Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 8051, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38052872

ABSTRACT

Gene editing strategies for cystic fibrosis are challenged by the complex barrier properties of airway epithelia. We previously reported that the amphiphilic S10 shuttle peptide non-covalently combined with CRISPR-associated (Cas) ribonucleoprotein (RNP) enabled editing of human and mouse airway epithelial cells. Here, we derive the S315 peptide as an improvement over S10 in delivering base editor RNP. Following intratracheal aerosol delivery of Cy5-labeled peptide in rhesus macaques, we confirm delivery throughout the respiratory tract. Subsequently, we target CCR5 with co-administration of ABE8e-Cas9 RNP and S315. We achieve editing efficiencies of up-to 5.3% in rhesus airway epithelia. Moreover, we document persistence of edited epithelia for up to 12 months in mice. Finally, delivery of ABE8e-Cas9 targeting the CFTR R553X mutation restores anion channel function in cultured human airway epithelia. These results demonstrate the therapeutic potential of base editor delivery with S315 to functionally correct the CFTR R553X mutation in respiratory epithelia.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator , Epithelial Cells , Animals , Humans , Mice , Macaca mulatta/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/metabolism , Respiratory Mucosa/metabolism , Ribonucleoproteins/metabolism , Peptides/genetics , CRISPR-Cas Systems
2.
Mol Ther ; 31(4): 1074-1087, 2023 Apr 05.
Article in English | MEDLINE | ID: mdl-36793210

ABSTRACT

While a number of methods exist to investigate CRISPR off-target (OT) editing, few have been compared head-to-head in primary cells after clinically relevant editing processes. Therefore, we compared in silico tools (COSMID, CCTop, and Cas-OFFinder) and empirical methods (CHANGE-Seq, CIRCLE-Seq, DISCOVER-Seq, GUIDE-Seq, and SITE-Seq) after ex vivo hematopoietic stem and progenitor cell (HSPC) editing. We performed editing using 11 different gRNAs complexed with Cas9 protein (high-fidelity [HiFi] or wild-type versions), then performed targeted next-generation sequencing of nominated OT sites identified by in silico and empirical methods. We identified an average of less than one OT site per guide RNA (gRNA) and all OT sites generated using HiFi Cas9 and a 20-nt gRNA were identified by all OT detection methods with the exception of SITE-seq. This resulted in high sensitivity for the majority of OT nomination tools and COSMID, DISCOVER-Seq, and GUIDE-Seq attained the highest positive predictive value (PPV). We found that empirical methods did not identify OT sites that were not also identified by bioinformatic methods. This study supports that refined bioinformatic algorithms could be developed that maintain both high sensitivity and PPV, thereby enabling more efficient identification of potential OT sites without compromising a thorough examination for any given gRNA.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Antigens, CD34 , CRISPR-Associated Protein 9/genetics , Gene Editing/methods , Hematopoietic Stem Cells/metabolism , RNA, Guide, CRISPR-Cas Systems
3.
iScience ; 25(6): 104374, 2022 Jun 17.
Article in English | MEDLINE | ID: mdl-35633935

ABSTRACT

Background: A point mutation in sickle cell disease (SCD) alters one amino acid in the ß-globin subunit of hemoglobin, with resultant anemia and multiorgan damage that typically shortens lifespan by decades. Because SCD is caused by a single mutation, and hematopoietic stem cells (HSCs) can be harvested, manipulated, and returned to an individual, it is an attractive target for gene correction. Results: An optimized Cas9 ribonucleoprotein (RNP) with an ssDNA oligonucleotide donor together generated correction of at least one ß-globin allele in more than 30% of long-term engrafting human HSCs. After adopting a high-fidelity Cas9 variant, efficient correction with minimal off-target events also was observed. In vivo erythroid differentiation markedly enriches for corrected ß-globin alleles, indicating that erythroblasts carrying one or more corrected alleles have a survival advantage. Significance: These findings indicate that the sickle mutation can be corrected in autologous HSCs with an optimized protocol suitable for clinical translation.

4.
Sci Rep ; 11(1): 19482, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34593942

ABSTRACT

CRISPR-Cas proteins are RNA-guided nucleases used to introduce double-stranded breaks (DSBs) at targeted genomic loci. DSBs are repaired by endogenous cellular pathways such as non-homologous end joining (NHEJ) and homology-directed repair (HDR). Providing an exogenous DNA template during repair allows for the intentional, precise incorporation of a desired mutation via the HDR pathway. However, rates of repair by HDR are often slow compared to the more rapid but less accurate NHEJ-mediated repair. Here, we describe comprehensive design considerations and optimized methods for highly efficient HDR using single-stranded oligodeoxynucleotide (ssODN) donor templates for several CRISPR-Cas systems including S.p. Cas9, S.p. Cas9 D10A nickase, and A.s. Cas12a delivered as ribonucleoprotein (RNP) complexes. Features relating to guide RNA selection, donor strand preference, and incorporation of blocking mutations in the donor template to prevent re-cleavage were investigated and were implemented in a novel online tool for HDR donor template design. These findings allow for high frequencies of precise repair utilizing HDR in multiple mammalian cell lines. Tool availability: https://www.idtdna.com/HDR.


Subject(s)
Bacterial Proteins/metabolism , CRISPR-Associated Protein 9/metabolism , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems , Endodeoxyribonucleases/metabolism , Gene Editing , Recombinational DNA Repair , Cell Line , Humans , Mutation , RNA, Guide, Kinetoplastida/genetics
5.
Nat Commun ; 12(1): 3042, 2021 05 24.
Article in English | MEDLINE | ID: mdl-34031394

ABSTRACT

Controlling off-target editing activity is one of the central challenges in making CRISPR technology accurate and applicable in medical practice. Current algorithms for analyzing off-target activity do not provide statistical quantification, are not sufficiently sensitive in separating signal from noise in experiments with low editing rates, and do not address the detection of translocations. Here we present CRISPECTOR, a software tool that supports the detection and quantification of on- and off-target genome-editing activity from NGS data using paired treatment/control CRISPR experiments. In particular, CRISPECTOR facilitates the statistical analysis of NGS data from multiplex-PCR comparative experiments to detect and quantify adverse translocation events. We validate the observed results and show independent evidence of the occurrence of translocations in human cell lines, after genome editing. Our methodology is based on a statistical model comparison approach leading to better false-negative rates in sites with weak yet significant off-target activity.


Subject(s)
CRISPR-Cas Systems , Computational Biology/methods , Gene Editing/methods , Algorithms , DNA-Binding Proteins/genetics , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Nuclear Proteins/genetics , Software , Transcription Factors/genetics
6.
Mol Ther Methods Clin Dev ; 21: 478-491, 2021 Jun 11.
Article in English | MEDLINE | ID: mdl-33981780

ABSTRACT

CRISPR systems enable targeted genome editing in a wide variety of organisms by introducing single- or double-strand DNA breaks, which are repaired using endogenous molecular pathways. Characterization of on- and off-target editing events from CRISPR proteins can be evaluated using targeted genome resequencing. We characterized DNA repair fingerprints that result from non-homologous end joining (NHEJ) after double-stranded breaks (DSBs) were introduced by Cas9 or Cas12a for >500 paired treatment/control experiments. We found that building biological understanding of the repair into a novel analysis tool (CRISPAltRations) improved the quality of the results. We validated our software using simulated, targeted amplicon sequencing data (11 guide RNAs [gRNAs] and 603 on- and off-target locations) and demonstrated that CRISPAltRations outperforms other publicly available software tools in accurately annotating CRISPR-associated indels and homology-directed repair (HDR) events. We enable non-bioinformaticians to use CRISPAltRations by developing a web-accessible, cloud-hosted deployment, which allows rapid batch processing of samples in a graphical user interface (GUI) and complies with HIPAA security standards. By ensuring that our software is thoroughly tested, version controlled, and supported with a user interface (UI), we enable resequencing analysis of CRISPR genome editing experiments to researchers no matter their skill in bioinformatics.

7.
Nat Med ; 27(4): 677-687, 2021 04.
Article in English | MEDLINE | ID: mdl-33737751

ABSTRACT

ß-Thalassemia pathology is due not only to loss of ß-globin (HBB), but also to erythrotoxic accumulation and aggregation of the ß-globin-binding partner, α-globin (HBA1/2). Here we describe a Cas9/AAV6-mediated genome editing strategy that can replace the entire HBA1 gene with a full-length HBB transgene in ß-thalassemia-derived hematopoietic stem and progenitor cells (HSPCs), which is sufficient to normalize ß-globin:α-globin messenger RNA and protein ratios and restore functional adult hemoglobin tetramers in patient-derived red blood cells. Edited HSPCs were capable of long-term and bilineage hematopoietic reconstitution in mice, establishing proof of concept for replacement of HBA1 with HBB as a novel therapeutic strategy for curing ß-thalassemia.


Subject(s)
Genetic Therapy , Hematopoietic Stem Cells/metabolism , Hemoglobins/metabolism , alpha-Globins/genetics , beta-Globins/genetics , beta-Thalassemia/genetics , beta-Thalassemia/therapy , Anemia, Sickle Cell/pathology , Animals , Antigens, CD34/metabolism , Dependovirus/genetics , Erythrocytes/metabolism , Gene Editing , Genes, Reporter , Genetic Loci , Hematopoietic Stem Cell Transplantation , Humans , Mice , Promoter Regions, Genetic/genetics , RNA, Guide, Kinetoplastida/genetics
8.
Blood ; 137(5): 624-636, 2021 02 04.
Article in English | MEDLINE | ID: mdl-32902645

ABSTRACT

Immune checkpoint therapy has resulted in remarkable improvements in the outcome for certain cancers. To broaden the clinical impact of checkpoint targeting, we devised a strategy that couples targeting of the cytokine-inducible Src homology 2-containing (CIS) protein, a key negative regulator of interleukin 15 (IL-15) signaling, with fourth-generation "armored" chimeric antigen receptor (CAR) engineering of cord blood-derived natural killer (NK) cells. This combined strategy boosted NK cell effector function through enhancing the Akt/mTORC1 axis and c-MYC signaling, resulting in increased aerobic glycolysis. When tested in a lymphoma mouse model, this combined approach improved NK cell antitumor activity more than either alteration alone, eradicating lymphoma xenografts without signs of any measurable toxicity. We conclude that targeting a cytokine checkpoint further enhances the antitumor activity of IL-15-secreting armored CAR-NK cells by promoting their metabolic fitness and antitumor activity. This combined approach represents a promising milestone in the development of the next generation of NK cells for cancer immunotherapy.


Subject(s)
Fetal Blood/cytology , Immunotherapy, Adoptive , Interleukin-15/genetics , Killer Cells, Natural/drug effects , Neoplasm Proteins/antagonists & inhibitors , Suppressor of Cytokine Signaling Proteins/antagonists & inhibitors , Aerobiosis , Animals , Antigens, CD19/immunology , Burkitt Lymphoma/pathology , Burkitt Lymphoma/therapy , CRISPR-Cas Systems , Cell Line, Tumor , Gene Knockout Techniques , Glycolysis , Humans , Immune Checkpoint Inhibitors/pharmacology , Interleukin-15/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/transplantation , Mechanistic Target of Rapamycin Complex 1/physiology , Mice , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Proto-Oncogene Proteins c-akt/physiology , Receptors, Chimeric Antigen , Signal Transduction/physiology , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/physiology , Xenograft Model Antitumor Assays
9.
Blood Adv ; 4(14): 3357-3367, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32717029

ABSTRACT

Virus-specific T cells have proven highly effective for the treatment of severe and drug-refractory infections after hematopoietic stem cell transplant (HSCT). However, the efficacy of these cells is hindered by the use of glucocorticoids, often given to patients for the management of complications such as graft-versus-host disease. To address this limitation, we have developed a novel strategy for the rapid generation of good manufacturing practice (GMP)-grade glucocorticoid-resistant multivirus-specific T cells (VSTs) using clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9) gene-editing technology. We have shown that deleting the nuclear receptor subfamily 3 group C member 1 (NR3C1; the gene encoding for the glucocorticoid receptor) renders VSTs resistant to the lymphocytotoxic effect of glucocorticoids. NR3C1-knockout (KO) VSTs kill their targets and proliferate successfully in the presence of high doses of dexamethasone both in vitro and in vivo. Moreover, we developed a protocol for the rapid generation of GMP-grade NR3C1 KO VSTs with high on-target activity and minimal off-target editing. These genetically engineered VSTs promise to be a novel approach for the treatment of patients with life-threatening viral infections post-HSCT on glucocorticoid therapy.


Subject(s)
CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing , Humans , Receptors, Glucocorticoid/genetics , T-Lymphocytes
10.
Mol Ther Methods Clin Dev ; 17: 1097-1107, 2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32478125

ABSTRACT

Genome editing of human cluster of differentiation 34+ (CD34+) hematopoietic stem and progenitor cells (HSPCs) holds great therapeutic potential. This study aimed to optimize on-target, ex vivo genome editing using the CRISPR-Cas9 system in CD34+ HSPCs and to create a clear workflow for precise identification of off-target effects. Modified synthetic guide RNAs (gRNAs), either 2-part gRNA or single-guide RNA (sgRNA), were delivered to CD34+ HSPCs as part of ribonucleoprotein (RNP) complexes, targeting therapeutically relevant genes. The addition of an Alt-R electroporation enhancer (EE), a short, single-stranded oligodeoxynucleotide (ssODN), significantly increased editing efficiency in CD34+ HSPCs. Notably, similar editing improvement was observed when excess gRNA over Cas9 protein was used, providing a DNA-free alternative suitable for therapeutic applications. Furthermore, we demonstrated that sgRNA may be preferable over 2-part gRNA in a locus-specific manner. Finally, we present a clear experimental framework suitable for the unbiased identification of bona fide off-target sites by Genome-Wide, Unbiased Identification of Double-Strand Breaks (DSBs) Enabled by Sequencing (GUIDE-seq), as well as subsequent editing quantification in CD34+ HSPCs using rhAmpSeq. These findings may facilitate the implementation of genome editing in CD34+ HSPCs for research and therapy and can be adapted for other hematopoietic cells.

11.
Nat Med ; 24(8): 1216-1224, 2018 08.
Article in English | MEDLINE | ID: mdl-30082871

ABSTRACT

Translation of the CRISPR-Cas9 system to human therapeutics holds high promise. However, specificity remains a concern especially when modifying stem cell populations. We show that existing rationally engineered Cas9 high-fidelity variants have reduced on-target activity when using the therapeutically relevant ribonucleoprotein (RNP) delivery method. Therefore, we devised an unbiased bacterial screen to isolate variants that retain activity in the RNP format. Introduction of a single point mutation, p.R691A, in Cas9 (high-fidelity (HiFi) Cas9) retained the high on-target activity of Cas9 while reducing off-target editing. HiFi Cas9 induces robust AAV6-mediated gene targeting at five therapeutically relevant loci (HBB, IL2RG, CCR5, HEXB, and TRAC) in human CD34+ hematopoietic stem and progenitor cells (HSPCs) as well as primary T cells. We also show that HiFi Cas9 mediates high-level correction of the sickle cell disease (SCD)-causing p.E6V mutation in HSPCs derived from patients with SCD. We anticipate that HiFi Cas9 will have wide utility for both basic science and therapeutic genome-editing applications.


Subject(s)
CRISPR-Associated Protein 9/genetics , Gene Editing , Hematopoietic Stem Cells/metabolism , Mutation/genetics , Ribonucleoproteins/metabolism , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Antigens, CD34/metabolism , Base Sequence , Escherichia coli , HEK293 Cells , Humans
12.
Methods ; 121-122: 16-28, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28351759

ABSTRACT

Genome editing using the CRISPR/Cas9 system requires the presence of guide RNAs bound to the Cas9 endonuclease as a ribonucleoprotein (RNP) complex in cells, which cleaves the host cell genome at sites specified by the guide RNAs. New genetic material may be introduced during repair of the double-stranded break via homology dependent repair (HDR) if suitable DNA templates are delivered with the CRISPR components. Early methods used plasmid or viral vectors to make these components in the host cell, however newer approaches using recombinant Cas9 protein with synthetic guide RNAs introduced directly as an RNP complex into cells shows faster onset of action with fewer off-target effects. This approach also enables use of chemically modified synthetic guide RNAs that have improved nuclease stability and reduces the risk of triggering an innate immune response in the host cell. This article provides detailed methods for genome editing using the RNP approach with synthetic guide RNAs using lipofection or electroporation in mammalian cells or using microinjection in murine zygotes, with or without addition of a single-stranded HDR template DNA.


Subject(s)
Bacterial Proteins/genetics , CRISPR-Cas Systems , Endonucleases/genetics , Gene Editing/methods , Gene Transfer Techniques , RNA, Guide, Kinetoplastida/genetics , Ribonucleoproteins/genetics , Animals , Bacterial Proteins/metabolism , Base Sequence , CRISPR-Associated Protein 9 , Clustered Regularly Interspaced Short Palindromic Repeats , DNA/genetics , DNA/metabolism , Electroporation , Endonucleases/metabolism , Gene Targeting/methods , Genome , HEK293 Cells , Humans , Jurkat Cells , Lipids/chemistry , Mice , Microinjections , RNA, Guide, Kinetoplastida/chemical synthesis , RNA, Guide, Kinetoplastida/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinational DNA Repair , Ribonucleoproteins/metabolism , Zygote/cytology , Zygote/metabolism
13.
Mol Ther ; 22(1): 92-101, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24089139

ABSTRACT

Despite progress in identifying molecular drivers of cancer, it has been difficult to translate this knowledge into new therapies, because many of the causal proteins cannot be inhibited by conventional small molecule therapeutics. RNA interference (RNAi), which uses small RNAs to inhibit gene expression, provides a promising alternative to reach traditionally undruggable protein targets by shutting off their expression at the messenger RNA (mRNA) level. Challenges for realizing the potential of RNAi have included identifying the appropriate genes to target and achieving sufficient knockdown in tumors. We have developed high-potency Dicer-substrate short-interfering RNAs (DsiRNAs) targeting ß-catenin and delivered these in vivo using lipid nanoparticles, resulting in significant reduction of ß-catenin expression in liver cancer models. Reduction of ß-catenin strongly reduced tumor burden, alone or in combination with sorafenib and as effectively as DsiRNAs that target mitotic genes such as PLK1 and KIF11. ß-catenin knockdown also strongly reduced the expression of ß-catenin-regulated genes, including MYC, providing a potential mechanism for tumor inhibition. These results validate ß-catenin as a target for liver cancer therapy and demonstrate the promise of RNAi in general and DsiRNAs in particular for reaching traditionally undruggable cancer targets.


Subject(s)
Liver Neoplasms/genetics , Liver Neoplasms/pathology , RNA, Small Interfering/genetics , beta Catenin/genetics , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Expression , Gene Knockdown Techniques , Humans , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Male , Mice , Nanoparticles/administration & dosage , Nanoparticles/chemistry , RNA Interference , RNA, Small Interfering/chemistry , RNA, Small Interfering/metabolism , Ribonuclease III/metabolism , Tumor Burden/genetics , Xenograft Model Antitumor Assays , beta Catenin/metabolism
14.
Mol Ther ; 20(3): 483-512, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22186795

ABSTRACT

RNA interference (RNAi) has been extensively employed for in vivo research since its use was first demonstrated in mammalian cells 10 years ago. Design rules have improved, and it is now routinely possible to obtain reagents that suppress expression of any gene desired. At the same time, increased understanding of the molecular basis of unwanted side effects has led to the development of chemical modification strategies that mitigate these concerns. Delivery remains the single greatest hurdle to widespread adoption of in vivo RNAi methods. However, exciting advances have been made and new delivery systems under development may help to overcome these barriers. This review discusses advances in RNAi biochemistry and biology that impact in vivo use and provides an overview of select publications that demonstrate interesting applications of these principles. Emphasis is placed on work with synthetic, small interfering RNAs (siRNAs) published since the first installment of this review which appeared in 2006.


Subject(s)
RNA Interference , RNA, Small Interfering/therapeutic use , Animals , Drug Delivery Systems/trends , Humans , RNA, Small Interfering/chemistry , RNA, Small Interfering/pharmacology
15.
Methods Mol Biol ; 574: 155-71, 2009.
Article in English | MEDLINE | ID: mdl-19685307

ABSTRACT

Bioluminescent imaging (BLI) coupled with hydrodynamic (HD) dosing of luciferase-expressing plasmid DNA (pDNA) has proven to be a powerful method for quantitatively benchmarking non-viral gene expression in the liver. The expression of luciferase or knockdown of luciferase by RNA interference (RNAi) in the liver is quantifiable over five-orders of magnitude in living mice. The photon emission data derived from BLI can be converted to the absolute amount of luciferase expression by comparison with a standard curve developed using luciferase as a primary standard. Quantitative BLI is also applicable to luciferase expression in other tissues, such as skeletal muscle, following intramuscular (IM) dosing and electroporation (EP) of pDNA. The primary advantages of using quantitative BLI in mouse liver and muscle are the sensitivity of the assay, the speed and ease of making measurements, the precision and linearity of the dose-response curves, and the ability to conduct serial sampling of gene expression over many days or months while eliminating the need to euthanize animals.


Subject(s)
Gene Expression , Gene Knockdown Techniques , RNA Interference , Animals , Electroporation , Luciferases/genetics , Mice , RNA, Small Interfering
16.
Expert Opin Biol Ther ; 7(6): 799-808, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17555366

ABSTRACT

Despite nearly two decades of research, the successful application of systemically delivered non-viral gene therapies to treat human disease is still limited by poor transfection efficiency. The major barriers in the circulation and in the cell that limit transfection efficiency have been identified and the field has entered a phase of design and testing of more sophisticated carrier systems that attempt to circumvent these barriers. These studies are increasingly conducted in vivo using rapid quantitative measures of gene transfer efficiency as a guide. Although there has been steady progress in developing DNA nanoparticles that navigate the circulation, enter the target cell and escape lysosomal targeting, the final goal of efficiently traversing the nuclear membrane remains the most significant challenge. The ultimate goal is to develop elegant delivery systems that work in concert to deliver DNA from the needle to the nucleus.


Subject(s)
Cell Nucleus/metabolism , DNA/metabolism , Nanoparticles , Transfection , Active Transport, Cell Nucleus , Animals , Cell Line , DNA/chemistry , Dendrimers/chemistry , Endocytosis , Endosomes/metabolism , Genes, Reporter , Humans , Lipids/chemistry , Luciferases/genetics , Luciferases/metabolism , Proteins/chemistry , Reproducibility of Results , Transfection/methods
17.
J Pharm Sci ; 96(11): 2922-30, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17518360

ABSTRACT

The intracellular delivery of small interfering RNA (siRNA) is a therapeutic strategy to transiently block gene expression. Two silencing RNA strategies utilize either synthetic double stranded RNA or plasmid DNA encoding a short hairpin RNA (shRNA). In the present study, we have quantitatively compared the potency of siRNA (siLuc1) and shRNA (pShagLuc) mediated knockdown of luciferase expression in vivo using hydrodynamic dosing and bioluminescence imaging (BLI). Following hydrodynamic coadministration of siLuc1 or pShagLuc with a plasmid encoding luciferase (pGL3), mice were analyzed for transgene expression by BLI. The knockdown of luciferase expression by siLuc1 or pShagLuc was observed at 3 h and persisted for 3 days. The potency of siLuc1 and pShagLuc was equivalent with maximal effect at 10 microg coadministered with 1 microg of pGL3 resulting in >80% knockdown. Combined dosing of siLuc1 and pShagluc (5 microg each) with 1 microg of pGL3 resulted in >99% knockdown. Analysis of the data established that shRNA was significantly more potent than siRNA at mediating knockdown when compared on a mole basis. The combination of hydrodynamic dosing and BLI to measure siRNA or shRNA mediated knockdown of luciferase provide an attractive in vivo quantitative method to test formulations that target the liver.


Subject(s)
Luciferases, Firefly/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA/administration & dosage , RNA/genetics , Transfection/methods , 3T3 Cells , Animals , CHO Cells , Cricetinae , Cricetulus , Gene Silencing , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Liver/enzymology , Liver/physiology , Luciferases, Firefly/biosynthesis , Luciferases, Firefly/deficiency , Luminescent Measurements , Mice , Nucleic Acid Conformation , RNA Interference
18.
Bioconjug Chem ; 18(2): 371-8, 2007.
Article in English | MEDLINE | ID: mdl-17373767

ABSTRACT

PEGylated glycoproteins (PGPs) were synthesized by copolymerizing a Cys-terminated PEG-peptide, glycopeptide, and melittin peptide. Compositionally unique PGPs were prepared by varying the ratio of PEG-peptide (20-90%) and melittin (0-70%) with a constant amount of glycopeptide (10%). The PGPs were purified by RP-HPLC, and characterized for molecular weight and polydispersity by GPC-HPLC and SDS-PAGE and for composition by RP-HPLC following reduction to form monomeric peptides. PGPs formed DNA condensates of 200-300 nm in diameter that were administered to mice via the tail vein. Biodistribution studies confirmed their primary targeting to liver hepatocytes with a DNA metabolic half-life of 1 h. Upon stimulation by hydrodynamic dosing with saline, PGP DNA (5 microg) mediated luciferase expression in the liver detected by bioluminescence imaging (BLI) after 24 h. The level of gene expression mediated by PGP DNA was 5000-fold less than direct hydrodynamic dosing of an equivalent amount of DNA and was independent of the mol percent of melittin incorporated into the polymer, but dependent on the presence of galactose on PGP. The results establish the ability to prepare three-component gene delivery polymers that function in vivo. Further design improvements in fusogenic peptides for gene delivery and for the simultaneous use of a nuclear targeting strategy will be necessary to approach levels of expression mediated by the direct hydrodynamic dosing of DNA.


Subject(s)
DNA/administration & dosage , Drug Delivery Systems , Gene Targeting , Glycopeptides/chemistry , Glycoproteins/pharmacology , Peptide Fragments/chemistry , Polyethylene Glycols/chemistry , Animals , Chromatography, High Pressure Liquid , DNA/chemistry , Drug Carriers , Gene Expression , Gene Transfer Techniques , Glycoproteins/chemistry , Glycoproteins/genetics , Liver/cytology , Liver/metabolism , Luciferases/genetics , Luciferases/metabolism , Melitten , Mice , Mice, Inbred ICR , Molecular Weight , Plasmids , Tissue Distribution , Transfection
19.
Anal Biochem ; 355(1): 90-4, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16737677

ABSTRACT

Bioluminescent imaging (BLI) is a widely used in vivo method to determine the location and relative intensity of luciferase expression in mice. Luciferase expression is observed following an i.p. dose of d-luciferin, resulting in bioluminescence that is detected in anesthetized mice by a charge-coupled device camera. To establish whether BLI could be used as a quantitative measurement of non-viral-mediated luciferase expression, precise quantities of plasmid DNA encoding the luciferase gene were hydrodynamically dosed in mice. The results established a linear correlation between the DNA dose and the BLI response measured in liver which spanned five orders of magnitude. The level of luciferase expression was found to be a direct function of d-luciferin dose. The time course of luciferase expression and the influence of multidosing of substrate were measured by BLI. The recovery of luciferase from the liver of hydrodynamically dosed mice allowed calibration of the BLI measurements. The results establish BLI's limit-of-detection at 20 pg of luciferase per liver following a hydrodynamic dose of 100 pg of plasmid DNA. These results demonstrate that BLI is both sensitive and linear and should allow for the direct comparison of the efficiency of gene transfer vectors that target the liver.


Subject(s)
Gene Expression/genetics , Luminescent Measurements/methods , Transgenes/genetics , Animals , Liver/metabolism , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Transgenic , Plasmids/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...