MOLECULAR & CELLULAR NEUROBIOLOGY 
Master Course Cognitive Neuroscience - Radboud University, Nijmegen

 

Chapter 5: Molecular biological research methodology

           Molecular biology and Recombinant DNA technology Detection of DNA, RNA and protein Generation of gene expression atlases of the CNS
           Techniques used in Molecular Biology    Detection of RNA Gene transfer - transgenic animals
           Genetic transmission    In situ hybridization Optogenetics
           Genetic mapping    PCR Cloning
           Genomic and cDNA libraries    Microarray and RNA-seq analysis Stem cells
   Bioinformatics - data analysis    CRISPR-cas genome editing
  ChIP-chip/seq  

 

 

CRISPR-cas9 genome editing

 

As we saw above and in other sections, technologies for making and manipulating DNA have enabled advances in biology ever since the discovery of the DNA double helix. Early approaches to introduce site-specific modifications in the genomes of cells and organisms relied on the principle of site-specific recognition of DNA sequences by oligonucleotides, small molecules, self-splicing introns or site-directed nucleases (e.g. TALENs). The field of biology is now experiencing a transformative phase with the advent of the molecular toolbox for mammalian genome engineer­ing: easily programmable RNA-guided nucleases, which are derived from micro­bial adaptive immune systems. Gene-editing technologies in the form of Clus­tered Regularly Interspaced Short Palindromic Repeat (CRISPR)–CRISPR-associated protein (Cas) systems stand poised to enable fast and accurate alterations of genomic information in mammalian model systems and human tissues.

 

The CRISPR-Cas9 technology originates from type II CRISPR-Cas systems, which provide bacteria with adaptive immunity to viruses and plasmids. After identification of the CRISPR/Cas9 system in bacteria, a series of elegant biochemical studies distilled the essential site-specific deoxyribonucleic acid cleavage activity to only two components: 1) an RNA guide sequence and 2) a DNA endonuclease (see figure below). Based on these observations, yet another group of investigators re-engineered these components to function in mammalian cells, and it was discovered subsequently that the CRISPR/Cas9 system could be manipulated to generate either random mutations or targeted repair. Aside from cultured cells, the CRISPR/Cas9 system functions efficiently to modify the genome of fertilized mouse zygotes, and this approach has been adopted rapidly as an efficient method for creating genetically altered mice.

 

Figure: The CRISPR (Clustered Regularly Interspaced Palindromic Repeat)/Cas9 system. A, The CRISPR/Cas9 system can be distilled to two essential components: 1) a ribonucleic acid guide sequence to target host deoxyribonucleic acid (DNA) containing a protospacer adjacent motif (PAM) and 2) the Cas9 endonuclease enzyme. B, DNA cleavage involves target recognition amid the complex genomic milieu followed by DNA strand separation and precise enzymatic scission of the DNA backbone. C, After DNA cleavage, nonhomologous end joining (NHEJ) can potentially lead to restoration of an out-of-frame transcript without needing a DNA template. Alternatively, the homology-directed repair (HDR) pathway can use an error-free template to correct a gene mutation.

 

Cas9 is an endonuclease that uses a guide sequence within an RNA duplex to form base pairs with DNA target sequences, enabling Cas9 to introduce a site-specific double-strand break in the DNA (see figure below). The RNA duplex(tracrRNA:crRNA) was engineered as a single guide RNA (sgRNA) that retains two critical features: a sequence at the 5’ side that determines the DNA target site by Watson-Crick base-pairing and a duplex RNA structure at the 3’ side that binds to Cas9. This finding created a simple two-component system in which changes in the guide sequence of the sgRNA program Cas9 to target any DNA sequence of interest. The simplicity of CRISPR-Cas9 programming, together with a unique DNA cleaving mechanism, the capacity for multiplexed target recognition, and the existence of many natural type II CRISPR-Cas system variants, has enabled remarkable developments using this cost-effective and easy-to-use technology to precisely and efficiently target, edit, modify, regulate, and mark genomic loci of a wide array of cells and organisms (see figures below).

 

The power of the technology includes the systematic analysis of gene functions in mammalian cells, study genomic rearrangements and the progression of cancers or other diseases, and correct genetic mutations responsible for inherited disorders. The CRISPR/Cas9 system has thus evolved quickly from the initial discovery of an adaptive immune system in bacteria to a 2-component genome editing tool to a global disease-modification strategy. CRISPR/Cas9 technology has also revolutionized how gene perturbation experiments are conducted at a genome-wide scale and has enabled the unprecedented creation of genetically altered nonhuman primates for preclinical studies. Eventual therapeutic translation, however, will require identification of appropriate disease targets and development of robust methods for introducing CRISPR/Cas9 components into specific cell-types without off-cell-type and off-target effects. Despite these immediate hurdles, CRISPR/Cas9 technology is poised to revolutionize disease management in ways that were once difficult to imagine.

 

 

Figure: The Cas9 enzyme (blue) generates breaks in double-stranded DNA by using its two catalytic centers (blades) to cleave each strand of a DNA target site (gold) next to a PAM sequence (red) and matching the 20-nucleotide sequence (orange) of the single guide RNA (sgRNA). The sgRNA includes a dual-RNA sequence derived from CRISPR RNA (light green) and a separate transcript (tracrRNA, dark green) that binds and stabilizes the Cas9 protein. Cas9-sgRNA–mediated DNA cleavage produces a blunt double-stranded break that triggers repair enzymes to disrupt or replace DNA sequences at or near the cleavage site. Catalytically inactive forms of Cas9 can also be used for programmable regulation of

transcription and visualization of genomic loci.

 

Figure: CRISPR–Cas in the generation of cellular models and large-scale screens. CRISPR–Cas gene editing can be used to generate isogenic cell lines for drug target validation, mechanistic analysis and patient stratification studies. Isogenic cell lines can also be used to generate organoids, which are particularly useful for modelling differentiation and self-organization processes. Large-scale single guide RNA (sgRNA) libraries can be used for high-throughput pooled or high-content arrayed screens, either in unmodified or in CRISPR–Cas-edited cell lines. RNPs, ribonucleoproteins.

 

Figure: Applications of CRISPR–Cas in in vivo screens and the generation of animal models. a. Ex vivo editing can be used to generate a library of modified cells for transplantation into recipient animals. Alternatively, editing reagents can be delivered to host animal tissues directly for somatic in situ editing. b.| CRISPR–Cas has also revolutionized the generation of transgenic animal models through facile editing of embryonic stem (ES) cells for traditional gene targeting and by enabling direct zygote editing in most species. Zygote editing can be done ex vivo by electroporating or microinjecting zygotes with CRISPR–Cas constructs in the form of plasmids, RNA preparations or ribonucleoproteins (RNPs). AAV, adeno-associated virus; sgRNA, single guide RNA. 

 

Figure: Applications in biomedicine and biotechnology. Developments include establishment of screens for target identification, human gene therapy by gene repair and gene disruption, gene disruption of viral sequences, and programmable RNA targeting.

 

Next page: Neurodevelopment Go back to: Stem cells