Abstract
Neurodevelopment is a highly organized and complex process involving lasting and often irreversible changes in the central nervous system. Inherited disorders of neurotransmission (IDNT) are a group of genetic disorders where neurotransmission is primarily affected, resulting in abnormal brain development from early life, manifest as neurodevelopmental disorders (NDDs) and other chronic conditions. In principle, IDNT (particularly those of monogenic causes) are amenable to gene replacement therapy via precise genetic correction. However, practical challenges for gene replacement therapy remain major hurdles for its translation from bench to bedside. We discuss key considerations for the development of gene replacement therapies for IDNT. As an example, we describe our ongoing work on gene replacement therapy for succinic semialdehyde dehydrogenase deficiency (SSADHD), a GABA catabolic disorder.
Introduction
Inherited disorders of neurotransmission (IDNT) are a specialized group of genetic disorders characterized by defects in the biosynthesis, recycling, or transport of neurotransmitters or relevant channelopathies1 (Table 1). IDNT often associate with defects in neurodevelopment, with clinical symptoms manifest in neonatal or infantile stages2. and life-long symptoms including intellectual disability3,4, movement disorders5,6, and epilepsy2,7. Traditional treatment options for IDNT are often limited to symptom alleviation rather than amending the underlying genetic cause8. Although in certain cases conditions are treatable and controllable9, relevant therapies are likely to require life-long administration and provide limited efficacy. Encouragingly, emerging treatment paradigms for IDNT are shifting from symptom-based, supportive, or palliative approaches to disease-modifying, directly targeting the disease-causing mechanisms10.
Table 1.
List of IDNT, genetic information, and disease-associated phenotypes.
| Disease categories | OMIM gene number | HGNC gene symbol | Molecular function | Genetic inheritance trait | Disease phenotype | CDS size (bp) from NCBI | Gene replacement therapy developmental stage |
|---|---|---|---|---|---|---|---|
| Monoamine synthesis | 600225 | GCH1 | GTP cyclohydrolase I | Autosomal recessive | Hyperphenylalaninemia | 753 | Clinical trial142,143 |
| 612719 | PTS | 6-pyruvoyl-tetrahydropterin synthase | Autosomal recessive | Hyperphenylalaninemia | 438 | Proof-of-concept144 | |
| 612676 | QDPR | Quinoid dihydropteridine reductase | Autosomal recessive | Hyperphenylalaninemia | 735 | No known studies | |
| 182125 | SPR | Sepiapterin reductase | Autosomal recessive | Dopa-responsive dystonia | 786 | No known studies | |
| 191290 | TH | Tyrosine hydroxylase | Autosomal recessive | Segawa syndrome | 1587 | Clinical Trial142,143 | |
| 107930 | DDC | Aromatic L-amino acid decarboxylase | Autosomal recessive | AADC deficiency | 1443 | On Europe and UK market145 | |
| 609312 | DBH | Dopamine beta-hydroxylase | Autosomal recessive | Orthostatic hypotension | 1056 | No known studies | |
| Monoamine catabolism | 309850/309860 | MAOA | Monoamine oxidase | X-linked recessive | Brunner syndrome | 1584 | No known studies |
| Monoamine transport | 126455 | SLC6A3 | Dopamine transporter | Autosomal recessive | Infantile Parkinsonism-dystonia (Dopamine transporter deficiency) | 1863 | Proof-of-concept146,147 |
| 193001 | SLC18A2 | Vesicular monoamine transporter | Autosomal recessive | Infantile Parkinsonism-dystonia | 1545 | No known studies | |
| GABA catabolism | 137150 | ABAT | GABA-transaminase | Autosomal recessive | GABA-T deficiency | 1503 | No known studies |
| 610045 | ALDH5A1 | Succinic semialdehyde dehydrogenase | Autosomal recessive | SSADHD | 1608 | Proof-of-concept (Lee et al., in prep) | |
| GABA receptors | 137160 | GABRA1 | GABA(A) receptor, alpha 1 | Autosomal dominant | Developmental and epileptic encephalopathy | 1371 | No known studies |
| 137190 | GABRB1 | GABA(A) receptor, beta 1 | Autosomal dominant | Developmental and epileptic encephalopathy | 1425 | No known studies | |
| 600232 | GABRB2 | GABA(A) receptor, beta 2 | Autosomal dominant | Developmental and epileptic encephalopathy | 1425 | No known studies | |
| 137192 | GABRB3 | GABA(A) receptor, beta 3 | Autosomal dominant | Developmental and epileptic encephalopathy | 1422 | No known studies | |
| 137164 | GABRG2 | GABA(A) receptor, gamma 2 | Autosomal dominant | Developmental and epileptic encephalopathy | 1404 | No known studies | |
| GABA transport | 137165 | SLC6A1 | GABA transporter | Autosomal dominant | Myoclonic-atonic epilepsy | 1800 | Proof-of-concept (Steven Gray, personal communication) |
| Glycine receptors | 138491 | GLRA1 | Glycine receptor | Autosomal recessive/dominant | Hyperekplexia | 1101 | No known studies |
| 138492 | GLRB | Glycine receptor | Autosomal recessive | Hyperekplexia | 1494 | No known studies | |
| Glycine transport | 604159 | SLC6A5 | Glycine transporter | Autosomal recessive/dominant | Hyperekplexia | 2474 | No known studies |
| 601019 | SLC6A9 | Glycine transporter | Autosomal recessive | Glycine encephalopathy | 2121 | No known studies | |
| Glutamate receptors | 138249 | GRIN1 | Glutamate receptor, NMDA type | Autosomal recessive | Developmental and epileptic encephalopathy | 2817 | No known studies |
| 138253 | GRIN2A | Glutamate receptor, NMDA type | Autosomal dominant | Epilepsy, focal, with speech disorder | 3846 | No known studies | |
| 138252 | GRIN2B | Glutamate receptor, NMDA type | Autosomal dominant | Developmental and epileptic encephalopathy | 4455 | No known studies | |
| 602717 | GRIN2D | Glutamate receptor, NMDA type | Autosomal dominant | Developmental and epileptic encephalopathy | 4011 | No known studies | |
| Metabotropic receptors | 604473 | GRM1 | Glutamate receptor, metabotropic | Autosomal recessive/dominant | Spinocerebellar ataxia | 3585 | No known studies |
| 604096 | GRM6 | Glutamate receptor, metabotropic | Autosomal recessive | Night blindness, congenital stationary | 2634 | No known studies | |
| Channelopathies | 182389 | SCN1A | Sodium channel, voltage-gated | Autosomal dominant | Dravet syndrome | 6000 | Proof-of-concept148, clinical trial ongoing |
| 182390 | SCN2A | Sodium channel, voltage-gated | Autosomal dominant | Developmental and epileptic encephalopathy | 6018 | No known studies | |
| 600702 | SCN8A | Sodium channel, voltage-gated | Autosomal dominant | Developmental and epileptic encephalopathy | 5199 | No known studies | |
| 176258 | KCNC1 | Potassium channel, voltage-gated | Autosomal recessive | Progressive myoclonic epilepsy | 1536 | No known studies |
Recent advances in genetic medicine, bioengineering, and viral vector technology enable the translational deployment of molecularly-based therapy, such as gene replacement therapy11–13 Gene replacement therapy involves the delivery of a functioning gene into relevant cells and tissues, replacing or compensating for a disease-causing genetic origin, thereby correcting the underlying genetic defect. Viral vectors are often used for such therapeutic delivery. The commonly used viral vectors are listed in Table 2, where their basic properties, including payload capacity, transgene integration characteristics, and immunogenicity, are compared.
Table 2.
Comparison of commonly used viral vectors for gene replacement therapy.
| Adenovirus | Adeno-associated virus | Lentivirus (LV) | |
|---|---|---|---|
| Size | ~100 nm | ~25 nm | ~100 nm |
| Genome | dsDNA | ssDNA | ssRNA |
| Packaging capacity | ~37 kb29 | ~4.6 kb (single-stranded) ~2.3 kb (self-complementary) |
~8 kb |
| Transduction | Dividing and non-dividing cells | Dividing and non-dividing cells | Dividing and non-dividing cells |
| Transduction efficiency in non-human primate neural tissues | Low149 | High (>70% for AAV9)52,150,151 | High (up to ~70% using a second generation LV vector)152 |
| Expression | Transient | Stable | Stable |
| Genome integration in brain | Very low chance of integration27,153,154 | Confirmed with identified site for wild-type AAV, very low chance of integration for recombinant rAAV40,155 | Confirmed integration156 |
| Immunogenicity | High | Moderate | Low |
Viral-mediated gene replacement therapy has gained traction in biomedical research in recent years, as the rapid growth of relevant research illustrated by a PubMed search for ‘Gene Replacement Therapy’ between 1985 and 2023 revealing a substantial increase in the number of published articles within the last decade (Figure 1A). Notably, most studies are directed toward neuromuscular disorders, while those related to neurodevelopment or neurotransmitter disorders in the central nervous system remain a minority (Figure 1B). While encouraging recent trends indicate that gene replacement therapy will one day become a prevalent form of IDNT treatment, challenges remain to be overcome for successfully translating gene replacement therapy from bench to bedside.
Figure 1. PubMed search results for “gene replacement therapy.”.

(A) The number of published articles listed in the PubMed database after a keyword search of gene replacement therapy from 1980 to 2023. (B) Published articles in 2023 is classified into different disorder categories.
This article will discuss key principles and considerations for developing gene replacement therapy for IDNT. First, genetic inheritance traits determine gene replacement strategy. For example, autosomal dominant diseases are more challenging than autosomal recessive diseases because the dominant mutations might require specific silencing in addition to functioning gene supplementation. Mutant proteins might interfere with the ectopically introduced therapeutic functional proteins, often generating their own immunogenic responses. In certain situations, knock-down of mutant proteins might be necessary for functional restoration of protein products. Second, the size of the gene of interest and its expression profile will determine the choice of viral vector and relevant delivery strategies. Due to the limited size of viral vector payload capacity, the gene of interest might be too large to be accommodated in a single viral vector. In such scenarios, a mini-gene alternative approach (e.g., a truncated gene encoding only the functional domains) may be a viable strategy. There are also dual-vector systems that reconstitute a functioning gene from multiple components. Third, the expression profile of the gene of interest will limit the choice of vector delivery route. For example, for a gene expressed in certain brain regions, a local intracortical injection rather than brain-wide coverage might be sufficient and optimal. Fourth, the availability of a viable disease-relevant model will be necessary to investigate target engagement and phenotypic reversibility upon gene replacement. Therapeutic parameters relevant for toxicity and efficacy are vital information that can only be evaluated empirically using an animal or cell-based model exhibiting disease-relevant phenotypes. For bench-to-bedside translational drug discovery, the availability of such experimental disease models is critical in providing valuable information for dose finding, toxicity, and disease reversibility information. As an example of how these principles might apply to the translation of gene replacement therapy for IDNT, we utilize a recently proposed framework for evaluating neurodevelopmental disorders for gene therapy14, describing our ongoing efforts in developing gene replacement for succinic semialdehyde dehydrogenase deficiency (SSADHD).
Mendelian inheritance traits
The pathogenic mechanisms underlying IDNT include dominant or recessive traits. The inheritance trait of a certain genetic disorder is crucial for the design of corresponding therapeutic approach15–17. Autosomal recessive inheritance requires variants in both copies of the allele. Mutational content of the gene in autosomal recessive disorders could be identical (homozygous mutants) or different (compound heterozygous), resulting in defective or absence of protein products. For recessive disorders, loss-of-function mutations might be amenable to directly introducing a functional gene in gene replacement therapy (Figure 2A). On the other hand, autosomal dominant inheritance entails a disease-causing variant copy of an allele, in which a genetic trait ‘dominates’ over a normal gene copy. In autosomal dominant disorders, one copy of the gene is pathogenic while the other copy is functional, which renders gene targeted approach particularly challenging18. An example of gene therapy for autosomal dominant disorder is an ablate-and-supplement strategy employed by Meng and colleagues for Retinitis Pigmentosa (Figure 2B)19. In the case of mutations leading to early stop codon or other mutations resulting in little or no protein produced, gene replacement therapy or other relevant strategies aimed at augmenting gene products might be sufficient and effective. For example, in Dravet syndrome (DS)-related epileptic encephalopathy where autosomal dominant SCN1A gene mutations result in voltage-gated sodium channel Nav1.1 haploinsufficiency20, an antisense oligonucleotide (ASO) approach called Targeted Augmentation of Nuclear Gene Output (TANGO) rescues seizure and sudden death in a relevant mouse model21. Importantly, a Phase1/2a human trial of TANGO-based therapy in patients with DS demonstrated tolerability and favorable therapeutic results (dose-dependent and lasting reductions in convulsive seizure frequency) (Press Release from Stoke Therapeutics, July 2023). However, there are situations where endogenous protein products might interfere with the exogenously supplemented normal-functioning protein products. This is particularly relevant for proteins that require the assembly of multimers, where each component contributes to the protein products’ functional activities. For example, in SLC6A1-related disorders, protein products from a variant allele might interfere with endoplasmic reticulum trafficking22. In such situations, the functional activities of hybrid protein products assembled from mutated endogenous and normal-functioning ectopic proteins will require careful characterization and empirical functional measures. Functional characterization in such heterologous recombinant proteins23 will offer mechanistic insights into the efficacy of gene supplementation therapy. Furthermore, crystal structures derived from X-ray crystallography24 and molecular dynamics simulations25 of each polypeptide chain in the protein complex resultant from normal and variant peptides might improve our understanding at atomic levels. Overall, additional functional and biophysical approaches may help to understand functional integrity of such protein constitutes or complex formed from functioning ‘wild type’ and mutant proteins. Similar approaches dealing with autosomal dominant disorders, including selective silencing via anti-sense oligonucleotide (ASO)26, might be necessary to dampen the dominating negative effects of endogenous mutant proteins in such situations.
Figure 2. Gene therapy strategies for autosomal recessive and autosomal dominant disorders.

(A) In autosomal recessive disorders, the underlying etiology is due to the presence of disease-causing DNA mutations in both alleles resulting in non-functional protein products. A viable gene therapy strategy involves functional gene replacement via a viral-mediated approach. (B) In autosomal dominant disorders, disease-causing DNA mutations are found in one allele, resulting in dominant mutant protein products. A corresponding gene therapy strategy requires allele-specific silencing of the dominant mutations at the mRNA messenger level eliminating the dominant mutant proteins, sometimes requiring functional gene supplementation via a viral-mediated approach.
Gene size, expression profile, and routes of administration consideration
Gene replacement therapy utilizes bioengineered viral vector molecules carrying a functioning copy of the disease-relevant gene for delivery producing therapeutic effects. Notable viral vectors gaining recent traction for regulatory approval in clinical trials and eventual clinical use include adenovirus, adeno-associated virus (AAV), and lentivirus27. Table 2 (adapted from Tosolini and Sleigh28) summarizes their properties, including payload capacity, transgene stability, and immunogenicity of these prevalent vectors. Adenovirus infects post-mitotic cells and has large cargo capacity up to 37 kb29, but it triggers strong inflammatory reactions in the brain30,31. Lentiviral vectors offer long-term and stable gene expression to post-mitotic cells including neurons, but their genome-integrating property presents a realistic risk for safety32. On the other hand, AAV integration has been shown to be mediated by its viral gene Rep33, which encodes Rep proteins targeting the Adeno-Associated Virus Integration Site 1 (AAVS1) locus in human chromosome 1934,35. In principle, recombinant AAV (rAAV) lacking viral gene Rep enables transgenes to form circular concatemers that persist as episomes in the nucleus of transduced cells, significantly minimizing the chance of genome integration36,37. Nevertheless, long-term genome integration by AAV and evidence of subsequent insertional mutagenesis remain an active research area requiring long-term assessment38. Noticeably, recent meta-analysis of rAAV clinical trials revealed 90% of cases involving the CNS achieved transgene durability in follow-up visits39. Nevertheless, AAV is limited by a small packaging capacity of less than 5 kb. Alternative strategies accommodating a larger gene include a micro gene encoding a truncated but functional protein and co-administration of a dual-vector system reconstituting a full-length gene40. Besides viral vectors, non-viral-based strategies including biodegradable polymers, lipids, or other inorganic nanoparticles as alternative vehicle materials for gene therapy are under rapid development41. Non-viral-based vectors have large payload capacity and low immunogenicity but lack cell target specificity in general42. Overall, these strategies require a deep understanding of the protein’s structure-function relationship and empirical testing in relevant disease models for therapeutic efficacy.
Another key consideration is the tropism of viral vectors enabling the necessary targeting of cells and tissues relevant for the expression profile of the gene of interest43, in combination with route of administration to achieve physiologically relevant transgene delivery44. Certain genes are expressed in a broad range of cell types, or covering multiple brain regions, whereas others are expressed in specific cell types within confined tissue origins. For broadly expressed genes, a delivery strategy enabling wide transgene coverage might be necessary45. Viral vectors can be administered via intrathecal (IT) injection, i.e., targeting the subarachnoid space bypassing the blood-brain barrier (BBB) via lumbar puncture46,47, or intra-cisterna magna (ICM) injection48–50. Brain-wide coverage might be better achieved through delivery into cerebrospinal fluid (CSF) in the ventricles by intracerebroventricular (ICV) injection51,52. Intravenous (IV) infusion is another option with clear advantage of less invasiveness compared to the abovementioned delivery methods53, but it requires therapeutic agents with superb BBB penetrance for brain coverage. The development of new AAV capsid with such high BBB penetrating property is an active research area54–56, including the use of novel molecularly-based screening platforms for high throughput capsid selection57,58. On the other hand, intraparenchymal delivery might be ideal for local delivery targeting specific groups of neurons or brain structures relevant to the disorders due to underlying etiology44,59. However, the spread of viral vector is physically limited and is not ideal for brain-wide coverage60. Overall, the choice of the administration routes requires simultaneous considerations of viral vector BBB penetrance, cell targeting, and the extent of brain coverage. Their associated risks in terms of dosing, immune response, and corresponding immunosuppressive strategies must be carefully considered61–64.
Besides the considerations of transgene packaging into viable vectors and disease-relevant cell or tissue targeting accessible via corresponding delivery procedures, another critical factor is the expression profile of transgene expression. Ideally, therapeutic efficacy and associated risks of gene replacement therapy are related to the transgene expression dosage, i.e., the delivered gene dosage should be within a certain range that produces sustainable therapeutic effects, but not excessive (i.e., overdose) such that overexpression toxicity might overshadow any therapeutic advantage. This may be achieved partly by carefully dosing the delivered vector65, but another key factor is the molecular regulatory element or ‘promoter’ incorporated into the vector payload66. The choices of ‘promoters’ in gene replacement therapy candidates range from small but constitutive active promoters to self-regulatory elements providing feedback control67,68. Synthetic promoters might be used to target specific cell types in the CNS69. An alternative strategy that has not been commonly adopted is the use of custom design promoters encompassing specific gene regulatory elements in the endogenous promoter70. Molecular cloning of certain length DNA sequences (e.g., 2 kb) upstream of the transcriptional start of a given gene is often needed to characterize a given gene’s promoter and associated regulatory elements71. Truncational analyses of such upstream sequences tethered with a reporter gene assay (e.g., luciferase reporter) are often used strategies to fine-tune the inclusion or exclusion of DNA sequences as the promoter. However, traditional promoter studies often exploit heterologous recombinant cell-based systems such as fibroblasts for excellent transfection efficiency and reporter assay compatibility, but the cellular signaling and genetic contents are often biased or irrelevant for physiologic transgene expression. A fundamentally different approach to designing a promoter would Involve an unbiased assessment of transgene expression in relevant cell types (e.g., single cell RNA-seq), combined with bioinformatics72 and machine learning approach73–75 to ‘mine’ patterns of DNA sequences correlating all genes’ expression profiles across cell types (Figure 3). Ideally, these regulatory elements or promoters should drive a given gene’s functional expression resembling its endogenous profile across developmental stages. These custom promoters are tailored to specific genes, and therefore require empirical testing using relevant disease models, including those with human genomic relevance (e.g., patient derived cells). Remarkably, machine learning approaches are being deployed in the rational design and identification of next-generation viral vectors for higher transduction efficiency, improved tropism, and low toxicity76–78. The use of novel machine learning algorithms such as large language models79 might further facilitate such rational design of viral vector capsid80 and therapeutic payload81.
Figure 3. Proposed machine learning approach optimizing gene-specific promoter sequences.

A novel approach to optimizing the gene-specific promoter sequence of any given gene involves curating single-cell RNA-seq data and applying neural network analysis to seek common patterns of gene expressions across cell types and developmental stages. The optimized promoter sequence will then be used guiding the custom design of vector. The resulting gene replacement therapy candidate can be evaluated in disease-relevant models.
Disease model availability
Successful translational efforts from bench to bedside require empirical testing of any given treatment paradigm or drug candidate at a preclinical stage minimizing risks and maximizing therapeutic efficacy. In principle, disease models containing genetic variant(s) relevant for the disorder should resemble the underlying pathophysiology at a molecular level, displaying a disease-relevant phenotype for preclinical testing. Disease models are broadly classified as in vitro (in a test tube or culture dish) and in vivo (in a living organism), which have their corresponding strengths and limitations. For example, induced pluripotent stem cells (iPSCs) derived from patient samples such as skin biopsy are useful in vitro models for experimental drug testing82,83. iPSCs have the capacity to differentiate into specific cell types via molecular programming in a culture dish. iPSC-derived cell models are therefore versatile, and they contain genetic content identical to patients. Isogenic controls are often included allowing the gene of interest to be studied specifically in an otherwise identical genetic background. The patients’ genomic and mutational content is particularly important for gene replacement therapy because the functional proteins (from the therapeutic payload delivered by the vector) and the mutant proteins (from patients) might interact and affect each other at a functional level. Mutant proteins might also occupy relevant cellular signaling or trafficking machinery84 necessary for the therapeutic proteins to function. Recently, iPSCs are coupled with organoid technology allowing cells to grow in 3 dimensions forming complex structures resembling tissue-like organization and function85,86. This new approach further enables the modeling of cell-cell interactions, intrinsic/extrinsic cell signaling factors, and cell migration which were not feasible to be studied in a dish87,88. While in vitro disease models offer unique opportunity to investigate cellular function and properties relevant for disease mechanisms, nevertheless they are limited by the lack of complexity and intrinsic anatomy of an organism, and no disease-relevant behavior at an organismal level can be performed in any cell culture or organoid models.
On the other hand, in vivo disease modelling can be achieved by precise genetic modifications in a model organism, complementing in vitro models in certain aspects. Ideally, disease models should have a well-established and well-controlled genetic background, and reasonably short life cycle (e.g., within twelve months) allowing practical experimental paradigm and testing duration. A commonly used organism for in vivo disease modelling is Mus musculus (house mouse)89. Such mammalian disease models that recapitulate the underlying pathophysiology and disease-relevant phenotypes are particularly valuable for preclinical testing, because key treatment parameters can be investigated and fine-tuned to produce the best therapeutic outcomes in terms of phenotypic reversal. Mice also exhibit behaviors including sensory function, motor function and coordination, cognition, and social communication allowing studies in these complex functions which are often disease relevant90–92. An in vivo disease model also provides an opportunity to study pharmacokinetics (the study of how an organism affects a drug) and pharmacodynamics (the study of how the drug affects the organism), as well as testing clinically relevant delivery routes and their associated biodistribution93,94.
Strategies for in vivo disease modelling include the total disruption of a given gene via locus-specific disruption (e.g., knock-out), or precise insertion of disease-causing variant-mimicking sequences (e.g., knock-in)95. Alternatively, gene excision can be achieved via Cre-loxP strategy96. This strategy typically involves the development of two independent genetic lines (e.g., mouse lines), with one line harboring a pair of ‘loxP’ sites flanking functional domain(s) of a given gene, and the other line expressing Cre recombinase (typically driven by specific promoter for tissue-specific expression)97. When these two independent lines are bred together, the resultant offspring expresses Cre recombinase in specific tissues, disrupting a given gene via Cre-lox recombination achieving tissue-specific or cell type-specific deletion of gene function98. This strategy is particularly useful for modeling IDNT disease etiology where a given gene’s cell type-specific role can be investigated in detail. Relevant for gene replacement therapy in IDNT, a variation of the Cre-loxP strategy called ‘lox-STOP’ can be employed. Here, a genetic cassette harboring a STOP codon flanked by a pair of loxP sites will be inserted into a given gene, disrupting its baseline function99. This results in essentially a gene ‘knock-out’ situation, but the STOP codon can be readily removed upon the introduction of Cre (e.g. via genetic breeding with a Cre-expressing line, or the injection of a Cre-expressing viral vector)100. This system allows precise reactivation of a given gene, mimicking a gene restoration scenario. Clinically relevant parameters for gene replacement therapy including rate, time, and cell-specificity can be investigated using such animal models.
Importantly, in vivo disease modeling is subject to limitations. For example, disease-causing variants of a given gene in patients might not cause the same phenotype in the model organism due to species-specific genetic compensation or other variance requiring careful design and experimental planning to circumvent potential pitfalls101. The disease relevant phenotype in the model organism might be too severe (e.g., embryonic or neonatal lethal) or not manifested at all. In such situations, a combination of knock-out and hypomorphic variant (partial reduction of gene product) might be useful to fine-tune gene dosage recapitulating disease phenotype102,103.
Current progress in developing gene replacement therapy for SSADHD
Clinical background and preclinical considerations for gene replacement therapy
SSADHD is a rare autosomal recessive disorder due to loss-of-function mutations of the gene ALDH5A1 essential for GABA catabolism104. The genetic cause of SSADHD is monogenic, although multiple variants have been reported among patients105. The size of the human ALDH5A1 gene coding region is 1.6 kb106, a size compatible with commonly used viral vectors (Table 2). The underlying pathophysiology of SSADHD is due to the pathologic accumulation of GABA and its metabolite γ-hydroxybutyrate (GHB), the biochemical hallmarks of this disorder107. SSADHD symptoms include developmental delay, intellectual disability, and autistic behaviors typically detectable at an early stage (2–3 years of age)108. A natural history SSADHD study is ongoing, and thus far revealed a 15% incidence of sudden unexpected death in epilepsy (SUDEP) in this patient population among other developmental and behavioral morbidities109. Current treatment options for SSADHD are symptomatic with limited efficacy110. Gene replacement therapy is a potential disease-modifying treatment for SSADHD but is currently unavailable111,112. The broad expression profile of ALDH5A1 in the brain suggests that a brain-wide targeted approach is likely necessary.
To develop gene replacement therapy for SSADHD, we evaluated the current preclinical and clinical landscape of this disorder following our previously proposed framework14. Given our current research progress, combined with a highly engaged family community and readily accessible patient population, we estimated a composite GTS (gene target suitability) score of 34 for SSADHD (out of a maximum score of 40), indicating overall very positive favorability among some of the prime examples of neurodevelopmental disorders currently under gene therapy development (Table 3). Ongoing work on gene replacement therapy for SSADHD focuses on disease-relevant modeling and clinical biomarkers, including electroencephalography (EEG) and transcranial magnetic stimulation (TMS)113,114.
Table 3.
The GENE TARGET framework evaluation for SSADHD.
| Category | Description | Current status for SSADHD | Score |
|---|---|---|---|
| G | Genetic mechanism understood | Autosomal recessive mutation causing ALDH5A1 loss of function | 6/6 |
| E | Early diagnosis typical | Mean diagnosis age ~6.6 years | 1/2 |
| N | Natural history documented | Natural History Study ongoing | 2/3 |
| E | Endpoints validated | EEG and TMS-based under development | 1/3 |
| T | Tools targeting relevant tissues available | Brain and liver targeting viral vector tools available | 5/6 |
| A | Availability of alternative effective treatment | No disease-modifying treatment available | 2/2 |
| R | Reversibility demonstrated | Phenotypic reversibility demonstrated in a mouse model | 4/4 |
| G | Gene is tolerant to dosage changes | Effective gene dosage threshold and range exist | 3/4 |
| E | Ethical principles considered | Moderate to severe level of disability often requiring life-long personal care, risk of epilepsy, and SUDEP | 6/6 |
| T | Target population is engaged and accessible | Well-organized clinics and engaged individual/family communities worldwide | 4/4 |
| Total score | 34/40 |
At the disease modeling front, we utilized the ‘lox-STOP’ strategy as described above, creating a novel SSADH mouse model allowing gene restoration ‘on demand’112. This new aldh5a1lox-STOP mouse model enables systematic investigation of three key parameters (i.e., rate, age, and cell-specificity) for successful phenotypic rescue. As a step toward clinical translation, current efforts also include the development of a custom-designed AAV vector encompassing an ALDH5A1-specific promoter tethered with the human ALDH5A1 coding sequence. In addition, human iPSC models are also under parallel development, which will provide a powerful testing platform with human genomic relevance. Furthermore, functional SSADH enzyme exists as tetramers115. The intracellular fate of endogenous mutant proteins (i.e., intracellular trafficking and recycling) and the molecular interaction between patients’ endogenous mutant proteins and the functioning proteins provided by the gene replacement therapy payload might play a key role in determining treatment efficacy, which can be tested empirically by a range of functional assays and biophysical methods116.
While ongoing experiments assessing the safety and efficacy of a candidate gene replacement therapy yield preliminary favorable results117,118, compensatory changes at a molecular level might have taken place during early brain development in SSADHD (Figure 4). Broadly, compensatory changes that occur during neurodevelopment are common among IDNT and are realistic concerns because unwanted adverse effects might result from suboptimal gene restoration. For example, restoring the GABA-degrading enzyme in a setting of down-regulated GABA receptors in SSADHD might inadvertently lead to a hypo-GABAergic condition, increasing the chance of seizures and brain injury111. Here, we describe two potential mechanisms relevant to compensatory changes that might fine-tune GABAergic inhibition.
Figure 4. Potential GABA-related compensatory mechanisms underlying SSADH deficiency.

A schematic diagram showing the conceptual molecular changes in SSADH deficiency where GABA catabolism is impaired. At the presynaptic level, pathologic accumulation of GABA and its metabolite GHB might be accompanied by reduced GABA synthesis. At the postsynaptic site, synaptic GABAA receptors (particularly of those γ-subunit containing receptors) might be removed from cell surface due to enhanced internalization.
Compensatory GABAergic synapse changes
GABAergic synapses in the brain consist of presynaptic and postsynaptic domains, which are highly regulated during development. At the pre-synaptic site, neurotransmitter release is mediated by a complex system involving syntaxin119, synuclein120, synaptotagmin121, synaptobrevin122, and synaptophysin123. Distinct developmental events involving relevant protein components are highly coordinated and often arranged sequentially. For example, vesicle pools are first formed and refined during synapse formation events124,125. Vesicle release undergoes transitional changes from spontaneous events to evoked release upon activity-dependent mechanisms126. Calcium-mediated vesicle fusion is then further coupled in an activity-dependent manner, strengthening synapse integrity127. At the postsynaptic site, GABA receptors are found in hetero-pentameric structures consisting of a typical 2α/2β/1γ composition128. In rodent early postnatal life, GABA receptors are predominantly α2/3-containing129,130. This expression pattern is replaced with α1-containing GABA receptors as the brain matures131,132. It is known that α2/3 subunits contribute to a slow kinetics of GABA current, while α1 subunits mediate fast decay. Strikingly, this subunit developmental switch precisely coordinates GABA current dynamics throughout development133. These findings have broad indications related to the function and computation of local neural circuits affecting higher-level functions. Interestingly, in pathologic conditions such as epilepsy, this GABA receptor subunit compositional switch is often delayed134. Patients with SSADHD have reduced GABA receptor expression135, highlighting an altered GABA developmental trajectory in these patients. Beyond the synaptic regions, the extrasynaptic GABAergic system mediating tonic inhibition is predominantly mediated by δ-containing GABA receptors136. Overall, the dynamic changes of presynaptic, postsynaptic, and extrasynaptic regions involved in GABAergic neurotransmission in SSADHD remain an active research topic that likely provides insights into de-risking and fine-tuning gene replacement strategy.
Chloride homeostasis disruptions
Fast GABAergic inhibition depends on the neuronal transmembrane chloride gradient regulated by chloride transporters137. The neuronal-specific potassium chloride co-transporter KCC2 is a key player in chloride homeostasis in mature neurons, and thus plays a crucial role in maintaining the efficacy of GABA-mediated inhibition138. In the early developmental stage, however, KCC2 expression is relatively low compared to the chloride intruder sodium-potassium chloride co-transporter NKCC1, rendering GABA less inhibitory or potentially depolarizing in premature neurons139. Interestingly, GABA itself plays a role in promoting KCC2 expression, implying that this developmental switch from depolarizing to hyperpolarizing GABA current is an autonomous process140. KCC2 activity also plays a key role in inhibitory neuron migration, a fundamental process in functional brain circuit development141. In a condition such as SSADHD, where pathologic GABA accumulation begins in early life, chloride homeostasis is likely significantly affected, resulting in a profound and sustained pathologic phenotype. The reversibility of these changes will be necessary for successful gene replacement therapy in SSADHD.
Concluding remarks: a generalizing principle beyond SSADHD
Given the recent rapid advancement of genetic medicine and viral technology, gene replacement therapy for inherited disorders of neurotransmission (IDNT) (especially those of monogenic origin) quickly emerged as a realistic treatment option. The translational research of gene replacement therapy is multi-disciplinary, requiring combined efforts from various areas of expertise, including bioengineering, molecular biology, in vitro and in vivo disease modeling, pharmacology, clinical care, and regulatory experience. Here, we discuss the practical aspects of developing gene replacement therapy for IDNT, focusing on the involved genetic content (i.e., inheritance, gene size, expression profile), as well as the availability of disease models allowing the explicit testing of key parameters for maximum therapeutic efficacy and minimizing risk. We use our ongoing program of gene replacement therapy in SSADHD as an example, demonstrating our thought process and describing experimental efforts for agile bench-to-bedside translation. While current work toward SSADHD treatment is in progress, we believe our experience provides necessary insights and guiding principles generalizable for initial efforts to develop gene replacement therapy for other indications within the IDNT family.
Funding
This research was funded by the SSADH Association and Translational Research Program (TRP) at Boston Children’s Hospital (to HHCL, AR), NIH R21NS121858 (to AR), NIH R01HD09114203 (to PLP), and the Intellectual and Development Disabilities Research Center (IDDRC) at Boston Children’s Hospital (P50HD105351) for research support infrastructures. HHCL is also supported by the Rosamund Stone Zander Translational Neuroscience Center (RSZ TNC) at Boston Children’s Hospital.
Conflict of interest statement
HHCL, PLP, and AR are co-inventors of the US Patent “Gene therapy in succinic semialdehyde dehydrogenase deficiency (SSADHD).” HHCL and AR are co-founders and have equity in Galibra Neuroscience. Galibra Neuroscience did not sponsor this work. AR also has equity in Neuromotion and PrevEp, and reports grant support or consulting relationships with Autifony Therapeutics, CRE medical, Encoded, Modulight, Neuroelectrics, Neurorex, Roche and Ovid, MS reports grant support from Biogen, Astellas, Bridgebio, and Aucta. He has served on Scientific Advisory Boards for Roche, SpringWorks Therapeutics, Jaguar Therapeutics and Alkermes.
Data availability statement
Data presented in this review is accessible from public databases PubMed (https://pubmed.ncbi.nlm.nih.gov/) and OMIM (https://www.omim.org/).
Reference
- 1.Tristan-Noguero A & Garcia-Cazorla A Synaptic metabolism: a new approach to inborn errors of neurotransmission. J Inherit Metab Dis 41, 1065–1075, doi: 10.1007/s10545-018-0235-7 (2018). [DOI] [PubMed] [Google Scholar]
- 2.Saudubray JM & Garcia-Cazorla A An overview of inborn errors of metabolism affecting the brain: from neurodevelopment to neurodegenerative disorders. Dialogues Clin Neurosci 20, 301–325, doi: 10.31887/DCNS.2018.20.4/jmsaudubray (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Warmerdam HAG et al. A Scoping Review of Inborn Errors of Metabolism Causing Progressive Intellectual and Neurologic Deterioration (PIND). Front Neurol 10, 1369, doi: 10.3389/fneur.2019.01369 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.van Karnebeek CD & Stockler S Treatable inborn errors of metabolism causing intellectual disability: a systematic literature review. Mol Genet Metab 105, 368–381, doi: 10.1016/j.ymgme.2011.11.191 (2012). [DOI] [PubMed] [Google Scholar]
- 5.Garcia-Cazorla A et al. Inborn errors of metabolism and motor disturbances in children. J Inherit Metab Dis 32, 618–629, doi: 10.1007/s10545-009-1194-9 (2009). [DOI] [PubMed] [Google Scholar]
- 6.Eggink H et al. Rare inborn errors of metabolism with movement disorders: a case study to evaluate the impact upon quality of life and adaptive functioning. Orphanet J Rare Dis 9, 177, doi: 10.1186/s13023-014-0177-6 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Mastrangelo M Epilepsy in inherited neurotransmitter disorders: Spotlights on pathophysiology and clinical management. Metab Brain Dis 36, 29–43, doi: 10.1007/s11011-020-00635-x (2021). [DOI] [PubMed] [Google Scholar]
- 8.Leach EL, Shevell M, Bowden K, Stockler-Ipsiroglu S & van Karnebeek CD Treatable inborn errors of metabolism presenting as cerebral palsy mimics: systematic literature review. Orphanet J Rare Dis 9, 197, doi: 10.1186/s13023-014-0197-2 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Hoytema van Konijnenburg EMM et al. Treatable inherited metabolic disorders causing intellectual disability: 2021 review and digital app. Orphanet J Rare Dis 16, 170, doi: 10.1186/s13023-021-01727-2 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Pearl PL, Tokatly-Latzer I, Lee HH & Rotenberg A New Therapeutic Approaches to Inherited Metabolic Pediatric Epilepsies. Neurology, doi: 10.1212/WNL.0000000000207133 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Petrich J, Marchese D, Jenkins C, Storey M & Blind J Gene Replacement Therapy: A Primer for the Health-system Pharmacist. J Pharm Pract 33, 846–855, doi: 10.1177/0897190019854962 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Bulaklak K & Gersbach CA The once and future gene therapy. Nat Commun 11, 5820, doi: 10.1038/s41467-020-19505-2 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Davidson BL et al. Gene-based therapeutics for rare genetic neurodevelopmental psychiatric disorders. Mol Ther 30, 2416–2428, doi: 10.1016/j.ymthe.2022.05.014 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Chopra M et al. GENE TARGET: A framework for evaluating Mendelian neurodevelopmental disorders for gene therapy. Mol Ther Methods Clin Dev 27, 32–46, doi: 10.1016/j.omtm.2022.08.007 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Carvill GL, Matheny T, Hesselberth J & Demarest S Haploinsufficiency, Dominant Negative, and Gain-of-Function Mechanisms in Epilepsy: Matching Therapeutic Approach to the Pathophysiology. Neurotherapeutics 18, 1500–1514, doi: 10.1007/s13311-021-01137-z (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Pelletier R, Caron SO & Puymirat J RNA based gene therapy for dominantly inherited diseases. Curr Gene Ther 6, 131–146, doi: 10.2174/156652306775515592 (2006). [DOI] [PubMed] [Google Scholar]
- 17.Millington-Ward S et al. Suppression and replacement gene therapy for autosomal dominant disease in a murine model of dominant retinitis pigmentosa. Mol Ther 19, 642–649, doi: 10.1038/mt.2010.293 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Kwon D Failure of genetic therapies for Huntington’s devastates community. Nature 593, 180, doi: 10.1038/d41586-021-01177-7 (2021). [DOI] [PubMed] [Google Scholar]
- 19.Meng D, Ragi SD & Tsang SH Therapy in Rhodopsin-Mediated Autosomal Dominant Retinitis Pigmentosa. Mol Ther 28, 2139–2149, doi: 10.1016/j.ymthe.2020.08.012 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Bechi G et al. Pure haploinsufficiency for Dravet syndrome Na(V)1.1 (SCN1A) sodium channel truncating mutations. Epilepsia 53, 87–100, doi: 10.1111/j.1528-1167.2011.03346.x (2012). [DOI] [PubMed] [Google Scholar]
- 21.Han Z et al. Antisense oligonucleotides increase Scn1a expression and reduce seizures and SUDEP incidence in a mouse model of Dravet syndrome. Sci Transl Med 12, doi: 10.1126/scitranslmed.aaz6100 (2020). [DOI] [PubMed] [Google Scholar]
- 22.Wang J et al. Endoplasmic reticulum retention and degradation of a mutation in SLC6A1 associated with epilepsy and autism. Mol Brain 13, 76, doi: 10.1186/s13041-020-00612-6 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Marvin JS et al. A genetically encoded fluorescent sensor for in vivo imaging of GABA. Nat Methods 16, 763–770, doi: 10.1038/s41592-019-0471-2 (2019). [DOI] [PubMed] [Google Scholar]
- 24.Papageorgiou AC X-Ray Crystallography in Structure-Function Characterization of Therapeutic Enzymes. Adv Exp Med Biol 1148, 81–103, doi: 10.1007/978-981-13-7709-9_4 (2019). [DOI] [PubMed] [Google Scholar]
- 25.Hollingsworth SA & Dror RO Molecular Dynamics Simulation for All. Neuron 99, 1129–1143, doi: 10.1016/j.neuron.2018.08.011 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Watts JK & Corey DR Silencing disease genes in the laboratory and the clinic. J Pathol 226, 365–379, doi: 10.1002/path.2993 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bulcha JT, Wang Y, Ma H, Tai PWL & Gao G Viral vector platforms within the gene therapy landscape. Signal Transduct Target Ther 6, 53, doi: 10.1038/s41392-021-00487-6 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Tosolini AP & Sleigh JN Intramuscular Delivery of Gene Therapy for Targeting the Nervous System. Front Mol Neurosci 13, 129, doi: 10.3389/fnmol.2020.00129 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Ricobaraza A, Gonzalez-Aparicio M, Mora-Jimenez L, Lumbreras S & Hernandez-Alcoceba R High-Capacity Adenoviral Vectors: Expanding the Scope of Gene Therapy. Int J Mol Sci 21, doi: 10.3390/ijms21103643 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Barkats M et al. Adenovirus in the brain: recent advances of gene therapy for neurodegenerative diseases. Prog Neurobiol 55, 333–341, doi: 10.1016/s0301-0082(98)00028-8 (1998). [DOI] [PubMed] [Google Scholar]
- 31.Byrnes AP, Rusby JE, Wood MJ & Charlton HM Adenovirus gene transfer causes inflammation in the brain. Neuroscience 66, 1015–1024, doi: 10.1016/0306-4522(95)00068-t (1995). [DOI] [PubMed] [Google Scholar]
- 32.Connolly JB Lentiviruses in gene therapy clinical research. Gene Ther 9, 1730–1734, doi: 10.1038/sj.gt.3301893 (2002). [DOI] [PubMed] [Google Scholar]
- 33.Weitzman MD, Kyostio SR, Kotin RM & Owens RA Adeno-associated virus (AAV) Rep proteins mediate complex formation between AAV DNA and its integration site in human DNA. Proc Natl Acad Sci U S A 91, 5808–5812, doi: 10.1073/pnas.91.13.5808 (1994). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Hamilton H, Gomos J, Berns KI & Falck-Pedersen E Adeno-associated virus site-specific integration and AAVS1 disruption. J Virol 78, 7874–7882, doi: 10.1128/JVI.78.15.7874-7882.2004 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Kotin RM, Linden RM & Berns KI Characterization of a preferred site on human chromosome 19q for integration of adeno-associated virus DNA by non-homologous recombination. EMBO J 11, 5071–5078, doi: 10.1002/j.1460-2075.1992.tb05614.x (1992). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Naso MF, Tomkowicz B, Perry WL 3rd & Strohl WR Adeno-Associated Virus (AAV) as a Vector for Gene Therapy. BioDrugs 31, 317–334, doi: 10.1007/s40259-017-0234-5 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Choi VW, McCarty DM & Samulski RJ Host cell DNA repair pathways in adeno-associated viral genome processing. J Virol 80, 10346–10356, doi: 10.1128/JVI.00841-06 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Sabatino DE et al. Evaluating the state of the science for adeno-associated virus integration: An integrated perspective. Mol Ther 30, 2646–2663, doi: 10.1016/j.ymthe.2022.06.004 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Shen W, Liu S & Ou L rAAV immunogenicity, toxicity, and durability in 255 clinical trials: A meta-analysis. Front Immunol 13, 1001263, doi: 10.3389/fimmu.2022.1001263 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Wang D, Tai PWL & Gao G Adeno-associated virus vector as a platform for gene therapy delivery. Nat Rev Drug Discov 18, 358–378, doi: 10.1038/s41573-019-0012-9 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Zu H & Gao D Non-viral Vectors in Gene Therapy: Recent Development, Challenges, and Prospects. AAPS J 23, 78, doi: 10.1208/s12248-021-00608-7 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Wang C et al. Emerging non-viral vectors for gene delivery. J Nanobiotechnology 21, 272, doi: 10.1186/s12951-023-02044-5 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Jang MJ et al. Spatial transcriptomics for profiling the tropism of viral vectors in tissues. Nat Biotechnol, doi: 10.1038/s41587-022-01648-w (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Zhou K, Han J, Wang Y, Zhang Y & Zhu C Routes of administration for adeno-associated viruses carrying gene therapies for brain diseases. Front Mol Neurosci 15, 988914, doi: 10.3389/fnmol.2022.988914 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Chen L et al. Recent progress in targeted delivery vectors based on biomimetic nanoparticles. Signal Transduct Target Ther 6, 225, doi: 10.1038/s41392-021-00631-2 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Bey K et al. Efficient CNS targeting in adult mice by intrathecal infusion of single-stranded AAV9-GFP for gene therapy of neurological disorders. Gene Ther 24, 325–332, doi: 10.1038/gt.2017.18 (2017). [DOI] [PubMed] [Google Scholar]
- 47.Hinderer C et al. Translational Feasibility of Lumbar Puncture for Intrathecal AAV Administration. Mol Ther Methods Clin Dev 17, 969–974, doi: 10.1016/j.omtm.2020.04.012 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Lutters B & Koehler PJ A road less travelled: the centenary of cisterna magna puncture. Brain 143, 2858–2862, doi: 10.1093/brain/awaa254 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Marchi PM, Marrone L & Azzouz M Delivery of therapeutic AAV9 vectors via cisterna magna to treat neurological disorders. Trends Mol Med 28, 79–80, doi: 10.1016/j.molmed.2021.09.007 (2022). [DOI] [PubMed] [Google Scholar]
- 50.Hinderer C et al. Widespread gene transfer in the central nervous system of cynomolgus macaques following delivery of AAV9 into the cisterna magna. Mol Ther Methods Clin Dev 1, 14051, doi: 10.1038/mtm.2014.51 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Galvan A et al. Intracerebroventricular Administration of AAV9-PHP.B SYN1-EmGFP Induces Widespread Transgene Expression in the Mouse and Monkey Central Nervous System. Hum Gene Ther 32, 599–615, doi: 10.1089/hum.2020.301 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Bey K et al. Intra-CSF AAV9 and AAVrh10 Administration in Nonhuman Primates: Promising Routes and Vectors for Which Neurological Diseases? Mol Ther Methods Clin Dev 17, 771–784, doi: 10.1016/j.omtm.2020.04.001 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Gessler DJ, Tai PWL, Li J & Gao G Intravenous Infusion of AAV for Widespread Gene Delivery to the Nervous System. Methods Mol Biol 1950, 143–163, doi: 10.1007/978-1-4939-9139-6_8 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Chuapoco MR et al. Adeno-associated viral vectors for functional intravenous gene transfer throughout the non-human primate brain. Nat Nanotechnol 18, 1241–1251, doi: 10.1038/s41565-023-01419-x (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Goertsen D et al. AAV capsid variants with brain-wide transgene expression and decreased liver targeting after intravenous delivery in mouse and marmoset. Nat Neurosci 25, 106–115, doi: 10.1038/s41593-021-00969-4 (2022). [DOI] [PubMed] [Google Scholar]
- 56.Yao Y et al. Variants of the adeno-associated virus serotype 9 with enhanced penetration of the blood-brain barrier in rodents and primates. Nat Biomed Eng 6, 1257–1271, doi: 10.1038/s41551-022-00938-7 (2022). [DOI] [PubMed] [Google Scholar]
- 57.Nonnenmacher M et al. Rapid evolution of blood-brain-barrier-penetrating AAV capsids by RNA-driven biopanning. Mol Ther Methods Clin Dev 20, 366–378, doi: 10.1016/j.omtm.2020.12.006 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Deverman BE et al. Cre-dependent selection yields AAV variants for widespread gene transfer to the adult brain. Nat Biotechnol 34, 204–209, doi: 10.1038/nbt.3440 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Salegio EA, Samaranch L, Kells AP, Forsayeth J & Bankiewicz K Guided delivery of adeno-associated viral vectors into the primate brain. Adv Drug Deliv Rev 64, 598–604, doi: 10.1016/j.addr.2011.10.005 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Gray SJ, Woodard KT & Samulski RJ Viral vectors and delivery strategies for CNS gene therapy. Ther Deliv 1, 517–534, doi: 10.4155/tde.10.50 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Choudhury SR et al. Viral vectors for therapy of neurologic diseases. Neuropharmacology 120, 63–80, doi: 10.1016/j.neuropharm.2016.02.013 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Fischell JM & Fishman PS A Multifaceted Approach to Optimizing AAV Delivery to the Brain for the Treatment of Neurodegenerative Diseases. Front Neurosci 15, 747726, doi: 10.3389/fnins.2021.747726 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Perez BA, Shutterly A, Chan YK, Byrne BJ & Corti M Management of Neuroinflammatory Responses to AAV-Mediated Gene Therapies for Neurodegenerative Diseases. Brain Sci 10, doi: 10.3390/brainsci10020119 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Prasad S et al. Immune Responses and Immunosuppressive Strategies for Adeno-Associated Virus-Based Gene Therapy for Treatment of Central Nervous System Disorders: Current Knowledge and Approaches. Hum Gene Ther 33, 1228–1245, doi: 10.1089/hum.2022.138 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Tang F, Wong H & Ng CM Rational Clinical Dose Selection of Adeno-Associated Virus-Mediated Gene Therapy Based on Allometric Principles. Clin Pharmacol Ther 110, 803–807, doi: 10.1002/cpt.2269 (2021). [DOI] [PubMed] [Google Scholar]
- 66.Nieuwenhuis B et al. Optimization of adeno-associated viral vector-mediated transduction of the corticospinal tract: comparison of four promoters. Gene Ther 28, 56–74, doi: 10.1038/s41434-020-0169-1 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Gray SJ et al. Optimizing promoters for recombinant adeno-associated virus-mediated gene expression in the peripheral and central nervous system using self-complementary vectors. Hum Gene Ther 22, 1143–1153, doi: 10.1089/hum.2010.245 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Choi JH et al. Optimization of AAV expression cassettes to improve packaging capacity and transgene expression in neurons. Mol Brain 7, 17, doi: 10.1186/1756-6606-7-17 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Juttner J et al. Targeting neuronal and glial cell types with synthetic promoter AAVs in mice, non-human primates and humans. Nat Neurosci 22, 1345–1356, doi: 10.1038/s41593-019-0431-2 (2019). [DOI] [PubMed] [Google Scholar]
- 70.Duba-Kiss R, Niibori Y & Hampson DR GABAergic Gene Regulatory Elements Used in Adeno-Associated Viral Vectors. Front Neurol 12, 745159, doi: 10.3389/fneur.2021.745159 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Doane AS & Elemento O Regulatory elements in molecular networks. Wiley Interdiscip Rev Syst Biol Med 9, doi: 10.1002/wsbm.1374 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Preissl S, Gaulton KJ & Ren B Characterizing cis-regulatory elements using single-cell epigenomics. Nat Rev Genet 24, 21–43, doi: 10.1038/s41576-022-00509-1 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Li Y, Shi W & Wasserman WW Genome-wide prediction of cis-regulatory regions using supervised deep learning methods. BMC Bioinformatics 19, 202, doi: 10.1186/s12859-018-2187-1 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Smith GD, Ching WH, Cornejo-Paramo P & Wong ES Decoding enhancer complexity with machine learning and high-throughput discovery. Genome Biol 24, 116, doi: 10.1186/s13059-023-02955-4 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Butt AH, Alkhalifah T, Alturise F & Khan YD A machine learning technique for identifying DNA enhancer regions utilizing CIS-regulatory element patterns. Sci Rep 12, 15183, doi: 10.1038/s41598-022-19099-3 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Becker J, Fakhiri J & Grimm D Fantastic AAV Gene Therapy Vectors and How to Find Them-Random Diversification, Rational Design and Machine Learning. Pathogens 11, doi: 10.3390/pathogens11070756 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Han Z et al. Computer-Aided Directed Evolution Generates Novel AAV Variants with High Transduction Efficiency. Viruses 15, doi: 10.3390/v15040848 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Ghauri MS & Ou L AAV Engineering for Improving Tropism to the Central Nervous System. Biology (Basel) 12, doi: 10.3390/biology12020186 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Liu J et al. Large language models in bioinformatics: applications and perspectives. ArXiv (2024). [Google Scholar]
- 80.Wec AZ et al. Overcoming Immunological Challenges Limiting Capsid-Mediated Gene Therapy With Machine Learning. Front Immunol 12, 674021, doi: 10.3389/fimmu.2021.674021 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Toufiq M et al. Harnessing large language models (LLMs) for candidate gene prioritization and selection. J Transl Med 21, 728, doi: 10.1186/s12967-023-04576-8 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Elitt MS, Barbar L & Tesar PJ Drug screening for human genetic diseases using iPSC models. Hum Mol Genet 27, R89–R98, doi: 10.1093/hmg/ddy186 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Farkhondeh A et al. Induced pluripotent stem cells for neural drug discovery. Drug Discov Today 24, 992–999, doi: 10.1016/j.drudis.2019.01.007 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Howell GJ, Holloway ZG, Cobbold C, Monaco AP & Ponnambalam S Cell biology of membrane trafficking in human disease. Int Rev Cytol 252, 1–69, doi: 10.1016/S0074-7696(06)52005-4 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Turhan AG et al. iPSC-Derived Organoids as Therapeutic Models in Regenerative Medicine and Oncology. Front Med (Lausanne) 8, 728543, doi: 10.3389/fmed.2021.728543 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Kim J, Koo BK & Knoblich JA Human organoids: model systems for human biology and medicine. Nat Rev Mol Cell Biol 21, 571–584, doi: 10.1038/s41580-020-0259-3 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Bose S, Clevers H & Shen X Promises and Challenges of Organoid-Guided Precision Medicine. Med 2, 1011–1026, doi: 10.1016/j.medj.2021.08.005 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Huang Y et al. Research Progress, Challenges, and Breakthroughs of Organoids as Disease Models. Front Cell Dev Biol 9, 740574, doi: 10.3389/fcell.2021.740574 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Phifer-Rixey M & Nachman MW Insights into mammalian biology from the wild house mouse Mus musculus. Elife 4, doi: 10.7554/eLife.05959 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Hanell A & Marklund N Structured evaluation of rodent behavioral tests used in drug discovery research. Front Behav Neurosci 8, 252, doi: 10.3389/fnbeh.2014.00252 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Simmons DH, Titley HK, Hansel C & Mason P Behavioral Tests for Mouse Models of Autism: An Argument for the Inclusion of Cerebellum-Controlled Motor Behaviors. Neuroscience 462, 303–319, doi: 10.1016/j.neuroscience.2020.05.010 (2021). [DOI] [PubMed] [Google Scholar]
- 92.Crawley JN Mouse behavioral assays relevant to the symptoms of autism. Brain Pathol 17, 448–459, doi: 10.1111/j.1750-3639.2007.00096.x (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Ball K et al. Characterizing the Pharmacokinetics and Biodistribution of Therapeutic Proteins: An Industry White Paper. Drug Metab Dispos 50, 858–866, doi: 10.1124/dmd.121.000463 (2022). [DOI] [PubMed] [Google Scholar]
- 94.Chen N, Sun K, Chemuturi NV, Cho H & Xia CQ The Perspective of DMPK on Recombinant Adeno-Associated Virus-Based Gene Therapy: Past Learning, Current Support, and Future Contribution. AAPS J 24, 31, doi: 10.1208/s12248-021-00678-7 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Doyle A, McGarry MP, Lee NA & Lee JJ The construction of transgenic and gene knockout/knockin mouse models of human disease. Transgenic Res 21, 327–349, doi: 10.1007/s11248-011-9537-3 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Kim H, Kim M, Im SK & Fang S Mouse Cre-LoxP system: general principles to determine tissue-specific roles of target genes. Lab Anim Res 34, 147–159, doi: 10.5625/lar.2018.34.4.147 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Wang X Cre transgenic mouse lines. Methods Mol Biol 561, 265–273, doi: 10.1007/978-1-60327-019-9_17 (2009). [DOI] [PubMed] [Google Scholar]
- 98.Harris JA et al. Anatomical characterization of Cre driver mice for neural circuit mapping and manipulation. Front Neural Circuits 8, 76, doi: 10.3389/fncir.2014.00076 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Dragatsis I & Zeitlin S A method for the generation of conditional gene repair mutations in mice. Nucleic Acids Res 29, E10, doi: 10.1093/nar/29.3.e10 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Sarno E & Robison AJ Emerging role of viral vectors for circuit-specific gene interrogation and manipulation in rodent brain. Pharmacol Biochem Behav 174, 2–8, doi: 10.1016/j.pbb.2018.04.008 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Gurumurthy CB, Saunders TL & Ohtsuka M Designing and generating a mouse model: frequently asked questions. J Biomed Res 35, 76–90, doi: 10.7555/JBR.35.20200197 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Pollizzi K et al. A hypomorphic allele of Tsc2 highlights the role of TSC1/TSC2 in signaling to AKT and models mild human TSC2 alleles. Hum Mol Genet 18, 2378–2387, doi: 10.1093/hmg/ddp176 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Kelly E et al. mGluR5 Modulation of Behavioral and Epileptic Phenotypes in a Mouse Model of Tuberous Sclerosis Complex. Neuropsychopharmacology 43, 1457–1465, doi: 10.1038/npp.2017.295 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Akaboshi S et al. Mutational spectrum of the succinate semialdehyde dehydrogenase (ALDH5A1) gene and functional analysis of 27 novel disease-causing mutations in patients with SSADH deficiency. Hum Mutat 22, 442–450, doi: 10.1002/humu.10288 (2003). [DOI] [PubMed] [Google Scholar]
- 105.DiBacco ML et al. Novel ALDH5A1 variants and genotype: Phenotype correlation in SSADH deficiency. Neurology 95, e2675–e2682, doi: 10.1212/WNL.0000000000010730 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Blasi P et al. Structure of human succinic semialdehyde dehydrogenase gene: identification of promoter region and alternatively processed isoforms. Mol Genet Metab 76, 348–362, doi: 10.1016/s1096-7192(02)00105-1 (2002). [DOI] [PubMed] [Google Scholar]
- 107.Pearl PL et al. Succinic semialdehyde dehydrogenase deficiency: lessons from mice and men. J Inherit Metab Dis 32, 343–352, doi: 10.1007/s10545-009-1034-y (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Pearl PL, Wiwattanadittakul N, Roullet JB & Gibson KM in GeneReviews((R)) (eds Adam MP et al.) (1993).
- 109.Pearl PL, DiBacco ML, Roullet JB & Gibson KM Proceedings of the International SSADH Deficiency 2020 Conference. J Child Neurol 36, 1151–1152, doi: 10.1177/08830738211061930 (2021). [DOI] [PubMed] [Google Scholar]
- 110.Schreiber JM et al. A Randomized Controlled Trial of SGS-742, a gamma-aminobutyric acid B (GABA-B) Receptor Antagonist, for Succinic Semialdehyde Dehydrogenase Deficiency. J Child Neurol 36, 1189–1199, doi: 10.1177/08830738211012804 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Lee HHC, Pearl PL & Rotenberg A Enzyme Replacement Therapy for Succinic Semialdehyde Dehydrogenase Deficiency: Relevance in gamma-Aminobutyric Acid Plasticity. J Child Neurol 36, 1200–1209, doi: 10.1177/0883073821993000 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Lee HHC, McGinty GE, Pearl PL & Rotenberg A Understanding the Molecular Mechanisms of Succinic Semialdehyde Dehydrogenase Deficiency (SSADHD): Towards the Development of SSADH-Targeted Medicine. Int J Mol Sci 23, doi: 10.3390/ijms23052606 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Tsuboyama M et al. Transcranial Magnetic Stimulation in Succinic Semialdehyde Dehydrogenase Deficiency: A Measure of Maturational Trajectory of Cortical Excitability. J Child Neurol 36, 1169–1176, doi: 10.1177/08830738211008735 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Pearl PL et al. Succinic Semialdehyde Dehydrogenase Deficiency: Review of the Natural History Study. J Child Neurol 36, 1153–1161, doi: 10.1177/0883073820981262 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Langendorf CG et al. The X-ray crystal structure of Escherichia coli succinic semialdehyde dehydrogenase; structural insights into NADP+/enzyme interactions. PLoS One 5, e9280, doi: 10.1371/journal.pone.0009280 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Kim KJ et al. Succinic semialdehyde dehydrogenase: biochemical-molecular-clinical disease mechanisms, redox regulation, and functional significance. Antioxid Redox Signal 15, 691–718, doi: 10.1089/ars.2010.3470 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Lee HHC et al. Phenotype Reversal in Succinic Semialdehyde Dehydrogenase Deficiency. American Epilepsy Society Annual Meeting Abstract 1.136 (2022). [Google Scholar]
- 118.Liebhardt A et al. Reduced Epilepsy Morbidity and Mortality upon Gene Replacement Therapy in Succinic Semialdehyde Dehydrogenase Deficiency (SSADHD). American Epilepsy Society Annual Meeting Abstract 1.109 (2023). [Google Scholar]
- 119.Bennett MK, Calakos N & Scheller RH Syntaxin: a synaptic protein implicated in docking of synaptic vesicles at presynaptic active zones. Science 257, 255–259, doi: 10.1126/science.1321498 (1992). [DOI] [PubMed] [Google Scholar]
- 120.Lashuel HA, Overk CR, Oueslati A & Masliah E The many faces of alpha-synuclein: from structure and toxicity to therapeutic target. Nat Rev Neurosci 14, 38–48, doi: 10.1038/nrn3406 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Wolfes AC & Dean C The diversity of synaptotagmin isoforms. Curr Opin Neurobiol 63, 198–209, doi: 10.1016/j.conb.2020.04.006 (2020). [DOI] [PubMed] [Google Scholar]
- 122.White DN & Stowell MHB Room for Two: The Synaptophysin/Synaptobrevin Complex. Front Synaptic Neurosci 13, 740318, doi: 10.3389/fnsyn.2021.740318 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Kwon SE & Chapman ER Synaptophysin regulates the kinetics of synaptic vesicle endocytosis in central neurons. Neuron 70, 847–854, doi: 10.1016/j.neuron.2011.04.001 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Mozhayeva MG, Sara Y, Liu X & Kavalali ET Development of vesicle pools during maturation of hippocampal synapses. J Neurosci 22, 654–665, doi: 10.1523/JNEUROSCI.22-03-00654.2002 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Rose T, Schoenenberger P, Jezek K & Oertner TG Developmental refinement of vesicle cycling at Schaffer collateral synapses. Neuron 77, 1109–1121, doi: 10.1016/j.neuron.2013.01.021 (2013). [DOI] [PubMed] [Google Scholar]
- 126.Andreae LC, Fredj NB & Burrone J Independent vesicle pools underlie different modes of release during neuronal development. J Neurosci 32, 1867–1874, doi: 10.1523/JNEUROSCI.5181-11.2012 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Yamashita T, Eguchi K, Saitoh N, von Gersdorff H & Takahashi T Developmental shift to a mechanism of synaptic vesicle endocytosis requiring nanodomain Ca2+. Nat Neurosci 13, 838–844, doi: 10.1038/nn.2576 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Jacob TC, Moss SJ & Jurd R GABA(A) receptor trafficking and its role in the dynamic modulation of neuronal inhibition. Nat Rev Neurosci 9, 331–343, doi: 10.1038/nrn2370 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Laurie DJ, Wisden W & Seeburg PH The distribution of thirteen GABAA receptor subunit mRNAs in the rat brain. III. Embryonic and postnatal development. J Neurosci 12, 4151–4172, doi: 10.1523/JNEUROSCI.12-11-04151.1992 (1992). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Maric D et al. Anatomical gradients in proliferation and differentiation of embryonic rat CNS accessed by buoyant density fractionation: alpha 3, beta 3 and gamma 2 GABAA receptor subunit co-expression by post-mitotic neocortical neurons correlates directly with cell buoyancy. Eur J Neurosci 9, 507–522, doi: 10.1111/j.1460-9568.1997.tb01628.x (1997). [DOI] [PubMed] [Google Scholar]
- 131.Fritschy JM, Paysan J, Enna A & Mohler H Switch in the expression of rat GABAA-receptor subtypes during postnatal development: an immunohistochemical study. J Neurosci 14, 5302–5324, doi: 10.1523/JNEUROSCI.14-09-05302.1994 (1994). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Chen L, Yang C & Mower GD Developmental changes in the expression of GABA(A) receptor subunits (alpha(1), alpha(2), alpha(3)) in the cat visual cortex and the effects of dark rearing. Brain Res Mol Brain Res 88, 135–143, doi: 10.1016/s0169-328x(01)00042-0 (2001). [DOI] [PubMed] [Google Scholar]
- 133.Bosman LW, Heinen K, Spijker S & Brussaard AB Mice lacking the major adult GABAA receptor subtype have normal number of synapses, but retain juvenile IPSC kinetics until adulthood. J Neurophysiol 94, 338–346, doi: 10.1152/jn.00084.2005 (2005). [DOI] [PubMed] [Google Scholar]
- 134.Loup F, Wieser HG, Yonekawa Y, Aguzzi A & Fritschy JM Selective alterations in GABAA receptor subtypes in human temporal lobe epilepsy. J Neurosci 20, 5401–5419, doi: 10.1523/JNEUROSCI.20-14-05401.2000 (2000). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Pearl PL et al. Decreased GABA-A binding on FMZ-PET in succinic semialdehyde dehydrogenase deficiency. Neurology 73, 423–429, doi: 10.1212/WNL.0b013e3181b163a5 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Brickley SG & Mody I Extrasynaptic GABA(A) receptors: their function in the CNS and implications for disease. Neuron 73, 23–34, doi: 10.1016/j.neuron.2011.12.012 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Ben-Ari Y Excitatory actions of gaba during development: the nature of the nurture. Nat Rev Neurosci 3, 728–739, doi: 10.1038/nrn920 (2002). [DOI] [PubMed] [Google Scholar]
- 138.Lee HH, Deeb TZ, Walker JA, Davies PA & Moss SJ NMDA receptor activity downregulates KCC2 resulting in depolarizing GABAA receptor-mediated currents. Nat Neurosci 14, 736–743, doi: 10.1038/nn.2806 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Rivera C et al. The K+/Cl- co-transporter KCC2 renders GABA hyperpolarizing during neuronal maturation. Nature 397, 251–255, doi: 10.1038/16697 (1999). [DOI] [PubMed] [Google Scholar]
- 140.Ganguly K, Schinder AF, Wong ST & Poo M GABA itself promotes the developmental switch of neuronal GABAergic responses from excitation to inhibition. Cell 105, 521–532, doi: 10.1016/s0092-8674(01)00341-5 (2001). [DOI] [PubMed] [Google Scholar]
- 141.Bortone D & Polleux F KCC2 expression promotes the termination of cortical interneuron migration in a voltage-sensitive calcium-dependent manner. Neuron 62, 53–71, doi: 10.1016/j.neuron.2009.01.034 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Palfi S et al. Long-term safety and tolerability of ProSavin, a lentiviral vector-based gene therapy for Parkinson’s disease: a dose escalation, open-label, phase 1/2 trial. Lancet 383, 1138–1146, doi: 10.1016/S0140-6736(13)61939-X (2014). [DOI] [PubMed] [Google Scholar]
- 143.Palfi S et al. Long-Term Follow-Up of a Phase I/II Study of ProSavin, a Lentiviral Vector Gene Therapy for Parkinson’s Disease. Hum Gene Ther Clin Dev 29, 148–155, doi: 10.1089/humc.2018.081 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Ding Z et al. Correction of murine PKU following AAV-mediated intramuscular expression of a complete phenylalanine hydroxylating system. Mol Ther 16, 673–681, doi: 10.1038/mt.2008.17 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Keam SJ Eladocagene Exuparvovec: First Approval. Drugs 82, 1427–1432, doi: 10.1007/s40265-022-01775-3 (2022). [DOI] [PubMed] [Google Scholar]
- 146.Illiano P et al. Recombinant Adeno-Associated Virus-mediated rescue of function in a mouse model of Dopamine Transporter Deficiency Syndrome. Sci Rep 7, 46280, doi: 10.1038/srep46280 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Ng J et al. Gene therapy restores dopamine transporter expression and ameliorates pathology in iPSC and mouse models of infantile parkinsonism. Sci Transl Med 13, doi: 10.1126/scitranslmed.aaw1564 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Mora-Jimenez L et al. Transfer of SCN1A to the brain of adolescent mouse model of Dravet syndrome improves epileptic, motor, and behavioral manifestations. Mol Ther Nucleic Acids 25, 585–602, doi: 10.1016/j.omtn.2021.08.003 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Bohn MC et al. Adenovirus-mediated transgene expression in nonhuman primate brain. Hum Gene Ther 10, 1175–1184, doi: 10.1089/10430349950018166 (1999). [DOI] [PubMed] [Google Scholar]
- 150.Green F et al. Axonal transport of AAV9 in nonhuman primate brain. Gene Ther 23, 520–526, doi: 10.1038/gt.2016.24 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Mathiesen SN, Lock JL, Schoderboeck L, Abraham WC & Hughes SM CNS Transduction Benefits of AAV-PHP.eB over AAV9 Are Dependent on Administration Route and Mouse Strain. Mol Ther Methods Clin Dev 19, 447–458, doi: 10.1016/j.omtm.2020.10.011 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Humbel M et al. Maximizing lentiviral vector gene transfer in the CNS. Gene Ther 28, 75–88, doi: 10.1038/s41434-020-0172-6 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Southgate T, Kroeger KM, Liu C, Lowenstein PR & Castro MG Gene transfer into neural cells in vitro using adenoviral vectors. Curr Protoc Neurosci Chapter 4, Unit 4 23, doi: 10.1002/0471142301.ns0423s45 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Mitani K & Kubo S Adenovirus as an integrating vector. Curr Gene Ther 2, 135–144, doi: 10.2174/1566523024605591 (2002). [DOI] [PubMed] [Google Scholar]
- 155.Sabatino DE & McCarty DM Topics in AAV integration come front and center at ASGCT AAV Integration Roundtable. Mol Ther 29, 3319–3320, doi: 10.1016/j.ymthe.2021.10.024 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Jakobsson J & Lundberg C Lentiviral vectors for use in the central nervous system. Mol Ther 13, 484–493, doi: 10.1016/j.ymthe.2005.11.012 (2006). [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data presented in this review is accessible from public databases PubMed (https://pubmed.ncbi.nlm.nih.gov/) and OMIM (https://www.omim.org/).
