Y.-Q.; Ma, B.-F.; Zhao, L.-R.; Tatom, J.B.; Li, B.; Jiang, L.-X.; Klein, R.L.; Duan, W.-M. AAV9-Mediated Erythropoietin Gene Delivery into the Brain Protects Nigral Dopaminergic Neurons in a Rat Model of Parkinson’s Disease. Gene Ther. 2010, 17, 83–94. [Google Scholar] [CrossRef]
  80. Cresto, N.; Gaillard, M.-C.; Gardier, C.; Gubinelli, F.; Diguet, E.; Bellet, D.; Legroux, L.; Mitja, J.; Auregan, G.; Guillermier, M.; et al. The C-Terminal Domain of LRRK2 with the G2019S Mutation Is Sufficient to Produce Neurodegeneration of Dopaminergic Neurons in Vivo. NeuroBiol. Dis. 2020, 134, 104614. [Google Scholar] [CrossRef]
  81. Albaugh, D.L.; Smith, Y.; Galvan, A. Comparative Analyses of Transgene Expression Patterns after Intra-striatal Injections of RAAV2-retro in Rats and Rhesus Monkeys: A Light and Electron Microscopic Study. Eur. J. Neurosci. 2020, 52, 4824–4839. [Google Scholar] [CrossRef]
  82. Tordo, J.; O’Leary, C.; Antunes, A.S.L.M.; Palomar, N.; Aldrin-Kirk, P.; Basche, M.; Bennett, A.; D’Souza, Z.; Gleitz, H.; Godwin, A.; et al. A Novel Adeno-Associated Virus Capsid with Enhanced Neurotropism Corrects a Lysosomal Transmembrane Enzyme Deficiency. Brain 2018, 141, 2014–2031. [Google Scholar] [CrossRef] [Green Version]
  83. Naidoo, J.; Stanek, L.M.; Ohno, K.; Trewman, S.; Samaranch, L.; Hadaczek, P.; O’Riordan, C.; Sullivan, J.; San Sebastian, W.; Bringas, J.R.; et al. Extensive Transduction and Enhanced Spread of a Modified AAV2 Capsid in the Non-Human Primate CNS. Mol. Ther. 2018, 26, 2418–2430. [Google Scholar] [CrossRef] [Green Version]
  84. Davidsson, M.; Wang, G.; Aldrin-Kirk, P.; Cardoso, T.; Nolbrant, S.; Hartnor, M.; Mudannayake, J.; Parmar, M.; Björklund, T. A Systematic Capsid Evolution Approach Performed in Vivo for the Design of AAV Vectors with Tailored Properties and Tropism. Proc. Natl. Acad. Sci. USA 2019. [Google Scholar] [CrossRef] [Green Version]
  85. Hammond, S.L.; Leek, A.N.; Richman, E.H.; Tjalkens, R.B. Cellular Selectivity of AAV Serotypes for Gene Delivery in Neurons and Astrocytes by Neonatal Intracerebroventricular Injection. PLoS ONE 2017, 12, e0188830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Gray, S.J.; Nagabhushan Kalburgi, S.; McCown, T.J.; Jude Samulski, R. Global CNS Gene Delivery and Evasion of Anti-AAV-Neutralizing Antibodies by Intrathecal AAV Administration in Non-Human Primates. Gene Ther. 2013, 20, 450–459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Vulchanova, L.; Schuster, D.J.; Belur, L.R.; Riedl, M.S.; Podetz-Pedersen, K.M.; Kitto, K.F.; Wilcox, G.L.; McIvor, R.S.; Fairbanks, C.A. Differential Adeno-Associated Virus Mediated Gene Transfer to Sensory Neurons Following Intrathecal Delivery by Direct Lumbar Puncture. Mol. Pain 2010, 6, 31. [Google Scholar] [CrossRef] [Green Version]
  88. Towne, C.; Pertin, M.; Beggah, A.T.; Aebischer, P.; Decosterd, I. Recombinant Adeno-Associated Virus Serotype 6 (RAAV2/6)-Mediated Gene Transfer to Nociceptive Neurons through Different Routes of Delivery. Mol. Pain 2009, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Samaranch, L.; Salegio, E.A.; San Sebastian, W.; Kells, A.P.; Bringas, J.R.; Forsayeth, J.; Bankiewicz, K.S. Strong Cortical and Spinal Cord Transduction after AAV7 and AAV9 Delivery into the Cerebrospinal Fluid of Nonhuman Primates. Hum. Gene Ther. 2013, 24, 526–532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Amado, D.A.; Rieders, J.M.; Diatta, F.; Hernandez-Con, P.; Singer, A.; Mak, J.T.; Zhang, J.; Lancaster, E.; Davidson, B.L.; Chen-Plotkin, A.S. AAV-Mediated Progranulin Delivery to a Mouse Model of Progranulin Deficiency Causes T Cell-Mediated Toxicity. Mol. Ther. 2019, 27, 465–478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Bey, K.; Ciron, C.; Dubreil, L.; Deniaud, J.; Ledevin, M.; Cristini, J.; Blouin, V.; Aubourg, P.; Colle, M.-A. Efficient CNS Targeting in Adult Mice by Intrathecal Infusion of Single-Stranded AAV9-GFP for Gene Therapy of Neurological Disorders. Gene Ther. 2017, 24, 325–332. [Google Scholar] [CrossRef] [PubMed]
  92. Chatterjee, D.; Marmion, D.J.; McBride, J.L.; Manfredsson, F.P.; Butler, D.; Messer, A.; Kordower, J.H. Enhanced CNS Transduction from AAV.PHP.EB Infusion into the Cisterna Magna of Older Adult Rats Compared to AAV9. Gene Ther. 2021. [Google Scholar] [CrossRef]
  93. Ohno, K.; Samaranch, L.; Hadaczek, P.; Bringas, J.R.; Allen, P.C.; Sudhakar, V.; Stockinger, D.E.; Snieckus, C.; Campagna, M.V.; San Sebastian, W.; et al. Kinetics and MR-Based Monitoring of AAV9 Vector Delivery into Cerebrospinal Fluid of Nonhuman Primates. Mol. Ther. Methods Clin. Dev. 2019, 13, 47–54. [Google Scholar] [CrossRef] [Green Version]
  94. Samaranch, L.; Salegio, E.A.; San Sebastian, W.; Kells, A.P.; Foust, K.D.; Bringas, J.R.; Lamarre, C.; Forsayeth, J.; Kaspar, B.K.; Bankiewicz, K.S. Adeno-Associated Virus Serotype 9 Transduction in the Central Nervous System of Nonhuman Primates. Hum. Gene Ther. 2012, 23, 382–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Hinderer, C.; Bell, P.; Vite, C.H.; Louboutin, J.-P.; Grant, R.; Bote, E.; Yu, H.; Pukenas, B.; Hurst, R.; Wilson, J.M. Widespread Gene Transfer in the Central Nervous System of Cynomolgus Macaques Following Delivery of AAV9 into the Cisterna Magna. Mol. Ther. Methods Clin. Dev. 2014, 1, 14051. [Google Scholar] [CrossRef] [PubMed]
  96. Bey, K.; Deniaud, J.; Dubreil, L.; Joussemet, B.; Cristini, J.; Ciron, C.; Hordeaux, J.; Le Boulc’h, M.; Marche, K.; Maquigneau, M.; et al. Intra-CSF AAV9 and AAVrh10 Administration in Nonhuman Primates: Promising Routes and Vectors for Which Neurological Diseases? Mol. Ther. Methods Clin. Dev. 2020, 17, 771–784. [Google Scholar] [CrossRef]
  97. Hordeaux, J.; Dubreil, L.; Deniaud, J.; Iacobelli, F.; Moreau, S.; Ledevin, M.; Le Guiner, C.; Blouin, V.; Le Duff, J.; Mendes-Madeira, A.; et al. Efficient Central Nervous System AAVrh10-Mediated Intrathecal Gene Transfer in Adult and Neonate Rats. Gene Ther. 2015, 22, 316–324. [Google Scholar] [CrossRef]
  98. Liguore, W.A.; Domire, J.S.; Button, D.; Wang, Y.; Dufour, B.D.; Srinivasan, S.; McBride, J.L. AAV-PHP.B Administration Results in a Differential Pattern of CNS Biodistribution in Non-Human Primates Compared with Mice. Mol. Ther. 2019, 27, 2018–2037. [Google Scholar] [CrossRef]
  99. Galvan, A.; Petkau, T.L.; Hill, A.M.; Korecki, A.J.; Lu, G.; Choi, D.; Rahman, K.; Simpson, E.; Leavitt, B.R.; Smith, Y. Intracerebroventricular Administration of AAV9-PHP.B SYN1-EmGFP Induces Widespread Transgene Expression in the Mouse and Monkey CNS. Hum. Gene Ther. 2021. [Google Scholar] [CrossRef]
  100. Iwata, N.; Sekiguchi, M.; Hattori, Y.; Takahashi, A.; Asai, M.; Ji, B.; Higuchi, M.; Staufenbiel, M.; Muramatsu, S.; Saido, T.C. Global Brain Delivery of Neprilysin Gene by Intravascular Administration of AAV Vector in Mice. Sci. Rep. 2013, 3, 1472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Rincon, M.Y.; de Vin, F.; Duqué, S.I.; Fripont, S.; Castaldo, S.A.; Bouhuijzen-Wenger, J.; Holt, M.G. Widespread Transduction of Astrocytes and Neurons in the Mouse Central Nervous System after Systemic Delivery of a Self-Complementary AAV-PHP.B Vector. Gene Ther. 2018, 25, 83–92. [Google Scholar] [CrossRef] [PubMed]
  102. Choudhury, S.R.; Harris, A.F.; Cabral, D.J.; Keeler, A.M.; Sapp, E.; Ferreira, J.S.; Gray-Edwards, H.L.; Johnson, J.A.; Johnson, A.K.; Su, Q.; et al. Widespread Central Nervous System Gene Transfer and Silencing After Systemic Delivery of Novel AAV-AS Vector. Mol. Ther. 2016, 24, 726–735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Lau, C.-H.; Ho, J.W.-T.; Lo, P.K.; Tin, C. Targeted Transgene Activation in the Brain Tissue by Systemic Delivery of Engineered AAV1 Expressing CRISPRa. Mol. Ther. Nucleic Acids 2019, 16, 637–649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Challis, R.C.; Ravindra Kumar, S.; Chan, K.Y.; Challis, C.; Beadle, K.; Jang, M.J.; Kim, H.M.; Rajendran, P.S.; Tompkins, J.D.; Shivkumar, K.; et al. Systemic AAV Vectors for Widespread and Targeted Gene Delivery in Rodents. Nat. Protoc. 2019, 14, 379–414. [Google Scholar] [CrossRef]
  105. Morabito, G.; Giannelli, S.G.; Ordazzo, G.; Bido, S.; Castoldi, V.; Indrigo, M.; Cabassi, T.; Cattaneo, S.; Luoni, M.; Cancellieri, C.; et al. AAV-PHP.B-Mediated Global-Scale Expression in the Mouse Nervous System Enables GBA1 Gene Therapy for Wide Protection from Synucleinopathy. Mol. Ther. 2017, 25, 2727–2742. [Google Scholar] [CrossRef] [Green Version]
  106. Miyanohara, A.; Kamizato, K.; Juhas, S.; Juhasova, J.; Navarro, M.; Marsala, S.; Lukacova, N.; Hruska-Plochan, M.; Curtis, E.; Gabel, B.; et al. Potent Spinal Parenchymal AAV9-Mediated Gene Delivery by Subpial Injection in Adult Rats and Pigs. Mol. Ther. Methods Clin. Dev. 2016, 3, 16046. [Google Scholar] [CrossRef] [Green Version]
  107. Bravo-Hernandez, M.; Tadokoro, T.; Navarro, M.R.; Platoshyn, O.; Kobayashi, Y.; Marsala, S.; Miyanohara, A.; Juhas, S.; Juhasova, J.; Skalnikova, H.; et al. Spinal Subpial Delivery of AAV9 Enables Widespread Gene Silencing and Blocks Motoneuron Degeneration in ALS. Nat. Med. 2020, 26, 118–130. [Google Scholar] [CrossRef] [PubMed]
  108. Williams, C.L.; Uytingco, C.R.; Green, W.W.; McIntyre, J.C.; Ukhanov, K.; Zimmerman, A.D.; Shively, D.T.; Zhang, L.; Nishimura, D.Y.; Sheffield, V.C.; et al. Gene Therapeutic Reversal of Peripheral Olfactory Impairment in Bardet-Biedl Syndrome. Mol. Ther. 2017, 25, 904–916. [Google Scholar] [CrossRef] [Green Version]
  109. Hocquemiller, M.; Giersch, L.; Audrain, M.; Parker, S.; Cartier, N. Adeno-Associated Virus-Based Gene Therapy for CNS Diseases. Hum. Gene Ther. 2016, 27, 478–496. [Google Scholar] [CrossRef] [Green Version]
  110. Choudhury, S.R.; Hudry, E.; Maguire, C.A.; Sena-Esteves, M.; Breakefield, X.O.; Grandi, P. Viral Vectors for Therapy of Neurologic Diseases. Neuropharmacology 2017, 120, 63–80. [Google Scholar] [CrossRef] [Green Version]
  111. Lonser, R.R.; Akhter, A.S.; Zabek, M.; Elder, J.B.; Bankiewicz, K.S. Direct Convective Delivery of Adeno-Associated Virus Gene Therapy for Treatment of Neurological Disorders. J. Neurosurg 2020, 1–13. [Google Scholar] [CrossRef]
  112. Lundstrom, K. Viral Vectors in Gene Therapy. Diseases 2018, 6, 42. [Google Scholar] [CrossRef] [Green Version]
  113. Samaranch, L.; Blits, B.; San Sebastian, W.; Hadaczek, P.; Bringas, J.; Sudhakar, V.; Macayan, M.; Pivirotto, P.J.; Petry, H.; Bankiewicz, K.S. MR-Guided Parenchymal Delivery of Adeno-Associated Viral Vector Serotype 5 in Non-Human Primate Brain. Gene Ther. 2017, 24, 253–261. [Google Scholar] [CrossRef]
  114. Christine, C.W.; Bankiewicz, K.S.; Van Laar, A.D.; Richardson, R.M.; Ravina, B.; Kells, A.P.; Boot, B.; Martin, A.J.; Nutt, J.; Thompson, M.E.; et al. Magnetic Resonance Imaging-Guided Phase 1 Trial of Putaminal AADC Gene Therapy for Parkinson’s Disease. Ann. Neurol. 2019, 85, 704–714. [Google Scholar] [CrossRef] [Green Version]
  115. Muramatsu, S.; Fujimoto, K.; Kato, S.; Mizukami, H.; Asari, S.; Ikeguchi, K.; Kawakami, T.; Urabe, M.; Kume, A.; Sato, T.; et al. A Phase I Study of Aromatic L-Amino Acid Decarboxylase Gene Therapy for Parkinson’s Disease. Mol. Ther. 2010, 18, 1731–1735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. McFarthing, K.; Prakash, N.; Simuni, T. Clinical trial highlights: 1. Gene therapy for parkinson’s, 2. Phase 3 study in focus-intec pharma’s accordion pill, 3. Clinical trials resources. J. Parkinsons Dis. 2019, 9, 251–264. [Google Scholar] [CrossRef] [Green Version]
  117. Nutt, J.G.; Curtze, C.; Hiller, A.; Anderson, S.; Larson, P.S.; Van Laar, A.D.; Richardson, R.M.; Thompson, M.E.; Sedkov, A.; Leinonen, M.; et al. Aromatic L-Amino Acid Decarboxylase Gene Therapy Enhances Levodopa Response in Parkinson’s Disease. Mov. Disord. 2020, 35, 851–858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Airaksinen, M.S.; Saarma, M. The GDNF Family: Signalling, Biological Functions and Therapeutic Value. Nat. Rev. Neurosci. 2002, 3, 383–394. [Google Scholar] [CrossRef]
  119. Bäckman, C.M.; Shan, L.; Zhang, Y.J.; Hoffer, B.J.; Leonard, S.; Troncoso, J.C.; Vonsatel, P.; Tomac, A.C. Gene Expression Patterns for GDNF and Its Receptors in the Human Putamen Affected by Parkinson’s Disease: A Real-Time PCR Study. Mol. Cell Endocrinol. 2006, 252, 160–166. [Google Scholar] [CrossRef]
  120. Björklund, A.; Kirik, D.; Rosenblad, C.; Georgievska, B.; Lundberg, C.; Mandel, R.J. Towards a Neuroprotective Gene Therapy for Parkinson’s Disease: Use of Adenovirus, AAV and Lentivirus Vectors for Gene Transfer of GDNF to the Nigrostriatal System in the Rat Parkinson Model. Brain Res. 2000, 886, 82–98. [Google Scholar] [CrossRef]
  121. LeWitt, P.A.; Rezai, A.R.; Leehey, M.A.; Ojemann, S.G.; Flaherty, A.W.; Eskandar, E.N.; Kostyk, S.K.; Thomas, K.; Sarkar, A.; Siddiqui, M.S.; et al. AAV2-GAD Gene Therapy for Advanced Parkinson’s Disease: A Double-Blind, Sham-Surgery Controlled, Randomised Trial. Lancet Neurol. 2011, 10, 309–319. [Google Scholar] [CrossRef]
  122. Kaplitt, M.G.; Feigin, A.; Tang, C.; Fitzsimons, H.L.; Mattis, P.; Lawlor, P.A.; Bland, R.J.; Young, D.; Strybing, K.; Eidelberg, D.; et al. Safety and Tolerability of Gene Therapy with an Adeno-Associated Virus (AAV) Borne GAD Gene for Parkinson’s Disease: An Open Label, Phase I Trial. Lancet 2007, 369, 2097–2105. [Google Scholar] [CrossRef]
  123. Feigin, A.; Kaplitt, M.G.; Tang, C.; Lin, T.; Mattis, P.; Dhawan, V.; During, M.J.; Eidelberg, D. Modulation of Metabolic Brain Networks after Subthalamic Gene Therapy for Parkinson’s Disease. Proc. Natl. Acad. Sci. USA 2007, 104, 19559–19564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Niethammer, M.; Tang, C.C.; LeWitt, P.A.; Rezai, A.R.; Leehey, M.A.; Ojemann, S.G.; Flaherty, A.W.; Eskandar, E.N.; Kostyk, S.K.; Sarkar, A.; et al. Long-Term Follow-up of a Randomized AAV2-GAD Gene Therapy Trial for Parkinson’s Disease. JCI Insight 2017, 2, e90133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Luo, J.; Kaplitt, M.G.; Fitzsimons, H.L.; Zuzga, D.S.; Liu, Y.; Oshinsky, M.L.; During, M.J. Subthalamic GAD Gene Therapy in a Parkinson’s Disease Rat Model. Science 2002, 298, 425–429. [Google Scholar] [CrossRef]
  126. Marks, W.J.; Ostrem, J.L.; Verhagen, L.; Starr, P.A.; Larson, P.S.; Bakay, R.A.; Taylor, R.; Cahn-Weiner, D.A.; Stoessl, A.J.; Olanow, C.W.; et al. Safety and Tolerability of Intraputaminal Delivery of CERE-120 (Adeno-Associated Virus Serotype 2-Neurturin) to Patients with Idiopathic Parkinson’s Disease: An Open-Label, Phase I Trial. Lancet Neurol. 2008, 7, 400–408. [Google Scholar] [CrossRef]
  127. Bartus, R.T.; Baumann, T.L.; Siffert, J.; Herzog, C.D.; Alterman, R.; Boulis, N.; Turner, D.A.; Stacy, M.; Lang, A.E.; Lozano, A.M.; et al. Safety/Feasibility of Targeting the Substantia Nigra with AAV2-Neurturin in Parkinson Patients. Neurology 2013, 80, 1698–1701. [Google Scholar] [CrossRef] [Green Version]
  128. Green, F.; Samaranch, L.; Zhang, H.S.; Manning-Bog, A.; Meyer, K.; Forsayeth, J.; Bankiewicz, K.S. Axonal Transport of AAV9 in Nonhuman Primate Brain. Gene Ther. 2016, 23, 520–526. [Google Scholar] [CrossRef] [Green Version]
  129. Tervo, D.G.R.; Hwang, B.-Y.; Viswanathan, S.; Gaj, T.; Lavzin, M.; Ritola, K.D.; 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. [Google Scholar] [CrossRef] [Green Version]
  130. Cohen-Pfeffer, J.L.; Gururangan, S.; Lester, T.; Lim, D.A.; Shaywitz, A.J.; Westphal, M.; Slavc, I. Intracerebroventricular Delivery as a Safe, Long-Term Route of Drug Administration. Pediatr. Neurol. 2017, 67, 23–35. [Google Scholar] [CrossRef] [Green Version]
  131. Hinderer, C.; Bell, P.; Katz, N.; Vite, C.H.; Louboutin, J.-P.; Bote, E.; Yu, H.; Zhu, Y.; Casal, M.L.; Bagel, J.; et al. Evaluation of Intrathecal Routes of Administration for Adeno-Associated Viral Vectors in Large Animals. Hum. Gene Ther. 2018, 29, 15–24. [Google Scholar] [CrossRef]
  132. Yang, B.; Li, S.; Wang, H.; Guo, Y.; Gessler, D.J.; Cao, C.; Su, Q.; Kramer, J.; Zhong, L.; Ahmed, S.S.; et al. Global CNS Transduction of Adult Mice by Intravenously Delivered RAAVrh.8 and RAAVrh.10 and Nonhuman Primates by RAAVrh.10. Mol. Ther. 2014, 22, 1299–1309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Manfredsson, F.P.; Rising, A.C.; Mandel, R.J. AAV9: A Potential Blood-Brain Barrier Buster. Mol. Ther. 2009, 17, 403–405. [Google Scholar] [CrossRef] [PubMed]
  134. Forsayeth, J.R.; Bankiewicz, K.S. AAV9: Over the Fence and into the Woods. Mol. Ther. 2011, 19, 1006–1007. [Google Scholar] [CrossRef] [PubMed]
  135. Deverman, B.E.; Pravdo, P.L.; Simpson, B.P.; Kumar, S.R.; Chan, K.Y.; Banerjee, A.; Wu, W.-L.; Yang, B.; Huber, N.; Pasca, S.P.; et al. Cre-Dependent Selection Yields AAV Variants for Widespread Gene Transfer to the Adult Brain. Nat. Biotechnol. 2016, 34, 204–209. [Google Scholar] [CrossRef]
  136. Hordeaux, J.; Wang, Q.; Katz, N.; Buza, E.L.; Bell, P.; Wilson, J.M. The Neurotropic Properties of AAV-PHP.B Are Limited to C57BL/6J. Mice. Mol. Ther. 2018, 26, 664–668. [Google Scholar] [CrossRef] [PubMed]
  137. Hudry, E.; Vandenberghe, L.H. Therapeutic AAV Gene Transfer to the Nervous System: A Clinical Reality. Neuron 2019, 101, 839–862. [Google Scholar] [CrossRef] [Green Version]
  138. Jackson, K.L.; Dayton, R.D.; Klein, R.L. AAV9 Supports Wide-Scale Transduction of the CNS and TDP-43 Disease Modeling in Adult Rats. Mol. Ther. Methods Clin. Dev. 2015, 2, 15036. [Google Scholar] [CrossRef]
  139. Dayton, R.D.; Grames, M.S.; Klein, R.L. More Expansive Gene Transfer to the Rat CNS: AAV PHP.EB Vector Dose-Response and Comparison to AAV PHP.B. Gene Ther. 2018, 25, 392–400. [Google Scholar] [CrossRef]
  140. Huang, Q.; Chan, K.Y.; Tobey, I.G.; Chan, Y.A.; Poterba, T.; Boutros, C.L.; Balazs, A.B.; Daneman, R.; Bloom, J.M.; Seed, C.; et al. Delivering Genes across the Blood-Brain Barrier: LY6A, a Novel Cellular Receptor for AAV-PHP.B Capsids. PLoS ONE 2019, 14, e0225206. [Google Scholar] [CrossRef] [Green Version]
  141. Batista, A.R.; King, O.D.; Reardon, C.P.; Davis, C.; Shankaracharya; Philip, V.; Gray-Edwards, H.; Aronin, N.; Lutz, C.; Landers, J.; et al. Ly6a Differential Expression in Blood-Brain Barrier Is Responsible for Strain Specific Central Nervous System Transduction Profile of AAV-PHP.B. Hum. Gene Ther. 2020, 31, 90–102. [Google Scholar] [CrossRef] [Green Version]
  142. Spindler, K.R.; Welton, A.R.; Lim, E.S.; Duvvuru, S.; Althaus, I.W.; Imperiale, J.E.; Daoud, A.I.; Chesler, E.J. The Major Locus for Mouse Adenovirus Susceptibility Maps to Genes of the Hematopoietic Cell Surface-Expressed LY6 Family. J. Immunol. 2010, 184, 3055–3062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Matsuzaki, Y.; Konno, A.; Mochizuki, R.; Shinohara, Y.; Nitta, K.; Okada, Y.; Hirai, H. Intravenous Administration of the Adeno-Associated Virus-PHP.B Capsid Fails to Upregulate Transduction Efficiency in the Marmoset Brain. Neurosci. Lett. 2018, 665, 182–188. [Google Scholar] [CrossRef] [PubMed]
  144. Uytingco, C.R.; Martens, J.R. Intranasal Delivery of Adenoviral and AAV Vectors for Transduction of the Mammalian Peripheral Olfactory System. Methods Mol. Biol. 2019, 1950, 283–297. [Google Scholar] [CrossRef]
  145. Imbert, C.; Bezard, E.; Guitraud, S.; Boraud, T.; Gross, C.E. Comparison of Eight Clinical Rating Scales Used for the Assessment of MPTP-Induced Parkinsonism in the Macaque Monkey. J. Neurosci. Methods 2000, 96, 71–76. [Google Scholar] [CrossRef]
  146. Eslamboli, A.; Romero-Ramos, M.; Burger, C.; Bjorklund, T.; Muzyczka, N.; Mandel, R.J.; Baker, H.; Ridley, R.M.; Kirik, D. Long-Term Consequences of Human Alpha-Synuclein Overexpression in the Primate Ventral Midbrain. Brain 2007, 130, 799–815. [Google Scholar] [CrossRef]
  147. Lanciego, J.L.; Luquin, N.; Obeso, J.A. Functional Neuroanatomy of the Basal Ganglia. Cold Spring Harb. Perspect Med. 2012, 2, a009621. [Google Scholar] [CrossRef] [PubMed]
  148. Marmion, D.J.; Kordower, J.H. α-Synuclein Nonhuman Primate Models of Parkinson’s Disease. J. Neural. Transm. 2018, 125, 385–400. [Google Scholar] [CrossRef]
  149. Spillantini, M.G.; Crowther, R.A.; Jakes, R.; Hasegawa, M.; Goedert, M. Alpha-Synuclein in Filamentous Inclusions of Lewy Bodies from Parkinson’s Disease and Dementia with Lewy Bodies. Proc. Natl. Acad. Sci. USA 1998, 95, 6469–6473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Visanji, N.P.; Brotchie, J.M.; Kalia, L.V.; Koprich, J.B.; Tandon, A.; Watts, J.C.; Lang, A.E. α-Synuclein-Based Animal Models of Parkinson’s Disease: Challenges and Opportunities in a New Era. Trends Neurosci. 2016, 39, 750–762. [Google Scholar] [CrossRef]
  151. Kirik, D.; Annett, L.E.; Burger, C.; Muzyczka, N.; Mandel, R.J.; Björklund, A. Nigrostriatal Alpha-Synucleinopathy Induced by Viral Vector-Mediated Overexpression of Human Alpha-Synuclein: A New Primate Model of Parkinson’s Disease. Proc. Natl. Acad. Sci. USA 2003, 100, 2884–2889. [Google Scholar] [CrossRef] [Green Version]
  152. Lo Bianco, C.; Ridet, J.-L.; Schneider, B.L.; Deglon, N.; Aebischer, P. Alpha -Synucleinopathy and Selective Dopaminergic Neuron Loss in a Rat Lentiviral-Based Model of Parkinson’s Disease. Proc. Natl. Acad. Sci. USA 2002, 99, 10813–10818. [Google Scholar] [CrossRef] [Green Version]
  153. Kirik, D.; Rosenblad, C.; Burger, C.; Lundberg, C.; Johansen, T.E.; Muzyczka, N.; Mandel, R.J.; Björklund, A. Parkinson-like Neurodegeneration Induced by Targeted Overexpression of Alpha-Synuclein in the Nigrostriatal System. J. Neurosci. 2002, 22, 2780–2791. [Google Scholar] [CrossRef]
  154. Koprich, J.B.; Johnston, T.H.; Reyes, M.G.; Sun, X.; Brotchie, J.M. Expression of Human A53T Alpha-Synuclein in the Rat Substantia Nigra Using a Novel AAV1/2 Vector Produces a Rapidly Evolving Pathology with Protein Aggregation, Dystrophic Neurite Architecture and Nigrostriatal Degeneration with Potential to Model the Pathology of Parkinson’s Disease. Mol. Neurodegener. 2010, 5, 43. [Google Scholar] [CrossRef] [Green Version]
  155. Koprich, J.B.; Johnston, T.H.; Huot, P.; Reyes, M.G.; Espinosa, M.; Brotchie, J.M. Progressive Neurodegeneration or Endogenous Compensation in an Animal Model of Parkinson’s Disease Produced by Decreasing Doses of Alpha-Synuclein. PLoS ONE 2011, 6, e17698. [Google Scholar] [CrossRef]
  156. Koprich, J.B.; Johnston, T.H.; Reyes, G.; Omana, V.; Brotchie, J.M. Towards a Non-Human Primate Model of Alpha-Synucleinopathy for Development of Therapeutics for Parkinson’s Disease: Optimization of AAV1/2 Delivery Parameters to Drive Sustained Expression of Alpha Synuclein and Dopaminergic Degeneration in Macaque. PLoS ONE 2016, 11, e0167235. [Google Scholar] [CrossRef]
  157. Sucunza, D.; Rico, A.J.; Roda, E.; Collantes, M.; González-Aseguinolaza, G.; Rodríguez-Pérez, A.I.; Peñuelas, I.; Vázquez, A.; Labandeira-García, J.L.; Broccoli, V.; et al. Glucocerebrosidase Gene Therapy Induces Alpha-Synuclein Clearance and Neuroprotection of Midbrain Dopaminergic Neurons in Mice and Macaques. Int. J. Mol. Sci. 2021, 22, 4825. [Google Scholar] [CrossRef]
  158. Yang, W.; Wang, G.; Wang, C.-E.; Guo, X.; Yin, P.; Gao, J.; Tu, Z.; Wang, Z.; Wu, J.; Hu, X.; et al. Mutant Alpha-Synuclein Causes Age-Dependent Neuropathology in Monkey Brain. J. Neurosci. 2015, 35, 8345–8358. [Google Scholar] [CrossRef] [Green Version]
  159. Braak, E.; Sandmann-Keil, D.; Rüb, U.; Gai, W.P.; de Vos, R.A.; Steur, E.N.; Arai, K.; Braak, H. Alpha-Synuclein Immunopositive Parkinson’s Disease-Related Inclusion Bodies in Lower Brain Stem Nuclei. Acta Neuropathol. 2001, 101, 195–201. [Google Scholar] [CrossRef] [PubMed]
  160. Carballo-Carbajal, I.; Laguna, A.; Romero-Giménez, J.; Cuadros, T.; Bové, J.; Martinez-Vicente, M.; Parent, A.; Gonzalez-Sepulveda, M.; Peñuelas, N.; Torra, A.; et al. Brain Tyrosinase Overexpression Implicates Age-Dependent Neuromelanin Production in Parkinson’s Disease Pathogenesis. Nat. Commun. 2019, 10, 973. [Google Scholar] [CrossRef] [Green Version]
  161. Zimprich, A.; Biskup, S.; Leitner, P.; Lichtner, P.; Farrer, M.; Lincoln, S.; Kachergus, J.; Hulihan, M.; Uitti, R.J.; Calne, D.B.; et al. Mutations in LRRK2 Cause Autosomal-Dominant Parkinsonism with Pleomorphic Pathology. Neuron 2004, 44, 601–607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Dusonchet, J.; Kochubey, O.; Stafa, K.; Young, S.M.; Zufferey, R.; Moore, D.J.; Schneider, B.L.; Aebischer, P. A Rat Model of Progressive Nigral Neurodegeneration Induced by the Parkinson’s Disease-Associated G2019S Mutation in LRRK2. J. Neurosci. 2011, 31, 907–912. [Google Scholar] [CrossRef] [Green Version]
  163. Lee, B.D.; Shin, J.-H.; VanKampen, J.; Petrucelli, L.; West, A.B.; Ko, H.S.; Lee, Y.-I.; Maguire-Zeiss, K.A.; Bowers, W.J.; Federoff, H.J.; et al. Inhibitors of Leucine-Rich Repeat Kinase-2 Protect against Models of Parkinson’s Disease. Nat. Med. 2010, 16, 998–1000. [Google Scholar] [CrossRef] [Green Version]
  164. Haque, M.E.; Mount, M.P.; Safarpour, F.; Abdel-Messih, E.; Callaghan, S.; Mazerolle, C.; Kitada, T.; Slack, R.S.; Wallace, V.; Shen, J.; et al. Inactivation of Pink1 Gene in Vivo Sensitizes Dopamine-Producing Neurons to 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine (MPTP) and Can Be Rescued by Autosomal Recessive Parkinson Disease Genes, Parkin or DJ-1. J. Biol. Chem. 2012, 287, 23162–23170. [Google Scholar] [CrossRef] [Green Version]
  165. Wagner, J.A.; Messner, A.H.; Moran, M.L.; Daifuku, R.; Kouyama, K.; Desch, J.K.; Manley, S.; Norbash, A.M.; Conrad, C.K.; Friborg, S.; et al. Safety and Biological Efficacy of an Adeno-Associated Virus Vector-Cystic Fibrosis Transmembrane Regulator (AAV-CFTR) in the Cystic Fibrosis Maxillary Sinus. Laryngoscope 1999, 109, 266–274. [Google Scholar] [CrossRef]
  166. Mandel, T.E.; Koulmanda, M.; Cozzi, E.; Waterworth, P.; Tolan, M.; Langford, G.; White, D.J. Transplantation of Normal and DAF-Transgenic Fetal Pig Pancreas into Cynomolgus Monkeys. Transplant. Proc. 1997, 29, 940. [Google Scholar] [CrossRef]
  167. Bartus, R.T.; Weinberg, M.S.; Samulski, R.J. Parkinson’s Disease Gene Therapy: Success by Design Meets Failure by Efficacy. Mol. Ther. 2014, 22, 487–497. [Google Scholar] [CrossRef] [Green Version]
  168. Bankiewicz, K.S.; Eberling, J.L.; Kohutnicka, M.; Jagust, W.; Pivirotto, P.; Bringas, J.; Cunningham, J.; Budinger, T.F.; Harvey-White, J. Convection-Enhanced Delivery of AAV Vector in Parkinsonian Monkeys; in Vivo Detection of Gene Expression and Restoration of Dopaminergic Function Using pro-Drug Approach. Exp. Neurol. 2000, 164, 2–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Christine, C.W.; Starr, P.A.; Larson, P.S.; Eberling, J.L.; Jagust, W.J.; Hawkins, R.A.; VanBrocklin, H.F.; Wright, J.F.; Bankiewicz, K.S.; Aminoff, M.J. Safety and Tolerability of Putaminal AADC Gene Therapy for Parkinson Disease. Neurology 2009, 73, 1662–1669. [Google Scholar] [CrossRef] [Green Version]
  170. Heiss, J.D.; Lungu, C.; Hammoud, D.A.; Herscovitch, P.; Ehrlich, D.J.; Argersinger, D.P.; Sinharay, S.; Scott, G.; Wu, T.; Federoff, H.J.; et al. Trial of Magnetic Resonance-Guided Putaminal Gene Therapy for Advanced Parkinson’s Disease. Mov. Disord. 2019, 34, 1073–1078. [Google Scholar] [CrossRef]
  171. Warren Olanow, C.; Bartus, R.T.; Baumann, T.L.; Factor, S.; Boulis, N.; Stacy, M.; Turner, D.A.; Marks, W.; Larson, P.; Starr, P.A.; et al. Gene Delivery of Neurturin to Putamen and Substantia Nigra in Parkinson Disease: A Double-Blind, Randomized, Controlled Trial. Ann. Neurol. 2015, 78, 248–257. [Google Scholar] [CrossRef]
  172. Erlander, M.G.; Tillakaratne, N.J.; Feldblum, S.; Patel, N.; Tobin, A.J. Two Genes Encode Distinct Glutamate Decarboxylases. Neuron 1991, 7, 91–100. [Google Scholar] [CrossRef]
  173. Sidransky, E.; Nalls, M.A.; Aasly, J.O.; Aharon-Peretz, J.; Annesi, G.; Barbosa, E.R.; Bar-Shira, A.; Berg, D.; Bras, J.; Brice, A.; et al. Multicenter Analysis of Glucocerebrosidase Mutations in Parkinson’s Disease. N. Engl. J. Med. 2009, 361, 1651–1661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Rocha, E.M.; Smith, G.A.; Park, E.; Cao, H.; Brown, E.; Hayes, M.A.; Beagan, J.; McLean, J.R.; Izen, S.C.; Perez-Torres, E.; et al. Glucocerebrosidase Gene Therapy Prevents α-Synucleinopathy of Midbrain Dopamine Neurons. NeuroBiol. Dis. 2015, 82, 495–503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Most commonly used AAV delivery routes for PD. Although the intraparenchymal approach is by far the choice most often used, AAV deliveries can also be implemented through intra-CSF (intraventricular, intracisternal, and intrathecal), intravenous, and subpial routes.
Figure 1. Most commonly used AAV delivery routes for PD. Although the intraparenchymal approach is by far the choice most often used, AAV deliveries can also be implemented through intra-CSF (intraventricular, intracisternal, and intrathecal), intravenous, and subpial routes.
Ijms 22 06389 g001
Figure 2. Schematic representation of CSF circulation. CSF is firstly secreted by choroid plexuses in each lateral ventricle (1) flowing through the interventricular Monro’s foramen into the third ventricle, where choroid plexuses (3) also add more CSF. From the third ventricle, the CSF flows through the cerebral aqueduct (4) down to the fourth ventricle, where more CSF is also added (5). Next, CSF leaves the ventricular system through two lateral apertures and one median aperture, known as foramina of Luschka and Magendie, respectively (6). CSF fills the subarachnoid space covering the external surfaces of the brain and spinal cord (7). At arachnoid villi, CSF is reabsorbed into peripheral venous blood through dural venous sinuses (8).
Figure 2. Schematic representation of CSF circulation. CSF is firstly secreted by choroid plexuses in each lateral ventricle (1) flowing through the interventricular Monro’s foramen into the third ventricle, where choroid plexuses (3) also add more CSF. From the third ventricle, the CSF flows through the cerebral aqueduct (4) down to the fourth ventricle, where more CSF is also added (5). Next, CSF leaves the ventricular system through two lateral apertures and one median aperture, known as foramina of Luschka and Magendie, respectively (6). CSF fills the subarachnoid space covering the external surfaces of the brain and spinal cord (7). At arachnoid villi, CSF is reabsorbed into peripheral venous blood through dural venous sinuses (8).
Ijms 22 06389 g002
Table 1. Most commonly used AAV delivery routes in the context of Parkinson’s disease.
Table 1. Most commonly used AAV delivery routes in the context of Parkinson’s disease.
Delivery RoutesAAV SerotypesAnimal SpeciesReferences
IntraparenchymalN.A.Pig[23]
AAV1Rat[24]
AAV2Mouse[25,26,27,28,29,30,31]
Rat[24,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48]
NHP (M. fascicularis)[47,49]
NHP (M. mulatta)[50,51,52,53]
AAV5Mouse[54,55,56]
Rat[24,32,57,58]
AAV6Mouse[59]
Rat[60,61,62,63,64]
AAV7Mouse[65]
Rat[65]
AAV8Rat[61,66,67,68]
AAV9Mouse[69,70,71,72,73]
Rat[61,72,74,75,76,77,78,79,80]
NHP (M. mulatta)[72]
AAV2-retroNHP (M. mulatta)[50,81]
AAV-TTMouse[82]
Rat[82]
AAV-HBKONHP (M. mulatta)[83]
AAV-MNMRat[84]
Intra-CSFAAV2.1Mouse[85]
AAV2.5NHP (M. fascicularis)[86]
AAV5Mouse[87]
AAV6Mouse[88]
AAV7NHP (M. fascicularis)[89]
AAV8Mouse[87]
AAV9Mouse[85,90,91]
Rat[92]
NHP (M. mulatta)[93,94]
NHP (M. fascicularis)[86,89,94,95,96]
AAVDJ8Mouse[85]
AAVrh10Mouse[91]
Rat[97]
NHP (M. fascicularis)[96]
AAV-PHP.BMouse[98,99]
NHP (M.mulatta)[98,99].
IntravenousAAV9Mouse[100,101]
NHP (M. fasciularis)[94]
NHP (M. mulatta)[94]
AAV-ASMouse[102]
AAV1-PHP.BMouse[103]
AAV-PHP.BMouse[98,101,104,105]
NHP (M. mulatta)[98]
AAV-PHP.eBMouse[104]
AAV-PHP.SMouse[104]
SubpialAAV9Rat[106]
Pig[106,107]
NHP (M. fasciularis)[107]
Table 2. Clinical trials for PD (http://www.genetherapynet.com/clinical-trials.html, last access 14/06/2021). AADC: human aromatic L-amino acid decarboxylase; NRTN: Neurturin; STN: Subtalamic Nucleus; IP: Intraparenchymal; IC: Intracisternal; CM: Cisterna Magna.
Table 2. Clinical trials for PD (http://www.genetherapynet.com/clinical-trials.html, last access 14/06/2021). AADC: human aromatic L-amino acid decarboxylase; NRTN: Neurturin; STN: Subtalamic Nucleus; IP: Intraparenchymal; IC: Intracisternal; CM: Cisterna Magna.
Clinical Trial IdentifierDurationPhaseGeneAAV SerotypeDelivery RoutesRegionStatusReferences
NCT019735432013–2020IAADCAAV2IPPutamenCompleted[114]
NCT024185982015–2018I/IIAADCAAV2IPPutamenTerminated[115]
NCT030651922017–2021IAADCAAV2IPPutamenActive, not recruitingN.A. (Neurocrine Biosciences).
NCT035624942018–2022IIAADCAAV2IPN.A.Recruiting[116]
NCT037334962018–2026N.A.AADCAAV2IPPutamenEnrolling, by invitation[51,114,117]
NCT041675402020–2022IGDNFAAV2IPPutamenRecruitingN.A. (Brain Neurotherapy Bio, Inc.)
NCT016215812013–2022IGDNFAAV2IPPutamenActive, not recruiting[116,118,119,120]
NCT006438902008–2010IIGADAAV2IPSTNTerminated[121,122,123,124]
NCT001951432003–2005IGADAAV2IPSTNCompleted[122,125]
NCT013015732011–2012N.A.GADAAV2IPSTNTerminatedN.A. (Neurologix, Inc.)
NCT002528502005–2007INRTNAAV2IPPutamenCompleted[126]
NCT009855172009–2017I/IINRTNAAV2IPPutamenCompleted[127]
NCT041275782020–2027I/IIGBA1AAV9ICCMRecruitingN.A. (Prevail Therapeutics)
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Fajardo-Serrano, A.; Rico, A.J.; Roda, E.; Honrubia, A.; Arrieta, S.; Ariznabarreta, G.; Chocarro, J.; Lorenzo-Ramos, E.; Pejenaute, A.; Vázquez, A.; et al. Adeno-Associated Viral Vectors as Versatile Tools for Parkinson’s Research, Both for Disease Modeling Purposes and for Therapeutic Uses. Int. J. Mol. Sci. 2021, 22, 6389. https://doi.org/10.3390/ijms22126389

AMA Style

Fajardo-Serrano A, Rico AJ, Roda E, Honrubia A, Arrieta S, Ariznabarreta G, Chocarro J, Lorenzo-Ramos E, Pejenaute A, Vázquez A, et al. Adeno-Associated Viral Vectors as Versatile Tools for Parkinson’s Research, Both for Disease Modeling Purposes and for Therapeutic Uses. International Journal of Molecular Sciences. 2021; 22(12):6389. https://doi.org/10.3390/ijms22126389

Chicago/Turabian Style

Fajardo-Serrano, Ana, Alberto J. Rico, Elvira Roda, Adriana Honrubia, Sandra Arrieta, Goiaz Ariznabarreta, Julia Chocarro, Elena Lorenzo-Ramos, Alvaro Pejenaute, Alfonso Vázquez, and et al. 2021. "Adeno-Associated Viral Vectors as Versatile Tools for Parkinson’s Research, Both for Disease Modeling Purposes and for Therapeutic Uses" International Journal of Molecular Sciences 22, no. 12: 6389. https://doi.org/10.3390/ijms22126389

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop