what new technologies allowed humans to study cells? who invented it?
Curr Stem Cell Rep. 2016; 2(one): ix–20.
A Wide Overview and Review of CRISPR-Cas Technology and Stem Cells
Simon N. Waddington
aneGene Transfer Engineering science Grouping, Establish for Women'south Health, Academy College London, 86-96 Chenies Mews, London, Britain
iiAntiviral Gene Therapy Research Unit of measurement, Faculty of Health Sciences, University of the Witswatersrand, Johannesburg, S Africa
Riccardo Privolizzi
iGene Transfer Engineering Group, Constitute for Women'south Health, Academy College London, 86-96 Chenies Mews, London, United kingdom
Rajvinder Karda
3Kinesthesia of Medicine, Department of Surgery and Cancer, Imperial College London, London, Great britain
Helen C. O'Neill
fourPreimplantation Genetics and Embryology Group, Constitute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX UK
Abstruse
The top of four decades of research, induced pluripotent stem cells (iPSCs), and genome editing with the advent of clustered, regularly interspaced, curt palindromic repeats (CRISPR) now promise to take drug development and regenerative medicine to new levels and to enable the interrogation of affliction mechanisms with a hitherto unimaginable level of model fidelity. Autumn 2014 witnessed the first patient receiving iPSCs differentiated into retinal pigmented epithelium to care for macular degeneration. Technologies such as 3D bioprinting may now exploit these advances to manufacture organs in a dish. Equally enticing every bit these prospects are, these technologies need a deeper agreement, which will lead to improvements in their safety and efficacy. For case, precise and more efficient reprogramming for iPSC production is a requisite for wider clinical adoption. Improving awareness of the roles of long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) and genomic epigenetic status volition contribute to the achievement of these aims. Similarly, increased efficiency, avoidance of astray effects, and expansion of bachelor target sequences are disquisitional to the uptake of genome editing technology. In this review, we survey the historical evolution of genetic manipulation and stem cells. We explore the potential of genetic manipulation of iPSCs for in vitro disease modeling, generation of new animal models, and clinical applicability. We highlight the aspects that define CRISPR-Cas every bit a breakthrough engineering, look at gene correction, and consider some important ethical and societal implications of this approach.
Keywords: Genome engineering, Gene therapy, CRISPR-Cas, iPSCs, Man Genome Editing
Introduction
The time has come, it may be said,
To dream of many things;
Of genes—and life—and human cells—
Of Medicine—and kings—
Edward L Tatum, Perspectives in biological science and medicine 1966 [1]
In 2015, Science Magazine recognized "CRISPR genome editing" as breakthrough of the yr. Six years earlier, the "Render of Gene Therapy" was recognized every bit a runner upwardly, and in 2008, the winning accolade was awarded to "Cellular Reprogramming." These combined technologies are not only showing slap-up promise just also are really starting to deliver on bully promises. Yet they are not new; their births tin each exist traced dorsum several decades. To provide context on today's achievements, and possibly to predict the forthcoming trajectory, it is worth because their developmental timelines (summarized in Fig.1).
A flow diagram illustrating the generation of stalk cell inquiry, transgenesis, genetic engineering, and gene transfer technology. This time line is carve up into three major engineering science streams which converged to establish the utilise of CRISPR-Cas engineering science and stem cells
In 1941, Edward Tatum and George Beadle used the fungus Neurospora to make it at their paradigmatic "one gene one enzyme" hypothesis [ii]; for this, they were jointly awarded the Nobel Prize in Physiology with Joshua Lederberg in 1958. In 1961, Marshall Nirenberg and Heinrich Matthei began to decipher the relationship between codons and amino acids [3]. Mindful of these advances, Edward Tatum tentatively predicted regenerative medicine and gene therapy:
Hence, information technology can exist suggested that the offset successful genetic engineering science will be done with the patient's own cells, for instance, liver cells, grown in civilisation. The desired new gene volition be introduced, by directed mutation, from normal cells of another donor by transduction or past straight DNA transfer. The rare prison cell with the desired change will then be selected, grown into a mass civilization, and reimplanted in the patient's liver.
Precedents for the introduction or transfer of genes from one cell to some other exist in microbial systems and are now being tried with mammalian cells in culture…If this tin can be done successfully … information technology will facilitate the evolution of a mammalian somatic prison cell genetics. It volition also bring us considerably closer to successful genetic engineering [1].
The development of prison cell lines in the early 1960s tested the notion that strange Dna could be permanently and stably introduced into mammalian cells with both functionality and heritability. Conceptually, these studies were derived from work on pneumococci carried out past Avery, MacLeod, and McCarty ii decades earlier, showing that DNA was involved in bacterial transformation [4].
It became clear past the mid-late 1960s that genetic transformation by exogenous Deoxyribonucleic acid was more efficient than yet suspected. Work on viral Dna in SV40-transformed cells showed that viral genomes likewise had the capacity to be covalently and stably transformed into target cells [five, vi]. These experiments preceded the era of recombinant DNA, then it was unclear how viruses may be modified to express or contain foreign genes and be used as therapeutic agents. Rogers and co-workers were the first to use viruses to transfer foreign genes into human cells [7]. However, their handling of two girls with hyperargininemia failed to accomplish any therapeutic issue [8].
At this fourth dimension, the commencement tools for recombinant DNA engineering were beingness assembled; the appearance of mammalian genetic modification was shut, and scientists recognized the need to ascertain both safety and regulation [9]. A prime example of the demand to enforce this regulation was the Cline experiment in 1979, which led to the introduction of the man globin cistron into murine bone marrow cells. This partially repopulated the bone marrow of irradiated mice with the genetically modified marrow cells. This technique, using calcium phosphate transfection, was prematurely applied on human bone marrow cells and transfused into ii thalassemia patients in 1980. The handling was not successful and was done without the approval of the FDA [10, 11].
Michael Wigler and colleagues (amongst others) had previously used calcium phosphate transfection to deliver a fragment of viral DNA containing the thymidine kinase gene into a mouse cell line. Nonetheless, this was woefully inefficient, with successful transfection in fewer than i in 100,000 cells [12]. By directly intra-nuclear injection of DNA using a glass micropipette, Mario Capecchi was able to better hugely upon the efficiency, achieving a success rate of i in 5 cells [13]. This set the scene for the generation of transgenic mice, which was achieved past four dissimilar groups within two years [xiv–17]. One of these studies demonstrated the insertion of the rabbit β-globin factor and germ line transmission into the resulting mouse strain [xvi]. This technology provided no ability to target the genomic insertions, which remained completely random. However, Capecchi's team noted that inserted DNA oft assumed a concatemeric form, concluding this may exist a product of homologous recombination [18]. This hinted at the prospect that it might be possible to target exogenous Dna to specific genomic loci, which is precisely what Oliver Smithies' team achieved in 1988. They flanked the prospective DNA insert with sequences homologous to the homo β-globin locus and used electroporation to deliver this to a hybrid human cell, observing specific insertion into this locus [19].
In 1961, the same year that Nirenberg and Matthei were decrypting codon usage, James Till and Ernest McCulloch first demonstrated the existence of multipotent stem cells: os marrow-derived cells capable of clonal expansion, colony formation, and self-renewal [20]. 2 decades afterward, pioneering work by Martin Evans and Matthew Kaufman revealed that early mouse embryos contain pluripotent cells—cells that have the ability to become any cell in the torso (except the placenta). This so-called prison cell potency was divers when they were transplanted into blastocysts of mice of a different strain and produced chimeric animals. By meticulous command of isolation timing and culture atmospheric condition, they were able to maintain these cells for the first time and observe in vitro differentiation and teratoma formation when implanted into immune-scarce mice [21]. The formation of a teratoma, which contains multiple tissue components, signified the ability of these cells to develop into more than one germ layer. Shortly afterwards, Gail Martin generated like data using a teratocarcinoma feeder prison cell line to maintain these stem cells. She gave these cells the proper noun with which we are and then familiar now; "embryonic stalk cells" [22]. In 1984, Martin Evans and colleagues demonstrated that they could generate germ line chimeric mice by the introduction of embryonic stem cells, from established lines, into mouse blastocysts before reimplantation [23].
The close of the 1980s saw accomplishment of two major goals; firstly, Kirk Thomas and Mario Capecchi described site-directed mutagenesis of the hypoxanthine phosphoribosyl transferase gene in mouse embryonic stalk cells using electroporation and homologous recombination [24]. Secondly, in 1989, Smithies and team took this technology to a natural conclusion, by generating germ line chimeric mice derived from embryonic stem cells which had undergone site-directed mutagenesis using homologous recombination [25]. For a decade, the application of this technology to generate mouse models of human affliction was embraced enthusiastically by the biomedical research community. Then, in 1998, the potential of this technology was taken to a new level, when James Thompson and colleagues generated the beginning human embryonic stem cell lines from blastocysts. These could exist maintained for months in an undifferentiated country using a mouse embryonic fibroblast feeder line simply were capable of generating cells of endodermal, mesodermal, and ectodermal lineages with a teratoma analysis [26]. V years subsequently, James Thompson and Thomas Zwaka demonstrated site-directed mutagenesis of homo embryonic stem cells using homologous recombination and electroporation [27].
1 major technological stream missing from this narrative, so far, has been that of viral vector-based factor transfer technology. More than fifteen years afterward Edward Tatum had suggested that viruses could be used to deliver genetic cloth for therapeutic benefit [1], three quite different virus-based vectors were being adult—those based on adenoviruses, retroviruses and adeno-associated viruses (AAVs). Four groups were involved in the construction of retroviral vectors. They demonstrated gene delivery and genomic integration into mammalian cells at efficiency vastly superior to non-viral methods, such as calcium phosphate transfection and glass pipette microinjection [28–31]. Separately, groups were developing adenovirus-based vectors; these vectors showed hope for highly efficient delivery, the chapters for delivering longer DNA sequences, and potent expression of the desired protein [32–34]. In improver, Paul Hermonat and Nicholas Muzyczka adult vectors based upon AAV, which served every bit tools not only for the study of factor role and regulation, but became invaluable gene therapy vectors [35]. Past 1992, numerous studies had demonstrated the delivery of therapeutically relevant human genes to rodent somatic tissues and human gene therapy trials were being initiated. A review article in the journal Nature entitled "Human gene therapy comes of historic period" typified the overweening enthusiasm of the field [36] and some brash confronting hyperbole, which risked damaging the entire subject field [37].
It was 2002 before the results of the commencement successful human cistron therapy trial were published. Infants with Ten-linked severe combined immunodeficiency (SCID) received autologous hematopoietic stalk prison cell transplants; these cells had been transduced with a retrovirus vector conveying functional common gamma concatenation gene. Reconstitution of immune office was reported in iv of the five patients [38]. However, the post-obit twelvemonth, serious adverse events of leukemogenesis arising from this factor therapy were reported [39]; the field was being viewed with increasing skepticism. Therapeutic cistron delivery required more preclinical piece of work on vector safety, targeting, and efficacy, and for the remainder of the decade, this was performed in adverse financial weather condition, as grant funding and industrial recruitment flattened [40].
Notwithstanding, as a primal research tool, retrovirus vectors were soon to prove their immense utility. A retrovirus library of 24 transcription factor genes was used to transduce mouse embryonic fibroblasts; these genes were each selected as playing a potential role in stem cell pluripotency. It is testament to the transduction efficiency of this vector that simultaneous delivery of 24 separate genes was feasible. Past withdrawing or combining unlike factors, four of these were found necessary and sufficient to revert mouse embryonic fibroblasts to a pluripotent stem jail cell state. These were Oct3/iv, Sox2, c-Myc, and Klf4, too known equally the Yamanaka factors [41]. Generation of the resulting induced pluripotent stalk cells (iPSCs) won, for Shinya Yamanaka, a share of the 2012 Nobel Prize in Physiology or Medicine. A year later, Yamanaka and colleagues demonstrated that germ line transgenic mice could be generated from iPSCs [42]. Both his team [43] and that of James Thomson [44] proceeded to generate iPSCs from human fibroblasts. In 2014, the first patient received autologous iPSCs, differentiated into retinal pigmented epithelium, to treat age-related macular degeneration. Numerous other clinical trials using embryonic stem cells were becoming bachelor from 2010 onwards (reviewed in [45••]). The need to address the reproducibility and scaling difficulties in iPSC production was recognized by Daniel Paull and colleagues who developed a robotic arrangement for derivation, label, and differentiation of iPSCs [46]. The automation of jail cell production has advanced fifty-fifty further as seen with the recent production of bioprinted mini-livers from iPSCs [47].
Stem cell development has seen continuous growth and improvement since its inception. Originally, iPSCs were generated by infecting cells with a cocktail of retroviral vectors; since these vectors integrate into the host genome, persistent expression of these factors and insertional mutagenesis increment the risk of oncogenesis. Therefore, the means of expressing reprogramming factors for sufficient duration, in the correct sequence, and in optimal amounts, are being adult. For example, Warren and colleagues designed synthetic mRNA to deliver reprogramming factors transiently still efficiently [48]. Other strategies are reviewed past Singh and co-workers [49]. It has been shown from unmarried-cell analyses that reprogramming consists of early stochastic and subsequent hierarchical events [50]. More than contempo studies take delineated the epigenetic changes over the course of reprogramming [51]; the dynamics of long not-coding RNA (lncRNA) expression [52] and the interaction between microRNAs (miRNAs) and the core reprogramming factors [53]. These, and other such studies, will exist valuable in informing how to maximize the efficiency and safe of reprogramming.
Concurrent with these tremendous advances in stalk cell biological science, gene therapy researchers were investigating multiple strategies to meliorate vector safe. As with the X-SCID trial, which led to leukemia, a 2d trial for Wiskott-Aldrich syndrome also caused leukemia in seven out of ten patients. Both trials availed of gamma retrovirus vectors, in this case for ex vivo hematopoietic stem cell gene therapy [54]. Gamma retroviral vectors integrate in a semi-random manner, and the expression cassette tin affect genes proximal to the insertion sites, likely accounting for the leukemogenesis.
These previous efforts focused on the incorporation of DNA via viral or vector-based delivery methods in order to express a missing protein where the endogenous genome fails to do so. In recent years, an increased understanding of nuclease function has enabled more direct DNA editing. These methods aim to restore normal factor part in situ, which reduces the risks associated with random integration, as genes are controlled using endogenous regulatory elements [55].
Crucially, targeted genome editing requires the double-stranded cleavage of Dna at the genomic locus to be modified. These double-stranded breaks (DSBs) are induced by nucleases and tin can be repaired by one of two mechanisms conserved beyond multiple organisms and cell types (Fig.2): non-homologous finish-joining (NHEJ) and homology-directed repair (HDR) [56].
An illustration of genome editing with CRISPR-Cas9. The knock-out approach results in a loss of office of the target DNA double strand breaks by non-homologous cease-joining. The knock-in results in an insertion at the repair site which exploits endogenous homology-directed repair
Historically, factor-specific targeting has been express to mouse embryonic stem cells. Since the discovery of iPSCs, many advances have been made in the field, with the successful differentiation into several specific cell types and institution of patient-derived affliction models [41, 57]. Establishment of a specific mutation/disease model has, until recently, relied upon traditionally low efficiency homologous recombination protocols or upon RNA interference (RNAi) (Tabular array 1). Platforms such as meganucleases, zinc finger nucleases (ZFNs), and transcription activator-like effector nucleases (TALENs) have since been developed. These rely on poly peptide-based systems for nuclease-directed DSBs, effectively inducing breaks that stimulate NHEJ or HDR at the specified genomic locations [58]. Such developments have permitted efficient genome editing in transformed and chief cells that were previously thought to be out of the scope of such genetic manipulation [59]. Indeed, TALENs have been used to efficiently generate mutant alleles in homo pluripotent stalk cells (hPSCs) of 15 unlike genes, as a means of performing affliction modeling [60].
Tabular array ane
Glossary of terms
| AAV (adeno-associated virus) | A viral vector system used for factor commitment. |
| Chimera | A unmarried organism composed of cells from different zygotes. |
| Germ line therapy | Insertion of Dna into germ line cells (egg or sperm) so that the offspring will accept the inserted factor. |
| gRNA | Guide RNA. |
| Hematopoietic stem cells | Unspecialized precursor cells that volition develop into mature blood cells. |
| Pluripotent stem cells | Stem cells that can become all cell types found in an implanted embryo, fetus, or developed organism (excluding trophoblast and placenta). |
| Recombinant Deoxyribonucleic acid | A novel Dna sequence formed by the joining, usually in vitro, of two non-homologous Dna molecules. |
| Retroviral vector | A disabled RNA virus in which the viral genes have been replaced with engineered sequences. |
| RuvC | An endonuclease domain named for an Due east. coli protein involved in Dna repair. |
| sgRNA | Single guide RNA. |
| Stem cells | Cells with the ability to separate for indefinite periods in civilization and to give ascension to specialized cells. |
| tracrRNA, trRNA | Trans-activating crRNA. |
While these site-specific nuclease technologies accept fabricated important advances in gene therapy, each has its own fix of associated advantages and disadvantages, such every bit cost and difficulty of synthesis [61]. The CRISPR-Cas (curt for clustered, regularly interspaced, brusk palindromic repeats/CRISPR-associated) technology has gained wide success in the global scientific community, where its ability as "molecular scissors" has revolutionized the prospects of genome editing [62, 63]. Since 2012, the CRISPR-Cas system has shown potential to satisfy the shortcomings of its predecessors by adapting a naturally occurring mechanism from prokaryotic to eukaryotic systems [64–67]. The combination of the ii powerful technologies of iPSCs and CRISPR-Cas is kickoff to revolutionize genetic research and boost the field of precision medicine.
In considering the convergent stories of stem cell research, transgenesis, genetic engineering, and gene transfer technology, several trends emerge: (i) Each intersection of these technologies has allowed the generation of models with increasing fidelity to the human disease; this has proved valuable both in interrogating the underlying pathology and in the development and testing of therapies. (ii) Genetic engineering has improved by many orders of magnitude, in both efficiency and precision. (iii) These technologies have transitioned from existence tools of bones enquiry to having actual, or at least potential, clinical application. (4) Improving the safety of these tools, in the laboratory and more recently in the clinic, has remained an indelible imperative.
CRISPR/Cas Machinery and Mechanisms of Action
CRISPR systems, together with cas genes, are highly diverse mechanisms of adaptable immunity used by many leaner and archaea to protect themselves from invading viruses, plasmids, and other foreign nucleic acids [68–71]. CRISPRs consist of a succession of highly conserved curt repeated sequences (23–44 bp in length) separated by similarly sized "spacers." These spacers are unique sequences usually originating from phage or plasmid DNA [72]. These adaptive systems tin can learn to recognize specific features of invading pathogens. The addition of these motifs to the host genome allows for the recognition and destruction of subsequent invasions from genetically similar pathogens. Kickoff observed in Escherichia coli in 1980, CRISPR loci take now been institute in 84 % of sequenced archaeal genomes and approximately 45 % of bacterial genomes [73, 74]. Their function was confirmed in Streptococcus thermophilus in 2007, with the sit-in that resistance against a bacteriophage could be acquired by the integration of virus fragments into the CRISPR locus [68].
Side by side to the CRISPRs are a set up of CRISPR-associated (cas) genes that code for proteins essential for CRISPR activeness. Comparative genomics of bacterial and archaeal genomes have suggested upwards of 45 cas gene families. The only two of these genes present in all 45 families are cas1 and cas2; both of which are involved in spacer acquisition [75, 76]. In that location are 3 major types of CRISPR-Cas systems (type I, type 2, and type III), which tin exist further divided into ten different subtypes. Each form contains different sets of genes, repeat patterns, and species ranges.
The CRISPR/Cas system of immunity is comprised of iii steps; adaptation, expression, and interference. The adaption stage involves the recognition and cleavage of a protospacer from invading DNA by the cas genes. The subsequent insertion (acquisition) of foreign DNA (spacers) into the CRISPR locus is besides referred to as spacer acquisition or immunization. The expression phase refers to the expression of relevant cas genes and their proteins leading to the transcription of the CRISPR array into a long RNA molecule called the forerunner CRISPR RNA (pre-crRNA). Cas proteins and other accompaniment factors then process this further into brusque mature crRNA. In the terminal interference stage, this mature crRNA and other cas proteins recognize foreign nucleic acrid and destroy it. This is also referred to as the immunity stage, which these mechanisms mimic [77].
Both the expression and interference stage occur differently in each of the CRISPR systems. In blazon I systems, Cas6e/Cas6f cut at the junction of single-stranded RNA (ssRNA) and double-stranded RNA (dsRNA) formed by hairpin loops. Trans-activating (tracr) RNAs are used in blazon II systems to form dsRNA, cleaved by Cas9 and RNaseIII. Type 3 systems use a Cas6 homolog in the direct repeat for cleavage and do not require hairpin loops [74].
With each integration of invading Deoxyribonucleic acid, spacer repeat units are formed. The preference of the host to recognize and create spacer precursors (protospacers) from specific sequences along the invading genome is determined past the protospacer next motifs (PAMs) [78]. PAMs are short DNA sequences (3–5 bp) that differ between the variants of CRISPR and have been shown to exist important for conquering in blazon I and blazon II, but not type III systems [79]. The process of spacer acquisition occurs in a directional manner whereby new spacers are preferentially added at one side of the CRISPR (the leader sequence) [lxxx, 81]. The leader sequence contains promoter elements, bounden sites, and elements important for spacer integration. The cumulative addition of spacers containing foreign nucleotide sequences therefore acts as a chronological record of the bequeathed viral invasions since protection is then inherited past the offspring [82].
A full general theme can be followed across all iii systems of CRISPR-mediated immunity. In all systems, the CRISPR locus is transcribed to generate a RNA-guide protein, Cas ribonucleases process the RNA guide to form a CRISPR ribonucleoprotein (crRNP) complex. This leads to the formation of a long master transcript, known as the pre-crRNA, which can contain secondary structures, called hairpins, if palindromic sequences be within the CRISPR sequence. These pre-crRNA sequences are and so candy into smaller units, which correspond to the spacer and repeats regions [83].
In August 2012, Jennifer Doudna and Emmanuelle Charpentier co-authored a key study demonstrating the technical potential of CRISPR-Cas to cut and splice genes with extreme ease and efficiency [84]. Due to its loftier degree of allegiance and comparatively simple construction, CRISPR-Cas is now widely used in genome editing. CRISPR-Cas genome editing is a blazon II CRISPR system; this organisation includes Cas9, crRNA, trans-activating crRNA (tracrRNA), and ii template options for DNA repair; non-homologous end joining (NHEJ) or homology directed repair (HDR). 2 nuclease domains confer cleavage ability to Cas9: the HNH domain cleaves the complementary Deoxyribonucleic acid strand and the RuvC-like domain cleaves the non-complementary strand [77]. A simple illustration of this is provided in Fig.2.
To date, every facet of the CRISPR-Cas arrangement has been contradistinct and improved in terms of its technical application. Starting with improved endonuclease function, an culling to Cas9, called Cpf1, was discovered in the bacterial genera Prevotella and Francisella. Interestingly, Cpf1 is a single RNA-guided enzyme that does not crave tracrRNA and generates staggered DSBs with a 4–v-nt overhang distal to a 5′ T-rich PAM [85••]. Firstly, Cas9 in its natural grade requires two RNAs and generates cleavage products with blunt ends, which are less easy to work with, equally DNA sequences could insert in either end. Cpf1, withal, makes staggered cuts that generate a 5′ overhang, which improves the precision of DNA insertions. Secondly, unlike Cas9, Cpf1 cuts at a site distal to the gene, preserving the seed region. This is essential for target recognition if hereafter editing is required. Thirdly, because Cpf1 is smaller and does not require a tracrRNA, information technology may exist easier to deliver to cells.
Awarding of CRISPR-Cas in Genome Editing
The therapeutic potential of CRISPR-Cas has already been demonstrated in many instances. Cas9 has been applied immunologically as an antimicrobial agent and has been adult to specifically target antibody resistant in highly virulent strains of leaner [77, 86]. Gene therapy applications have besides been demonstrated for monogenic diseases. Cells from human patients with cystic fibrosis showed functional repair of the CFTR cistron in vitro in cultured intestinal stem jail cell organoids using CRISPR-Cas [86]. The lacking gene causing hereditary tyrosinemia was corrected in mice later on hydrodynamic injection of CRISPR components. This led to an expansion of mutation-corrected hepatocytes in vivo and resulted in a rescued phenotype in developed mice [87]. Advancing from therapeutic treatment, as described, to preventative techniques, muscular dystrophy was prevented when germ line mediated editing of mice with Duchenne muscular was carried out [88].
The treatment of viral infections such as HIV and hepatitis B [89] has also been demonstrated using Cas9. In the first example, iPSCs were generated, and through genome editing, were made to be homozygous for a mutation, which confers HIV-1 resistance. In this study, wild-type iPSCs were modified using TALENS and CRISPR-Cas [90, 91••].
An important milestone, was the offset genetic modifications to exist carried out in primate embryos. Hither, CRISPR was carried out in 1-cell embryos to successfully generate modified cynomolgus monkeys [92]. This is the closest of the creature models in similarity to humans and tin requite evidence for how the system might behave in human being embryos. This could be used to potentially forestall non-complex hereditary diseases, such as single factor defects. However, without full predictability of the off-target effects, germ line cistron editing remains ethically and scientifically unsafe.
In terms of delivery, the CRISPR-Cas system has been directly applied to human cells by co-commitment of plasmids that encode Cas9 expression together with the necessary crRNA components [64, 93]. Recent identification of smaller Cas proteins may enable and raise the combination of this engineering science with vectors that have gained increasing success for their safety profile and efficiency, such every bit AAV vectors [94]. Due to their relatively low immunogenicity, AAVs are commonly chosen for in vivo factor delivery for in somatic cistron therapy [95].
Advantages, Disadvantages, and Potential Applications
The primary advantages of the CRISPR-Cas system are its ability to genetically modify an organism without leaving any foreign Dna behind and its versatility and simplicity of programming. Unlike the reprogramming of its predecessors, ZFNs and TALENs, which require editing of Dna-interacting domains located at different sites on the Deoxyribonucleic acid-binding scaffolds, CRISPR-Cas systems changes are simply executed on the recombinant RNA sequences [62, 96]. Ease of use, low cost, high speed, multiplexing potential, and equal or higher specific DNA targeting power have secured its popularity and success across the global scientific customs [65, 96, 97]. Gupta and colleagues provide a useful review comparing ZFN, TALEN, and CRISPR-Cas technologies [98].
Nevertheless, equally for many other emerging technologies, delivery of the CRISPR-Cas components to the target cells remains 1 of the chief bug [63]. The express Deoxyribonucleic acid packaging capacity of AAV vectors, however, is being addressed with the development of shorter gRNA-coding sequences and the identification of smaller Cas endonucleases, as mentioned above. A 2nd trouble concerns the number of programmable bases in Cas9, which is limited to 20 and whose specificity is subject to the PAM sequence'southward position: if this is not within ten bases from the base target, the targeting frequency is greatly diminished [63, 96]. This event is being addressed by extending the PAM preferences of Cas9 and identifying new CRISPR endonucleases [85••, 99]. The concerns over Cas9 re-cut after successful introduction of the desired modification may be solved by exploitation of synthetic CRISPR RNAs (scrRNAs): these offering controlled silencing options through natural decay of the scrRNA itself, injection of a "sponge" sequence complementary to the scrRNA or tracrRNA, or injection of another scrRNA directed against the Cas9 cistron [97]. Principle concerns that remain are the insufficient target specificity and the potential for astray events, which are difficult to prevent and may be undetectable for current low-cost broadly used tools likewise as for more expensive and less attainable whole-genome sequencing [100••]. In particular, an emergence of tools for the prediction of off-target events and gRNA design has ensued—these are reviewed past Graham and Root [87]. The latest evolution of a modified Cas9 variant shows remarkably fewer off-target effects while retaining total site-specific action and illustrates the aplenty scope for the optimization of the organization. Here, use of a SpCas9-HF1 (for loftier-fidelity) variant resulted in the occurrence of off-target events undetectable by genome-wide interruption capture and targeted sequencing methods [101].
Besides these practical aspects, the major drawback the CRISPR-Cas system faces is restrictive legislation. The power of this factor-editing tool has caused concerns to wider society, due to the potential for irrevocable alteration of future generations, if used in germ line modifications.
Upstanding Considerations and Conclusions
Concerns about the genetic alteration of the human species from both the scientific and lay community started to rise slowly but progressively after the advent of recombinant DNA technology [102]. The call for the recent International Summit of Genome Editing in Washington 2015 echoes that of the first Asilomar conference in 1975. Both called for a moratorium on experiments due to fear that the technologies would be used for experiments which lift the ethical threshold with the potential to modify human evolution.
With the rapid advancement of CRISPR-Cas systems towards application the for treatment of diseases such as Duchenne muscular dystrophy, hemoglobinopathies, Leber congenital amaurosis, and HIV infection, ethical implications are at present being discussed [63]. In detail, the first report on the utilise of CRISPR-Cas9 on man tripronuclear zygotes was published in 2015. These zygotes take one oocyte nucleus and 2 sperm nuclei and are therefore unable to develop into viable embryos. It was this work that prompted a worldwide moratorium by both biologists and ethicists on human germ line genetic technology [103]. Although this study showed low homologous recombination events of the human β-globin (HBB) gene, for which the CRISPR-Cas9 system was designed, likewise as mosaicism and off-target cleavage at diverse sites, it was the beginning fourth dimension CRISPR-Cas9 effectively broken endogenous genes on human embryos [104••]. Information technology was proposed that dynamic guidelines, involving the wider gild, should evolve with the progression of the scientific knowledge and should be established rather than imposing a moratorium on the advancement of such a promising technology [105]. This proclamation was in keeping with a previous statement by the Hinxton Group, that concerns on possible human applications of these techniques should not inhibit the advancement of scientifically defensible basic research, especially as this very enquiry will exist the cardinal to gaining such noesis [106]. It did hold that human embryos or germ line cells subject to gene editing should not be used to establish pregnancies and produced a list of the current technical and moral problems, upon resolution of which, clinical somatic and germ line applications could exist envisaged to eradicate devastating inheritable diseases [105]. Fear that i of CRISPR-Cas' major advantages, its power for "democratization of gene targeting" [107] could be dangerous if used by the wrong people to "enhance" population minorities and reignite an involvement for eugenics, perchance addressed by stiff international jurisdictions and global public engagement [105, 106, 108]. However, before this could be performed, a deeper understanding of appropriate models (both cellular and beast, also as human embryonic stem/iPS prison cell-derived germ line cells) to examination efficacy and safety and multigenerational furnishings, and optimization of genome editing tools to minimize off-target events will be warranted. These include the development of more accurate and sensitive tools to assess astray events and mosaicism [106]. Moreover, the types of cells and embryos that may be used in this context vary broadly and they need to be antiseptic to both the scientific and lay community to allow for constructive progression.
Not-viable preimplantation embryos from in vitro fertilization clinics may not satisfy the benchmark of scientific validity, as the endogenous DNA repair mechanisms in their cells may be altered, just also raise ethical issues. Viable, unused embryos from these clinics would technically exist more than suitable; yet, it could brandish high levels of mosaicism. Embryos specifically designed for research would be the most valuable in terms of research, only are the about ethically leap source of research material [106]. When considering such aspects, information technology should be kept in listen that human germ line editing may be the merely solution to cure genetic diseases that manifest before birth and systemically (e.g., cystic fibrosis) in simply one just very predominant tissue (due east.g., muscular dystrophy) or in not easily accessible tissues (e.grand., basal ganglia in Huntington's affliction). Patient-advancement groups put hope in the use of these novel alternatives [109]. Concerns take further been expressed well-nigh the polygenic control of many human traits, the current difficulty to predict short-term furnishings of a genetic mutation and its side effects, likewise as the apparent impossibility to anticipate its long-term effects in a putative future surroundings. To some, these points signal that the promise to definitively eradicate human genetic diseases is only an illusion [110].
Unlike other moments in scientific history, the CRISPR-Cas system has opened an era of changes, which may span from groundbreaking therapeutic applications to daunting fears of irreversible perturbation of human evolution. It is essential that broad informed discussion across all exponents of society continue, to find a balance that will progressively lead to a gild pivoted on the prioritization of human well-beingness and human rights.
Acknowledgments
Simon N. Waddington received funding from the European Research Council grant "Somabio."
Compliance with Ethical Standards
Conflict of Involvement
Simon N. Waddington, Riccardo Privolizzi, Rajvinder Karda, and Helen C. O'Neill declare that they accept no conflict of interest.
Man and Animal Rights and Informed Consent
This article does not contain any studies with man or animal subjects performed by whatsoever of the authors.
Footnotes
This article is function of the Topical Drove on Genome Editing
References
Papers of particular interest, published recently, have been highlighted equally: •• Of major importance
1. Tatum EL. Molecular biology, nucleic acids, and the time to come of medicine. Perspect Biol Med. 1966;10(ane):xix–32. doi: 10.1353/pbm.1966.0027. [PubMed] [CrossRef] [Google Scholar]
2. Beadle GW, Tatum EL. Genetic control of biochemical reactions in neurospora. Proc Natl Acad Sci U S A. 1941;27(eleven):499–506. doi: 10.1073/pnas.27.11.499. [PMC complimentary article] [PubMed] [CrossRef] [Google Scholar]
3. Nirenberg MW, Matthaei JH. The dependence of jail cell-costless protein synthesis in Due east. coli upon naturally occurring or synthetic polyribonucleotides. Proc Natl Acad Sci U South A. 1961;47(10):1588–602. doi: 10.1073/pnas.47.10.1588. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]
4. Avery OT, Macleod CM, McCarty Chiliad. Studies on the chemic nature of the substance inducing transformation of pneumococcal types: induction of transformation by a desoxyribonucleic acid fraction isolated from pneumococcus type Three. J Exp Med. 1944;79(two):137–58. doi: x.1084/jem.79.two.137. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
5. Sambrook J, Westphal H, Srinivasan PR, Dulbecco R. The integrated land of viral Dna in SV40-transformed cells. Proc Natl Acad Sci U South A. 1968;60(4):1288–95. doi: 10.1073/pnas.60.4.1288. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]
6. Colina M, Hillova J. Virus recovery in chicken cells tested with Rous sarcoma cell DNA. Nat New Biol. 1972;237(71):35–9. doi: x.1038/newbio237035a0. [PubMed] [CrossRef] [Google Scholar]
7. Rogers S, Lowenthal A, Terheggen HG, Columbo JP. Induction of arginase activity with the Shope papilloma virus in tissue culture cells from an argininemic patient. J Exp Med. 1973;137(iv):1091–6. doi: ten.1084/jem.137.4.1091. [PMC complimentary article] [PubMed] [CrossRef] [Google Scholar]
8. Terheggen HG, Lowenthal A, Lavinha F, Colombo JP, Rogers South. Unsuccessful trial of factor replacement in arginase deficiency. Zeitschrift fur Kinderheilkunde. 1975;119(1):1–3. doi: ten.1007/BF00464689. [PubMed] [CrossRef] [Google Scholar]
nine. Berg P, Baltimore D, Brenner South, Roblin RO, Vocaliser MF. Summary statement of the Asilomar conference on recombinant DNA molecules. Proc Natl Acad Sci U S A. 1975;72(half-dozen):1981–4. doi: 10.1073/pnas.72.6.1981. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
10. Cline MJ, Stang H, Mercola Thousand, et al. Gene transfer in intact animals. Nature. 1980;284(5755):422–five. doi: x.1038/284422a0. [PubMed] [CrossRef] [Google Scholar]
11. Mercola KE, Stang HD, Browne J, Salser West, Cline MJ. Insertion of a new factor of viral origin into bone marrow cells of mice. Scientific discipline. 1980;208(4447):1033–five. doi: 10.1126/scientific discipline.6246577. [PubMed] [CrossRef] [Google Scholar]
12. Wigler M, Silverstein S, Lee LS, Pellicer A, Cheng Y, Axel R. Transfer of purified herpes virus thymidine kinase cistron to cultured mouse cells. Cell. 1977;11(1):223–32. doi: 10.1016/0092-8674(77)90333-6. [PubMed] [CrossRef] [Google Scholar]
13. Capecchi MR. Loftier efficiency transformation by direct microinjection of Dna into cultured mammalian cells. Cell. 1980;22(two Pt 2):479–88. doi: 10.1016/0092-8674(80)90358-Ten. [PubMed] [CrossRef] [Google Scholar]
14. Gordon JW, Scangos GA, Plotkin DJ, Barbosa JA, Ruddle FH. Genetic transformation of mouse embryos by microinjection of purified Deoxyribonucleic acid. Proc Natl Acad Sci U S A. 1980;77(12):7380–4. doi: 10.1073/pnas.77.12.7380. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
15. Brinster RL, Chen HY, Trumbauer M, Senear AW, Warren R, Palmiter RD. Somatic expression of herpes thymidine kinase in mice post-obit injection of a fusion factor into eggs. Jail cell. 1981;27(i Pt ii):223–31. doi: x.1016/0092-8674(81)90376-seven. [PMC gratuitous article] [PubMed] [CrossRef] [Google Scholar]
16. Costantini F, Lacy E. Introduction of a rabbit beta-globin gene into the mouse germ line. Nature. 1981;294(5836):92–iv. doi: 10.1038/294092a0. [PubMed] [CrossRef] [Google Scholar]
17. Wagner EF, Stewart TA, Mintz B. The human beta-globin gene and a functional viral thymidine kinase gene in developing mice. Proc Natl Acad Sci U S A. 1981;78(8):5016–20. doi: 10.1073/pnas.78.8.5016. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
eighteen. Folger KR, Wong EA, Wahl G, Capecchi MR. Patterns of integration of DNA microinjected into cultured mammalian cells: evidence for homologous recombination between injected plasmid DNA molecules. Mol Jail cell Biol. 1982;2(11):1372–87. doi: x.1128/MCB.2.11.1372. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
xix. Smithies O, Gregg RG, Boggs SS, Koralewski MA, Kucherlapati RS. Insertion of Deoxyribonucleic acid sequences into the human chromosomal beta-globin locus past homologous recombination. Nature. 1985;317(6034):230–iv. doi: 10.1038/317230a0. [PubMed] [CrossRef] [Google Scholar]
20. Till JE, Mc CE. A direct measurement of the radiations sensitivity of normal mouse bone marrow cells. Radiat Res. 1961;14:213–22. doi: 10.2307/3570892. [PubMed] [CrossRef] [Google Scholar]
21. Evans MJ, Kaufman MH. Institution in culture of pluripotential cells from mouse embryos. Nature. 1981;292(5819):154–half-dozen. doi: 10.1038/292154a0. [PubMed] [CrossRef] [Google Scholar]
22. Martin GR. Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned past teratocarcinoma stalk cells. Proc Natl Acad Sci. 1981;78(12):7634–8. doi: x.1073/pnas.78.12.7634. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
23. Bradley A, Evans M, Kaufman MH, Robertson E. Formation of germ-line chimaeras from embryo-derived teratocarcinoma cell lines. Nature. 1984;309(5965):255–half dozen. doi: 10.1038/309255a0. [PubMed] [CrossRef] [Google Scholar]
24. Thomas KR, Capecchi MR. Site-directed mutagenesis by gene targeting in mouse embryo-derived stem cells. Cell. 1987;51(3):503–12. doi: x.1016/0092-8674(87)90646-5. [PubMed] [CrossRef] [Google Scholar]
25. Koller BH, Hagemann LJ, Doetschman T, et al. Germ-line transmission of a planned alteration made in a hypoxanthine phosphoribosyltransferase gene by homologous recombination in embryonic stem cells. Proc Natl Acad Sci U South A. 1989;86(22):8927–31. doi: 10.1073/pnas.86.22.8927. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]
26. Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. Embryonic stem jail cell lines derived from human blastocysts. Scientific discipline. 1998;282(5391):1145–7. doi: 10.1126/science.282.5391.1145. [PubMed] [CrossRef] [Google Scholar]
27. Zwaka TP, Thomson JA. Homologous recombination in human embryonic stem cells. Nat Biotechnol. 2003;21(3):319–21. doi: 10.1038/nbt788. [PubMed] [CrossRef] [Google Scholar]
28. Mann R, Mulligan RC, Baltimore D. Construction of a retrovirus packaging mutant and its use to produce helper-free defective retrovirus. Cell. 1983;33(1):153–nine. doi: 10.1016/0092-8674(83)90344-6. [PubMed] [CrossRef] [Google Scholar]
29. Tabin CJ, Hoffmann JW, Goff SP, Weinberg RA. Adaptation of a retrovirus as a eucaryotic vector transmitting the herpes simplex virus thymidine kinase gene. Mol Cell Biol. 1982;2(iv):426–36. doi: 10.1128/MCB.ii.4.426. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
xxx. Wei CM, Gibson K, Spear PG, Scolnick EM. Structure and isolation of a transmissible retrovirus containing the src gene of Harvey murine sarcoma virus and the thymidine kinase gene of herpes simplex virus type 1. J Virol. 1981;39(three):935–44. [PMC free article] [PubMed] [Google Scholar]
31. Shimotohno K, Temin HM. Formation of infectious progeny virus after insertion of herpes simplex thymidine kinase factor into DNA of an avian retrovirus. Cell. 1981;26(1 Pt one):67–77. doi: x.1016/0092-8674(81)90034-ix. [PubMed] [CrossRef] [Google Scholar]
32. Yamada Thousand, Lewis JA, Grodzicker T. Overproduction of the protein product of a nonselected foreign factor carried by an adenovirus vector. Proc Natl Acad Sci U Due south A. 1985;82(11):3567–71. doi: 10.1073/pnas.82.xi.3567. [PMC complimentary commodity] [PubMed] [CrossRef] [Google Scholar]
33. Berkner KL, Sharp PA. Expression of dihydrofolate reductase, and of the next EIb region, in an Ad5-dihydrofolate reductase recombinant virus. Nucleic Acids Res. 1984;12(4):1925–41. doi: 10.1093/nar/12.4.1925. [PMC gratis article] [PubMed] [CrossRef] [Google Scholar]
34. Thummel C, Tjian R, Grodzicker T. Expression of SV40 T antigen under control of adenovirus promoters. Cell. 1981;23(3):825–36. doi: 10.1016/0092-8674(81)90447-v. [PubMed] [CrossRef] [Google Scholar]
35. Hermonat PL, Muzyczka Northward. Use of adeno-associated virus as a mammalian DNA cloning vector: transduction of neomycin resistance into mammalian tissue civilization cells. Proc Natl Acad Sci U Southward A. 1984;81(twenty):6466–seventy. doi: 10.1073/pnas.81.xx.6466. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
36. Miller AD. Human cistron therapy comes of age. Nature. 1992;357(6378):455–60. doi: 10.1038/357455a0. [PubMed] [CrossRef] [Google Scholar]
37. Wadman Grand. Hyping results 'could harm' factor therapy. Nature. 1995;378(6558):655. [PubMed] [Google Scholar]
38. Hacein-Bey-Abina S, Le Deist F, Carlier F, et al. Sustained correction of X-linked severe combined immunodeficiency by ex vivo gene therapy. North Engl J Med. 2002;346(16):1185–93. doi: 10.1056/NEJMoa012616. [PubMed] [CrossRef] [Google Scholar]
39. Hacein-Bey-Abina S, von Kalle C, Schmidt M, et al. A serious adverse event after successful factor therapy for X-linked severe combined immunodeficiency. N Engl J Med. 2003;348(iii):255–half-dozen. doi: 10.1056/NEJM200301163480314. [PubMed] [CrossRef] [Google Scholar]
twoscore. Hoag H. Gene therapy ascension? Nature. 2005;435(7041):530–i. doi: x.1038/nj7041-530a. [PubMed] [CrossRef] [Google Scholar]
41. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures past defined factors. Prison cell. 2006;126(four):663–76. doi: 10.1016/j.cell.2006.07.024. [PubMed] [CrossRef] [Google Scholar]
42. Okita K, Ichisaka T, Yamanaka South. Generation of germline-competent induced pluripotent stem cells. Nature. 2007;448(7151):313–seven. doi: ten.1038/nature05934. [PubMed] [CrossRef] [Google Scholar]
43. Takahashi K, Tanabe Grand, Ohnuki K, et al. Consecration of pluripotent stem cells from adult man fibroblasts past divers factors. Cell. 2007;131(five):861–72. doi: 10.1016/j.prison cell.2007.11.019. [PubMed] [CrossRef] [Google Scholar]
44. Yu J, Vodyanik MA, Smuga-Otto M, et al. Induced pluripotent stalk cell lines derived from human somatic cells. Science. 2007;318(5858):1917–20. doi: 10.1126/scientific discipline.1151526. [PubMed] [CrossRef] [Google Scholar]
45.••. Ilic D, Devito L, Miere C, Codognotto Southward. Human being embryonic and induced pluripotent stem cells in clinical trials. Br Med Bull. 2015;116(ane):19–27. [PubMed] [Google Scholar]
46. Paull D, Sevilla A, Zhou H, et al. Automated, high-throughput derivation, label and differentiation of induced pluripotent stem cells. Nat Methods. 2015;12(9):885–92. doi: 10.1038/nmeth.3507. [PubMed] [CrossRef] [Google Scholar]
47. Faulkner-Jones A, Fyfe C, Cornelissen DJ, et al. Bioprinting of human pluripotent stem cells and their directed differentiation into hepatocyte-like cells for the generation of mini-livers in 3D. Biofabrication. 2015;vii(4):044102. doi: 10.1088/1758-5090/7/four/044102. [PubMed] [CrossRef] [Google Scholar]
48. Warren L, Manos PD, Ahfeldt T, et al. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Jail cell. 2010;seven(5):618–30. doi: 10.1016/j.stem.2010.08.012. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
49. Singh VK, Kalsan Thou, Kumar N, Saini A, Chandra R. Induced pluripotent stem cells: applications in regenerative medicine, disease modeling, and drug discovery. Front Cell Dev Biol. 2015;3:2. doi: ten.3389/fcell.2015.00002. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
50. Buganim Y, Faddah DA, Cheng AW, et al. Single-cell expression analyses during cellular reprogramming reveal an early stochastic and a late hierarchic stage. Cell. 2012;150(six):1209–22. doi: x.1016/j.jail cell.2012.08.023. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]
51. Hussein SM, Puri MC, Tonge PD, et al. Genome-wide label of the routes to pluripotency. Nature. 2014;516(7530):198–206. doi: 10.1038/nature14046. [PubMed] [CrossRef] [Google Scholar]
52. Kim DH, Marinov GK, Pepke S, et al. Unmarried-cell transcriptome analysis reveals dynamic changes in lncRNA expression during reprogramming. Cell Stem Cell. 2015;16(1):88–101. doi: 10.1016/j.stem.2014.11.005. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]
53. Wang A, He Q, Zhong Y. Systematically dissecting the global mechanism of miRNA functions in mouse pluripotent stem cells. BMC Genomics. 2015;16:490. doi: 10.1186/s12864-015-1706-y. [PMC gratuitous article] [PubMed] [CrossRef] [Google Scholar]
54. Braun CJ, Boztug K, Paruzynski A, et al. Factor therapy for Wiskott-Aldrich syndrome—long-term efficacy and genotoxicity. Sci Transl Med. 2014;half dozen(227):227ra33. doi: 10.1126/scitranslmed.3007280. [PubMed] [CrossRef] [Google Scholar]
55. Urnov FD, Miller JC, Lee YL, et al. Highly efficient endogenous human factor correction using designed zinc-finger nucleases. Nature. 2005;435(7042):646–51. doi: 10.1038/nature03556. [PubMed] [CrossRef] [Google Scholar]
56. Lieber MR. The machinery of double-strand Deoxyribonucleic acid pause repair by the nonhomologous Dna end-joining pathway. Annu Rev Biochem. 2010;79:181–211. doi: 10.1146/annurev.biochem.052308.093131. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]
57. Yamanaka Due south. Induced pluripotent stalk cells: by, present, and future. Cell Stem Cell. 2012;x(vi):678–84. doi: 10.1016/j.stalk.2012.05.005. [PubMed] [CrossRef] [Google Scholar]
58. Gaj T, Gersbach CA, Barbas CF., third ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering science. Trends Biotechnol. 2013;31(7):397–405. doi: 10.1016/j.tibtech.2013.04.004. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
59. Urnov FD, Rebar EJ, Holmes MC, Zhang HS, Gregory PD. Genome editing with engineered zinc finger nucleases. Nat Rev Genet. 2010;11(9):636–46. doi: x.1038/nrg2842. [PubMed] [CrossRef] [Google Scholar]
sixty. Ding Q, Lee YK, Schaefer EA, et al. A TALEN genome-editing system for generating human stalk cell-based disease models. Cell Stem Cell. 2013;12(2):238–51. doi: 10.1016/j.stem.2012.11.011. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]
61. Ding Q, Regan SN, Xia Y, Oostrom LA, Cowan CA, Musunuru Yard. Enhanced efficiency of human pluripotent stalk cell genome editing through replacing TALENs with CRISPRs. Jail cell Stem Prison cell. 2013;12(4):393–4. doi: ten.1016/j.stem.2013.03.006. [PMC costless commodity] [PubMed] [CrossRef] [Google Scholar]
62. Travis J. Making the cut. Science. 2015;350(6267):1456–seven. doi: 10.1126/scientific discipline.350.6267.1456. [PubMed] [CrossRef] [Google Scholar]
63. Keener AB. Delivering the goods: scientists seek a way to make CRISPR-Cas gene editing more targeted. Nat Med. 2015;21(eleven):1239–41. doi: x.1038/nm1115-1239. [PubMed] [CrossRef] [Google Scholar]
64. Cong L, Ran FA, Cox D, Lin Due south, Barretto R, Habib N et al. Multiplex genome applied science using CRISPR/Cas systems. Science. 2013. [PMC free article] [PubMed]
65. Mali P, Yang L, Esvelt KM, et al. RNA-guided human being genome engineering science via Cas9. Science. 2013;339(6121):823–half dozen. doi: x.1126/scientific discipline.1232033. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
66. Jinek M, East A, Cheng A, Lin S, Ma Due east, Doudna J. RNA-programmed genome editing in human cells. Elife. 2013;2:e00471. doi: 10.7554/eLife.00471. [PMC gratis article] [PubMed] [CrossRef] [Google Scholar]
67. Maggio I, Holkers Yard, Liu J, Janssen JM, Chen X, Goncalves MA. Adenoviral vector commitment of RNA-guided CRISPR/Cas9 nuclease complexes induces targeted mutagenesis in a various array of man cells. Sci Rep. 2014;4:5105. doi: 10.1038/srep05105. [PMC gratuitous commodity] [PubMed] [CrossRef] [Google Scholar]
68. Barrangou R, Fremaux C, Deveau H, et al. CRISPR provides acquired resistance against viruses in prokaryotes. Scientific discipline. 2007;315(5819):1709–12. doi: 10.1126/science.1138140. [PubMed] [CrossRef] [Google Scholar]
69. Fineran PC, Charpentier E. Retentiveness of viral infections past CRISPR-Cas adaptive immune systems: acquisition of new data. Virology. 2012;434(2):202–nine. doi: ten.1016/j.virol.2012.10.003. [PubMed] [CrossRef] [Google Scholar]
70. Horvath P, Barrangou R. CRISPR/Cas, the allowed arrangement of leaner and archaea. Science. 2010;327(5962):167–70. doi: 10.1126/science.1179555. [PubMed] [CrossRef] [Google Scholar]
71. Wiedenheft B, Sternberg SH, Doudna JA. RNA-guided genetic silencing systems in bacteria and archaea. Nature. 2012;482(7385):331–viii. doi: 10.1038/nature10886. [PubMed] [CrossRef] [Google Scholar]
72. Brouns SJ, Jore MM, Lundgren M, et al. Small-scale CRISPR RNAs guide antiviral defense in prokaryotes. Science. 2008;321(5891):960–4. doi: 10.1126/science.1159689. [PMC costless commodity] [PubMed] [CrossRef] [Google Scholar]
73. Ishino Y, Shinagawa H, Makino K, Amemura 1000, Nakata A. Nucleotide sequence of the iap factor, responsible for alkaline phosphatase isozyme conversion in Escherichia coli, and identification of the gene production. J Bacteriol. 1987;169(12):5429–33. [PMC free article] [PubMed] [Google Scholar]
74. Grissa I, Vergnaud Yard, Pourcel C. CRISPRFinder: a web tool to identify clustered regularly interspaced curt palindromic repeats. Nucleic Acids Res. 2007;35(Spider web Server issue):W52–seven. doi: 10.1093/nar/gkm360. [PMC costless article] [PubMed] [CrossRef] [Google Scholar]
75. Haft DH, Selengut J, Mongodin EF, Nelson KE. A guild of 45 CRISPR-associated (Cas) protein families and multiple CRISPR/Cas subtypes exist in prokaryotic genomes. PLoS Comput Biol. 2005;one(half-dozen):e60. doi: ten.1371/journal.pcbi.0010060. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
76. Swarts DC, Mosterd C, van Passel MW, Brouns SJ. CRISPR interference directs strand specific spacer acquisition. PLoS ONE. 2012;seven(4):e35888. doi: 10.1371/journal.pone.0035888. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]
77. Makarova KS, Grishin NV, Shabalina SA, Wolf YI, Koonin EV. A putative RNA-interference-based immune organization in prokaryotes: computational analysis of the predicted enzymatic mechanism, functional analogies with eukaryotic RNAi, and hypothetical mechanisms of action. Biol Straight. 2006;one:seven. doi: x.1186/1745-6150-1-7. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]
78. Mojica FJ, Diez-Villasenor C, Garcia-Martinez J, Almendros C. Short motif sequences determine the targets of the prokaryotic CRISPR defense force system. Microbiology (Reading, England). 2009;155(Pt 3):733–forty. [PubMed]
79. Yosef I, Goren MG, Qimron U. Proteins and Dna elements essential for the CRISPR accommodation process in Escherichia coli. Nucleic Acids Res. 2012;40(12):5569–76. doi: x.1093/nar/gks216. [PMC costless article] [PubMed] [CrossRef] [Google Scholar]
lxxx. Pourcel C, Salvignol G, Vergnaud G. CRISPR elements in Yersinia pestis larn new repeats past preferential uptake of bacteriophage DNA, and provide additional tools for evolutionary studies. Microbiology. 2005;151(Pt 3):653–63. doi: 10.1099/mic.0.27437-0. [PubMed] [CrossRef] [Google Scholar]
81. Tyson GW, Banfield JF. Chop-chop evolving CRISPRs implicated in acquired resistance of microorganisms to viruses. Environ Microbiol. 2008;10(i):200–7. [PubMed] [Google Scholar]
82. Heidelberg JF, Nelson WC, Schoenfeld T, Bhaya D. Germ warfare in a microbial mat customs: CRISPRs provide insights into the co-evolution of host and viral genomes. PLoS ONE. 2009;four(1):e4169. doi: 10.1371/journal.pone.0004169. [PMC gratuitous article] [PubMed] [CrossRef] [Google Scholar]
83. Rath D, Amlinger L, Rath A, Lundgren 1000. The CRISPR-Cas immune system: biology, mechanisms and applications. Biochimie. 2015;117:119–28. doi: 10.1016/j.biochi.2015.03.025. [PubMed] [CrossRef] [Google Scholar]
84. Jinek Thou, Chylinski Thousand, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual-RNA-guided Deoxyribonucleic acid endonuclease in adaptive bacterial amnesty. Science. 2012;337(6096):816–21. doi: 10.1126/science.1225829. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
85.••. Zetsche B, Gootenberg JS, Abudayyeh OO, et al. Cpf1 is a single RNA-guided endonuclease of a class two CRISPR-Cas system. Cell. 2015;163(3):759–71. doi: 10.1016/j.cell.2015.09.038. [PMC complimentary article] [PubMed] [CrossRef] [Google Scholar]
86. Schwank G, Koo BK, Sasselli V, et al. Functional repair of CFTR past CRISPR/Cas9 in abdominal stem cell organoids of cystic fibrosis patients. Cell Stalk Cell. 2013;xiii(6):653–viii. doi: 10.1016/j.stalk.2013.xi.002. [PubMed] [CrossRef] [Google Scholar]
87. Yin H, Xue W, Chen S, et al. Genome editing with Cas9 in developed mice corrects a disease mutation and phenotype. Nat Biotechnol. 2014;32(vi):551–3. doi: ten.1038/nbt.2884. [PMC free commodity] [PubMed] [CrossRef] [Google Scholar]
88. Long C, McAnally JR, Shelton JM, Mireault AA, Bassel-Duby R, Olson EN. Prevention of muscular dystrophy in mice by CRISPR/Cas9-mediated editing of germline Dna. Science. 2014;345(6201):1184–8. doi: 10.1126/science.1254445. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
89. Zhen S, Hua 50, Liu YH, et al. Harnessing the clustered regularly interspaced curt palindromic echo (CRISPR)/CRISPR-associated Cas9 system to disrupt the hepatitis B virus. Gene Ther. 2015;22(5):404–12. doi: 10.1038/gt.2015.2. [PubMed] [CrossRef] [Google Scholar]
90. Hu W, Kaminski R, Yang F, et al. RNA-directed gene editing specifically eradicates latent and prevents new HIV-one infection. Proc Natl Acad Sci U S A. 2014;111(31):11461–6. doi: 10.1073/pnas.1405186111. [PMC complimentary article] [PubMed] [CrossRef] [Google Scholar]
91.••. Ye L, Wang J, Beyer AI, et al. Seamless modification of wild-type induced pluripotent stem cells to the natural CCR5Delta32 mutation confers resistance to HIV infection. Proc Natl Acad Sci U Southward A. 2014;111(26):9591–half dozen. doi: 10.1073/pnas.1407473111. [PMC gratis commodity] [PubMed] [CrossRef] [Google Scholar]
92. Niu Y, Shen B, Cui Y, et al. Generation of gene-modified cynomolgus monkey via Cas9/RNA-mediated cistron targeting in one-cell embryos. Cell. 2014;156(four):836–43. doi: 10.1016/j.jail cell.2014.01.027. [PubMed] [CrossRef] [Google Scholar]
93. Liu F, Barrangou R, Gerner-Smidt P, Ribot EM, Knabel SJ, Dudley EG. Novel virulence gene and amassed regularly interspaced short palindromic repeat (CRISPR) multilocus sequence typing scheme for subtyping of the major serovars of Salmonella enterica subsp. enterica. Appl Environ Microbiol. 2011;77(half dozen):1946–56. doi: 10.1128/AEM.02625-10. [PMC complimentary article] [PubMed] [CrossRef] [Google Scholar]
94. Friedland AE, Baral R, Singhal P, et al. Label of Staphylococcus aureus Cas9: a smaller Cas9 for all-in-one adeno-associated virus delivery and paired nickase applications. Genome Biol. 2015;16:257. doi: 10.1186/s13059-015-0817-8. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
95. Ran FA, Cong L, Yan WX, Scott DA, Gootenberg JS, Kriz AJ et al. In vivo genome editing using Staphylococcus aureus Cas9. Nature. 2015. [PMC free article] [PubMed]
96. Strauβ A, Lahaye T. Zinc fingers, TAL effectors, or Cas9-based DNA bounden proteins: what's best for targeting desired genome loci? Mol Plant. 2013;6(five):1384–7. doi: ten.1093/mp/sst075. [PubMed] [CrossRef] [Google Scholar]
97. Rahdar Yard, McMahon MA, Prakash TP, Swayze EE, Bennett CF, Cleveland DW. Synthetic CRISPR RNA-Cas9-guided genome editing in human cells. Proc Natl Acad Sci U Southward A. 2015;112(51):E7110–7. [PMC free article] [PubMed] [Google Scholar]
98. Gupta RM, Musunuru One thousand. Expanding the genetic editing tool kit: ZFNs, TALENs, and CRISPR-Cas9. J Clin Invest. 2014;124(10):4154–61. doi: ten.1172/JCI72992. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
99. Kleinstiver BP, Prew MS, Tsai SQ, et al. Engineered CRISPR-Cas9 nucleases with altered PAM specificities. Nature. 2015;523(7561):481–five. doi: 10.1038/nature14592. [PMC gratuitous commodity] [PubMed] [CrossRef] [Google Scholar]
100.••. Graham DB, Root DE. Resources for the design of CRISPR gene editing experiments. Genome Biol. 2015;sixteen:260. doi: x.1186/s13059-015-0823-x. [PMC costless commodity] [PubMed] [CrossRef] [Google Scholar]
101. Kleinstiver BP, Pattanayak 5, Prew MS, Tsai SQ, Nguyen NT, Zheng Z et al. High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects. Nature. 2016. [PMC gratis article] [PubMed]
102. Berg P, Singer MF. The recombinant DNA controversy: twenty years later. Proc Natl Acad Sci U S A. 1995;92(20):9011–three. doi: 10.1073/pnas.92.20.9011. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
103. Hurlbut JB. Limits of responsibility: genome editing, asilomar, and the politics of deliberation. Hastings Cent Rep. 2015;45(5):11–iv. doi: 10.1002/hast.484. [PubMed] [CrossRef] [Google Scholar]
104.••. Liang P, Xu Y, Zhang X, et al. CRISPR/Cas9-mediated cistron editing in human tripronuclear zygotes. Protein Cell. 2015;half dozen(five):363–72. doi: x.1007/s13238-015-0153-v. [PMC gratuitous article] [PubMed] [CrossRef] [Google Scholar]
105. Statement by the co-sponsoring presidents of the peak on man gene editing. 2015.
106. Chan Southward, Donovan PJ, Douglas T, et al. Genome editing technologies and human germline genetic modification: the Hinxton group consensus statement. Am J Bioeth. 2015;15(12):42–7. doi: x.1080/15265161.2015.1103814. [PubMed] [CrossRef] [Google Scholar]
107. Hotta A, Yamanaka Due south. From genomics to gene therapy: induced pluripotent stem cells run across genome editing. Annu Rev Genet. 2015;49:47–70. doi: 10.1146/annurev-genet-112414-054926. [PubMed] [CrossRef] [Google Scholar]
108. Krishan G, Kanchan T, Singh B. Human genome editing and ethical considerations. Sci Eng Ethics. 2015. [PubMed]
109. Porteus MH, Dann CT. Genome editing of the germline: broadening the word. Mol Ther. 2015;23(6):980–two. doi: 10.1038/mt.2015.83. [PMC complimentary article] [PubMed] [CrossRef] [Google Scholar]
110. Morange Chiliad. Genetic modification of the human germ line: the reasons why this project has no hereafter. C R Biol. 2015;338(eight–9):554–8. doi: 10.1016/j.crvi.2015.07.005. [PubMed] [CrossRef] [Google Scholar]
Source: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4913977/
0 Response to "what new technologies allowed humans to study cells? who invented it?"
Post a Comment