The CRISPR/Cas9 system is one of the most extensively researched and developed gene editing tools, primarily consisting of Cas9 and single-guide RNA (sgRNA), which is formed by trans-activating CRISPR RNA (tracrRNA) and CRISPR RNA (crRNA). CRISPR-Cas9 technology has a wide range of applications and holds significant promise for gene function research, the construction of model organisms, and gene therapy, including treatments for cancers, liver diseases, and cardiovascular diseases.
Cas9 possesses DNA cleavage domains, including HNH (His/Asn/His) and RuvC (recombinase UVC). The RuvC domain cleaves the opposite strand of double-stranded DNA (dsDNA), while the HNH domain cleaves the complementary CRISPR RNA (crRNA) strand.
Under the guidance of sgRNA, Cas9 proteins can achieve targeted cleavage of specific genes, resulting in double-stranded breaks (DSBs) in the DNA. These breaks are repaired through either cell-autonomous non-homologous end joining (NHEJ) or homologous recombination (HR), leading to the knockout, insertion, or mutational modification of the target genes.
Fig.1 Overview of the CRISPR-Cas9 mechanism of action. (Bora J, et al., 2023)
The specific mechanism of action can be categorized into three distinct stages.
Stages | Descriptions |
---|---|
Stage I | The acquisition of highly variable spacer regions in CRISPR: When bacteria and archaea containing CRISPR are invaded by exogenous phages or plasmids, they acquire foreign DNA fragments that are inserted into the spacer region. |
Stage II | The expression of the CRISPR motif occurs when foreign nucleic acids that are homologous to the spacer region re-enter the bacteria. This process activates the transcription of the CRISPR array, leading to the production of pre-crRNA. Simultaneously, tracrRNA, which is complementary to the pre-crRNA sequence, is also transcribed. After transcription, tracrRNA first binds to the Cas9 protein. The complementary base pairing between pre-crRNA and tracrRNA forms double-stranded RNA, which subsequently binds to Cas9 to create a complex. After double-stranded RNA binds to Cas9, RNase III synthesizes pre-crRNA in the primary process. In the secondary process, Cas9 can eliminate redundant repetitive sequences and spacer sequences. Following these two processes, the crRNA matures and acquires the ability to target DNA strands. |
Stage III | Exploitation of CRISPR/Cas System Activity (Targeted Interference): If re-infected with homologous DNA, the bacteria will initiate transcription of the CRISPR region. Following a series of processing and maturation steps to produce single-guide RNA (sgRNA), the sgRNA directs Cas9 to cleave and disrupt the DNA strand in the homologous spacer region, resulting in a double-strand break (DSB). The cell then repairs the target gene through non-homologous end joining (NHEJ) or homologous recombination (HR). |
Viral vectors commonly utilized in CRISPR-Cas9 systems include adeno-associated virus (AAV), lentivirus, and baculovirus vectors.
Liposome-based nanoparticles (LNPs) are regarded as promising vehicles for the delivery of CRISPR/Cas9.
PLGA, chitosan, and other molecules commonly utilized to construct polymer nanoparticle shells enhance the efficiency of cellular uptake of these polymers.
Since exosomes directly package sgRNA and Cas9, effectively reducing off-target side effects during transport, they represent promising vehicles for the delivery of the CRISPR/dCas9 system.
Gold nanoparticles exhibit excellent stability and biocompatibility, and they can be easily modified on their surfaces, making them ideal carriers for the delivery of gene therapies.
CRISPR/Cas9 technology is extensively utilized in gene therapy applications for nucleic acid formulations. It effectively repairs or replaces disease-causing genes through precisely targeted gene editing, thereby addressing genetic disorders. This technology enhances both the efficiency and accuracy of gene therapy while significantly minimizing off-target effects during treatment, ensuring the safety and efficacy of the intervention.
In nucleic acid drug development, CRISPR/Cas9 technology serves as a precise gene-editing tool that can efficiently identify potential drug targets. This technology not only shortens the drug development timeline but also enhances the accuracy of targeted therapies and contributes to the advancement of personalized medicine.
Fig.2 Applications of CRISPR/Cas9 technology. (CD Formulation)
CRISPR/Cas9 technology allows scientists to precisely knock out or modify specific genes in cells and animal models to study the functions of these genes and their associated pathological mechanisms. For instance, by knocking out a particular gene, researchers can observe the physiological changes that occur due to the loss of expression of that gene, thereby elucidating its role in the organism.
Another significant application of CRISPR-Cas9 technology is in the treatment of genetic diseases. By directly correcting disease-causing gene mutations in a patient's genome, these conditions can be treated effectively. For instance, CRISPR has been utilized to correct mutations associated with beta thalassemia and sickle cell disease in in vitro models and preclinical trials.
CRISPR-Cas9 technology has demonstrated significant potential in the field of microbial engineering, particularly in modifying microorganisms through gene editing to produce biofuels, pharmaceuticals, and other valuable chemicals. This technology enables the precise regulation of microbial metabolic pathways, optimizing the generation of desired products.
In the field of biopharmaceuticals, CRISPR-Cas9 technology can be utilized to modify genes in mammalian cells, thereby optimizing protein expression and modification during drug production. This capability is especially crucial for the production of biologics with complex structures, such as antibody-based therapies.
References