Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters











Publication year range
1.
Sci Rep ; 11(1): 17801, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34493749

ABSTRACT

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.


Subject(s)
Adhesins, Escherichia coli/genetics , CRISPR-Cas Systems , Escherichia coli Infections/microbiology , Fimbriae Proteins/genetics , Gene Editing/methods , Quantum Dots/administration & dosage , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/genetics , Animals , Bacterial Adhesion/drug effects , Biofilms/drug effects , CRISPR-Associated Protein 9/administration & dosage , CRISPR-Associated Protein 9/genetics , Caenorhabditis elegans/microbiology , Carbon , Drug Delivery Systems , Escherichia coli K12/drug effects , Escherichia coli K12/genetics , HeLa Cells , Hemagglutination/drug effects , Humans , Mannose/pharmacology , Pharmaceutical Vehicles , Quantum Dots/toxicity , RNA, Guide, Kinetoplastida/administration & dosage , RNA, Guide, Kinetoplastida/genetics , THP-1 Cells , Uropathogenic Escherichia coli/drug effects , Uropathogenic Escherichia coli/isolation & purification , Uropathogenic Escherichia coli/pathogenicity , Virulence/genetics
3.
Exp Biol Med (Maywood) ; 246(14): 1643-1649, 2021 07.
Article in English | MEDLINE | ID: mdl-33899542

ABSTRACT

The year 2020 witnessed an unpredictable pandemic situation due to novel coronavirus (COVID-19) outbreaks. This condition can be more severe if the patient has comorbidities. Failure of viable treatment for such viral infection caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is due to lack of identification. Thus, modern and productive biotechnology-based tools are being used to manipulate target genes by introducing the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas (CRISPR-associated) system. Moreover, it has now been used as a tool to inhibit viral replication. Hence, it can be hypothesized that the CRISPR/Cas system can be a viable tool to target both the SARS-CoV-2 genome with specific target RNA sequence and host factors to destroy the SARS-CoV-2 community via inhibition of viral replication and infection. Moreover, comorbidities and COVID-19 escalate the rate of mortality globally, and as a result, we have faced this pandemic. CRISPR/Cas-mediated genetic manipulation to knockdown viral sequences may be a preventive strategy against such pandemic caused by SARS-CoV-2. Furthermore, prophylactic antiviral CRISPR in human cells (PAC-MAN) along with CRISPR/Cas13d efficiently degrades the specific RNA sequence to inhibit viral replication. Therefore, we suggest that CRISPR/Cas system with PAC-MAN could be a useful tool to fight against such a global pandemic caused by SARS-CoV-2. This is an alternative preventive approach of management against the pandemic to destroy the target sequence of RNA in SARS-CoV-2 by viral inhibition.


Subject(s)
Antiviral Agents/pharmacology , COVID-19/virology , Gene Transfer Techniques , Host-Pathogen Interactions/physiology , SARS-CoV-2/genetics , Antiviral Agents/administration & dosage , COVID-19/epidemiology , COVID-19/genetics , CRISPR-Cas Systems , Gene Editing/methods , Host-Pathogen Interactions/genetics , Humans , RNA, Guide, Kinetoplastida/administration & dosage , RNA, Guide, Kinetoplastida/pharmacology , SARS-CoV-2/pathogenicity
4.
Cancer Res ; 81(5): 1332-1346, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33372040

ABSTRACT

Although the cyclin-dependent kinases CDK4 and CDK6 play fundamental roles in cancer, the specific pathways and downstream targets by which they exert their tumorigenic effects remain elusive. In this study, we uncover distinct and novel functions for these kinases in regulating tumor formation and metastatic colonization in various solid tumors, including those of the breast, prostate, and pancreas. Combining in vivo CRISPR-based CDK4 and CDK6 gene editing with pharmacologic inhibition approaches in orthotopic transplantation and patient-derived xenograft preclinical models, we defined clear functions for CDK4 and CDK6 in facilitating tumor growth and progression in metastatic cancers. Transcriptomic profiling of CDK4/6 CRISPR knockouts in breast cancer revealed these two kinases to regulate cancer progression through distinct mechanisms. CDK4 regulated prometastatic inflammatory cytokine signaling, whereas CDK6 mainly controlled DNA replication and repair processes. Inhibition of CDK6 but not CDK4 resulted in defective DNA repair and increased DNA damage. Multiple CDK6 DNA replication/repair genes were not only associated with cancer subtype, grades, and poor clinical outcomes, but also facilitated primary tumor growth and metastasis in vivo. CRISPR-based genomic deletion of CDK6 efficiently blocked tumor formation and progression in preestablished cell- and patient-derived xenograft preclinical models of breast cancer, providing a potential novel targeted therapy for these deadly tumors. SIGNIFICANCE: In-depth transcriptomic analysis identifies cyclin-dependent kinases CDK4 and CDK6 as regulators of metastasis through distinct signaling pathways and reveals the DNA replication/repair pathway as central in promoting these effects.


Subject(s)
Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 6/genetics , DNA Repair/physiology , DNA Replication/physiology , Neoplasms/pathology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Male , Mice, SCID , Neoplasms/genetics , RNA, Guide, Kinetoplastida/administration & dosage , RNA, Guide, Kinetoplastida/pharmacology , Xenograft Model Antitumor Assays
5.
Sci Rep ; 10(1): 18583, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33122788

ABSTRACT

The ability to ablate a gene in a given tissue by generating a conditional knockout (cKO) is crucial for determining its function in the targeted tissue. Such tissue-specific ablation is even more critical when the gene's conventional knockout (KO) is lethal, which precludes studying the consequences of its deletion in other tissues. Therefore, here we describe a successful strategy that generated a Matrix Gla floxed mouse (Mgp.floxed) by the CRISPR/Cas9 system, that subsequently allowed the generation of cKOs by local viral delivery of the Cre-recombinase enzyme. MGP is a well-established inhibitor of calcification gene, highly expressed in arteries' smooth muscle cells and chondrocytes. MGP is also one of the most abundant genes in the trabecular meshwork, the eye tissue responsible for maintenance of intraocular pressure (IOP) and development of Glaucoma. Our strategy entailed one-step injection of two gRNAs, Cas9 protein and a long-single-stranded-circular DNA donor vector (lsscDNA, 6.7 kb) containing two loxP sites in cis and 900-700 bp 5'/3' homology arms. Ocular intracameral injection of Mgp.floxed mice with a Cre-adenovirus, led to an Mgp.TMcKO mouse which developed elevated IOP. Our study discovered a new role for the Mgp gene as a keeper of physiological IOP in the eye.


Subject(s)
Calcium-Binding Proteins/physiology , Extracellular Matrix Proteins/physiology , Eye/physiopathology , Intraocular Pressure , Trabecular Meshwork/physiopathology , Animals , Base Sequence , Female , Glaucoma/physiopathology , Integrases/metabolism , Mice , Mice, Knockout , RNA, Guide, Kinetoplastida/administration & dosage , Matrix Gla Protein
6.
J Cell Mol Med ; 24(18): 10512-10524, 2020 09.
Article in English | MEDLINE | ID: mdl-32691935

ABSTRACT

Dysregulation of long non-coding RNAs (lncRNAs) has been implicated in many cancer developments. Previous studies showed that lncRNA LINC00941 was aberrantly expressed in oral squamous cell carcinoma (OSCC). However, its role in OSCC development remains elusive. In this study, we demonstrated that in OSCC cells, EP300 activates LINC00941 transcription through up-regulating its promoter H3K27ac modification. Up-regulated LINC00941 in turn activates CAPRIN2 expression by looping to CAPRIN2 promoter. Functional assays suggest that both LINC00941 and CAPRIN2 play pivotal roles in promoting OSCC cell proliferation and colony formation. In vivo assay further confirmed the role of LINC00941 in promoting OSCC cell tumour formation. Lastly, we showed that the role of LINC00941 and CAPRIN2 in OSCC progression was mediated through activating the canonical WNT/ß-catenin signaling pathway. Thus, LINC00941/CAPRIN2/ WNT/ß-catenin signaling pathway provides new therapeutic targets for OSCC treatment.


Subject(s)
Carcinoma, Squamous Cell/pathology , Mouth Neoplasms/pathology , Neoplasm Proteins/physiology , RNA, Long Noncoding/metabolism , RNA, Neoplasm/physiology , RNA-Binding Proteins/physiology , Wnt Signaling Pathway/physiology , Animals , CRISPR-Cas Systems , Carcinoma, Squamous Cell/genetics , Cell Division , Cells, Cultured , DNA, Neoplasm/genetics , DNA, Neoplasm/ultrastructure , Disease Progression , E1A-Associated p300 Protein/physiology , Gene Expression Regulation, Neoplastic , Genes, Reporter , Histone Code , Keratinocytes , Mice , Mice, Inbred BALB C , Mice, Nude , Mouth Neoplasms/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Transplantation , Promoter Regions, Genetic/genetics , RNA, Guide, Kinetoplastida/administration & dosage , RNA, Guide, Kinetoplastida/genetics , RNA, Long Noncoding/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/genetics , Recombinant Proteins/metabolism , Tumor Stem Cell Assay , Up-Regulation , Wnt Signaling Pathway/genetics
7.
EBioMedicine ; 58: 102897, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32711250

ABSTRACT

BACKGROUND: Gene therapy has held promises for treating specific genetic diseases. However, the key to clinical application depends on effective gene delivery. METHODS: Using a large animal model, we developed two pharmaceutical formulations for gene delivery in the pigs' vagina, which were made up of poly (ß-amino ester) (PBAE)-plasmid polyplex nanoparticles (NPs) based two gel materials, modified montmorillonite (mMMT) and hectorite (HTT). FINDINGS: By conducting flow cytometry of the cervical cells, we found that PBAE-GFP-NPs-mMMT gel was more efficient than PBAE-GFP-NPs-HTT gel in delivering exogenous DNA intravaginally. Next, we designed specific CRISPR/SpCas9 sgRNAs targeting porcine endogenous retroviruses (PERVs) and evaluated the genome editing efficacy in vivo. We discovered that PERV copy number in vaginal epithelium could be significantly reduced by the local delivery of the PBAE-SpCas9/sgRNA NPs-mMMT gel. Comparable genome editing results were also obtained by high-fidelity version of SpCas9, SpCas9-HF1 and eSpCas9, in the mMMT gel. Further, we confirmed that the expression of topically delivered SpCas9 was limited to the vagina/cervix and did not diffuse to nearby organs, which was relatively safe with low toxicity. INTERPRETATION: Our data suggested that the PBAE-NPs mMMT vaginal gel is an effective preparation for local gene therapy, yielding insights into novel therapeutic approaches to sexually transmitted disease in the genital tract. FUNDING: This work was supported by the National Science and Technology Major Project of the Ministry of science and technology of China (No. 2018ZX10301402); the National Natural Science Foundation of China (81761148025, 81871473 and 81402158); Guangzhou Science and Technology Programme (No. 201704020093); National Ten Thousand Plan-Young Top Talents of China, Fundamental Research Funds for the Central Universities (17ykzd15 and 19ykyjs07); Three Big Constructions-Supercomputing Application Cultivation Projects sponsored by National Supercomputer Center In Guangzhou; the National Research FFoundation (NRF) South Africa under BRICS Multilateral Joint Call for Proposals; grant 17-54-80078 from the Russian Foundation for Basic Research.


Subject(s)
Cervix Uteri/cytology , Endogenous Retroviruses/genetics , Gene Dosage/drug effects , Polymers/chemistry , RNA, Guide, Kinetoplastida/administration & dosage , Administration, Intravaginal , Animals , Bentonite/chemistry , CRISPR-Cas Systems , Cells, Cultured , Cervix Uteri/chemistry , Endogenous Retroviruses/drug effects , Female , Gene Editing , Genetic Therapy , Mice , Models, Animal , Nanoparticles , Plasmids/administration & dosage , Plasmids/genetics , Silicates/chemistry , Swine , Vaginal Creams, Foams, and Jellies
8.
Methods Mol Biol ; 2050: 121-126, 2020.
Article in English | MEDLINE | ID: mdl-31468486

ABSTRACT

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.


Subject(s)
CRISPR-Associated Protein 9/administration & dosage , Electroporation/methods , RNA, Guide, Kinetoplastida/administration & dosage , Zygote/growth & development , Animals , Animals, Genetically Modified/growth & development , CRISPR-Cas Systems , Female , Male , Mice , Zygote/chemistry
9.
J Am Chem Soc ; 141(48): 19032-19037, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31729871

ABSTRACT

Precisely assembled DNA nanostructures are promising candidates for the delivery of biomolecule-based therapeutics. Herein, we introduce a facile strategy for the construction of a branched DNA-based nanoplatform for codelivery of gene editing (sgRNA/Cas9, targeting DNA in the nucleus) and gene silencing (antisense, targeting mRNA in the cytoplasm) components for synergistic tumor therapy in vitro and in vivo. In our design, the branched DNA structure can efficiently load a sgRNA/Cas9/antisense complex targeting a tumor-associated gene, PLK1, through DNA self-assembly. With the incorporation of an active targeting aptamer and an endosomal escape peptide by host-guest interaction, the biocompatible DNA nanoplatform demonstrates efficient inhibition of tumor growth without apparent systemic toxicity. This multifunctional DNA nanocarrier provides a new strategy for the development of gene therapeutics.


Subject(s)
Breast Neoplasms/therapy , CRISPR-Cas Systems , DNA/chemistry , Gene Editing/methods , RNA, Antisense/administration & dosage , RNA, Guide, Kinetoplastida/administration & dosage , Animals , Breast Neoplasms/genetics , Cell Cycle Proteins/genetics , Female , Genetic Therapy/methods , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Nanostructures/chemistry , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , RNA, Antisense/genetics , RNA, Antisense/therapeutic use , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/therapeutic use , Polo-Like Kinase 1
10.
Sci Rep ; 9(1): 14900, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31624292

ABSTRACT

Genetic mosaicism is the presence of more than two alleles on an individual and it is commonly observed following CRISPR microinjection of zygotes. This phenomenon appears when DNA replication precedes CRISPR-mediated genome edition and it is undesirable because it reduces greatly the odds for direct KO generation by randomly generated indels. In this study, we have developed alternative protocols to reduce mosaicism rates following CRISPR-mediated genome edition in bovine. In a preliminary study we observed by EdU incorporation that DNA replication has already occurred at the conventional microinjection time (20 hpi). Aiming to reduce mosaicism appearance, we have developed three alternative microinjection protocols: early zygote microinjection (10 hpi RNA) or oocyte microinjection before fertilization with either RNA or Ribonucleoprotein delivery (0 hpi RNA or 0 hpi RNP). All three alternative microinjection protocols resulted in similar blastocyst and genome edition rates compared to the conventional 20 hpi group, whereas mosaicism rates were significantly reduced in all early delivery groups (~10-30% of edited embryos being mosaic depending on the loci) compared to conventional 20 hpi microinjection (100% mosaicism rate). These strategies constitute an efficient way to reduce the number of indels, increasing the odds for direct KO generation.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing/methods , Gene Knockout Techniques/methods , Mosaicism , Animals , Blastocyst , Cattle , DNA Replication , Female , Genotyping Techniques , INDEL Mutation , Microinjections/methods , Models, Animal , Oocytes , RNA, Guide, Kinetoplastida/administration & dosage , RNA, Guide, Kinetoplastida/genetics , Ribonucleoproteins/administration & dosage , Ribonucleoproteins/genetics , Zygote
11.
Sci Rep ; 9(1): 14713, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31604975

ABSTRACT

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.


Subject(s)
Chromosome Inversion/genetics , Chromosomes/genetics , Electroporation/methods , Genetic Engineering/methods , Zygote , Alleles , Animals , CRISPR-Associated Protein 9/administration & dosage , CRISPR-Cas Systems , Female , Genome , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Microinjections , RNA, Guide, Kinetoplastida/administration & dosage
12.
Nat Nanotechnol ; 14(10): 974-980, 2019 10.
Article in English | MEDLINE | ID: mdl-31501532

ABSTRACT

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.


Subject(s)
CRISPR-Associated Protein 9/administration & dosage , CRISPR-Cas Systems , Gene Editing , Nanocapsules/chemistry , RNA, Guide, Kinetoplastida/administration & dosage , Animals , CRISPR-Associated Protein 9/genetics , Glutathione/chemistry , HEK293 Cells , Humans , Mice , Polymers/chemistry , RNA, Guide, Kinetoplastida/genetics
13.
Genome Res ; 29(9): 1442-1452, 2019 09.
Article in English | MEDLINE | ID: mdl-31467027

ABSTRACT

Obesity is an increasing pathophysiological problem in developed societies. Despite all major progress in understanding molecular mechanisms of obesity, currently available anti-obesity drugs have shown limited efficacy with severe side effects. CRISPR interference (CRISPRi) mechanism based on catalytically dead Cas9 (dCas9) and single guide RNA (sgRNA) was combined with a targeted nonviral gene delivery system to treat obesity and obesity-induced type 2 diabetes. A fusion peptide targeting a vascular and cellular marker of adipose tissue, prohibitin, was developed by conjugation of adipocyte targeting sequence (CKGGRAKDC) to 9-mer arginine (ATS-9R). (dCas9/sgFabp4) + ATS-9R oligoplexes showed effective condensation and selective delivery into mature adipocytes. Targeted delivery of the CRISPRi system against Fabp4 to white adipocytes by ATS-9R induced effective silencing of Fabp4, resulting in reduction of body weight and inflammation and restoration of hepatic steatosis in obese mice. This RNA-guided DNA recognition platform provides a simple and safe approach to regress and treat obesity and obesity-induced metabolic syndromes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Fatty Acid-Binding Proteins/genetics , Fatty Liver/drug therapy , Obesity/drug therapy , RNA, Guide, Kinetoplastida/administration & dosage , 3T3 Cells , Adipocytes, White/chemistry , Adipocytes, White/cytology , Animals , CRISPR-Cas Systems , Cytokines/metabolism , Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Fatty Acid-Binding Proteins/antagonists & inhibitors , Fatty Liver/genetics , Gene Expression Regulation , Gene Knockdown Techniques , Insulin Resistance , Mice , Molecular Targeted Therapy , Obesity/genetics , RNA, Guide, Kinetoplastida/pharmacology
14.
Nat Protoc ; 14(8): 2452-2482, 2019 08.
Article in English | MEDLINE | ID: mdl-31341289

ABSTRACT

Methods to create genetically engineered mice involve three major steps: harvesting embryos from one set of females, microinjection of reagents into embryos ex vivo and their surgical transfer to another set of females. Although tedious, these methods have been used for more than three decades to create mouse models. We recently developed a method named GONAD (genome editing via oviductal nucleic acids delivery), which bypasses these steps. GONAD involves injection of CRISPR components (Cas9 mRNA and guide RNA (gRNA)) into the oviducts of pregnant females 1.5 d post conception, followed by in vivo electroporation to deliver the components into the zygotes in situ. Using GONAD, we demonstrated that target genes can be disrupted and analyzed at different stages of mouse embryonic development. Subsequently, we developed improved GONAD (i-GONAD) by delivering CRISPR ribonucleoproteins (RNPs; Cas9 protein or Cpf1 protein and gRNA) into day-0.7 pregnant mice, which made it suitable for routine generation of knockout and large-deletion mouse models. i-GONAD can also generate knock-in models containing up to 1-kb inserts when single-stranded DNA (ssDNA) repair templates are supplied. i-GONAD offers other advantages: it does not require vasectomized males and pseudo-pregnant females, the females used for i-GONAD are not sacrificed and can be used for other experiments, it can be easily adopted in laboratories lacking sophisticated microinjection equipment, and can be implemented by researchers skilled in small-animal surgery but lacking embryo-handling skills. Here, we provide a step-by-step protocol for establishing the i-GONAD method. The protocol takes ∼6 weeks to generate the founder mice.


Subject(s)
CRISPR-Cas Systems/genetics , Electroporation/methods , Gene Editing/methods , Animals , Female , Male , Mice , Microinjections , Oviducts/physiology , Pregnancy , RNA, Guide, Kinetoplastida/administration & dosage , RNA, Guide, Kinetoplastida/genetics , RNA, Messenger/administration & dosage , RNA, Messenger/genetics
15.
Adv Mater ; 31(33): e1902575, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31215123

ABSTRACT

A main challenge to broaden the biomedical application of CRISPR/Cas9 (clustered regularly interspaced short palindromic repeat (CRISPR) associated protein 9) genome editing technique is the delivery of Cas9 nuclease and single-guide RNA (sgRNA) into the specific cell and organ. An effective and very fast CRISPR/Cas9 genome editing in vitro and in vivo enabled by bioreducible lipid/Cas9 messenger RNA (mRNA) nanoparticle is reported. BAMEA-O16B, a lipid nanoparticle integrated with disulfide bonds, can efficiently deliver Cas9 mRNA and sgRNA into cells while releasing RNA in response to the reductive intracellular environment for genome editing as fast as 24 h post mRNA delivery. It is demonstrated that the simultaneous delivery of Cas9 mRNA and sgRNA using BAMEA-O16B knocks out green fluorescent protein (GFP) expression of human embryonic kidney cells with efficiency up to 90%. Moreover, the intravenous injection of BAMEA-O16B/Cas9 mRNA/sgRNA nanoparticle effectively accumulates in hepatocytes, and knocks down proprotein convertase subtilisin/kexin type 9 level in mouse serum down to 20% of nontreatment. The leading lipid nanoparticle, BAMEA-O16B, represents one of the most efficient CRISPR/Cas9 delivery nanocarriers reported so far, and it can broaden the therapeutic promise of mRNA and CRISPR/Cas9 technique further.


Subject(s)
CRISPR-Associated Protein 9/genetics , Gene Editing/methods , Lipids/chemistry , Nanoparticles/chemistry , RNA, Guide, Kinetoplastida/chemistry , RNA, Messenger/chemistry , Animals , Biological Transport , Cell Line, Tumor , Gene Knockdown Techniques/methods , Gene Transfer Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Oxidation-Reduction , Proprotein Convertase 9/genetics , Proprotein Convertase 9/metabolism , RNA, Guide, Kinetoplastida/administration & dosage , RNA, Messenger/administration & dosage
16.
Sci Rep ; 9(1): 3928, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30850620

ABSTRACT

CRISPR-Cas9/gRNA exhibits therapeutic efficacy against latent human immunodeficiency virus (HIV) genome but the delivery of this therapeutic cargo to the brain remains as a challenge. In this research, for the first time, we demonstrated magnetically guided non-invasive delivery of a nano-formulation (NF), composed of Cas9/gRNA bound with magneto-electric nanoparticles (MENPs), across the blood-brain barrier (BBB) to inhibit latent HIV-1 infection in microglial (hµglia)/HIV (HC69) cells. An optimized ac-magnetic field of 60 Oe was applied on NF to release Cas9/gRNA from MENPs surface and to facilitate NF cell uptake resulting in intracellular release and inhibition of HIV. The outcomes suggested that developed NF reduced HIV-LTR expression significantly in comparison to unbound Cas9/gRNA in HIV latent hµglia/HIV (HC69) cells. These findings were also validated qualitatively using fluorescence microscopy to assess NF efficacy against latent HIV in the microglia cells. We believe that CNS delivery of NF (CRISPR/Cas9-gRNA-MENPs) across the BBB certainly will have clinical utility as future personalized nanomedicine to manage neuroHIV/AIDS.


Subject(s)
Blood-Brain Barrier/virology , HIV Infections/therapy , HIV Infections/virology , HIV-1 , RNA, Guide, Kinetoplastida/administration & dosage , CRISPR-Cas Systems , Cells, Cultured , Drug Delivery Systems , Gene Editing/methods , HIV-1/genetics , Humans , In Vitro Techniques , Magnetite Nanoparticles/administration & dosage , RNA, Guide, Kinetoplastida/genetics , Virus Latency
17.
J Nanobiotechnology ; 17(1): 19, 2019 Jan 29.
Article in English | MEDLINE | ID: mdl-30696428

ABSTRACT

BACKGROUND: Protein-based Cas9 in vivo gene editing therapeutics have practical limitations owing to their instability and low efficacy. To overcome these obstacles and improve stability, we designed a nanocarrier primarily consisting of lecithin that can efficiently target liver disease and encapsulate complexes of Cas9 with a single-stranded guide RNA (sgRNA) ribonucleoprotein (Cas9-RNP) through polymer fusion self-assembly. RESULTS: In this study, we optimized an sgRNA sequence specifically for dipeptidyl peptidase-4 gene (DPP-4) to modulate the function of glucagon-like peptide 1. We then injected our nanocarrier Cas9-RNP complexes directly into type 2 diabetes mellitus (T2DM) db/db mice, which disrupted the expression of DPP-4 gene in T2DM mice with remarkable efficacy. The decline in DPP-4 enzyme activity was also accompanied by normalized blood glucose levels, insulin response, and reduced liver and kidney damage. These outcomes were found to be similar to those of sitagliptin, the current chemical DPP-4 inhibition therapy drug which requires recurrent doses. CONCLUSIONS: Our results demonstrate that a nano-liposomal carrier system with therapeutic Cas9-RNP has great potential as a platform to improve genomic editing therapies for human liver diseases.


Subject(s)
CRISPR-Cas Systems , Diabetes Mellitus, Type 2/therapy , Dipeptidyl Peptidase 4/genetics , Drug Delivery Systems , Genetic Therapy/methods , Lecithins , Liposomes , Animals , Blood Glucose/drug effects , Cell Line , Dipeptidyl Peptidase 4/metabolism , Gene Editing , Gene Targeting , Glucagon-Like Peptide 1/blood , Humans , Lecithins/administration & dosage , Lecithins/chemistry , Liposomes/administration & dosage , Liposomes/chemistry , Mice , Mice, Knockout , RNA, Guide, Kinetoplastida/administration & dosage , RNA, Guide, Kinetoplastida/chemistry , RNA, Guide, Kinetoplastida/genetics
18.
IUBMB Life ; 71(7): 835-844, 2019 07.
Article in English | MEDLINE | ID: mdl-30635953

ABSTRACT

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.


Subject(s)
Fetus/physiology , Gene Editing , Green Fluorescent Proteins/genetics , Heart/embryology , Mutation , Myocytes, Cardiac/physiology , RNA, Guide, Kinetoplastida/genetics , Animals , CRISPR-Associated Protein 9/administration & dosage , CRISPR-Associated Protein 9/genetics , Cells, Cultured , Female , Gene Transfer Techniques , Genetic Vectors , Genome , Heart/physiology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Myocytes, Cardiac/cytology , Plasmids , RNA, Guide, Kinetoplastida/administration & dosage , Transgenes
19.
Sci Rep ; 8(1): 12059, 2018 08 13.
Article in English | MEDLINE | ID: mdl-30104681

ABSTRACT

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.


Subject(s)
CRISPR-Cas Systems/genetics , Electroporation/methods , Fallopian Tubes , Gene Editing/methods , Animals , CRISPR-Associated Protein 9/administration & dosage , CRISPR-Associated Protein 9/genetics , Embryo, Mammalian , Female , Male , Mutation , PAX6 Transcription Factor/genetics , Pregnancy , RNA, Guide, Kinetoplastida/administration & dosage , RNA, Guide, Kinetoplastida/genetics , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Rats, Transgenic
20.
Dev Cell ; 45(4): 526-536.e5, 2018 05 21.
Article in English | MEDLINE | ID: mdl-29787711

ABSTRACT

The targeting efficiency of knockin sequences via homologous recombination (HR) is generally low. Here we describe a method we call Tild-CRISPR (targeted integration with linearized dsDNA-CRISPR), a targeting strategy in which a PCR-amplified or precisely enzyme-cut transgene donor with 800-bp homology arms is injected with Cas9 mRNA and single guide RNA into mouse zygotes. Compared with existing targeting strategies, this method achieved much higher knockin efficiency in mouse embryos, as well as brain tissue. Importantly, the Tild-CRISPR method also yielded up to 12-fold higher knockin efficiency than HR-based methods in human embryos, making it suitable for studying gene functions in vivo and developing potential gene therapies.


Subject(s)
CRISPR-Cas Systems , DNA/administration & dosage , Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Gene Knock-In Techniques/methods , RNA, Guide, Kinetoplastida/administration & dosage , Animals , Cells, Cultured , Electroporation , Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Female , Fertilization in Vitro , Homologous Recombination , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred ICR , Zygote/growth & development , Zygote/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL