Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Biotechnol ; 40(8): 1285-1294, 2022 08.
Article in English | MEDLINE | ID: mdl-35393561

ABSTRACT

Homologous recombination (HR)-based gene therapy using adeno-associated viruses (AAV-HR) without nucleases has several advantages over classic gene therapy, especially the potential for permanent transgene expression. However, the low efficiency of AAV-HR remains a major limitation. Here, we tested a series of small-molecule compounds and found that ribonucleotide reductase (RNR) inhibitors substantially enhance AAV-HR efficiency in mouse and human liver cell lines approximately threefold. Short-term administration of the RNR inhibitor fludarabine increased the in vivo efficiency of both non-nuclease- and CRISPR/Cas9-mediated AAV-HR two- to sevenfold in the murine liver, without causing overt toxicity. Fludarabine administration induced transient DNA damage signaling in both proliferating and quiescent hepatocytes. Notably, the majority of AAV-HR events occurred in non-proliferating hepatocytes in both fludarabine-treated and control mice, suggesting that the induction of transient DNA repair signaling in non-dividing hepatocytes was responsible for enhancing AAV-HR efficiency in mice. These results suggest that use of a clinically approved RNR inhibitor can potentiate AAV-HR-based genome-editing therapeutics.


Subject(s)
CRISPR-Cas Systems , Genetic Vectors , Animals , CRISPR-Cas Systems/genetics , Dependovirus/genetics , Endonucleases/genetics , Gene Editing/methods , Homologous Recombination , Humans , Mice , Vidarabine/analogs & derivatives
2.
Mol Ther ; 29(3): 1016-1027, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33678249

ABSTRACT

Recombinant adeno-associated virus (rAAV) vectors have the unique property of being able to perform genomic targeted integration (TI) without inducing a double-strand break (DSB). In order to improve our understanding of the mechanism behind TI mediated by AAV and improve its efficiency, we performed an unbiased genetic screen in human cells using a promoterless AAV-homologous recombination (AAV-HR) vector system. We identified that the inhibition of the Fanconi anemia complementation group M (FANCM) protein enhanced AAV-HR-mediated TI efficiencies in different cultured human cells by ∼6- to 9-fold. The combined knockdown of the FANCM and two proteins also associated with the FANCM complex, RecQ-mediated genome instability 1 (RMI1) and Bloom DNA helicase (BLM) from the BLM-topoisomerase IIIα (TOP3A)-RMI (BTR) dissolvase complex (RMI1, having also been identified in our screen), led to the enhancement of AAV-HR-mediated TI up to ∼17 times. AAV-HR-mediated TI in the presence of a nuclease (CRISPR-Cas9) was also increased by ∼1.5- to 2-fold in FANCM and RMI1 knockout cells, respectively. Furthermore, knockdown of FANCM in human CD34+ hematopoietic stem and progenitor cells (HSPCs) increased AAV-HR-mediated TI by ∼3.5-fold. This study expands our knowledge on the mechanisms related to AAV-mediated TI, and it highlights new pathways that might be manipulated for future improvements in AAV-HR-mediated TI.


Subject(s)
CRISPR-Cas Systems , DNA Helicases/antagonists & inhibitors , DNA-Binding Proteins/antagonists & inhibitors , Dependovirus/genetics , Gene Editing , Hematopoietic Stem Cells/metabolism , RecQ Helicases/antagonists & inhibitors , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genetic Vectors , HeLa Cells , Hematopoietic Stem Cells/cytology , Homologous Recombination , Humans , RecQ Helicases/genetics , RecQ Helicases/metabolism
3.
Hum Gene Ther ; 31(9-10): 553-564, 2020 05.
Article in English | MEDLINE | ID: mdl-32024384

ABSTRACT

Despite early successes using recombinant adeno-associated virus (rAAV) vectors in clinical gene therapy trials, limitations remain making additional advancements a necessity. Some of the challenges include variable levels of pre-existing neutralizing antibodies and poor transduction in specific target tissues and/or diseases. In addition, readministration of an rAAV vector is in general not possible due to the immune response against the capsid. Recombinant adeno-associated virus (AAV) vectors with novel capsids can be isolated in nature or developed through different directed evolution strategies. However, in most cases, the process of AAV selection is not well understood and new strategies are required to define the best parameters to develop more efficient and functional rAAV capsids. Therefore, the use of barcoding for AAV capsid libraries, which can be screened by high-throughput sequencing, provides a powerful tool to track AAV capsid evolution and potentially improve AAV capsid library screens. In this study, we examined how different parameters affect the screen of two different AAV libraries in two human cell types. We uncovered new and unexpected insights in how to maximize the likelihood of obtaining AAV variants with the desired properties. The major findings of the study are the following. (1) Inclusion of helper-virus for AAV replication can selectively propagate variants that can replicate to higher titers, but are not necessarily better at transduction. (2) Competition between AAVs with specific capsids can take place in cells that have been infected with different AAVs. (3) The use of low multiplicity of infections for infection results in more variation between screens and is not optimal at selecting the most desired capsids. (4) Using multiple rounds of selection can be counterproductive. We conclude that each of these parameters should be taken into consideration when screening AAV libraries for enhanced properties of interest.


Subject(s)
Capsid/metabolism , Dependovirus/genetics , Genetic Vectors/genetics , Biodiversity , Cell Line , Directed Molecular Evolution , Gene Library , Gene Transfer Techniques , Genome, Viral , HaCaT Cells , Helper Viruses , High-Throughput Nucleotide Sequencing , Humans , Reproducibility of Results , Transduction, Genetic , Virus Replication
4.
JCI Insight ; 4(22)2019 11 14.
Article in English | MEDLINE | ID: mdl-31723052

ABSTRACT

While gene transfer using recombinant adeno-associated viral (rAAV) vectors has shown success in some clinical trials, there remain many tissues that are not well transduced. Because of the recent success in reprogramming islet-derived cells into functional ß cells in animal models, we constructed 2 highly complex barcoded replication competent capsid shuffled libraries and selected for high-transducing variants on primary human islets. We describe the generation of a chimeric AAV capsid (AAV-KP1) that facilitates transduction of primary human islet cells and human embryonic stem cell-derived ß cells with up to 10-fold higher efficiency compared with previously studied best-in-class AAV vectors. Remarkably, this chimeric capsid also enabled transduction of both mouse and human hepatocytes at very high levels in a humanized chimeric mouse model, thus providing a versatile vector that has the potential to be used in both preclinical testing and human clinical trials for liver-based diseases and diabetes.


Subject(s)
Capsid/chemistry , Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Animals , Cells, Cultured , Diabetes Mellitus , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Library , Gene Transfer Techniques , HEK293 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Mice
5.
Hepatology ; 62(2): 417-28, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26011400

ABSTRACT

UNLABELLED: Liver-targeted gene therapy based on recombinant adeno-associated viral vectors (rAAV) shows promising therapeutic efficacy in animal models and adult-focused clinical trials. This promise, however, is not directly translatable to the growing liver, where high rates of hepatocellular proliferation are accompanied by loss of episomal rAAV genomes and subsequently a loss in therapeutic efficacy. We have developed a hybrid rAAV/piggyBac transposon vector system combining the highly efficient liver-targeting properties of rAAV with stable piggyBac-mediated transposition of the transgene into the hepatocyte genome. Transposition efficiency was first tested using an enhanced green fluorescent protein expression cassette following delivery to newborn wild-type mice, with a 20-fold increase in stably gene-modified hepatocytes observed 4 weeks posttreatment compared to traditional rAAV gene delivery. We next modeled the therapeutic potential of the system in the context of severe urea cycle defects. A single treatment in the perinatal period was sufficient to confer robust and stable phenotype correction in the ornithine transcarbamylase-deficient Spf(ash) mouse and the neonatal lethal argininosuccinate synthetase knockout mouse. Finally, transposon integration patterns were analyzed, revealing 127,386 unique integration sites which conformed to previously published piggyBac data. CONCLUSION: Using a hybrid rAAV/piggyBac transposon vector system, we achieved stable therapeutic protection in two urea cycle defect mouse models; a clinically conceivable early application of this technology in the management of severe urea cycle defects could be as a bridging therapy while awaiting liver transplantation; further improvement of the system will result from the development of highly human liver-tropic capsids, the use of alternative strategies to achieve transient transposase expression, and engineered refinements in the safety profile of piggyBac transposase-mediated integration.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Genetic Vectors/pharmacology , Hyperammonemia/therapy , Urea/metabolism , Animals , Animals, Newborn , Disease Models, Animal , Gene Transfer Techniques , Humans , Hyperammonemia/diagnosis , Liver Diseases/therapy , Mice , Mice, Transgenic , Severity of Illness Index , Statistics, Nonparametric
6.
Int J Biochem Cell Biol ; 56: 141-52, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25449261

ABSTRACT

The number of genetic or acquired diseases of the liver treatable by organ transplantation is ever-increasing as transplantation techniques improve placing additional demands on an already limited organ supply. While cell and gene therapies are distinctly different modalities, they offer a synergistic alternative to organ transplant due to distinct architectural and physiological properties of the liver. The hepatic blood supply and fenestrated endothelial system affords relatively facile accessibility for cell and/or gene delivery. More importantly, however, the remarkable capacity of hepatocytes to proliferate and repopulate the liver creates opportunities for new treatments based on emerging technologies. This review will summarise current understanding of liver regeneration, describe clinical and experimental cell and gene therapeutic modalities and discuss critical challenges to translate these new technologies to wider clinical utility. This article is part of a Directed Issue entitled: "Regenerative Medicine: the challenge of translation".


Subject(s)
Hepatocytes/cytology , Liver Regeneration/physiology , Liver/cytology , Liver/physiology , Animals , Cell Proliferation , Genetic Therapy/methods , Humans , Liver Diseases/genetics , Liver Diseases/surgery , Liver Diseases/therapy , Liver Transplantation/methods , Regenerative Medicine/methods , Regenerative Medicine/trends
7.
Cell Stem Cell ; 14(1): 107-20, 2014 Jan 02.
Article in English | MEDLINE | ID: mdl-24139757

ABSTRACT

Mouse epiblast stem cells (EpiSCs) can be derived from a wide range of developmental stages. To characterize and compare EpiSCs with different origins, we derived a series of EpiSC lines from pregastrula stage to late-bud-stage mouse embryos. We found that the transcriptomes of these cells are hierarchically distinct from those of the embryonic stem cells, induced pluripotent stem cells (iPSCs), and epiblast/ectoderm. The EpiSCs display globally similar gene expression profiles irrespective of the original developmental stage of the source tissue. They are developmentally similar to the ectoderm of the late-gastrula-stage embryo and behave like anterior primitive streak cells when differentiated in vitro and in vivo. The EpiSC lines that we derived can also be categorized based on a correlation between gene expression signature and predisposition to differentiate into particular germ-layer derivatives. Our findings therefore highlight distinct identifying characteristics of EpiSCs and provide a foundation for further examination of EpiSC properties and potential.


Subject(s)
Cell Differentiation , Cell Lineage , Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Germ Layers/cytology , Pluripotent Stem Cells/cytology , Primitive Streak/cytology , Animals , Biomarkers/metabolism , Blotting, Western , Cell Proliferation , Cells, Cultured , Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gastrulation , Gene Expression Profiling , Germ Layers/metabolism , Immunoenzyme Techniques , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Pluripotent Stem Cells/metabolism , Primitive Streak/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
8.
BMC Res Notes ; 3: 242, 2010 Sep 21.
Article in English | MEDLINE | ID: mdl-20858277

ABSTRACT

BACKGROUND: Collybistin (CB), a neuron-specific guanine nucleotide exchange factor, has been implicated in targeting gephyrin-GABAA receptors clusters to inhibitory postsynaptic sites. However, little is known about additional CB partners and functions. FINDINGS: Here, we identified the p40 subunit of the eukaryotic translation initiation factor 3 (eIF3H) as a novel binding partner of CB, documenting the interaction in yeast, non-neuronal cell lines, and the brain. In addition, we demonstrated that gephyrin also interacts with eIF3H in non-neuronal cells and forms a complex with eIF3 in the brain. CONCLUSIONS: Together, our results suggest, for the first time, that CB and gephyrin associate with the translation initiation machinery, and lend further support to the previous evidence that gephyrin may act as a regulator of synaptic protein synthesis.

SELECTION OF CITATIONS
SEARCH DETAIL
...