RESUMO
The chemokine-like factor (CKLF)-like MARVEL transmembrane domain-containing family (CMTM) is a gene family that has been implicated in male reproduction. CMTM4 is an evolutionarily conserved member that is highly expressed in the testis. However, its function in male fertility remains unknown. Here, we demonstrate that CMTM4 is associated with spermatogenesis and sperm quality. Using Western blotting and immunohistochemical analyses, we found CMTM4 expression to be decreased in poor-quality human spermatozoa, old human testes, and testicular biopsies with nonobstructive azoospermia. Using CRISPR-Cas9 technology, we knocked out the Cmtm4 gene in mice. These Cmtm4 knockout (KO) mice showed reduced testicular daily sperm production, lower epididymal sperm motility and increased proportion of abnormally backward-curved sperm heads and bent sperm midpieces. These mice also had an evident sub-fertile phenotype, characterized by low pregnancy rates on prolonged breeding with wild type female mice, reduced in vitro fertilization efficiency and a reduced percentage of acrosome reactions. We then performed quantitative proteomic analysis of the testes, where we identified 139 proteins to be downregulated in Cmtm4-KO mice, 100 (71.9%) of which were related to sperm motility and acrosome reaction. The same proteomic analysis was performed on sperm, where we identified 3588 proteins with 409 being differentially regulated in Cmtm4-KO mice. Our enrichment analysis showed that upregulated proteins were enriched with nucleosomal DNA binding functions and the downregulated proteins were enriched with actin binding functions. These findings elucidate the roles of CMTM4 in male fertility and demonstrates its potential as a promising molecular candidate for sperm quality assessment and the diagnosis or treatment of male infertility.
Assuntos
Fertilidade , Proteínas com Domínio MARVEL/genética , Proteoma/metabolismo , Adulto , Animais , Sequência de Bases , Proteína 9 Associada à CRISPR/administração & dosagem , Feminino , Humanos , Marcação por Isótopo , Proteínas com Domínio MARVEL/metabolismo , Masculino , Camundongos Knockout , Microinjeções , Fenótipo , Proteômica , Espermatogênese , Espermatozoides , Testículo/metabolismoRESUMO
BACKGROUND & AIMS: Despite advances in gene editing technologies, generation of tissue-specific knockout mice is time-consuming. We used CRISPR/Cas9-mediated genome editing to disrupt genes in livers of adult mice in just a few months, which we refer to as somatic liver knockouts. METHODS: In this system, Fah-/- mice are given hydrodynamic tail vein injections of plasmids carrying CRISPR/Cas9 designed to excise exons in Hpd; the Hpd-edited hepatocytes have a survival advantage in these mice. Plasmids that target Hpd and a separate gene of interest can therefore be used to rapidly generate mice with liver-specific deletion of nearly any gene product. RESULTS: We used this system to create mice with liver-specific knockout of argininosuccinate lyase, which develop hyperammonemia, observed in humans with mutations in this gene. We also created mice with liver-specific knockout of ATP binding cassette subfamily B member 11, which encodes the bile salt export pump. We found that these mice have a biochemical phenotype similar to that of Abcb11-/- mice. We then used this system to knock out expression of 5 different enzymes involved in drug metabolism within the same mouse. CONCLUSIONS: This approach might be used to develop new models of liver diseases and study liver functions of genes that are required during development.
Assuntos
Argininossuccinato Liase/genética , Proteína 9 Associada à CRISPR/administração & dosagem , Sistemas CRISPR-Cas/genética , Edição de Genes/métodos , Fígado/enzimologia , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/genética , Animais , Modelos Animais de Doenças , Hepatócitos/enzimologia , Hepatócitos/fisiologia , Camundongos , Camundongos Knockout , Oxirredutases/genética , Fenótipo , Plasmídeos/genéticaRESUMO
Genome editing, as exemplified by CRISPR/Cas9, is now recognized as a powerful tool for the engineering of endogenous target genes. It employs only two components, namely, Cas9 in the form of DNA, mRNA, or protein; and guide RNA (gRNA), which is specific to a target gene. When these components are transferred to cells, they create insertion/deletion mutations (indels) within a target gene. Therefore, when fetuses within the uteri of pregnant murine females are exposed to these reagents, fetal cells incorporating them should show mutations in the target gene. To examine a possible genome editing of fetal cells in vivo, we intravenously administered a solution containing plasmid DNA-FuGENE complex to pregnant wild-type female mice [which had been successfully mated with enhanced green fluorescent protein (EGFP)-expressing male transgenic mice] on day 12.5 of gestation. The plasmid DNA induces the expression of gRNA, which was targeted at the EGFP cDNA, and that of the Cas9 gene. All fetuses in the pregnant females should express EGFP systemically, since they are heterozygous (Tg/+) for the transgene. Thus, the delivery of CRISPR system targeted at EGFP in the fetuses will cause a reduced expression of EGFP as a result of the genome editing of EGFP genomic sequence. Of the 24 fetuses isolated from three pregnant females 2 days after gene delivery, 3 were found to have reduced fluorescence in their hearts. Genotyping of the dissected hearts revealed the presence of the transgene construct (Cas9 gene) in all the samples. Furthermore, all the three samples exhibited mutations at the target loci, although normal cells were also present. Thus, transplacental delivery of gene editing components may be a useful tool for developing animal models with heart disorder for heart-related disease research, and gene therapy in congenital heart defects such as hypertrophic cardiomyopathy (HCM). © 2019 IUBMB Life, 9999(9999):1-10, 2019.
Assuntos
Feto/fisiologia , Edição de Genes , Proteínas de Fluorescência Verde/genética , Coração/embriologia , Mutação , Miócitos Cardíacos/fisiologia , RNA Guia de Cinetoplastídeos/genética , Animais , Proteína 9 Associada à CRISPR/administração & dosagem , Proteína 9 Associada à CRISPR/genética , Células Cultivadas , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Genoma , Coração/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/citologia , Plasmídeos , RNA Guia de Cinetoplastídeos/administração & dosagem , TransgenesRESUMO
CRISPR/Cas9 genome editing has gained rapidly increasing attentions in recent years, however, the translation of this biotechnology into therapy has been hindered by efficient delivery of CRISPR/Cas9 materials into target cells. Direct delivery of CRISPR/Cas9 system as a ribonucleoprotein (RNP) complex consisting of Cas9 protein and single guide RNA (sgRNA) has emerged as a powerful and widespread method for genome editing due to its advantages of transient genome editing and reduced off-target effects. In this review, we summarized the current Cas9 RNP delivery systems including physical approaches and synthetic carriers. The mechanisms and beneficial roles of these strategies in intracellular Cas9 RNP delivery were reviewed. Examples in the development of stimuli-responsive and targeted carriers for RNP delivery are highlighted. Finally, the challenges of current Cas9 RNP delivery systems and perspectives in rational design of next generation materials for this promising field will be discussed.
Assuntos
Proteína 9 Associada à CRISPR/administração & dosagem , Proteína 9 Associada à CRISPR/metabolismo , Sistemas CRISPR-Cas , Edição de Genes , RNA Guia de Cinetoplastídeos , Animais , Proteína 9 Associada à CRISPR/genética , Técnicas de Transferência de Genes , HumanosRESUMO
The clustered regularly interspaced short palindromic repeats (CRISPR)/associated protein 9 (CRISPR/Cas9) technology enables genome editing with high precision and versatility and has been widely utilized to combat viruses, bacteria, cancers, and genetic diseases. Nonviral nanocarriers can overcome several limitations of viral vehicles, including immunogenicity, inflammation, carcinogenicity, and low versatility, and thus represent promising platforms for CRISPR/Cas9 delivery. Herein, we for the first time develop the application of protamine-capped gold nanoclusters (protamine-AuNCs) as an effective nanocarrier for Cas9-sgRNA plasmid transport and release to achieve efficient genome editing. The protamine-AuNCs integrate the merits of AuNCs and protamine: AuNCs are able to promptly assemble with Cas9-sgRNA plasmids to allow efficient cellular delivery, while the cationic protamine facilitates the effective release of Cas9-sgRNA plasmids into the cellular nucleus. The AuNCs/Cas9-gRNA plasmid nanocomplexes can not only achieve successful gene editing in cells but also knock out the oncogenic gene for cancer therapy. Moreover, the AuNCs with excellent photoluminescence characteristics endow our nanoplatform with the functionality of bioimaging. Overall, our study provides strong evidence that demonstrates protamine-AuNCs as an effective CRISPR/Cas9 delivery tool for gene therapy.
Assuntos
Proteína 9 Associada à CRISPR/metabolismo , Sistemas CRISPR-Cas/fisiologia , Núcleo Celular/metabolismo , Edição de Genes/métodos , Ouro/metabolismo , Nanopartículas Metálicas , Proteína 9 Associada à CRISPR/administração & dosagem , Proteína 9 Associada à CRISPR/genética , Sistemas CRISPR-Cas/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Ouro/administração & dosagem , Humanos , Nanopartículas Metálicas/administração & dosagemRESUMO
Urinary tract infections (UTI) are the most common infectious diseases in the world. It is becoming increasingly tough to treat because of emergence of antibiotic resistance. So, there is an exigency to develop novel anti-virulence therapeutics to combat multi-drug resistance pathogenic strains. Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) discovery has revolutionized the gene editing technology for targeted approach. The greatest obstacle for CRISPR/Cas9 is cargo delivery systems and both viral and plasmid methods have disadvantages. Here, we report a highly efficient novel CRISPR based gene editing strategy, CRISPR-dots for targeting virulence factor Fimbrial Adhesion (papG gene), the bacterial adhesion molecule. Carbon quantum dots (CQD) were used as a delivery vehicle for Cas9 and gRNA into CFT073, a UPEC strain. CQDs were covalently conjugated to cas9 and papG-targeted guide RNA (gRNA) forming a nanocomplex CRISPR-dots (Cri-dots) as confirmed by DLS and transmission electron microscopy. Cri-dots-papG significantly targeted papG as demonstrated by decrease in the expression of papG.Further papG deficient UPEC had significantly reduced adherence ability and biofilm forming ability as demonstrated by fluorescence microscopy and scanning electron microscopy. Also, papG deficient UPEC had reduced virulence as shown by significantly increased survival of Caenorhabditis elegans (C. elegans) worms compared to UPEC. Our findings suggest that targeting of papG gene using Cri-dots nanocomplexes significantly reduced the pathogenicity of UPEC. Thus, Cri-dots nanocomplex offer a novel anti-bacterial strategy against multi-drug resistant UPEC.
Assuntos
Adesinas de Escherichia coli/genética , Sistemas CRISPR-Cas , Infecções por Escherichia coli/microbiologia , Proteínas de Fímbrias/genética , Edição de Genes/métodos , Pontos Quânticos/administração & dosagem , Infecções Urinárias/microbiologia , Escherichia coli Uropatogênica/genética , Animais , Aderência Bacteriana/efeitos dos fármacos , Biofilmes/efeitos dos fármacos , Proteína 9 Associada à CRISPR/administração & dosagem , Proteína 9 Associada à CRISPR/genética , Caenorhabditis elegans/microbiologia , Carbono , Sistemas de Liberação de Medicamentos , Escherichia coli K12/efeitos dos fármacos , Escherichia coli K12/genética , Células HeLa , Hemaglutinação/efeitos dos fármacos , Humanos , Manose/farmacologia , Veículos Farmacêuticos , Pontos Quânticos/toxicidade , RNA Guia de Cinetoplastídeos/administração & dosagem , RNA Guia de Cinetoplastídeos/genética , Células THP-1 , Escherichia coli Uropatogênica/efeitos dos fármacos , Escherichia coli Uropatogênica/isolamento & purificação , Escherichia coli Uropatogênica/patogenicidade , Virulência/genéticaRESUMO
The CRISPR/Cas9 system is a powerful tool for generation of genetically modified mice. In conventional protocols, Cas9 protein (or mRNA) and sgRNA are introduced into zygotes by microinjection. However, microinjection requires special skill and is too time-consuming to treat zygotes on a large scale. Recently, we have developed a simple electroporation method which generates genetically modified mice with high efficiency. Here, we describe our method GEEP (genome editing by electroporation of Cas9 protein). This method facilitates high-throughput genetic analysis of the mouse. This chapter describes the GEEP method to generate genetically modified mice.
Assuntos
Proteína 9 Associada à CRISPR/administração & dosagem , Eletroporação/métodos , RNA Guia de Cinetoplastídeos/administração & dosagem , Zigoto/crescimento & desenvolvimento , Animais , Animais Geneticamente Modificados/crescimento & desenvolvimento , Sistemas CRISPR-Cas , Feminino , Masculino , Camundongos , Zigoto/químicaRESUMO
Chronic hepatitis B virus (HBV) infection constitutes a global health issue with limited current therapeutic efficacy owing to the persistence of viral episomal DNA (cccDNA). The CRISPR/Cas9 system, a newly developed, powerful tool for genome editing and potential gene therapy, requires efficient delivery of CRISPR components for successful therapeutic application. Here, we investigated the effects of lentiviral- or adeno-associated virus 2 (AAV2) vector-mediated delivery of 3 guide (g)RNAs/Cas9 selected from 16 gRNAs. These significantly suppressed HBV replication in cells, with WJ11/Cas9 exhibiting highest efficacy and chosen for in vivo study. AAV2/WJ11-Cas9 also significantly inhibited HBV replication and significantly reduced cccDNA in the tested cells. Moreover, AAV2/WJ11-Cas9 enhanced entecavir effects when used in combination, indicative of different modes of action. Notably, in humanized chimeric mice, AAV2/WJ11-Cas9 significantly suppressed HBcAg, HBsAg, and HBV DNA along with cccDNA in the liver tissues without significant cytotoxicity; accordingly, next generation sequencing data showed no significant genomic mutations. To our knowledge, this represents the first evaluation of the CRISPR/Cas9 system using an HBV natural infection mode. Therefore, WJ11/Cas9 delivered by comparatively safer AAV2 vectors may provide a new therapeutic strategy for eliminating HBV infection and serve as an effective platform for curing chronic HBV infection.
Assuntos
Proteína 9 Associada à CRISPR/genética , Sistemas CRISPR-Cas , DNA Circular/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Vírus da Hepatite B/genética , RNA Guia de Cinetoplastídeos/genética , Adenoviridae/genética , Animais , Animais Geneticamente Modificados , Proteína 9 Associada à CRISPR/administração & dosagem , DNA Viral/genética , Guanina/análogos & derivados , Guanina/farmacologia , Células HEK293 , Células Hep G2 , Hepatite B/terapia , Hepatite B/virologia , Antígenos de Superfície da Hepatite B/genética , Vírus da Hepatite B/efeitos dos fármacos , Humanos , Masculino , Camundongos , Plasmídeos/genéticaRESUMO
The clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system has facilitated dramatic progress in the field of genome engineering. Whilst microinjection of the Cas9 protein and a single guide RNA (sgRNA) into mouse zygotes is a widespread method for producing genetically engineered mice, in vitro and in vivo electroporation (which are much more convenient strategies) have recently been developed. However, it remains unknown whether these electroporation methods are able to manipulate genomes at the chromosome level. In the present study, we used these techniques to introduce chromosomal inversions of several megabases (Mb) in length in mouse zygotes. Using in vitro electroporation, we successfully introduced a 7.67 Mb inversion, which is longer than any previously reported inversion produced using microinjection-based methods. Additionally, using in vivo electroporation, we also introduced a long chromosomal inversion by targeting an allele in F1 hybrid mice. To our knowledge, the present study is the first report of target-specific chromosomal inversions in mammalian zygotes using electroporation.
Assuntos
Inversão Cromossômica/genética , Cromossomos/genética , Eletroporação/métodos , Engenharia Genética/métodos , Zigoto , Alelos , Animais , Proteína 9 Associada à CRISPR/administração & dosagem , Sistemas CRISPR-Cas , Feminino , Genoma , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Microinjeções , RNA Guia de Cinetoplastídeos/administração & dosagemRESUMO
Delivery technologies for the CRISPR-Cas9 (CRISPR, clustered regularly interspaced short palindromic repeats) gene editing system often require viral vectors, which pose safety concerns for therapeutic genome editing1. Alternatively, cationic liposomal components or polymers can be used to encapsulate multiple CRISPR components into large particles (typically >100 nm diameter); however, such systems are limited by variability in the loading of the cargo. Here, we report the design of customizable synthetic nanoparticles for the delivery of Cas9 nuclease and a single-guide RNA (sgRNA) that enables the controlled stoichiometry of CRISPR components and limits the possible safety concerns in vivo. We describe the synthesis of a thin glutathione (GSH)-cleavable covalently crosslinked polymer coating, called a nanocapsule (NC), around a preassembled ribonucleoprotein (RNP) complex between a Cas9 nuclease and an sgRNA. The NC is synthesized by in situ polymerization, has a hydrodynamic diameter of 25 nm and can be customized via facile surface modification. NCs efficiently generate targeted gene edits in vitro without any apparent cytotoxicity. Furthermore, NCs produce robust gene editing in vivo in murine retinal pigment epithelium (RPE) tissue and skeletal muscle after local administration. This customizable NC nanoplatform efficiently delivers CRISPR RNP complexes for in vitro and in vivo somatic gene editing.
Assuntos
Proteína 9 Associada à CRISPR/administração & dosagem , Sistemas CRISPR-Cas , Edição de Genes , Nanocápsulas/química , RNA Guia de Cinetoplastídeos/administração & dosagem , Animais , Proteína 9 Associada à CRISPR/genética , Glutationa/química , Células HEK293 , Humanos , Camundongos , Polímeros/química , RNA Guia de Cinetoplastídeos/genéticaRESUMO
The CRISPR/Cas9 system is a revolutionary gene editing technology that combines simplicity of use and efficiency of mutagenesis. As this technology progresses toward human therapies, valid concerns including off-target mutations and immunogenicity must be addressed. One approach to address these issues is to minimize the presence of the CRISPR/Cas9 components by maintaining a tighter temporal control of Cas9 endonuclease and reducing the time period of activity. This has been achieved to some degree by delivering the CRISPR/Cas9 system via pre-formed Cas9 + gRNA ribonucleoprotein (RNP) complexes. In this review, we first discuss the molecular modifications that can be made using CRISPR/Cas9 and provide an overview of current methods for delivering Cas9 RNP complexes both in vitro and in vivo. We conclude with examples of how Cas9 RNP delivery may be used to target neuroinflammatory processes, namely in regard to viral infections of the central nervous system and neurodegenerative diseases. We propose that Cas9 RNP delivery is a viable approach when considering the CRISPR/Cas9 system for both experimentation and the treatment of disease. Graphical Abstract.
Assuntos
Proteína 9 Associada à CRISPR/genética , Sistemas CRISPR-Cas/fisiologia , Edição de Genes/métodos , Técnicas de Transferência de Genes , Ribonucleoproteínas/genética , Animais , Proteína 9 Associada à CRISPR/administração & dosagem , Proteína 9 Associada à CRISPR/metabolismo , Edição de Genes/tendências , Técnicas de Transferência de Genes/tendências , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/terapia , Ribonucleoproteínas/administração & dosagem , Ribonucleoproteínas/metabolismoRESUMO
The selective regulation of bacteria in complex microbial populations is key to controlling pathogenic bacteria. CRISPR nucleases can be programmed to kill bacteria, but require an efficient and broad-host range delivery system to be effective. Here, using an Escherichia coli and Salmonella enterica co-culture system, we show that plasmids based on the IncP RK2 conjugative system can be used as delivery vectors for a TevSpCas9 dual nuclease. Notably, a cis-acting plasmid that encodes the conjugation and CRISPR machinery conjugates from E. coli to S. enterica with high frequency compared to a trans system that separates conjugation and CRISPR machinery. In culture conditions that enhance cell-to-cell contact, conjugation rates approach 100% with the cis-acting plasmid. Targeting of single or multiplexed sgRNAs to non-essential genes results in high S. enterica killing efficiencies. Our data highlight the potential of cis-acting conjugative plasmids as a delivery system for CRISPR nucleases or other microbial-altering agents for targeted bacterial killing.
Assuntos
Anti-Infecciosos/administração & dosagem , Proteína 9 Associada à CRISPR/administração & dosagem , Conjugação Genética , Sistemas de Liberação de Medicamentos/métodos , Técnicas de Transferência de Genes , Biofilmes/efeitos dos fármacos , Proteína 9 Associada à CRISPR/genética , Técnicas de Cocultura , Escherichia coli/efeitos dos fármacos , Escherichia coli/genética , Testes de Sensibilidade Microbiana , Plasmídeos/genética , RNA Guia de Cinetoplastídeos/genética , Saccharomyces cerevisiae , Salmonella enterica/efeitos dos fármacos , Salmonella enterica/genéticaRESUMO
Majority of disease-modifying therapeutic targets are restricted to the intracellular space and are therefore not druggable using existing biologic modalities. The ability to efficiently deliver macromolecules inside target cells or tissues would greatly expand the current landscape of therapeutic targets for future generations of biologic drugs, but remains challenging. Here we report the use of extracellular vesicles, known as arrestin domain containing protein 1 [ARRDC1]-mediated microvesicles (ARMMs), for packaging and intracellular delivery of a myriad of macromolecules, including the tumor suppressor p53 protein, RNAs, and the genome-editing CRISPR-Cas9/guide RNA complex. We demonstrate selective recruitment of these macromolecules into ARMMs. When delivered intracellularly via ARMMs, these macromolecules are biologically active in recipient cells. P53 delivered via ARMMs induces DNA damage-dependent apoptosis in multiple tissues in mice. Together, our results provide proof-of-principle demonstration that ARMMs represent a highly versatile platform for packaging and intracellular delivery of therapeutic macromolecules.
Assuntos
Arrestinas/metabolismo , Portadores de Fármacos/metabolismo , Vesículas Extracelulares/metabolismo , Células A549 , Animais , Proteína 9 Associada à CRISPR/administração & dosagem , Proteínas de Fluorescência Verde , Células HEK293 , Humanos , Camundongos Knockout , RNA/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Proteína Supressora de Tumor p53/administração & dosagem , Proteína Supressora de Tumor p53/genéticaRESUMO
Zygote-microinjection or in vitro electroporation of isolated zygotes are now widely used methods to produce genome-edited mice. However, these technologies require laborious and time-consuming ex vivo handling of fertilized eggs, including zygote isolation, gene delivery into zygotes and embryo transfer into recipients. We recently developed an alternative method called improved genome-editing via oviductal nucleic acids delivery (i-GONAD), which does not require the above-mentioned ex vivo handing of zygotes, but instead involves intraoviductal instillation of genome-editing components, Cas9 protein and synthetic gRNAs, into the oviducts of pregnant females at the late 1-cell embryo stage under a dissecting microscope and subsequent electroporation. With this method, we succeeded in generating genome-edited mice at relatively high efficiencies (for example, knockout alleles were produced at ~97% efficiency). Here, we extended this improved technology to rats, and found that i-GONAD can create genome-edited rats in various strains, including Sprague Dawley and Lewis, and F1 hybrids (between Sprague Dawley and Brown Norway), with efficiencies of ~62% for indel mutations and ~9% for knock-ins. Thus, i-GONAD will be especially useful for the production of genome-edited rats in small laboratories where expensive micromanipulator systems and highly skilled personnel for embryo manipulation are unavailable.
Assuntos
Sistemas CRISPR-Cas/genética , Eletroporação/métodos , Tubas Uterinas , Edição de Genes/métodos , Animais , Proteína 9 Associada à CRISPR/administração & dosagem , Proteína 9 Associada à CRISPR/genética , Embrião de Mamíferos , Feminino , Masculino , Mutação , Fator de Transcrição PAX6/genética , Gravidez , RNA Guia de Cinetoplastídeos/administração & dosagem , RNA Guia de Cinetoplastídeos/genética , Ratos , Ratos Endogâmicos Lew , Ratos Sprague-Dawley , Ratos TransgênicosRESUMO
Genome-editing technologies have revolutionized the biomedical sciences by providing researchers with the ability to quickly and efficiently modify genes. While programmable nucleases can be introduced into cells using a variety of techniques, their delivery as purified proteins is an effective approach for limiting off-target effects. Here, we describe step-by-step procedures for manufacturing and delivering genome-modifying proteins-including Cas9 ribonucleoproteins (RNPs) and TALE and zinc-finger nucleases-into mammalian cells. Protocols for combining Cas9 RNP with naturally recombinogenic adeno-associated virus (AAV) donor vectors for the seamless insertion of transgenes by homology-directed genome editing are also provided.