Abstract
The clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system is an acquired immune system of many bacteria and archaea, comprising CRISPR loci, Cas genes, and its associated proteins. This system can recognize exogenous DNA and utilize the Cas9 protein's nuclease activity to break DNA double-strand and to achieve base insertion or deletion by subsequent DNA repair. In recent years, multiple laboratory and clinical studies have revealed the therapeutic role of the CRISPR/Cas9 system in neurological diseases. This article reviews the CRISPR/Cas9-mediated gene editing technology and its potential for clinical application against neurological diseases.
Keywords: CRISPR/Cas9, Gene editing, Mutation, Neurological diseases, Therapeutics
Introduction
CRISPR/Cas9 system belongs to the third generation of gene editing technology after zinc finger nucleases, transcription activation-like effect nucleases, and other techniques.1 CRISPR/Cas9 become the focus of current research with its simple and efficient features. This system allows DNA double-strand to break at corresponding targets by the nuclease activity of Cas9 proteins.2 Multiple gene editing techniques have been developed based on this principle, including gene knock-out, gene knock-in, transcriptional regulation, and base editing.3, 4, 5, 6 The basic principle of the CRISPR/Cas9 system is to identify the target gene by an artificially designed single guide RNA (sgRNA). The sgRNA guides the cleavage of DNA double strands by Cas9 and forms double-strand breaks (DSB) at the target site, after the DNA damage spontaneous repair occurs, during which gene knock-out or knock-in can be achieved.7
Nervous system diseases have complex etiology, are relatively difficult to treat, and seriously affect patients' quality of life. WHO predicts that by 2050, neurodegenerative diseases will replace cancer as the leading cause of death in humans.8 The CRISPR/Cas9 gene-editing technology can effectively treat neurological diseases, as confirmed by recent studies demonstrating the therapeutic value of the CRISPR/Cas9 system in Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), glioblastoma (GBM), epilepsy, amyotrophic lateral sclerosis (ALS), etc.9, 10, 11, 12, 13, 14
This review expounds on the mechanism of the CRISPR/Cas9 system and its mediated gene editing technology (Fig. 1). It summarizes its research progress associated with the pathogenesis and treatment of nervous system diseases and will provide new ideas for its prevention and treatment (Table 1).
Figure 1.
Mechanism of action of the CRISPR/Cas9 system. The process of the CRISPR/Cas9 system can be summarized in three stages: adaptation, expression, and interference. In the adaptation phase, bacteria capture the invading exogenous DNA as a pre-spacer sequence. Then, at the expression stage, bacteria produce crRNA-tracrRNA-Cas9 triplet complex by transcription. Finally, during the interference phase, when exogenous DNA invades again, the bacteria specifically recognize and eliminate the invaders. CRISPR, clustered regularly interspaced short palindromic repeats; Cas1(2,9), CRISPR associated-protein1 (2,9); PAM, protospacer-adjacent motif; pre-spacer, precursor spacer; crRNA, CRISPR RNA; pre-crRNA, pre-CRISPR RNA; tracrRNA, trans-activating crRNA; DSB, double-strand breaks.
Table 1.
Applications of the CRISPR/Cas9 system in nervous system diseases.
Pathology | Target gene | Model | Result | Reference |
---|---|---|---|---|
AD | Bace1 gene | 5XFAD mouse, APP knock-in (KI) mouse | APP↓, Aβ↓ | 68 |
APP gene | Mouse | Aβ↓ | 69 | |
APOE4 gene | Mouse | APOE4 allele→APOE3 allele | 56 | |
iPSCs | Tau hyperphosphorylation↓ | 73,74 | ||
ATP6V1 A gene | iPSCs | Neuronal activity↓ | 79 | |
PD | Vps35 D620N | Mouse | Mouse model of PD | 87 |
α-synuclein gene | Pig | Pig model of PD | 88 | |
Not mentioned | Monkey | Monkey model of PD | 90 | |
DNAJC6 | ESCs | Stem cell model of PD | 91,92 | |
Not mentioned | Human pluripotent stem cells | Human brain-like organoids model of PD | 93 | |
SNCA gene | Mouse | SNCA↓, α-synuclein↓ | 95,96 | |
VPS59 D620N | Cell | Mitosis↓ | 99 | |
HD | HHT gene | Pig | Pig model of HD | 107 |
HHT gene | HD140Q-Ki mice | Early neuropathological changes in the striatum↓ | 109 | |
GBM | TLE4, IKZF2, EIF5A, TMEM184B | CAR-T cell | Death of CAR-T cells↓ | 116 |
relA/p65, NPLOC4 | GBM cell | Death of GBM cells↑ | 116 | |
Gene of IFNγR signaling pathway | GBM cell | Drug resistance↑ | 117 | |
Epilepsy | KCNA1 gene | Mouse | Kv1.1↑, frequency of seizures↓ | 124 |
KCNB1 gene | Mouse | Kv2.1↑, abnormal EEG activity↑ | 127 | |
SCN1A gene | Mouse | Nav1.1↑, epileptic symptoms↓ | 131 | |
ALS | C9orf72 | Mouse | Synaptic dysfunction↓ | 143 |
SOD1 gene | SOD1G93A mutant neonatal mice | Quantity of motor neurons↑ Muscle strength↑ Survival rate↑ |
147,148 |
Structure of CRISPR/Cas9 system
CRISPR gene
The CRISPR gene is a repetitive sequence in the prokaryotic genome that is distributed in 40% of sequenced bacteria and 90% of sequenced archaea.15 CRISPR gene sequence mainly comprises leader, repeat, and spacer.16, 17, 18 The leader sequence is located upstream of the CRISPR gene, is relatively simple with high AT content, and acts as a promoter. The repeats are a palindromic sequence of about 20–50 bp long. The transcripts of the repeats form hairpin-like structures essential for maintaining RNA structure and functional integrity. The spacer sequence is an exogenous DNA sequence captured by bacteria, making it the product of bacterial adaptive immunity. When the bacteria detect these exogenous DNA sequences, the CRISPR/Cas system acts as a part of the immune system.
Cas gene and Cas9 protein
The Cas gene is located adjacent to the CRISPR gene, but a portion of the Cas is also discretely distributed elsewhere in the genome. Cas, the encoded product of the Cas gene, is a protein with nuclease activity and is a key component in bacterial autonomous immunity that binds to DNA at its corresponding targets. Based on the different roles of Cas in the immune mechanism, CRISPR/Cas system is divided into two categories. In the first category, Cas (type I, III, and IV) act through a cascade amplification mechanism, which usually produces the Cas complex. In the second category, Cas does not rely on a cascade amplification mechanism, and only a single Cas is involved, including type II and type V. Cas9 belongs to type II.19
Mechanism of CRISPR/Cas9 system
Adaptation
The first step is “adaptation”, which involves the acquisition of a spacer sequence. Phage or plasmid-derived exogenous DNA is integrated into the phosphate group of the CRISPR gene when bacteria are attacked.20 Therefore, it can be speculated that the position of the phosphate group end to the base end in the spacer sequence is consistent with the temporal order of exogenous DNA invasion. Protospacer adjacent motif (PAM) is a hallmark of the pre-spacer sequence and can be used to distinguish exogenous DNA and CRISPR genes.21 Cas1 and Cas2 are the critical proteins that take part in the adaptation stage. To perform their function, both proteins usually polymerize to form a Cas1-Cas2 complex.22,23 The Cas1-Cas2 complex cleaves the DNA sequence near PAM by utilizing it as the recognition object. The original spacer sequence is then inserted downstream of the leader sequence, and finally, the DNA completes the spontaneous repair. During the adaptation phase, bacteria generate specific immunity against the exogenous DNA, therefore when it encounters invaders containing the same genetic material again; it specifically identifies and destroys these invaders.
Expression
The leader sequence contains the promoter and spacer sequence integration elements of the CRISPR/Cas9 system. When the intruder is identified, the leader sequence regulates the CRISPR gene transcription to synthesize base-complementary pre-CRISPR RNA (pre-crRNA) and trans-activating crRNA (tracrRNA).24 Pre-crRNA, tracrRNA, and Cas9 constitute a complex. Subsequently, the pre-crRNA is sliced into smaller units by RNase III, namely CRISPR RNA (crRNA). crRNA corresponds to the corresponding spacer sequence and guides Cas9 to target the DNA cleavage site.25 crRNA-tracrRNA-Cas9 triplet complex is the basic structural and functional unit of the CRISPR/Cas9 system.26
Interference
PAM of exogenous DNA is first recognized by the Cas1-Cas2 complex. Subsequently, crRNA further guides the binding of the crRNA-tracrRNA-Cas9 triple complex to this exogenous DNA according to the principle of base complementary pairing. The crRNA then cooperates with a matching complement (protospacer) on an invading DNA to form the R-loop.27 In the R-loop structure, crRNA binds to one strand by complementary base pairing, and the other single strand maintains a free state. At this time, Cas9 exerts its endonuclease activity, generates a target cleavage site 3 nucleotides upstream of PAM, and forms a DSB, thereby eliminating the exogenous after a series of complex actions.7 The six domains of Cas9 have different mechanisms and functions. The NHN domain and the RuvC domain are responsible for targeted DNA cleavage. The NHN domain cuts the complementary DNA strands, regulated by single metal ions, and the RuvC domain cuts the non-complementary DNA strands, regulated by bimetallic ions.7,28
Gene editing technology based on CRISPR/Cas9 system
The sgRNA is a synthetic T-shaped RNA that guides Cas9 to bind the target DNA and can achieve editing for different genes instead of the crRNA.29,30 The basic mechanism of the CRISPR/Cas9 system involves guiding Cas9 to cleave DNA double strands after sgRNA recognizes the target gene and forms DSB at the corresponding site. After DNA damage, gene knock-out or knock-in occurs in the process of spontaneous repair, ultimately achieving gene editing.
Knock-out
Non-homologous end joining (NHEJ) is the most efficient and common cellular DNA repair mechanism.31 NHEJ repairs DSB by directly connecting the broken ends and performing minimal DNA end treatment.32 Upon NHEJ activation, bases are randomly inserted or deleted at the DNA ends, causing a complete genetic code disruption downstream of the DSB and stopping the target gene expression.32 Although the frequency of NHEJ occurrence is relatively high, its low accuracy often causes non-target editing, which greatly limits its application. To achieve precise target gene knockout, mutations in the NHN or the RuvC domains can be artificially produced. This allows Cas9 to act on only one DNA strand. To achieve DSB, two different sgRNA are designed to target DNA double strands, which significantly improves gene knockout specificity. It has been shown that the size of the Cas9-mediated deletion and its frequency are negatively correlated.33 For Cas9-induced DSBs, first, the PAM proximal end of DSB is released by Cas9, while its distal end remains attached to Cas9 for a long period.34,35 Ultimately, the PAM proximal DSB end is exposed for repair treatment, while the distal end is not repaired temporarily due to Cas9 binding. Cas9 can be removed from the distal end of PAM by RNA polymerase, but the specific mechanism is not clear.36
Knock-in
Homology-directed repair (HDR) is another DNA repair mechanism, which results in gene insertion. Unlike NHEJ, cells perform HDR using repair templates. DNA reconstruction occurs at the DSB site based on the repair template. HDR is a high-fidelity repair pathway, and repair templates often include genes that require the insertion of homologous sequences at the insertion site. Homologous sequence matching is a primary condition for HDR.31,37,38 Repair templates can either be endogenous DNA or artificially introduced exogenous genetic material such as plasmids.39 The incidence of HDR is much lower than NHEJ, but it has higher accuracy. Compared with NHEJ, HDR is restricted to the S/G2 phase of the cell cycle,40 which provides a theoretical basis for improving HDR efficiency. NHEJ inhibitors or HDR enhancers can be used to improve HDR efficiency, and the rationale for inhibitors or enhancers is to limit cell division to the S/G2 phase with increased HD, allowing the cells to undergo DNA repair by HDR rather than NHEJ.41,42 Furthermore, that chemical modification has been indicated to increase HDR incidence.43,44
Transcriptional regulation
Due to the poor specificity, Cas9 is more prone to off-target effects, increasing the possibility of non-target mutations45; to overcome this, dead Cas9 (dCas9) mutates the two domains NHN and RuvC to lose DNA cleavage activity,46 while retaining the ability to fuse to DNA mediated by sgRNA. The dCas9 can fuse to the transcriptional activation domain (ADS), including VP64, P65, and RTA, to stimulate transcriptional activation.47, 48, 49 Additionally, dCas9 inhibits transcription and gene expression by fusing to the promoter region or the transcriptional repressor Krab-dCas9.50 Thus, dCas9 differs from Cas9 as dCas9 stimulated transcriptional activation or repression is transient, reversible, and does not cause a permanent alteration or DNA genomic damage. Recently, researchers have constructed a dimer complex in which dCas9 fuses to FokI (dCas9-FokI) and achieve transcriptional regulation mediated by RNA; this has minimum off-target effects.51
Base editing
Single-base pair mutations are the leading causes of many inherited and acquired diseases.52 Gene editing both by HDR and NHEJ pathways has less specificity and efficiency, and once the mutations occur, they can cause great damage to the human body.53 Moreover, due to the specificity of HDR and NHEJ mechanisms, their mediated gene editing can only be applied to dividing cells, thus limiting the scope of disease treatment.54 Recently, a new Cas9-mediated base editing tool was introduced, which is of two types, cytosine base editing molecule (CBES) and adenine base editing molecule (ABES).52,55 The base editing tool consists of two key components: dCas9 which binds to DNA and enzymes that target base changes. CBES tool combines rat-derived cytosine deaminase apolipoprotein B mRNA editing enzyme catalytic subunit 1 (APOBEC1) with dCas9 for gene editing,56 whereas ABES combines deoxyadenosine deaminase with dCas9 for base editing.57 CBES and ABES can achieve four changes (A-G, G-A, C-T, and T-C) to achieve a single base editing.56,58 The efficiency of ABES in human cells is about 50%, while that of CBES remains unknown.57
Epigenome editing
The conventional CRISPR/Cas9 system can produce an irreversible DNA sequence alteration. Moreover, the overreliance on endogenous DNA repair mechanisms also increases the complexity and uncontrollability of artificial operations to a large extent. Recently, an epigenetic editor called CRISPRoff was reported. CRISPRoff methylates specific DNA fragments as guided by sgRNA. These methylated genes are then silenced or shut down, thereby epigenetic gene silencing is achieved.59 CRISPRoff does not alter the DNA sequence and is not limited by endogenous DNA repair mechanisms. Furthermore, CRISPRoff-stimulating methylation is reversible as it can be removed by an epigenetic editor called CRISPRon. It is worth noting that CRISPRoff can silence genes lacking CpG islands (CGIs), which were previously considered essential for DNA methylation.
CAPTURE
CRISPR affinity purification in situ of regulatory elements (CAPTURE) is a derivative technology of the CRISPR/Cas9 system. CAPTURE comprises specific sgRNA, biotin-labeled dCas9 (FB-dCas9), and biotin ligase BirA. Briefly, CAPTURE uses sgRNA to guide FB-dCas9 to the target DNA sequence element and then separates and purifies proteins, DNA, RNA, and other macromolecules at the corresponding sites by high-affinity streptomycin.60 It provides a new method for studying the function and specific regulatory factors of DNA non-coding regions. At present, CAPTURE has played a huge role in the regulation mechanism of the snRNA gene, mouse embryonic stem cells, and other fields.61,62 It can also provide new paths to discover novel drug targets.
Applications of CRISP/Cas9 system in nervous system diseases
Alzheimer's disease
One of the most common neurodegenerative diseases is AD, characterized by senile plaques [abnormal accumulation of extracellular amyloid-β (Aβ)] and neurofibrillary tangles (abnormal hyper-phosphorylation of intracellular tau protein).63 AD is also associated with extensive neuronal loss, hippocampal pyramidal cell granule vacuolar degeneration, and other pathological changes. No effective radical cure nor an efficient strategy to reverse AD progression exists. Traditional drug therapy is relatively effective over a long period. However, as the disease progresses, the effect of drug treatment decreases significantly. CRISPR/Cas9 system provides a new door for identifying novel ideas and methods for treating AD.
The metabolic abnormality of Aβ is manifested by the imbalance between the production and elimination of Aβ, subsequently causing its abnormal accumulation.64 Aβ is produced by amyloid precursor protein (APP) catalyzed by β-secretase 1 (Bace1) and γ-secretase, where Bace1 plays a key role. Therefore, any link of this metabolic pathway can reduce the content of Aβ, thereby reducing the pathological damage of AD.65 The CRISPR/Cas9 system has been used to model AD in a variety of cells in vitro,66,67 however, targeted editing of highly differentiated cells such as neurons has been difficult, making in vivo AD studies difficult. Recently, Park et al used CRISPR/Cas9 nano-complex (Cas9-sgRNA supplemented with two amphiphilic R7L10 peptides in different proportions) to introduce sgRNA containing Bace1 gene targeting sites into five familial Alzheimer's disease (5XFAD) and APP knock-in AD mouse models, respectively. A significant reduction was noted in Bace1 and Aβ. In addition, treatment mice performed better in the water maze test compared with control mice, indicating a shorter escape time to find the platform.68 These results indicate that CRISPR/Cas9 nano-complexes can effectively reduce the pathological manifestations and cognitive impairment caused by AD. As the upstream protein of Aβ, the expression of APP directly affects Aβ content. Knocking out APP allele by CRISPR/Cas9 can also reduce Aβ expression.69
Age is the most dangerous pathogenic factor of AD. Based on age and onset, AD is divided into early-onset AD (EOAD) and late-onset AD (LOAD, by 65 years old), where LOAD is more common.70 The Apolipoprotein E (APOE) gene is an important risk factor for LOAD. APOE has three subtypes: APOE2, APOE3, and APOE4. The APOE4 allele greatly increases the risk of carriers, whereas APOE2 has a protective effect.71,72 Using CRISPR/Cas9, Komor et al successfully transformed APOE4 into the APOE3 allele in mouse astrocytes by generating a C-T transformation at codon 158 via base editing techniques.56 Lin et al established induced pluripotent stem cells (iPSCs) comprising homozygous APOE4 alleles using CRISPR/Cas9 and demonstrated that the conversion of APOE4 to APOE3 can greatly improve AD-related pathological features.73 Based on this mechanism, studies have shown that the conversion of APOE4 to APOE3 can also reduce tau hyper-phosphorylation to some extent.74
Most current AD therapeutic studies are focused on AD-related proteins and genes such as APP and Aβ.75, 76, 77, 78 Wang et al conducted a large data comprehensive network analysis of multiple LOAD samples and found that neuronal gene subnetwork dysregulation was the main pathogenic factor of LOAD and identified ATP6V1 A as a key regulator. Wang et al targeted ATP6V1A genes by using dCas9 and found that its expression was reduced while neuronal activity was inhibited. This network modeling strategy provides a novel perspective on AD treatment.79
Parkinson's disease
Tremor paralysis or PD is a common neurodegenerative disease of the middle-aged and elderly and is ranked the second most common neurodegenerative disease globally.80 The main clinical manifestations of PD are static tremors, bradykinesia, myotonia, and postural imbalance.81 The major pathological changes associated with PD include the degenerative loss of the dopaminergic neurons in the substantia nigra and the emergence of eosinophilic inclusions, namely the Lewy body.82 Approximately 10% of all PD patients are familial and 90% are sporadic. Genetic susceptibility is the main reason in the familial type of PD, where the SNCA and LRRK2 gene mutations are autosomal dominant, and Parkin, PINK1, and DJ-1 gene mutations are autosomal recessive.83,84 The cause of sporadic AD is undetermined, it is presumed to be related to oxidative stress and other factors.85
The construction of the PD model has always been complex and difficult. The construction of traditional animal models has defects such as long cycles, low success rates, and low specificity.86 The CRISPR/Cas9 system, with its precise targeted editing function, provides conditions for animal modeling. Ishizu et al used the CRISPR/Cas9 technique to knock in the Vps35 D620N allele in mice that exhibited age-related substantia nigra-striatum neurodegeneration and had positive features similar to PD.87 Zhu et al successfully created the PD pig model by combining CRISPR/Cas9 with somatic cell nuclear transfer (SCNT).88 PINK1 gene mutations are the main cause of early-onset PD.89 However, neither the current PINK1 knock-out mouse model nor the pig model has fully demonstrated the typical PD-associated pathological changes. Chen et al injected sgRNA and Cas9-D10A nicking enzyme into zygotes of cynomolgus monkeys to establish a typical PD neuro-degenerated primate model.90 Cell models also help study the pathogenic mechanisms and molecular pathological alterations in PD. Wulansari et al established DNAJC6 gene mutation in human embryonic stem cells and observed the key pathological features of PD including dopaminergic neuronal degeneration and abnormal aggregation of α-synuclein91. Dolatabadi et al also constructed similar cellular models.92 Furthermore, organoid models have been reported. Jo et al used human pluripotent stem cells and CRISPR/Cas9 genetic engineering to construct human brain-like organoids in a 3D culture system, producing human PD-specific Louis bodies in organoids.93 Organoids provide a new idea for the construction of in vitro PD models.
SNCA gene mutation, specifically SNCA hypermethylation, critically participates in the pathogenesis of PD.94 Guhathakurta et al revealed that histone H3 lysine 4 trimethylation (H3K4me3) was recruited in the SNCA promoter of PD patients. The team used dCas9-mediated demethylase to specifically block SNCA promoter expression, thereby down-regulating α-synuclein.95 Gene knockout can also mitigate PD damage. Targeted knockout at A53T mutation sites on SNCA by CRISPR/Cas9 technique significantly reduced SNCA expression both in vitro and in vivo.96
Oxidative stress and mitochondrial damage have also been implicated as major factors in PD pathogenesis.97 PTEN-induced putative kinase 1 (PINK1) protein is important for the maintenance of normal morphology and mitochondrial function. The reduction or deletion of PINK1 disrupts the redox balance and promotes PD development.98 Ma et al constructed a PD cell model with the D620N mutation in VPS59, which ultimately affected PINK1-mediated mitosis,99 suggesting that the study of oxidative stress mechanism may be a breakthrough in the treatment of sporadic PD.
In addition to studying the bio-molecular mechanisms of PD, the CRISPR/Cas9 system can also be used for screening the therapeutic drugs against PD. Basu et al inserted a luciferase gene into SNCA by CRISPR/Cas9 to measure α-synuclein expression in living cells.100 This technology can achieve high-throughput drug screening, which greatly improves the efficiency of drug development and testing for PD or other neurodegenerative disorders.
Huntington's disease
Huntington's disease, also known as Huntington's chorea, is an inherited autosomal dominant disorder of the central nervous system. It is caused by an abnormal expansion of the CAG repeat of the Huntington's protein (HTT) gene on chromosome 4, resulting in a toxic gain of function.101,102 The severity of the clinical symptoms is proportional to the CAG copy number. No effective treatment for HD exists. Based on the characteristics of HD single gene inheritance, the CRISPR/Cas9 system is expected to become a promising treatment method.103
Animal models allow the study of pathogenesis and pathological changes associated with HD, therefore, rodents such as mice are commonly used as disease models.104,105 However, although these models can express mutant HHT, it lacks the neurodegeneration typical of HD and fails to comprehensively mimic the manifestation of human HD development at both molecular and behavioral phenotype levels.104,106 Yan et al have successfully developed the world's first HD knock-in pig model via CRISPR/Cas9 and inserted the CAG repeat sequence into the pig HHT gene. This model revealed typical pathological manifestations of striatal medium spiny neuron degeneration and dance-like involuntary movement in the behavioral phenotype. More importantly, these characteristics exhibit age-related similarities to humans and can be stably transmitted through the reproductive system.107 The study provides a basis for animal models of other neurological diseases.
CAG repeats knockout in the HHT gene using CRISPR/Cas9 reduced HHT expression.108,109 Notably, previous studies suggest that the HHT gene knockout by CRISPR/Cas9 causes mice death (not in adults) from acute pancreatitis.110 In a later study, Yang et al knocked out the HHT gene in HD140Q-Ki mice, reducing early neuropathological changes in the mouse striatum.109
Most current HD treatments mainly target the disease remission after onset. However, 50% of the striatal volume disappears before the symptoms even appear.111 Therefore, early detection and treatment before the onset are very critical. Liu et al used an inflow-based vascular-space-occupancy (IVASO) MRI to map the time-varying model of cerebral blood volume (CBVa) in small arteries of zQ175 mice. HTT in zQ175 mice was mutated and progressed slowly. As the disease progressed, CBVa gradually increased. This study revealed that mice treated with HTT sgRNA and Cas9 lacked striatal atrophy and behavioral phenotype alterations compared with the control group.112 Therefore, the CBVa model can be used as a sensitive noninvasive indicator for early intervention in HD.
Glioblastoma
Glioblastoma is a neuroepithelial tissue-derived neurodegenerative disorder. It is the most common, aggressive, and least-prognostic malignancy of the central nervous system.113 Currently, there are no effective treatments for GBM. Surgery combined with chemoradiotherapy is usually the therapeutic strategy applied, which only prolongs the median survival time of GBM by < 3 months. Chimeric antigen receptor T cell therapy (CAR-T) is a method that allows T cells to express the tumor chimeric antigen receptor (CAR) through gene editing technology to specifically identify and kill tumor cells.114 CAR-T is effective for the treatment of hematological malignancies but is relatively insensitive for solid tumors.115 Therefore, it is important to find therapeutic strategies for these tumors. Wang et al established a new high-throughput screening platform for high-throughput CRISPR gene knockout screening in CAR-T and GBM cells, respectively. The team identified four sites TLE4, IKZF2, EIF5A, and TMEM184B in CAR-T cells, and two relA/p65 and NPLOC4 in GBM cells. Knocking out these genes increased the GBM cell killing and reduced the death of CAR-T cells in vitro and in vivo.116 Furthermore, reducing GBM resistance to cell therapy is also one of the CAR-T strategies. Larson et al used CRISPR for genome-wide screening of GBM and found that gene deletion in the IFNγR signaling pathway increases drug resistance in GBM, which is associated with reduced adhesion of GBM cells to CAR-T cells.117
CRISPR/Cas9 is also used for targeted drug therapy of GBM. In the past, CRISPR/Cas9 therapy for tumors had low specificity, low editing efficiency, and drug resistance; however, Rosenblum et al reported a method for targeted delivery using lipid nanoparticles (LNP) coated with CRISPR/Cas9 (CRISPR-LNP). In vivo, intracerebral CRISPR-LNP injection in GBM mice reduced tumor volume by 50% and significantly improved mouse survival.118
With high efficiency for targeting tumor cells and extremely low drug resistance, CRISPR/Cas9 gene editing technology allow new avenues for GBM treatment, its proper use can spare patients the pain caused by radiotherapy and chemotherapy. Moreover, it provides new possibilities for treating malignant pan-tumors.
Epilepsy
Epilepsy is caused by the abnormal synchronized discharge of brain neurons; it is a clinical syndrome rather than an independent disease. The etiology of epilepsy is very complex, it can be caused by gene mutations or occur secondary to various neurological diseases, such as stroke, brain trauma, brain tumors, intracranial infections, etc.119,120 Treatment for epilepsy remains to be established. Drug therapy is not effective because of its high drug resistance; however, gene therapy may be a powerful new tool for its treatment.
Ion channels maintain normal neuronal structure and function.121 Mutation in genes encoding ion channels leading their abnormality, an important cause of epileptic seizures. Therefore, studying ion channel-related genes can be a key to overcoming epilepsy.122 Exogenous overexpression of KCNA1 reduces neuronal excitability,123 however, its effect on neurons remains unknown. Colasante et al combined a dCas9 targeting the promoter region of KCNA1 (encoding Kv1.1) and a sgRNA to construct the CRISPRa system (KCNA1-dCas9A).124 In the mice temporal lobe epilepsy model, KCNA1-dCas9A up-regulated Kv1.1 expression by activating the KCNA1 gene and reduced neuronal excitability and seizure frequency. More importantly, KCNA1-dCas9A also improved cognitive deficits in mice.124 However, the consistency of the temporal lobe epilepsy model with other types of models in terms of gene expression patterns remains unconfirmed.125,126 KCNB1 (encoding Kv2.1) is the causative gene of developmental and epileptic encephalopathies (DEE). Unlike KCNA1, mutations in KCNB1 can trigger epilepsy. Hawkins et al used CRISPR/Cas9 to knock out KCNB1 in mice and observed typical DEE manifestations such as abnormal EEG activity and hyperactivity.127 The model can be used for drug evaluation and screening to solve the problem of drug-resistant DEE.
Dravet syndrome (DS) is a hereditary epileptic encephalopathy. It is commonly triggered by a fever.128,129 Although DS is generally defined as a disease independent of epilepsy, the two are usually discussed together because of their similar pathogenesis and clinical manifestations. DS is also closely related to ion channels. The heterozygous deletion mutation of the SCN1A gene (encoding Nav1.1) is the main cause of DS.130 Colasante et al applied SCN1A-dCas9A to SCN1A mutant mice and revealed that SCN1A stimulation increased Nav1.1 levels and the excitability of intermediate inhibitory neurons. The epileptic symptoms of SCN1A-activated mice were also significantly reduced compared with the control group,131 suggesting a strategy to rescue DS and other fever-induced seizures.
Zebrafish are excellent animal models for studying gene function but are rarely utilized for human neurological diseases. Griffin et al established 40 zebrafish lines with childhood epilepsy via CRISPR/Cas9 and identified eight single-gene phenotypes after electrophysiological screening.132 However, this model cannot summarize the typical human epilepsy phenotype, which may be related to multiple epilepsy etiological mechanisms, such as multiple gene mutations, environmental factors, etc.133,134
Amyotrophic lateral sclerosis
Amyotrophic lateral sclerosis is characterized by the progressive loss of motor neurons in the central nervous system,135 it is also the most common motor neuron disease characterized by progressive muscle weakness that eventually leads to respiratory failure and death.135 Multiple genetic loci are associated with ALS, such as the C9orf72, SOD1, TARDBP, and FUS genes.136, 137, 138, 139 The CRISPR/Cas9 system is effective against these gene loci in vitro and in vivo.
A hexanucleotide repeat expansion of the C9orf72 sequence is the most critical and common cause of ALS, specifically the G4C2 repeat. Mutations cause a dipeptide protein clearance disorder, and excessive accumulation, which in turn produces neurotoxicity.140 A study showed that when IPSCs were inhibited by CRISPR/Cas9 in C9orf72 patients, dipeptide levels reduced dramatically.141 At the transcriptional level, CRISPR/Cas9 knockout of G4C2 repeats reduces RNA transcription and nuclear RNA foci.142 It was revealed that reducing the G4C2 repeat sequence can protect neurons. C9orf72 mutation causes synaptic neuronal dysfunction. In a recent study, Perkins et al used CRISPR/Cas9 to correct the C9orf72 mutation and successfully rescued synaptic dysfunction,143 providing new insights into the early mechanisms of ALS and new options for drug targets.
The SOD1 gene, located on chromosome 21, is the first and the most studied ALS-related gene discovered.144 The copper-zinc superoxide dismutase encoded by the SOD1 gene is very important for maintaining normal redox balance in cells. The mutation of the SOD1 gene can lead to mitochondrial dysfunction, oxidative stress, abnormal protein accumulation, cell inflammation, etc., and then promote ALS incidence and development.145,146 In a previous study, Staphylococcus aureus-derived Cas9 (SaCas9) and sgRNA targeting the SOD1 gene were injected via the facial vein in SOD1G93A mutant neonatal mice mediated by AVV9. The results revealed that mutant SOD1 significantly reduced, the number of motor neurons increased significantly, and the motor function was effectively protected.147 Based on this study, Duan et al injected a high dose (3.3 times the normal dose) of SaCas9 to observe the phenotype of SaCas9-modified mice and found that mice had increased muscle strength, reduced muscle atrophy, and significantly improved survival.148 The above two studies demonstrate the potential of SaCas9 to correct ALS both in neuropathology and in clinical phenotype. However, the small carrying capacity of AAV limits the SaCas9's clinical application. Lim et al invented a method to intrathecally inject AVV and CBES encoding split introns into SOD1G93A mice and observed reduced muscle atrophy and denervation.149 The CRISPR base editor largely solves the problem of AVV's limited carrying capacity, making it possible to use CRISPR/Cas9 for the clinical treatment of ALS.
Conclusions
As researchers delve deeper into the CRISPR/Cas9 system, its potential is being tapped continuously. The advent of the CRISPR/Cas9 system has greatly facilitated the study of neurological diseases. The application of CRISPR/Cas9 gene editing technology has allowed us to achieve more precise modification of pathogenic genes. It can also be used to establish cellular or animal models that closely resemble human disease phenotypes. At the neuropathological level, the application of CRISPR/Cas9 more clearly reveals the molecular mechanisms of the disease. Its application also provides new ideas for the treatment of other types of diseases, such as AIDS, malignant tumors, etc. However, there are still some shortcomings in this technology, such as the off-target effect, the specific sgRNA mechanism, and the immunity of the human body toward Cas9. The biggest concern is the potential ethical issues posed by CRISPR/Cas9. If the technology is misapplied to the human reproductive system, it can have unimaginable consequences.
Author contributions
All authors made a significant contribution to this review in the conception, study design, execution, acquisition of data, analysis, and interpretation; took part in drafting, revising, or critically reviewing the article; gave final approval of the version to be published; agreed on the journal to which the article has been submitted; and agreed to be accountable for all aspects of the work.
Conflict of interests
The authors report no conflict of interests in this work.
Funding
This study was funded by the National Natural Science Foundation of China (No. 82271747).
Footnotes
Peer review under responsibility of Chongqing Medical University.
Contributor Information
Jianghu Zhu, Email: zhujianghu@wmu.edu.cn.
Zhenlang Lin, Email: lzlprof2020@163.com.
Min Zhang, Email: zmzhangmin2018@163.com.
References
- 1.Zhang H.X., Zhang Y., Yin H. Genome editing with mRNA encoding ZFN, TALEN, and Cas9. Mol Ther. 2019;27(4):735–746. doi: 10.1016/j.ymthe.2019.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Mojica F.J.M., Díez-Villaseñor C., García-Martínez J., et al. Short motif sequences determine the targets of the prokaryotic CRISPR defence system. Microbiology (Read) 2009;155(Pt 3):733–740. doi: 10.1099/mic.0.023960-0. [DOI] [PubMed] [Google Scholar]
- 3.Zhu S., Li W., Liu J., et al. Genome-scale deletion screening of human long non-coding RNAs using a paired-guide RNA CRISPR-Cas9 library. Nat Biotechnol. 2016;34(12):1279–1286. doi: 10.1038/nbt.3715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Li J., Meng X., Zong Y., et al. Gene replacements and insertions in rice by intron targeting using CRISPR-Cas9. Native Plants. 2016;2:16139. doi: 10.1038/nplants.2016.139. [DOI] [PubMed] [Google Scholar]
- 5.Stachler A.E., Marchfelder A. Gene repression in haloarchaea using the CRISPR (clustered regularly interspaced short palindromic repeats)-Cas I-B system. J Biol Chem. 2016;291(29):15226–15242. doi: 10.1074/jbc.M116.724062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Joung J., Konermann S., Gootenberg J.S., et al. Genome-scale CRISPR-Cas9 knockout and transcriptional activation screening. Nat Protoc. 2017;12(4):828–863. doi: 10.1038/nprot.2017.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Jinek M., Chylinski K., Fonfara I., et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337(6096):816–821. doi: 10.1126/science.1225829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Menken M., Munsat T.L., Toole J.F. The global burden of disease study: implications for neurology. Arch Neurol. 2000;57(3):418–420. doi: 10.1001/archneur.57.3.418. [DOI] [PubMed] [Google Scholar]
- 9.Lu L., Yu X., Cai Y., et al. Application of CRISPR/Cas9 in Alzheimer's disease. Front Neurosci. 2021;15:803894. doi: 10.3389/fnins.2021.803894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Safari F., Hatam G., Behbahani A.B., et al. CRISPR system: a high-throughput toolbox for research and treatment of Parkinson's disease. Cell Mol Neurobiol. 2020;40(4):477–493. doi: 10.1007/s10571-019-00761-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Qin Y., Li S., Li X.J., et al. CRISPR-based genome-editing tools for Huntington's disease research and therapy. Neurosci Bull. 2022;38(11):1397–1408. doi: 10.1007/s12264-022-00880-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Al-Sammarraie N., Ray S.K. Applications of CRISPR-Cas9 technology to genome editing in glioblastoma multiforme. Cells. 2021;10(9):2342. doi: 10.3390/cells10092342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Carpenter J.C., Lignani G. Gene editing and modulation: the holy grail for the genetic epilepsies? Neurotherapeutics. 2021;18(3):1515–1523. doi: 10.1007/s13311-021-01081-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Yun Y., Ha Y. CRISPR/Cas9-mediated gene correction to understand ALS. Int J Mol Sci. 2020;21(11):3801. doi: 10.3390/ijms21113801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Sorek R., Kunin V., Hugenholtz P. CRISPR: a widespread system that provides acquired resistance against phages in bacteria and Archaea. Nat Rev Microbiol. 2008;6(3):181–186. doi: 10.1038/nrmicro1793. [DOI] [PubMed] [Google Scholar]
- 16.Lillestøl R.K., Shah S.A., Brügger K., et al. CRISPR families of the crenarchaeal genus Sulfolobus: bidirectional transcription and dynamic properties. Mol Microbiol. 2009;72(1):259–272. doi: 10.1111/j.1365-2958.2009.06641.x. [DOI] [PubMed] [Google Scholar]
- 17.Hale C., Kleppe K., Terns R.M., et al. Prokaryotic silencing (psi)RNAs in Pyrococcus furiosus. RNA. 2008;14(12):2572–2579. doi: 10.1261/rna.1246808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Carte J., Wang R., Li H., et al. Cas6 is an endoribonuclease that generates guide RNAs for invader defense in prokaryotes. Genes Dev. 2008;22(24):3489–3496. doi: 10.1101/gad.1742908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Chylinski K., Makarova K.S., Charpentier E., et al. Classification and evolution of type II CRISPR-Cas systems. Nucleic Acids Res. 2014;42(10):6091–6105. doi: 10.1093/nar/gku241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Makarova K.S., Haft D.H., Barrangou R., et al. Evolution and classification of the CRISPR-Cas systems. Nat Rev Microbiol. 2011;9(6):467–477. doi: 10.1038/nrmicro2577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Barrangou R., Fremaux C., Deveau H., et al. CRISPR provides acquired resistance against viruses in prokaryotes. Science. 2007;315(5819):1709–1712. doi: 10.1126/science.1138140. [DOI] [PubMed] [Google Scholar]
- 22.Wiedenheft B., Zhou K., Jinek M., et al. Structural basis for DNase activity of a conserved protein implicated in CRISPR-mediated genome defense. Structure. 2009;17(6):904–912. doi: 10.1016/j.str.2009.03.019. [DOI] [PubMed] [Google Scholar]
- 23.Beloglazova N., Brown G., Zimmerman M.D., et al. A novel family of sequence-specific endoribonucleases associated with the clustered regularly interspaced short palindromic repeats. J Biol Chem. 2008;283(29):20361–20371. doi: 10.1074/jbc.M803225200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Staals R.J., Agari Y., Maki-Yonekura S., et al. Structure and activity of the RNA-targeting type III-B CRISPR-Cas complex of Thermus thermophilus. Mol Cell. 2013;52(1):135–145. doi: 10.1016/j.molcel.2013.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Makarova K.S., Wolf Y.I., Alkhnbashi O.S., et al. An updated evolutionary classification of CRISPR–Cas systems. Nat Rev Microbiol. 2015;13(11):722–736. doi: 10.1038/nrmicro3569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Jinek M., Jiang F., Taylor D.W., et al. Structures of Cas9 endonucleases reveal RNA-mediated conformational activation. Science. 2014;343(6176):1247997. doi: 10.1126/science.1247997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Szczelkun M.D., Tikhomirova M.S., Sinkunas T., et al. Direct observation of R-loop formation by single RNA-guided Cas9 and Cascade effector complexes. Proc Natl Acad Sci U S A. 2014;111(27):9798–9803. doi: 10.1073/pnas.1402597111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Gasiunas G., Barrangou R., Horvath P., et al. Cas9-crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria. Proc Natl Acad Sci U S A. 2012;109(39):E2579–E2586. doi: 10.1073/pnas.1208507109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Jinek M., East A., Cheng A., et al. RNA-programmed genome editing in human cells. Elife. 2013;2 doi: 10.7554/eLife.00471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Mali P., Yang L., Esvelt K.M., et al. RNA-guided human genome engineering via Cas9. Science. 2013;339(6121):823–826. doi: 10.1126/science.1232033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Sung P., Klein H. Mechanism of homologous recombination: mediators and helicases take on regulatory functions. Nat Rev Mol Cell Biol. 2006;7(10):739–750. doi: 10.1038/nrm2008. [DOI] [PubMed] [Google Scholar]
- 32.Chang H.H.Y., Pannunzio N.R., Adachi N., et al. Non-homologous DNA end joining and alternative pathways to double-strand break repair. Nat Rev Mol Cell Biol. 2017;18(8):495–506. doi: 10.1038/nrm.2017.48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Canver M.C., Bauer D.E., Dass A., et al. Characterization of genomic deletion efficiency mediated by clustered regularly interspaced palindromic repeats (CRISPR)/Cas9 nuclease system in mammalian cells. J Biol Chem. 2014;289(31):21312–21324. doi: 10.1074/jbc.M114.564625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Liu Y., Zou R.S., He S., et al. Very fast CRISPR on demand. Science. 2020;368(6496):1265–1269. doi: 10.1126/science.aay8204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Wang A.S., Chen L.C., Wu R.A., et al. The histone chaperone FACT induces Cas9 multi-turnover behavior and modifies genome manipulation in human cells. Mol Cell. 2020;79(2):221–233.e5. doi: 10.1016/j.molcel.2020.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Clarke R., Heler R., MacDougall M.S., et al. Enhanced bacterial immunity and mammalian genome editing via RNA-polymerase-mediated dislodging of Cas9 from double-strand DNA breaks. Mol Cell. 2018;71(1):42–55.e8. doi: 10.1016/j.molcel.2018.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Ceccaldi R., Rondinelli B., D'Andrea A.D. Repair pathway choices and consequences at the double-strand break. Trends Cell Biol. 2016;26(1):52–64. doi: 10.1016/j.tcb.2015.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Scully R., Panday A., Elango R., et al. DNA double-strand break repair-pathway choice in somatic mammalian cells. Nat Rev Mol Cell Biol. 2019;20(11):698–714. doi: 10.1038/s41580-019-0152-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Yeh C.D., Richardson C.D., Corn J.E. Advances in genome editing through control of DNA repair pathways. Nat Cell Biol. 2019;21(12):1468–1478. doi: 10.1038/s41556-019-0425-z. [DOI] [PubMed] [Google Scholar]
- 40.Peterson S.E., Li Y., Chait B.T., et al. Cdk1 uncouples CtIP-dependent resection and Rad51 filament formation during M-phase double-strand break repair. J Cell Biol. 2011;194(5):705–720. doi: 10.1083/jcb.201103103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Song J., Yang D., Xu J., et al. RS-1 enhances CRISPR/Cas9- and TALEN-mediated knock-in efficiency. Nat Commun. 2016;7:10548. doi: 10.1038/ncomms10548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Maruyama T., Dougan S.K., Truttmann M.C., et al. Increasing the efficiency of precise genome editing with CRISPR-Cas9 by inhibition of nonhomologous end joining. Nat Biotechnol. 2015;33(5):538–542. doi: 10.1038/nbt.3190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Aylon Y., Liefshitz B., Kupiec M. The CDK regulates repair of double-strand breaks by homologous recombination during the cell cycle. EMBO J. 2004;23(24):4868–4875. doi: 10.1038/sj.emboj.7600469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ira G., Pellicioli A., Balijja A., et al. DNA end resection, homologous recombination and DNA damage checkpoint activation require CDK1. Nature. 2004;431(7011):1011–1017. doi: 10.1038/nature02964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Müller M., Lee C.M., Gasiunas G., et al. Streptococcus thermophilus CRISPR-Cas9 systems enable specific editing of the human genome. Mol Ther. 2016;24(3):636–644. doi: 10.1038/mt.2015.218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Qi L., Larson M., Gilbert L., et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell. 2013;152(5):1173–1183. doi: 10.1016/j.cell.2013.02.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Chavez A., Scheiman J., Vora S., et al. Highly efficient Cas9-mediated transcriptional programming. Nat Methods. 2015;12(4):326–328. doi: 10.1038/nmeth.3312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Gilbert L., Larson M., Morsut L., et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell. 2013;154(2):442–451. doi: 10.1016/j.cell.2013.06.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Mali P., Esvelt K.M., Church G.M. Cas9 as a versatile tool for engineering biology. Nat Methods. 2013;10(10):957–963. doi: 10.1038/nmeth.2649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Alerasool N., Segal D., Lee H., et al. An efficient KRAB domain for CRISPRi applications in human cells. Nat Methods. 2020;17(11):1093–1096. doi: 10.1038/s41592-020-0966-x. [DOI] [PubMed] [Google Scholar]
- 51.Saifaldeen M., Al-Ansari D.E., Ramotar D., et al. CRISPR FokI dead Cas9 system: principles and applications in genome engineering. Cells. 2020;9(11):2518. doi: 10.3390/cells9112518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Komor A.C., Badran A.H., Liu D.R. CRISPR-based technologies for the manipulation of eukaryotic genomes. Cell. 2017;168(1–2):20–36. doi: 10.1016/j.cell.2016.10.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Song F., Stieger K. Optimizing the DNA donor template for homology-directed repair of double-strand breaks. Mol Ther Nucleic Acids. 2017;7:53–60. doi: 10.1016/j.omtn.2017.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Bollen Y., Post J., Koo B.K., et al. How to create state-of-the-art genetic model systems: strategies for optimal CRISPR-mediated genome editing. Nucleic Acids Res. 2018;46(13):6435–6454. doi: 10.1093/nar/gky571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Nishida K., Arazoe T., Yachie N., et al. Targeted nucleotide editing using hybrid prokaryotic and vertebrate adaptive immune systems. Science. 2016;353(6305):aaf8729. doi: 10.1126/science.aaf8729. [DOI] [PubMed] [Google Scholar]
- 56.Komor A.C., Kim Y.B., Packer M.S., et al. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature. 2016;533(7603):420–424. doi: 10.1038/nature17946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Gaudelli N.M., Komor A.C., Rees H.A., et al. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature. 2017;551(7681):464–471. doi: 10.1038/nature24644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Li C., Zong Y., Wang Y., et al. Expanded base editing in rice and wheat using a Cas9-adenosine deaminase fusion. Genome Biol. 2018;19:59. doi: 10.1186/s13059-018-1443-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Nuñez J.K., Chen J., Pommier G.C., et al. Genome-wide programmable transcriptional memory by CRISPR-based epigenome editing. Cell. 2021;184(9):2503–2519.e17. doi: 10.1016/j.cell.2021.03.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Liu X., Zhang Y., Chen Y., et al. In situ capture of chromatin interactions by biotinylated dCas9. Cell. 2017;170(5):1028–1043.e19. doi: 10.1016/j.cell.2017.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Guiro J., Fagbemi M., Tellier M., et al. CAPTURE of the human U2 snRNA genes expands the repertoire of associated factors. Biomolecules. 2022;12(5):704. doi: 10.3390/biom12050704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Burramsetty A.K., Nishimura K., Kishimoto T., et al. Locus-specific isolation of the Nanog chromatin identifies regulators relevant to pluripotency of mouse embryonic stem cells and reprogramming of somatic cells. Int J Mol Sci. 2022;23(23):15242. doi: 10.3390/ijms232315242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Zetterberg H., Bendlin B.B. Biomarkers for Alzheimer's disease-preparing for a new era of disease-modifying therapies. Mol Psychiatr. 2021;26(1):296–308. doi: 10.1038/s41380-020-0721-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.LaFerla F.M., Oddo S. Alzheimer's disease: abeta, tau and synaptic dysfunction. Trends Mol Med. 2005;11(4):170–176. doi: 10.1016/j.molmed.2005.02.009. [DOI] [PubMed] [Google Scholar]
- 65.Hardy J., Selkoe D.J. The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science. 2002;297(5580):353–356. doi: 10.1126/science.1072994. [DOI] [PubMed] [Google Scholar]
- 66.Paquet D., Kwart D., Chen A., et al. Efficient introduction of specific homozygous and heterozygous mutations using CRISPR/Cas9. Nature. 2016;533(7601):125–129. doi: 10.1038/nature17664. [DOI] [PubMed] [Google Scholar]
- 67.Sun L., Zhou R., Yang G., et al. Analysis of 138 pathogenic mutations in presenilin-1 on the in vitro production of Aβ42 and Aβ40 peptides by γ-secretase. Proc Natl Acad Sci U S A. 2017;114(4):E476–E485. doi: 10.1073/pnas.1618657114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Park H., Oh J., Shim G., et al. In vivo neuronal gene editing via CRISPR-Cas9 amphiphilic nanocomplexes alleviates deficits in mouse models of Alzheimer's disease. Nat Neurosci. 2019;22(4):524–528. doi: 10.1038/s41593-019-0352-0. [DOI] [PubMed] [Google Scholar]
- 69.György B., Lööv C., Zaborowski M.P., et al. CRISPR/Cas9 mediated disruption of the Swedish APP allele as a therapeutic approach for early-onset Alzheimer's disease. Mol Ther Nucleic Acids. 2018;11:429–440. doi: 10.1016/j.omtn.2018.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Carr D.B., Goate A., Phil D., et al. Current concepts in the pathogenesis of Alzheimer's disease. Am J Med. 1997;103(3A):3S–10S. doi: 10.1016/s0002-9343(97)00262-3. [DOI] [PubMed] [Google Scholar]
- 71.Lambert J.C., Ibrahim-Verbaas C.A., Harold D., et al. Meta-analysis of 74, 046 individuals identifies 11 new susceptibility loci for Alzheimer's disease. Nat Genet. 2013;45(12):1452–1458. doi: 10.1038/ng.2802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Corder E.H., Saunders A.M., Strittmatter W.J., et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer's disease in late onset families. Science. 1993;261(5123):921–923. doi: 10.1126/science.8346443. [DOI] [PubMed] [Google Scholar]
- 73.Lin Y.T., Seo J., Gao F., et al. APOE4 causes widespread molecular and cellular alterations associated with Alzheimer's disease phenotypes in human iPSC-derived brain cell types. Neuron. 2018;98(6):1141–1154. doi: 10.1016/j.neuron.2018.05.008. e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Wadhwani A.R., Affaneh A., van Gulden S., et al. Neuronal apolipoprotein E4 increases cell death and phosphorylated tau release in Alzheimer disease. Ann Neurol. 2019;85(5):726–739. doi: 10.1002/ana.25455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.O'Brien R.J., Wong P.C. Amyloid precursor protein processing and Alzheimer's disease. Annu Rev Neurosci. 2011;34:185–204. doi: 10.1146/annurev-neuro-061010-113613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Saito T., Matsuba Y., Mihira N., et al. Single App knock-in mouse models of Alzheimer's disease. Nat Neurosci. 2014;17(5):661–663. doi: 10.1038/nn.3697. [DOI] [PubMed] [Google Scholar]
- 77.Viola K.L., Klein W.L. Amyloid β oligomers in Alzheimer's disease pathogenesis, treatment, and diagnosis. Acta Neuropathol. 2015;129(2):183–206. doi: 10.1007/s00401-015-1386-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Thal D.R., Walter J., Saido T.C., et al. Neuropathology and biochemistry of aβ and its aggregates in Alzheimer's disease. Acta Neuropathol. 2015;129(2):167–182. doi: 10.1007/s00401-014-1375-y. [DOI] [PubMed] [Google Scholar]
- 79.Wang M., Li A., Sekiya M., et al. Transformative network modeling of multi-omics data reveals detailed circuits, key regulators, and potential therapeutics for Alzheimer's disease. Neuron. 2021;109(2):257–272. doi: 10.1016/j.neuron.2020.11.002. e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Ascherio A., Schwarzschild M.A. The epidemiology of Parkinson's disease: risk factors and prevention. Lancet Neurol. 2016;15(12):1257–1272. doi: 10.1016/S1474-4422(16)30230-7. [DOI] [PubMed] [Google Scholar]
- 81.Hayes M.T. Parkinson's disease and Parkinsonism. Am J Med. 2019;132(7):802–807. doi: 10.1016/j.amjmed.2019.03.001. [DOI] [PubMed] [Google Scholar]
- 82.Taghavi S., Chaouni R., Tafakhori A., et al. A clinical and molecular genetic study of 50 families with autosomal recessive Parkinsonism revealed known and novel gene mutations. Mol Neurobiol. 2018;55(4):3477–3489. doi: 10.1007/s12035-017-0535-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Nuytemans K., Theuns J., Cruts M., et al. Genetic etiology of Parkinson disease associated with mutations in the SNCA, PARK2, PINK1, PARK7, and LRRK2 genes: a mutation update. Hum Mutat. 2010;31(7):763–780. doi: 10.1002/humu.21277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Scott L., Dawson V.L., Dawson T.M. Trumping neurodegeneration: targeting common pathways regulated by autosomal recessive Parkinson's disease genes. Exp Neurol. 2017;298:191–201. doi: 10.1016/j.expneurol.2017.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Nasrolahi A., Safari F., Farhoudi M., et al. Immune system and new avenues in Parkinson's disease research and treatment. Rev Neurosci. 2019;30(7):709–727. doi: 10.1515/revneuro-2018-0105. [DOI] [PubMed] [Google Scholar]
- 86.Dauer W., Przedborski S. Parkinson's disease: mechanisms and models. Neuron. 2003;39(6):889–909. doi: 10.1016/s0896-6273(03)00568-3. [DOI] [PubMed] [Google Scholar]
- 87.Ishizu N., Yui D., Hebisawa A., et al. Impaired striatal dopamine release in homozygous Vps35 D620N knock-in mice. Hum Mol Genet. 2016;25(20):4507–4517. doi: 10.1093/hmg/ddw279. [DOI] [PubMed] [Google Scholar]
- 88.Zhu X.X., Zhong Y.Z., Ge Y.W., et al. CRISPR/Cas9-mediated generation of Guangxi Bama minipigs harboring three mutations in α-synuclein causing Parkinson's disease. Sci Rep. 2018;8:12420. doi: 10.1038/s41598-018-30436-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Bonifati V., Breedveld G.J., Squitieri F., et al. Localization of autosomal recessive early-onset Parkinsonism to chromosome 1p36 (PARK7) in an independent dataset. Ann Neurol. 2002;51(2):253–256. doi: 10.1002/ana.10106. [DOI] [PubMed] [Google Scholar]
- 90.Chen Z.Z., Wang J.Y., Kang Y., et al. PINK1 gene mutation by pair truncated sgRNA/Cas9-D10A in cynomolgus monkeys. Zool Res. 2021;42(4):469–477. doi: 10.24272/j.issn.2095-8137.2021.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Wulansari N., Darsono W.H.W., Woo H.J., et al. Neurodevelopmental defects and neurodegenerative phenotypes in human brain organoids carrying Parkinson's disease-linked DNAJC6 mutations. Sci Adv. 2021;7(8) doi: 10.1126/sciadv.abb1540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Oh C.K., Dolatabadi N., Cieplak P., et al. S-nitrosylation of p62 inhibits autophagic flux to promote α-synuclein secretion and spread in Parkinson's disease and Lewy body dementia. J Neurosci. 2022;42(14):3011–3024. doi: 10.1523/JNEUROSCI.1508-21.2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Jo J., Yang L., Tran H.D., et al. Lewy body-like inclusions in human midbrain organoids carrying glucocerebrosidase and α-synuclein mutations. Ann Neurol. 2021;90(3):490–505. doi: 10.1002/ana.26166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Jowaed A., Schmitt I., Kaut O., et al. Methylation regulates alpha-synuclein expression and is decreased in Parkinson's disease patients' brains. J Neurosci. 2010;30(18):6355–6359. doi: 10.1523/JNEUROSCI.6119-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Guhathakurta S., Kim J., Adams L., et al. Targeted attenuation of elevated histone marks at SNCA alleviates α-synuclein in Parkinson's disease. EMBO Mol Med. 2021;13(2) doi: 10.15252/emmm.202012188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Yoon H.H., Ye S., Lim S., et al. CRISPR-Cas9 gene editing protects from the A53T-SNCA overexpression-induced pathology of Parkinson's disease in vivo. CRISPR J. 2022;5(1):95–108. doi: 10.1089/crispr.2021.0025. [DOI] [PubMed] [Google Scholar]
- 97.Ryan S., Dolatabadi N., Chan S., et al. Isogenic human iPSC Parkinson's model shows nitrosative stress-induced dysfunction in MEF2-PGC1α transcription. Cell. 2013;155(6):1351–1364. doi: 10.1016/j.cell.2013.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Delgado-Camprubi M., Esteras N., Soutar M.P., et al. Deficiency of Parkinson's disease-related gene Fbxo7 is associated with impaired mitochondrial metabolism by PARP activation. Cell Death Differ. 2017;24(1):120–131. doi: 10.1038/cdd.2016.104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Ma K.Y., Fokkens M.R., Reggiori F., et al. Parkinson's disease-associated VPS35 mutant reduces mitochondrial membrane potential and impairs PINK1/Parkin-mediated mitophagy. Transl Neurodegener. 2021;10:19. doi: 10.1186/s40035-021-00243-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Basu S., Adams L., Guhathakurta S., et al. A novel tool for monitoring endogenous alpha-synuclein transcription by NanoLuciferase tag insertion at the 3'end using CRISPR-Cas9 genome editing technique. Sci Rep. 2017;8:45883. doi: 10.1038/srep45883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Fields E., Vaughan E., Tripu D., et al. Gene targeting techniques for Huntington's disease. Ageing Res Rev. 2021;70:101385. doi: 10.1016/j.arr.2021.101385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Tabrizi S.J., Ghosh R., Leavitt B.R. Huntingtin lowering strategies for disease modification in Huntington's disease. Neuron. 2019;101(5):801–819. doi: 10.1016/j.neuron.2019.01.039. [DOI] [PubMed] [Google Scholar]
- 103.Ross C.A., Tabrizi S.J. Huntington's disease: from molecular pathogenesis to clinical treatment. Lancet Neurol. 2011;10(1):83–98. doi: 10.1016/S1474-4422(10)70245-3. [DOI] [PubMed] [Google Scholar]
- 104.Crook Z.R., Housman D., Huntington's disease Can mice lead the way to treatment? Neuron. 2011;69(3):423–435. doi: 10.1016/j.neuron.2010.12.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Farshim P.P., Bates G.P. Mouse models of Huntington's disease. Methods Mol Biol. 2018;1780:97–120. doi: 10.1007/978-1-4939-7825-0_6. [DOI] [PubMed] [Google Scholar]
- 106.Levine M.S., Cepeda C., Hickey M.A., et al. Genetic mouse models of Huntington's and Parkinson's diseases: illuminating but imperfect. Trends Neurosci. 2004;27(11):691–697. doi: 10.1016/j.tins.2004.08.008. [DOI] [PubMed] [Google Scholar]
- 107.Yan S., Tu Z., Liu Z., et al. A huntingtin knockin pig model recapitulates features of selective neurodegeneration in Huntington's disease. Cell. 2018;173(4):989–1002. doi: 10.1016/j.cell.2018.03.005. e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Dabrowska M., Juzwa W., Krzyzosiak W.J., et al. Precise excision of the CAG tract from the huntingtin gene by Cas9 nickases. Front Neurosci. 2018;12:75. doi: 10.3389/fnins.2018.00075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Yang S., Chang R., Yang H., et al. CRISPR/Cas9-mediated gene editing ameliorates neurotoxicity in mouse model of Huntington's disease. J Clin Invest. 2017;127(7):2719–2724. doi: 10.1172/JCI92087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Wang G., Liu X., Gaertig M.A., et al. Ablation of huntingtin in adult neurons is nondeleterious but its depletion in young mice causes acute pancreatitis. Proc Natl Acad Sci U S A. 2016;113(12):3359–3364. doi: 10.1073/pnas.1524575113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Tabrizi S.J., Scahill R.I., Durr A., et al. Biological and clinical changes in premanifest and early stage Huntington's disease in the TRACK-HD study: the 12-month longitudinal analysis. Lancet Neurol. 2011;10(1):31–42. doi: 10.1016/S1474-4422(10)70276-3. [DOI] [PubMed] [Google Scholar]
- 112.Liu H., Zhang C., Xu J., et al. Huntingtin silencing delays onset and slows progression of Huntington's disease: a biomarker study. Brain. 2021;144(10):3101–3113. doi: 10.1093/brain/awab190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Ostrom Q.T., Gittleman H., Farah P., et al. CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2006-2010. Neuro Oncol. 2013;15(Suppl 2):ii1–ii56. doi: 10.1093/neuonc/not151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Jackson H.J., Rafiq S., Brentjens R.J. Driving CAR T-cells forward. Nat Rev Clin Oncol. 2016;13(6):370–383. doi: 10.1038/nrclinonc.2016.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.June C.H., Sadelain M. Chimeric antigen receptor therapy. N Engl J Med. 2018;379(1):64–73. doi: 10.1056/NEJMra1706169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Wang D., Prager B.C., Gimple R.C., et al. CRISPR screening of CAR T cells and cancer stem cells reveals critical dependencies for cell-based therapies. Cancer Discov. 2021;11(5):1192–1211. doi: 10.1158/2159-8290.CD-20-1243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Larson R.C., Kann M.C., Bailey S.R., et al. CAR T cell killing requires the IFNγR pathway in solid but not liquid tumours. Nature. 2022;604(7906):563–570. doi: 10.1038/s41586-022-04585-5. [DOI] [PubMed] [Google Scholar]
- 118.Rosenblum D., Gutkin A., Kedmi R., et al. CRISPR-Cas9 genome editing using targeted lipid nanoparticles for cancer therapy. Sci Adv. 2020;6(47) doi: 10.1126/sciadv.abc9450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Tang F., Hartz A.M.S., Bauer B. Drug-resistant epilepsy: multiple hypotheses, few answers. Front Neurol. 2017;8:301. doi: 10.3389/fneur.2017.00301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Manford M. Recent advances in epilepsy. J Neurol. 2017;264(8):1811–1824. doi: 10.1007/s00415-017-8394-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Kefauver J.M., Ward A.B., Patapoutian A. Discoveries in structure and physiology of mechanically activated ion channels. Nature. 2020;587(7835):567–576. doi: 10.1038/s41586-020-2933-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Thijs R.D., Surges R., O'Brien T.J., et al. Epilepsy in adults. Lancet. 2019;393(10172):689–701. doi: 10.1016/S0140-6736(18)32596-0. [DOI] [PubMed] [Google Scholar]
- 123.Wykes R.C., Heeroma J.H., Mantoan L., et al. Optogenetic and potassium channel gene therapy in a rodent model of focal neocortical epilepsy. Sci Transl Med. 2012;4(161):161ra152. doi: 10.1126/scitranslmed.3004190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Colasante G., Qiu Y., Massimino L., et al. In vivo CRISPRa decreases seizures and rescues cognitive deficits in a rodent model of epilepsy. Brain. 2020;143(3):891–905. doi: 10.1093/brain/awaa045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Hansen K.F., Sakamoto K., Pelz C., et al. Profiling status epilepticus-induced changes in hippocampal RNA expression using high-throughput RNA sequencing. Sci Rep. 2014;4:6930. doi: 10.1038/srep06930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Hawkins N.A., Calhoun J.D., Huffman A.M., et al. Gene expression profiling in a mouse model of Dravet syndrome. Exp Neurol. 2019;311:247–256. doi: 10.1016/j.expneurol.2018.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Hawkins N.A., Misra S.N., Jurado M., et al. Epilepsy and neurobehavioral abnormalities in mice with a dominant-negative KCNB1 pathogenic variant. Neurobiol Dis. 2021;147:105141. doi: 10.1016/j.nbd.2020.105141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Lagae L. Dravet syndrome. Curr Opin Neurol. 2020;34(2):213–218. doi: 10.1097/WCO.0000000000000902. [DOI] [PubMed] [Google Scholar]
- 129.Dravet C. Dravet syndrome history. Dev Med Child Neurol. 2011;53(Suppl 2):1–6. doi: 10.1111/j.1469-8749.2011.03964.x. [DOI] [PubMed] [Google Scholar]
- 130.Cetica V., Chiari S., Mei D., et al. Clinical and genetic factors predicting Dravet syndrome in infants with SCN1A mutations. Neurology. 2017;88(11):1037–1044. doi: 10.1212/WNL.0000000000003716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Colasante G., Lignani G., Brusco S., et al. dCas9-based Scn1a gene activation restores inhibitory interneuron excitability and attenuates seizures in Dravet syndrome mice. Mol Ther. 2020;28(1):235–253. doi: 10.1016/j.ymthe.2019.08.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Griffin A., Carpenter C., Liu J., et al. Phenotypic analysis of catastrophic childhood epilepsy genes. Commun Biol. 2021;4:680. doi: 10.1038/s42003-021-02221-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Leu C., Stevelink R., Smith A.W., et al. Polygenic burden in focal and generalized epilepsies. Brain. 2019;142(11):3473–3481. doi: 10.1093/brain/awz292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Zhang Y.H., Burgess R., Malone J.P., et al. Genetic epilepsy with febrile seizures plus: refining the spectrum. Neurology. 2017;89(12):1210–1219. doi: 10.1212/WNL.0000000000004384. [DOI] [PubMed] [Google Scholar]
- 135.Hardiman O., Al-Chalabi A., Chio A., et al. Amyotrophic lateral sclerosis. Nat Rev Dis Prim. 2017;3:17071. doi: 10.1038/nrdp.2017.71. [DOI] [PubMed] [Google Scholar]
- 136.Rosen D.R. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature. 1993;364(6435):362. doi: 10.1038/364362c0. [DOI] [PubMed] [Google Scholar]
- 137.DeJesus-Hernandez M., MacKenzie I., Boeve B., et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron. 2011;72(2):245–256. doi: 10.1016/j.neuron.2011.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Sreedharan J., Blair I.P., Tripathi V.B., et al. TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science. 2008;319(5870):1668–1672. doi: 10.1126/science.1154584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Kwiatkowski T.J., Jr., Bosco D.A., Leclerc A.L., et al. Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science. 2009;323(5918):1205–1208. doi: 10.1126/science.1166066. [DOI] [PubMed] [Google Scholar]
- 140.Zhu Q., Jiang J., Gendron T.F., et al. Reduced C9ORF72 function exacerbates gain of toxicity from ALS/FTD-causing repeat expansion in C9orf72. Nat Neurosci. 2020;23(5):615–624. doi: 10.1038/s41593-020-0619-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Krishnan G., Zhang Y., Gu Y., et al. CRISPR deletion of the C9ORF72 promoter in ALS/FTD patient motor neurons abolishes production of dipeptide repeat proteins and rescues neurodegeneration. Acta Neuropathol. 2020;140(1):81–84. doi: 10.1007/s00401-020-02154-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Pinto B.S., Saxena T., Oliveira R., et al. Impeding transcription of expanded microsatellite repeats by deactivated Cas9. Mol Cell. 2017;68(3):479–490. doi: 10.1016/j.molcel.2017.09.033. e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Perkins E.M., Burr K., Banerjee P., et al. Altered network properties in C9ORF72 repeat expansion cortical neurons are due to synaptic dysfunction. Mol Neurodegener. 2021;16:13. doi: 10.1186/s13024-021-00433-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Pickles S., Vande Velde C. Misfolded SOD1 and ALS: zeroing in on mitochondria. Amyotroph Lateral Scler. 2012;13(4):333–340. doi: 10.3109/17482968.2012.648645. [DOI] [PubMed] [Google Scholar]
- 145.Brown R.H., Al-Chalabi A. Amyotrophic lateral sclerosis. N Engl J Med. 2017;377(2):162–172. doi: 10.1056/NEJMra1603471. [DOI] [PubMed] [Google Scholar]
- 146.Kato S., Kato M., Abe Y., et al. Redox system expression in the motor neurons in amyotrophic lateral sclerosis (ALS): immunohistochemical studies on sporadic ALS, superoxide dismutase 1 (SOD1)-mutated familial ALS, and SOD1-mutated ALS animal models. Acta Neuropathol. 2005;110(2):101–112. doi: 10.1007/s00401-005-1019-3. [DOI] [PubMed] [Google Scholar]
- 147.Gaj T., Ojala D.S., Ekman F.K., et al. In vivo genome editing improves motor function and extends survival in a mouse model of ALS. Sci Adv. 2017;3(12):eaar3952. doi: 10.1126/sciadv.aar3952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Duan W., Guo M., Yi L., et al. The deletion of mutant SOD1 via CRISPR/Cas9/sgRNA prolongs survival in an amyotrophic lateral sclerosis mouse model. Gene Ther. 2020;27(3–4):157–169. doi: 10.1038/s41434-019-0116-1. [DOI] [PubMed] [Google Scholar]
- 149.Lim C.K.W., Gapinske M., Brooks A.K., et al. Treatment of a mouse model of ALS by in vivo base editing. Mol Ther. 2020;28(4):1177–1189. doi: 10.1016/j.ymthe.2020.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]