Sigma Institute of Pharmacy, Bakrol, Vadodara 390009
Advances in genome editing technologies have transformed the prospects of treating neurological disorders, a leading global cause of disability and mortality. Programmable nucleases—including zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and, most notably, CRISPR–Cas systems—have enabled precise manipulation of disease-associated genes. Recent innovations such as base editing, prime editing, and RNA-targeting CRISPR variants further extend therapeutic potential, allowing targeted correction of pathogenic mutations, modulation of risk alleles, and reversible gene silencing. These strategies are being explored in neurodegenerative conditions such as Huntington’s disease, amyotrophic lateral sclerosis (ALS), Alzheimer’s disease, and Parkinson’s disease, as well as developmental and neuromuscular disorders like spinal muscular atrophy (SMA) and Rett syndrome. Preclinical studies demonstrate robust efficacy, ranging from restored motor function in SMA models to reduction of amyloid-? in Alzheimer’s disease. Nonetheless, challenges including safe delivery across the blood–brain barrier, off-target effects, immunogenicity, and ethical considerations remain substantial. Delivery innovations—such as engineered adeno-associated viruses (AAVs), lipid nanoparticles, exosomes, and focused ultrasound—offer new opportunities to overcome these barriers. This review critically examines the progress of gene editing for neurological disorders, with emphasis on preclinical advances, delivery strategies, clinical translation, and ethical dimensions. We argue that continued integration of high-fidelity editors, advanced delivery platforms, and rigorous clinical trial design will be essential for realising the promise of durable, one-time therapies for otherwise incurable brain diseases.
Neurological disorders encompass a wide spectrum of conditions affecting the central and peripheral nervous systems, including neurodegenerative diseases (e.g., Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis), neuromuscular syndromes (e.g., muscular dystrophies, spinal muscular atrophy), demyelinating conditions (e.g., multiple sclerosis), epilepsies, and neurodevelopmental syndromes such as Rett syndrome and autism spectrum disorder [1,2]. Collectively, they represent the leading cause of disability worldwide, with the Global Burden of Disease Study 2021 reporting that over 3.4 billion people were affected, contributing to more than 443 million disability-adjusted life years (DALYs) lost [3]. Dementia alone impacts more than 55 million individuals globally, with projections exceeding 150 million by 2050 [4]. These conditions disproportionately affect low- and middle-income countries, reflecting inequities in diagnosis and care [5,6].
Despite advances in symptomatic treatments (e.g., cholinesterase inhibitors for AD, dopamine replacement in PD), most neurological disorders remain intractable and progressive [7]. Conventional pharmacological approaches target downstream pathways but rarely address the underlying genetic or molecular causes.
In recent decades, molecular therapeutics—including antisense oligonucleotides (ASOs), RNA interference (RNAi), and viral vector-based gene supplementation—have provided proof of principle that genetic modulation can alter disease trajectories in neurological disorders [8]. The approval of onasemnogene abeparvovec (Zolgensma) for SMA illustrates the transformative potential of genetic therapies [9]. Yet, these strategies often modulate rather than correct disease-causing genes, require repeated administration, or carry risks of systemic toxicity [10].
Gene editing technologies have emerged as powerful tools capable of addressing these limitations. ZFNs and TALENs introduced the first programmable nucleases for site-specific DNA cleavage, but their complexity restricted broad application [11]. By contrast, the RNA-guided CRISPR–Cas9 system revolutionised genome engineering through its simplicity, efficiency, and adaptability [12]. More recently, precision editors including base editors (enabling single-nucleotide substitutions without double-strand breaks) [13], prime editors (allowing versatile “search-and-replace” genome editing) [14], and RNA-targeting CRISPR enzymes (e.g., Cas13) [15]—have expanded therapeutic possibilities beyond conventional gene disruption. Collectively, these platforms offer opportunities to not only inactivate pathogenic alleles but also repair, silence, or modulate gene expression in disease-relevant neural populations [16,17].
Figure 1 Prime Editing
Figure 2 Delivery Particle Genome
Figure 3 TALENs
This review provides a critical evaluation of the applications of gene editing in neurological disorders, highlighting advances in disease-specific models, innovations in delivery across the blood–brain barrier, and the evolving landscape of preclinical and early clinical trials. Special attention is given to both the promise (long-lasting, one-time curative potential) and the challenges (off-target effects, immunogenicity, ethical constraints) that define the translational path of these therapies.
2. Gene Editing Tools and Neurological Disorders Overview
2.1 Evolution of Genome Editing Technologies
The last three decades have witnessed remarkable advances in genome engineering. Early programmable nucleases—ZFNs and TALENs—enabled targeted double-strand breaks (DSBs) in DNA but required labor-intensive protein engineering [11]. The development of CRISPR–Cas9 in 2012 marked a turning point, allowing RNA-guided targeting with unprecedented simplicity and efficiency [12]. Beyond conventional CRISPR–Cas9, next-generation editors now provide finer control:
These platforms collectively extend therapeutic potential for neurological disorders [17].
2.2 Neurological Disorders: Scope and Burden
Neurological disorders—including AD, PD, HD, ALS, SMA, and Rett syndrome—affect billions globally [1–4]. The global burden continues to rise due to population aging, lifestyle factors, and improved diagnostics. By 2050, dementia cases alone may exceed 150 million [4]. The unmet need for disease-modifying treatments highlights the urgency of developing durable genome-based interventions [5–7].
2.3 Why Gene Editing is Transformative for Neurology
Several factors position gene editing as uniquely suited for neurological disorders:
2.4 Current Limitations and the Path Forward
Despite transformative potential, challenges remain significant:
Nevertheless, the accelerating pace of preclinical advances and the emergence of first-in-human gene-editing trials for neurological disorders underscore the field’s rapid transition from proof-of-concept to clinical reality [22].
3. Applications of Gene Editing in Neurological Disorders
3.1 Huntington’s Disease (HD)
Huntington’s disease (HD) is a progressive autosomal dominant neurodegenerative disorder caused by an abnormal expansion of CAG trinucleotide repeats in the HTT gene, which encodes mutant huntingtin protein [23]. This expansion leads to toxic protein aggregation, selective striatal neuronal loss, and clinical features including motor dysfunction, cognitive decline, and psychiatric symptoms. The monogenic etiology makes HD an attractive target for gene editing–based interventions [24].
Gene Editing Strategies
Several gene editing approaches are under investigation for HD:
Delivery Approaches
Efficient delivery to the central nervous system (CNS) remains a major challenge. Strategies include:
Preclinical and Translational Advances
Figure 4 Huntington's Disease
Figure 5 Gene Editing Tools
Critical Perspective
HD has emerged as a flagship candidate for CRISPR-based therapeutics due to its monogenic nature and well-characterized pathology. Preclinical results are promising, with clear demonstration of both gene correction and functional recovery. However, key challenges remain:
Despite these hurdles, ongoing technological progress in base and prime editing, alongside non-viral delivery innovations, makes HD one of the most promising neurological targets for first-in-human CRISPR therapies.
3.2 Amyotrophic Lateral Sclerosis (ALS)
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder characterized by the progressive degeneration of upper and lower motor neurons, leading to muscle weakness, paralysis, and death typically within 2–5 years of onset [32]. While most ALS cases are sporadic, ~10% are familial, frequently associated with mutations in SOD1, C9orf72, TARDBP, and FUS genes [33]. These mutations drive toxic protein aggregation, RNA foci, and neuroinflammation, making ALS a strong candidate for genetic intervention.
Gene Editing Strategies
Multiple gene editing approaches have been developed to address ALS pathogenesis:
Delivery Approaches
The blood–brain barrier poses a significant barrier to therapeutic access. Strategies include:
Preclinical and Translational Advances
Figure 6 ALS
Critical Perspective
Gene editing offers a promising avenue for ALS therapy, particularly in familial cases with known mutations. CRISPR-mediated SOD1 knockdown has shown consistent efficacy in preclinical models, while Cas13 RNA-targeting approaches may offer a safer, reversible alternative. Base and prime editors introduce precision correction capabilities, though delivery and efficiency remain key barriers.
Clinical translation is advancing rapidly: while ASOs have reached human trials, CRISPR-based ALS therapies remain preclinical, with first-in-human studies expected in the coming years [40]. Challenges include the heterogeneity of ALS genetics, the rapid progression of disease, and the need for early intervention before significant neuronal loss occurs. Nonetheless, ALS represents one of the most active and promising frontiers in neurological gene editing.
3.3 Alzheimer’s Disease (AD)
Alzheimer’s disease (AD) is the most common cause of dementia, affecting more than 55 million people worldwide and projected to surpass 150 million cases by 2050 [41]. Pathologically, AD is characterized by extracellular amyloid-β (Aβ) plaques, intracellular neurofibrillary tangles of hyperphosphorylated tau, synaptic dysfunction, and progressive neuronal loss [42]. Genetic risk factors play a significant role: early-onset familial AD is linked to mutations in APP, PSEN1, and PSEN2, while the APOE ε4 allele is the strongest genetic risk factor for sporadic late-onset AD [43]. These insights position gene editing as a promising approach for both monogenic and multifactorial forms of the disease.
Gene Editing Strategies
Delivery Approaches
Preclinical and Translational Advances
Figure 7 Pathophysiology Of Alzheimer's Disease
Critical Perspective
AD presents a unique challenge compared to monogenic diseases like HD or SMA, given its multifactorial etiology and late-onset progression. Nonetheless, gene editing offers several promising avenues:
Major barriers include the need for broad brain delivery, early intervention before extensive neurodegeneration, and balancing safety with irreversible edits. However, the combination of next-generation editors with emerging CNS delivery technologies positions AD as a leading candidate for translational genome editing research.
3.4 Parkinson’s Disease (PD)
Parkinson’s disease (PD) is the second most common neurodegenerative disorder, characterized by progressive loss of dopaminergic neurons in the substantia nigra, accumulation of α-synuclein aggregates (Lewy bodies), and motor symptoms including tremor, rigidity, and bradykinesia [52]. Genetic contributors include mutations in SNCA (α-synuclein), LRRK2, PARK7, PINK1, and GBA, making PD partially amenable to gene editing interventions [53].
Gene Editing Strategies
Preclinical Advances
Critical Perspective
Gene editing in PD offers both disease-modifying (mutation correction) and symptomatic (neuroprotective) strategies. However, heterogeneity of sporadic PD limits broad application. Promisingly, SNCA silencing and LRRK2 correction are advancing toward translational relevance.
3.5 Spinal Muscular Atrophy (SMA)
SMA is a neuromuscular disorder caused by loss-of-function mutations in SMN1, leading to reduced survival motor neuron (SMN) protein and motor neuron degeneration [58]. SMA is a monogenic disease, making it highly suitable for gene therapy and editing.
Gene Editing Strategies
Preclinical Advances
Figure 8 SMA
Figure 9 Pathophysiology Of SMA
3.6 Rett Syndrome
Rett syndrome is an X-linked neurodevelopmental disorder caused primarily by mutations in MECP2, leading to impaired synaptic function, intellectual disability, and autistic features [62].
Gene Editing Strategies
Preclinical Advances