[go: nahoru, domu]

Jump to content

Cas9: Difference between revisions

From Wikipedia, the free encyclopedia
Content deleted Content added
Helito (talk | contribs)
Tags: Mobile edit Mobile web edit
Helito (talk | contribs)
→‎Patent Landscape: merged to CRISPR gene editing page
Line 109: Line 109:
=== Possible solutions to bacterium cas-9 editing ===
=== Possible solutions to bacterium cas-9 editing ===
Recent studies have shown that with some minor modification to the cas-9 protein it is possible to bypass the “Restriction Modification System”. The process that achieves this is called “Photospacing” which means that the cas-9 system will copy a section of DNA from the host and insert it as a spacer gene into its own genome.<ref>Barrangou R, Fremaux C, Deveau H, Richards M, Boyaval P, Moineau S, Romero DA, Horvath P. 2007 CRISPR provides acquired resistance against viruses in prokaryotes. Science 315, 1709–1712.</ref> Though we do not fully understand why “photospacers” work we do understand what they do. When a prokaryote “senses” a part of its own genetic coding inside the foreign entity it is less likely to destroy the cas-9 insertion. This is known as the beginning of cas-9 immunity.<ref name="Fineran_2012">{{cite journal | vauthors = Fineran PC, Charpentier E | title = Memory of viral infections by CRISPR-Cas adaptive immune systems: acquisition of new information | journal = Virology | volume = 434 | issue = 2 | pages = 202–9 | date = December 2012 | pmid = 23123013 | doi = 10.1016/j.virol.2012.10.003 }}</ref>
Recent studies have shown that with some minor modification to the cas-9 protein it is possible to bypass the “Restriction Modification System”. The process that achieves this is called “Photospacing” which means that the cas-9 system will copy a section of DNA from the host and insert it as a spacer gene into its own genome.<ref>Barrangou R, Fremaux C, Deveau H, Richards M, Boyaval P, Moineau S, Romero DA, Horvath P. 2007 CRISPR provides acquired resistance against viruses in prokaryotes. Science 315, 1709–1712.</ref> Though we do not fully understand why “photospacers” work we do understand what they do. When a prokaryote “senses” a part of its own genetic coding inside the foreign entity it is less likely to destroy the cas-9 insertion. This is known as the beginning of cas-9 immunity.<ref name="Fineran_2012">{{cite journal | vauthors = Fineran PC, Charpentier E | title = Memory of viral infections by CRISPR-Cas adaptive immune systems: acquisition of new information | journal = Virology | volume = 434 | issue = 2 | pages = 202–9 | date = December 2012 | pmid = 23123013 | doi = 10.1016/j.virol.2012.10.003 }}</ref>

== Patent Landscape ==
As of November 2018, CRISPR-Cas9 patent landscape is dominated by universities and research institutes over commercial companies. US and Chinese academic institutions are actively pursuing research activities in CRISPR domain. Massachusetts Institute of Technology (MIT) holds the largest patent portfolio in CRISPR-Cas9 technology with 103 IP assets. Several new Chinese players like University of China Agricultural, University of Shanghai Jiao Tong, University of Northwest A&F, and Institute of Genetics & Developmental Biology, CAS have been key applicants from 2015 onward.


== See also ==
== See also ==

Revision as of 01:29, 18 February 2019

CRISPR-associated endonuclease Cas9
Crystal structure of S pyogenes Cas9 in complex with sgRNA and its target DNA at 2.5 A ˚ resolution.[1]
Identifiers
OrganismStreptococcus pyogenes M1
Symbolcas9
Alt. symbolsSpyCas9
Entrez901176
PDB4OO8
RefSeq (mRNA)NC_002737.2
RefSeq (Prot)NP_269215.1
UniProtQ99ZW2
Other data
EC number3.1.-.-
ChromosomeGenomic: 0.85 - 0.86 Mb
Search for
StructuresSwiss-model
DomainsInterPro

Cas9 (CRISPR associated protein 9) is an RNA-guided DNA endonuclease enzyme associated with the CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) adaptive immunity system in Streptococcus pyogenes, among other bacteria. S. pyogenes utilizes Cas9 to memorize[2] and later interrogate and cleave foreign DNA,[3] such as invading bacteriophage DNA or plasmid DNA. Cas9 performs this interrogation by unwinding foreign DNA and checking for sites complementary to the 20 basepair spacer region of the guide RNA. If the DNA substrate is complementary to the guide RNA, Cas9 cleaves the invading DNA. In this sense, the CRISPR-Cas9 mechanism has a number of parallels with the RNA interference (RNAi) mechanism in eukaryotes.

Apart from its original function in bacterial immunity, the Cas9 protein has been heavily utilized as a genome engineering tool to induce site-directed double strand breaks in DNA. These breaks can lead to gene inactivation or the introduction of heterologous genes through non-homologous end joining and homologous recombination respectively in many laboratory model organisms. Alongside zinc finger nucleases and TALEN proteins, Cas9 is becoming a prominent tool in the field of genome editing.

Cas9 has gained traction in recent years because it can cleave nearly any sequence complementary to the guide RNA.[3] Because the target specificity of Cas9 stems from the guide RNA:DNA complementarity and not modifications to the protein itself (like TALENs and Zinc-fingers), engineering Cas9 to target new DNA is straightforward.[4] Versions of Cas9 that bind but do not cleave cognate DNA can be used to localize transcriptional activator or repressors to specific DNA sequences in order to control transcriptional activation and repression.[5][6] While native Cas9 requires a guide RNA composed of two disparate RNAs that associate to make the guide – the CRISPR RNA (crRNA), and the trans-activating RNA (tracrRNA).,[3] Cas9 targeting has been simplified through the engineering of a chimeric single guide RNA (chiRNA). Scientists have suggested that Cas9-based gene drives may be capable of editing the genomes of entire populations of organisms.[7] In 2015, Cas9 was used to modify the genome of human embryos for the first time.[8]

CRISPR-mediated immunity

Introduction

To survive in a variety of challenging, inhospitable habitats that are filled with bacteriophages, bacteria and archea have evolved methods to evade and fend off predatory viruses. This includes the CRISPR system of adaptive immunity. In practice, CRISPR acts as a self-programmable restriction enzyme. CRISPR loci are composed of short, palindromic repeats that occur at regular intervals composed of alternate CRISPR repeats and variable CRISPR spacers between 24-48 nucleotides long. These CRISPR loci are usually accompanied by adjacent CRISPR-associated (cas) genes. In 2005, it was discovered by three separate groups that the spacer regions were homologous to foreign DNA elements, including plasmids and viruses. These reports provided the first biological evidence that CRISPRs might function as an immune system.

Cas9 has been used often as a genome-editing tool. Cas9 has been used in recent developments in preventing viruses from manipulating hosts’ DNA. Since the CRISPR-Cas9 was developed from bacterial genome systems, it can be used to target the genetic material in viruses. The use of the enzyme Cas9 can be a solution to many viral infections. Cas9 possesses the ability to target specific viruses by the targeting of specific strands of the viral genetic information. More specifically the Cas9 enzyme targets certain sections of the viral genome that prevents the virus from carrying out its normal function.[9] Cas9 has also been used to disrupt the detrimental strand of DNA and RNA that cause diseases and mutated strands of DNA. Cas9 has already showed promise in disrupting the effects of HIV-1. Cas9 has been shown to suppress the expression of the long terminal repeats in HIV-1. When introduced into the HIV-1 genome Cas9 has shown the ability to mutate strands of HIV-1.[10][11] Cas9 has also been used in the treatment of hepatitis b through targeting of the ends of certain of long terminal repeats in the hepatitis b viral genome.[12] In addition, Cas9 has already been used in human trials in the treatment of cystic fibrosis and oncogenic mutations in human pluripotent stem cells.[13] Cas9 has used to repair the mutations causing cataracts in mice.

Overview of CRISPR-Cas types and function

Fig. 2: The Stages of CRISPR immunity
Fig. 2: The Stages of CRISPR immunity

CRISPR-Cas systems are divided into three major types (type I, type II, and type III) and twelve subtypes, which are based on their genetic content and structural differences. However, the core defining features of all CRISPR-Cas systems are the cas genes and their proteins: cas1 and cas2 are universal across types and subtypes, while cas3, cas9, and cas10 are signature genes for type I, type II, and type III, respectively.

CRISPR-Cas defense stages

  1. AdaptationAdaptation involves recognition and integration of spacers between two adjacent repeats in the CRISPR locus. The “Protospacer” refers to the sequence on the viral genome that corresponds to the spacer. A short stretch of conserved nucleotides exists proximal to the protospacer, which is called the protospacer adjacent motif (PAM). The PAM is a recognition motif that is used to acquire the DNA fragment.[14] In type II, Cas9 recognizes the PAM during adaptation in order to ensure the acquisition of functional spacers.[2]
  2. CRISPR Processing – CRISPR expression includes the transcription of a primary transcript called a CRISPR RNA (pre-crRNA), which is transcribed from the CRISPR locus by RNA polymerase. Specific endoribonucleases then cleave the pre-crRNAs into small CRISPR RNAs (crRNAs).[15]
  3. Interference / Immunity – Interference involves the crRNAs within a multi-protein complex called CASCADE, which can recognize and specifically base-pair with regions of inserting complementary foreign DNA. The crRNA-foreign nucleic acid complex is then cleaved, however if there are mismatches between the spacer and the target DNA, or if there are mutations in the PAM, then cleavage will not be initiated. In the latter scenario, the foreign DNA is not targeted for attack by the cell, thus the replication of the virus proceeds and the host is not immune to viral infection. The interference stage can be mechanistically and temporally distinct from CRISPR acquisition and expression, yet for complete function as a defense system, all three phases must be functional.[16]

Stage 1: CRISPR spacer integration. Protospacers and protospacer-associated motifs (shown in red) are acquired at the “leader” end of a CRISPR array in the host DNA. The CRISPR array is composed of spacer sequences (shown in colored boxes) flanked by repeats (black diamonds). This process requires Cas1 and Cas2 (and Cas9 in type II[2]), which are encoded in the cas locus, which are usually located near the CRISPR array.

Stage 2: CRISPR expression. Pre-crRNA is transcribed starting at the leader region by the host RNA polymerase and then cleaved by Cas proteins into smaller crRNAs containing a single spacer and a partial repeat (shown as hairpin structure with colored spacers).

Stage 3: CRISPR interference. crRNA with a spacer that has strong complementarity to the incoming foreign DNA begins a cleavage event (depicted with scissors), which requires Cas proteins. DNA cleavage interferes with viral replication and provides immunity to the host. The interference stage can be functionally and temporarily distinct from CRISPR acquisition and expression (depicted by white line dividing the cell).

Transcription deactivation using dCas9

dCas9, also referred to as endonuclease deficient Cas9 can be utilized to edit gene expression when applied to the transcription binding site of the desired section of a gene. The optimal function of dCas9 is attributed to its mode of action. Gene expression is inhibited when nucleotides are no longer added to the RNA chain and therefore terminating elongation of that chain, and as a result affects the transcription process. This process occurs when dCas9 is mass-produced so it is able to affect the most amount of genes at any given time via a sequence specific guide RNA molecule. Since dCas9 appears to down regulate gene expression, this action is amplified even more when it is used in conjunction with repressive chromatin modifier domains.[17] The dCas9 protein has other functions outside of the regulation of gene expression. A promoter can be added to the dCas9 protein which allows them to work with each other to become efficient at beginning or stopping transcription at different sequences along a strand of DNA. These two proteins are specific in where they act on a gene. This is prevalent in certain types of prokaryotes when a promoter and dCas9 align themselves together to impede the ability of elongation of polymer of nucleotides coming together to form a transcribed piece of DNA. Without the promoter, the dCas9 protein does not have the same effect by itself or with a gene body.[18]

When examining the effects of repression of transcription further, H3K27, an amino acid component of a histone, becomes methylated through the interaction of dCas9 and a peptide called FOG1. Essentially, this interaction causes gene repression on the C + N terminal section of the amino acid complex at the specific junction of the gene, and as a result, terminates transcription.[19]

dCas9 also proves to be efficient when it comes to altering certain proteins that can create diseases. When the dCas9 attaches to a form of RNA called guide-RNA, it prevents the proliferation of repeating codons and DNA sequences that might be harmful to an organism's genome. Essentially, when multiple repeat codons are produced, it elicits a response, or recruits an abundance of dCas9 to combat the overproduction of those codons and results in the shut-down of transcription. dCas9 works synergistically with gRNA and directly affects the DNA polymerase II from continuing transcription.

Further explanation of how the dCas9 protein works can be found in their utilization of plant genomes by the regulation of gene production in plants to either increase or decrease certain characteristics. The CRISPR-CAS9 system has the ability to either upregulate or downregulate genes. The dCas9 proteins are a component of the CRISPR-CAS9 system and these proteins can repress certain areas of a plant gene. This happens when dCAS9 binds to repressor domains, and in the case of the plants, deactivation of a regulatory gene such as AtCSTF64, does occur.[20]

Bacteria are another focus of the usage of dCas9 proteins as well. Since eukaryotes have a larger DNA makeup and genome; the much smaller bacteria are easy to manipulate. As a result, eukaryotes use dCas9 to inhibit RNA polymerase from continuing the process of transcription of genetic material.[21]

Genomic editing in eukaryotic cells

While genomic editing in eukaryotic cells has been possible using various methods since the 1980s the methods employed had proved to be inefficient and impractical to implement on a larger scale. Genomic editing leads to irreversible changes to the gene. Working like genetic scissors, the Cas9 nuclease opens both strands of the targeted sequence of DNA to introduce the modification by one of two methods. Knock-in mutations, facilitated via Homology Directed Repair, is the traditional pathway of targeted genomic editing approaches.[22] This allows for the introduction of targeted DNA damage and repair. HDR employs the use of similar DNA sequences to drive the repair of the break via the incorporation of exogenous DNA to function as the repair template.[22] This method relies on the periodic and isolated occurrence of DNA damage at the target site in order for a repair to commence. Knock-out mutations caused by Cas9/CRISPR results in the repair of the double-strand break by means of NHEJ (Non-Homologous End Joining). NHEJ can often result random deletions or insertions at the repair site disrupting or altering gene functionality. Therefore, genomic engineering by CRISPR/Cas9 gives researches the ability to generated targeted random gene disruption. Because of this, the precision of genomic editing is a great concern. With the discovery of CRISPR and specifically the Cas9 nuclease molecule, efficient and highly selective editing is now a reality. Cas9 allows for a reliable method of creating a targeted break at a specific location as designated by the crRNA and tracrRna guide strands.[23] Cas9 derived from S. pyogenes has facilitated the targeted genomic modification in eukaryotic cells. The ease by which researches can insert Cas9 and template RNA in order to silence or cause point mutations on specific loci has proved invaluable to the quick and efficient mapping of genomic models and biological processes associated with various genes in a variety of eukaryotes. New engineered variant of the Cas9 nuclease have been developed that significantly reduce off-target manipulation. Called spCas9-HF1 (Streptococcus pyogenes Cas9 High Fidelity 1), has a success rate of modification in vivo of 85% and undetectable off-target manipulations as measured by genome wide break capture and targeted sequencing methods used to measure total genomic changes.[24][25]

Cellular modeling

Cas9 genomic modification has allowed for the quick and efficient generation of transgenic models within the field of genetics. Cas9 can be easily introduced into the target cells via plasmid transfection along with sgRNA in order to model the spread of diseases and the cell's response and defense to infection.[26] The ability of Cas9 to be introduced in vivo allows for the creation of more accurate models of gene function, mutation effects, all while avoiding the of off-target mutations typically observed with older methods of genetic engineering. The CRISPR and Cas9 revolution in genomic modeling doesn't only extent to mammals. Traditional genomic models such as Drosophila Melanogaster, one of the first model species, have seen further refinement in their resolution with the use of Cas9.[26] Cas9 uses cell-specific promoters allowing the a controlled use of the Cas9. Cas9 is an accurate method of treating diseases due to the targeting of the Cas9 enzyme only affecting certain cell types. The cells undergoing the Cas9 therapy can also be removed and reintroduced to provide amplified effects of the therapy.[9]

Structural studies of Cas9

Overview

Cas9 features a bi-lobed architecture with the guide RNA nestled between the alpha-helical lobe (blue; Fig. 2) and the nuclease lobe (cyan, orange and gray). These two lobes are connected through a single bridge helix. There are two nuclease domains located in the multi-domain nuclease lobe, the RuvC (gray) which cleaves the non-target DNA strand, and the HNH nuclease domain (cyan) that cleaves the target strand of DNA. The RuvC domain is encoded by sequentially disparate sites that interact in the tertiary structure to form the RuvC cleavage domain (See Figure 3).

Fig. 3: Crystal structure of Cas9 in the Apo form, as solved by M Jinek et al. in their 2014 Science paper. Structural rendition was performed using UCSF Chimera software.

A key feature of the target DNA is that it must contain a protospacer adjacent motif (PAM) consisting of the three-nucleotide sequence- NGG. This PAM is recognized by the PAM-interacting domain (PI domain, orange) located near the C-terminal end of Cas9. Cas9 undergoes distinct conformational changes between the apo, guide RNA bound, and guide RNA:DNA bound states, which are detailed below.

Crystal structures in detail

Cas9 recognizes the stem-loop architecture inherent in the CRISPR locus, which mediates the maturation of crRNA-tracrRNA ribonucleoprotein complex.[27] Cas9 in complex with CRISPR RNA (crRNA) and trans-activating crRNA (tracrRNA) further recognizes and degrades the target dsDNA.[28] In the co-crystal structure shown here (Fig. 4), the crRNA-tracrRNA complex is replaced by a chimeric single-guide RNA (sgRNA, in red) which has been proved to have the same function as the natural RNA complex.[3] The sgRNA base paired with target ssDNA is anchored by Cas9 as a T-shaped architecture. This crystal structure of the DNA-bound Cas9 enzyme reveals distinct conformational changes in the alpha-helical lobe with respect to the nuclease lobe, as well as the location of the HNH domain. The protein consists of a recognition lobe (REC) and a nuclease lobe (NUC). It should be noted that all regions except the HNH form tight interactions with each other and sgRNA-ssDNA complex, while the HNH domain forms few contacts with the rest of the protein. In another conformation of Cas9 complex observed in the crystal, the HNH domain is not visible. These structures suggest the conformational flexibility of HNH domain.

So far, three crystal structures have been studied and published. One representing a conformation of Cas9 in the apo state, and two representing Cas9 in the DNA bound state.

  1. Jinek et al. Structures of Cas9 endonucleases reveal RNA-mediated conformational activation. Science, Feb 2014 [29]
  2. Anders et al. Structural basis of PAM-dependent target DNA recognition by the Cas9 endonuclease. Nature Sept 2014 [30]
  3. Nishimasu et al. Crystal structure of Cas9 in complex with guide RNA and target DNA. Cell Feb 2014 [1]

The interactions between sgRNA and in cas9

CRISPR/Cas9

In sgRNA-Cas9 complex, based on the crystal structure, REC1, BH and PI domains have important contacts with backbone or bases in both repeat and spacer region.[1][30] Several Cas9 mutants including REC1 or REC2 domains deletion and residues mutations in BH have been tested. REC1 and BH related mutants show lower or none activity compared with wild type, which indicate these two domains are crucial for the sgRNA recognition at repeat sequence and stabilization of the whole complex. Although the interactions between spacer sequence and Cas9 as well as PI domain and repeat region need further studies, the co-crystal demonstrates clear interface between Cas9 and sgRNA.

Target digestion

Previous sequence analysis and biochemical studies have suggested Cas9 contain RNase H and HNH endonuclease homologous domains which are responsible for cleavages of two target DNA strands, respectively. These results are finally proved in the structure. Although the low sequence similarity, the sequence similar to RNase H has a RuvC fold (one member of RNase H family) and the HNH region folds as T4 Endo VII (one member of HNH endonuclease family). Previous works on Cas9 have demonstrated that HNH domain is responsible for complementary sequence cleavage of target DNA and RuvC is responsible for the non-complementary sequence.[31]

Problems bacteria pose to cas-9 editing

Most Archea and Bacteria stubbornly refuse to allow a cas-9 to edit their genome. This is because they can attach foreign DNA, that does not affect them, into their genome. Another way that these cells defy cas-9 is by process of “Restriction Modification System”. When a bacteriophage enters a bacteria or archea cell it is targeted by the RM system. The RM system then cuts the bacteriophages DNA into separate pieces by restriction enzymes and uses endonucleases to further destroy the strands of DNA. This poses a problem to cas-9 editing because the RM system also targets the foreign genes added by the cas-9 process.[32]

Applications of Cas9 to transcription tuning

Interference of transcription by dCas9

Due to the unique ability of Cas9 to bind to essentially any complement sequence in any genome, researchers wanted to use this enzyme to repress transcription of various genomic loci. To accomplish this, the two crucial catalytic residues of the RuvC and HNH domain can be mutated to alanine abolishing all endonuclease activity of Cas9. The resulting protein coined ‘dead’ Cas9 or ‘dCas9’ for short, can still tightly bind to dsDNA. This catalytically inactive Cas9 variant has been used for both mechanistic studies into Cas9 DNA interrogative binding and as a general programmable DNA binding RNA-Protein complex.

The interaction of dCas9 with target dsDNA is so tight that high molarity urea protein denaturant can not fully dissociate the dCas9 RNA-protein complex from dsDNA target.[33] dCas9 has been targeted with engineered single guide RNAs to transcription initiation sites of any loci where dCas9 can compete with RNA polymerase at promoters to halt transcription.[34] Also, dCas9 can be targeted to the coding region of loci such that inhibition of RNA Polymerase occurs during the elongation phase of transcription.[34] In Eukaryotes, silencing of gene expression can be extented by targeting dCas9 to enhancer sequences, where dCas9 can block assembly of transcription factors leading to silencing of specific gene expression.[6] Moreover, the guide RNAs provided to dCas9 can be designed to include specific mismatches to its complementary cognate sequence that will quantitatively weaken the interaction of dCas9 for its programmed cognate sequence allowing a researcher to tune the extent of gene silencing applied to a gene of interest.[34] This technology is similar in principle to RNAi such that gene expression is being modulated at the RNA level. However, the dCas9 approach has gained much traction as there exist less off-target effects and in general larger and more reproducible silencing effects through the use of dCas9 compared to RNAi screens.[35] Furthermore, because the dCas9 approach to gene silencing can be quantitatively controlled, a researcher can now precisely control the extent to which a gene of interest is repressed allowing more questions about gene regulation and gene stoichiometry to be answered.

Beyond direct binding of dCas9 to transcriptionally sensitive positions of loci, dCas9 can be fused to a variety of modulatory protein domains to carry out a myriad of functions. Recently, dCas9 has been fused to chromatin remodeling proteins(HDACs/HATs) to reorganize the chromatin structure around various loci.[34] This is an important in targeting various eukaryotic genes of interest as heterochromatin structures hinder Cas9 binding. Furthermore, because Cas9 can react to heterochromatin, it is theorized that this enzyme can be further applied to studying the chromatin structure of various loci.[34] Additionally, dCas9 has been employed in genome wide screens of gene repression. By employing large libraries of guide RNAs capable of targeting thousands of genes, genome wide genetic screens using dCas9 have been conducted.[36]

Another method for silencing transcription with Cas9 is to directly cleave mRNA products with the catalytically active Cas9 enzyme.[37] This approach is made possible by hybridizing ssDNA with a PAM complement sequence to ssRNA allowing for a dsDNA-RNA PAM site for Cas9 binding. This technology makes available the ability to isolate endogenous RNA transcripts in cells without the need to induce chemical modifications to RNA or RNA tagging methods.

Transcription activation by dCas9 fusion proteins

In contrast to silencing genes, dCas9 can also be used to activate genes when fused to transcription activating factors.[34] These factors include subunits of bacterial RNA Polymerase II and traditional transcription factors in eukaryotes. Recently, genome-wide screens of transcription activation have also been accomplished using dCas9 fusions named ‘CRISPRa’ for activation.[36]

Possible solutions to bacterium cas-9 editing

Recent studies have shown that with some minor modification to the cas-9 protein it is possible to bypass the “Restriction Modification System”. The process that achieves this is called “Photospacing” which means that the cas-9 system will copy a section of DNA from the host and insert it as a spacer gene into its own genome.[38] Though we do not fully understand why “photospacers” work we do understand what they do. When a prokaryote “senses” a part of its own genetic coding inside the foreign entity it is less likely to destroy the cas-9 insertion. This is known as the beginning of cas-9 immunity.[39]

See also

References

  1. ^ a b c Nishimasu H, Ran FA, Hsu PD, Konermann S, Shehata SI, Dohmae N, Ishitani R, Zhang F, Nureki O (February 2014). "Crystal structure of Cas9 in complex with guide RNA and target DNA". Cell. 156 (5): 935–49. doi:10.1016/j.cell.2014.02.001. PMC 4139937. PMID 24529477.
  2. ^ a b c Heler R, Samai P, Modell JW, Weiner C, Goldberg GW, Bikard D, Marraffini LA (March 2015). "Cas9 specifies functional viral targets during CRISPR-Cas adaptation". Nature. 519 (7542): 199–202. doi:10.1038/nature14245. PMC 4385744. PMID 25707807.
  3. ^ a b c d Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E (August 2012). "A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity". Science. 337 (6096): 816–21. doi:10.1126/science.1225829. PMID 22745249.
  4. ^ Mali P, Esvelt KM, Church GM (October 2013). "Cas9 as a versatile tool for engineering biology". Nature Methods. 10 (10): 957–63. doi:10.1038/nmeth.2649. PMC 4051438. PMID 24076990.
  5. ^ Mali P, Aach J, Stranges PB, Esvelt KM, Moosburner M, Kosuri S, Yang L, Church GM (September 2013). "CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering". Nature Biotechnology. 31 (9): 833–8. doi:10.1038/nbt.2675. PMC 3818127. PMID 23907171.
  6. ^ a b Gilbert LA, Larson MH, Morsut L, Liu Z, Brar GA, Torres SE, Stern-Ginossar N, Brandman O, Whitehead EH, Doudna JA, Lim WA, Weissman JS, Qi LS (July 2013). "CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes". Cell. 154 (2): 442–51. doi:10.1016/j.cell.2013.06.044. PMC 3770145. PMID 23849981.
  7. ^ Esvelt KM, Smidler AL, Catteruccia F, Church GM (July 2014). "Concerning RNA-guided gene drives for the alteration of wild populations". eLife. 3. doi:10.7554/eLife.03401. PMC 4117217. PMID 25035423.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  8. ^ Cyranoski, David; Reardon, Sara (22 April 2015). "Chinese scientists genetically modify human embryos". Nature. doi:10.1038/nature.2015.17378. {{cite journal}}: Unknown parameter |name-list-format= ignored (|name-list-style= suggested) (help)
  9. ^ a b Doudna, Jennifer A.; Mali, Prashant (2016). CRISPR-Cas : a laboratory manual. Doudna, Jennifer A.,, Mali, Prashant. Cold Spring Harbor, New York. ISBN 9781621821304. OCLC 922914104.{{cite book}}: CS1 maint: location missing publisher (link)
  10. ^ "CRISPR/Cas9 gene editing". doi:10.1036/1097-8542.yb150927. {{cite journal}}: Cite journal requires |journal= (help)
  11. ^ Ebina H, Misawa N, Kanemura Y, Koyanagi Y (2013-08-26). "Harnessing the CRISPR/Cas9 system to disrupt latent HIV-1 provirus". Scientific Reports. 3 (1): 2510. doi:10.1038/srep02510. PMC 3752613. PMID 23974631.
  12. ^ Li H, Sheng C, Wang S, Yang L, Liang Y, Huang Y, Liu H, Li P, Yang C, Yang X, Jia L, Xie J, Wang L, Hao R, Du X, Xu D, Zhou J, Li M, Sun Y, Tong Y, Li Q, Qiu S, Song H (2017-03-22). "Removal of Integrated Hepatitis B Virus DNA Using CRISPR-Cas9". Frontiers in Cellular and Infection Microbiology. 7: 91. doi:10.3389/fcimb.2017.00091. PMC 5360708. PMID 28382278.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  13. ^ Bogdanovich, Sasha (2016-02-10). "Gene Editing With Crispr/Cas9: A New Therapeutic Hope for Duchenne Muscular Dystrophy". MOJ Cell Science & Report. 3 (2). doi:10.15406/mojcsr.2016.03.00050. ISSN 2374-6912. {{cite journal}}: Unknown parameter |name-list-format= ignored (|name-list-style= suggested) (help)
  14. ^ Garneau JE, Dupuis MÈ, Villion M, Romero DA, Barrangou R, Boyaval P, Fremaux C, Horvath P, Magadán AH, Moineau S (November 2010). "The CRISPR/Cas bacterial immune system cleaves bacteriophage and plasmid DNA". Nature. 468 (7320): 67–71. CiteSeerX 10.1.1.451.9645. doi:10.1038/nature09523. PMID 21048762.
  15. ^ Horvath P, Barrangou R (January 2010). "CRISPR/Cas, the immune system of bacteria and archaea". Science. 327 (5962): 167–70. doi:10.1126/science.1179555. PMID 20056882.
  16. ^ Karginov FV, Hannon GJ (January 2010). "The CRISPR system: small RNA-guided defense in bacteria and archaea". Molecular Cell. 37 (1): 7–19. doi:10.1016/j.molcel.2009.12.033. PMC 2819186. PMID 20129051.
  17. ^ Jensen ED, Ferreira R, Jakočiūnas T, Arsovska D, Zhang J, Ding L, et al. (March 2017). "Transcriptional reprogramming in yeast using dCas9 and combinatorial gRNA strategies". Microbial Cell Factories. 16 (1): 46. doi:10.1186/s12934-017-0664-2. PMC 5353793. PMID 28298224.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  18. ^ Pinto BS, Saxena T, Oliveira R, Méndez-Gómez HR, Cleary JD, Denes LT, McConnell O, Arboleda J, Xia G, Swanson MS, Wang ET (November 2017). "Impeding Transcription of Expanded Microsatellite Repeats by Deactivated Cas9". Molecular Cell. 68 (3): 479–490.e5. doi:10.1016/j.molcel.2017.09.033. PMC 6013302. PMID 29056323.
  19. ^ O'Geen H, Ren C, Nicolet CM, Perez AA, Halmai J, Le VM, Mackay JP, Farnham PJ, Segal DJ (September 2017). "dCas9-based epigenome editing suggests acquisition of histone methylation is not sufficient for target gene repression". Nucleic Acids Research. 45 (17): 9901–9916. doi:10.1093/nar/gkx578. PMC 5622328. PMID 28973434.
  20. ^ Lowder LG, Paul JW, Qi Y (2017). Multiplexed Transcriptional Activation or Repression in Plants Using CRISPR-dCas9-Based Systems. Vol. 1629. pp. 167–184. doi:10.1007/978-1-4939-7125-1_12. ISBN 978-1-4939-7124-4. PMID 28623586. {{cite book}}: |journal= ignored (help)
  21. ^ Barrangou R, Horvath P (June 2017). "A decade of discovery: CRISPR functions and applications". Nature Microbiology. 2: 17092. doi:10.1038/nmicrobiol.2017.92. PMID 28581505.
  22. ^ a b Zhang JH, Adikaram P, Pandey M, Genis A, Simonds WF (April 2016). "Optimization of genome editing through CRISPR-Cas9 engineering". Bioengineered. 7 (3): 166–74. doi:10.1080/21655979.2016.1189039. PMC 4927198. PMID 27340770.
  23. ^ Zhang JH, Pandey M, Kahler JF, Loshakov A, Harris B, Dagur PK, Mo YY, Simonds WF (November 2014). "Improving the specificity and efficacy of CRISPR/CAS9 and gRNA through target specific DNA reporter". Journal of Biotechnology. 189: 1–8. doi:10.1016/j.jbiotec.2014.08.033. PMC 4252756. PMID 25193712.
  24. ^ Kleinstiver BP, Pattanayak V, Prew MS, Tsai SQ, Nguyen NT, Zheng Z, Joung JK (January 2016). "High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects". Nature. 529 (7587): 490–5. doi:10.1038/nature16526. PMC 4851738. PMID 26735016.
  25. ^ Hsu PD, Lander ES, Zhang F (June 2014). "Development and applications of CRISPR-Cas9 for genome engineering". Cell. 157 (6): 1262–78. doi:10.1016/j.cell.2014.05.010. PMC 4343198. PMID 24906146.
  26. ^ a b Dow LE (October 2015). "Modeling Disease In Vivo With CRISPR/Cas9". Trends in Molecular Medicine. 21 (10): 609–621. doi:10.1016/j.molmed.2015.07.006. PMC 4592741. PMID 26432018.
  27. ^ Wiedenheft B, Sternberg SH, Doudna JA (February 2012). "RNA-guided genetic silencing systems in bacteria and archaea". Nature. 482 (7385): 331–8. doi:10.1038/nature10886. PMID 22337052.
  28. ^ Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA, Zhang F (November 2013). "Genome engineering using the CRISPR-Cas9 system". Nature Protocols. 8 (11): 2281–2308. doi:10.1038/nprot.2013.143. PMC 3969860. PMID 24157548.
  29. ^ Jinek M, Jiang F, Taylor DW, Sternberg SH, Kaya E, Ma E, Anders C, Hauer M, Zhou K, Lin S, Kaplan M, Iavarone AT, Charpentier E, Nogales E, Doudna JA (March 2014). "Structures of Cas9 endonucleases reveal RNA-mediated conformational activation". Science. 343 (6176): 1247997. doi:10.1126/science.1247997. PMC 4184034. PMID 24505130.
  30. ^ a b Anders C, Niewoehner O, Duerst A, Jinek M (September 2014). "Structural basis of PAM-dependent target DNA recognition by the Cas9 endonuclease". Nature. 513 (7519): 569–73. doi:10.1038/nature13579. PMC 4176945. PMID 25079318.
  31. ^ Chen H, Bailey S (February 2016). "Structural biology. Cas9, poised for DNA cleavage". Science. 351 (6275): 811–2. doi:10.1126/science.aaf2089. PMID 26912877.
  32. ^ Kusano K, Naito T, Handa N, Kobayashi I (November 1995). "Restriction-modification systems as genomic parasites in competition for specific sequences". Proceedings of the National Academy of Sciences of the United States of America. 92 (24): 11095–9. doi:10.1073/pnas.92.24.11095. PMC 40578. PMID 7479944.
  33. ^ Sternberg SH, Redding S, Jinek M, Greene EC, Doudna JA (March 2014). "DNA interrogation by the CRISPR RNA-guided endonuclease Cas9". Nature. 507 (7490): 62–7. doi:10.1038/nature13011. PMC 4106473. PMID 24476820.
  34. ^ a b c d e f Bikard D, Jiang W, Samai P, Hochschild A, Zhang F, Marraffini LA (August 2013). "Programmable repression and activation of bacterial gene expression using an engineered CRISPR-Cas system". Nucleic Acids Research. 41 (15): 7429–37. doi:10.1093/nar/gkt520. PMC 3753641. PMID 23761437.
  35. ^ Heintze J, Luft C, Ketteler R (2013). "A CRISPR CASe for high-throughput silencing". Frontiers in Genetics. 4: 193. doi:10.3389/fgene.2013.00193. PMC 3791873. PMID 24109485.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  36. ^ a b Gilbert LA, Horlbeck MA, Adamson B, Villalta JE, Chen Y, Whitehead EH, Guimaraes C, Panning B, Ploegh HL, Bassik MC, Qi LS, Kampmann M, Weissman JS (October 2014). "Genome-Scale CRISPR-Mediated Control of Gene Repression and Activation". Cell. 159 (3): 647–61. doi:10.1016/j.cell.2014.09.029. PMC 4253859. PMID 25307932.
  37. ^ O'Connell MR, Oakes BL, Sternberg SH, East-Seletsky A, Kaplan M, Doudna JA (December 2014). "Programmable RNA recognition and cleavage by CRISPR/Cas9". Nature. 516 (7530): 263–6. doi:10.1038/nature13769. PMC 4268322. PMID 25274302.
  38. ^ Barrangou R, Fremaux C, Deveau H, Richards M, Boyaval P, Moineau S, Romero DA, Horvath P. 2007 CRISPR provides acquired resistance against viruses in prokaryotes. Science 315, 1709–1712.
  39. ^ Fineran PC, Charpentier E (December 2012). "Memory of viral infections by CRISPR-Cas adaptive immune systems: acquisition of new information". Virology. 434 (2): 202–9. doi:10.1016/j.virol.2012.10.003. PMID 23123013.

Further reading