Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Cell ; 184(22): 5653-5669.e25, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34672952

ABSTRACT

Cells repair DNA double-strand breaks (DSBs) through a complex set of pathways critical for maintaining genomic integrity. To systematically map these pathways, we developed a high-throughput screening approach called Repair-seq that measures the effects of thousands of genetic perturbations on mutations introduced at targeted DNA lesions. Using Repair-seq, we profiled DSB repair products induced by two programmable nucleases (Cas9 and Cas12a) in the presence or absence of oligonucleotides for homology-directed repair (HDR) after knockdown of 476 genes involved in DSB repair or associated processes. The resulting data enabled principled, data-driven inference of DSB end joining and HDR pathways. Systematic interrogation of this data uncovered unexpected relationships among DSB repair genes and demonstrated that repair outcomes with superficially similar sequence architectures can have markedly different genetic dependencies. This work provides a foundation for mapping DNA repair pathways and for optimizing genome editing across diverse modalities.


Subject(s)
DNA Breaks, Double-Stranded , Genomics , CRISPR-Associated Protein 9/metabolism , Cell Line , Cluster Analysis , DNA Repair/genetics , Gene Editing , Gene Expression Regulation , Genome, Human , Humans , Phenotype , RNA, Guide, Kinetoplastida/metabolism , Reproducibility of Results
2.
Mol Ther ; 32(10): 3356-3371, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-38981468

ABSTRACT

Recombinant adeno-associated virus (rAAV) vector gene delivery systems have demonstrated great promise in clinical trials but continue to face durability and dose-related challenges. Unlike rAAV gene therapy, integrating gene addition approaches can provide curative expression in mitotically active cells and pediatric populations. We explored a novel in vivo delivery approach based on an engineered transposase, Sleeping Beauty (SB100X), delivered as an mRNA within a lipid nanoparticle (LNP), in combination with an rAAV-delivered transposable transgene. This combinatorial approach achieved correction of ornithine transcarbamylase deficiency in the neonatal Spfash mouse model following a single delivery to dividing hepatocytes in the newborn liver. Correction remained stable into adulthood, while a conventional rAAV approach resulted in a return to the disease state. In non-human primates, integration by transposition, mediated by this technology, improved gene expression 10-fold over conventional rAAV-mediated gene transfer while requiring 5-fold less vector. Additionally, integration site analysis confirmed a random profile while specifically targeting TA dinucleotides across the genome. Together, these findings demonstrate that transposable elements can improve rAAV-delivered therapies by lowering the vector dose requirement and associated toxicity while expanding target cell types.


Subject(s)
Dependovirus , Genetic Vectors , Hepatocytes , Nanoparticles , RNA, Messenger , Transgenes , Transposases , Animals , Dependovirus/genetics , Mice , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Hepatocytes/metabolism , Transposases/genetics , Transposases/metabolism , Nanoparticles/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Genetic Therapy/methods , Humans , Gene Expression , Lipids/chemistry , Disease Models, Animal , Gene Transfer Techniques , Ornithine Carbamoyltransferase/genetics , Ornithine Carbamoyltransferase/metabolism , Liposomes
3.
J Gene Med ; 26(8): e3726, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39160647

ABSTRACT

BACKGROUND: Conventional adeno-associated viral (AAV) vectors, while highly effective in quiescent cells such as hepatocytes in the adult liver, confer less durable transgene expression in proliferating cells owing to episome loss. Sustained therapeutic success is therefore less likely in liver disorders requiring early intervention. We have previously developed a hybrid, dual virion approach, recombinant AAV (rAAV)/piggyBac transposon system capable of achieving stable gene transfer in proliferating hepatocytes at levels many fold above conventional AAV vectors. An alternative transposon system, Sleeping Beauty, has been widely used for ex vivo gene delivery; however liver-targeted delivery using a hybrid rAAV/Sleeping Beauty approach remains relatively unexplored. METHODS: We investigated the capacity of a Sleeping Beauty (SB)-based dual rAAV virion approach to achieve stable and efficient gene transfer to the newborn murine liver using transposable therapeutic cassettes encoding coagulation factor IX or ornithine transcarbamylase (OTC). RESULTS: At equivalent doses, rAAV/SB100X transduced hepatocytes with high efficiency, achieving stable expression into adulthood. Compared with conventional AAV, the proportion of hepatocytes transduced, and factor IX and OTC activity levels, were both markedly increased. The proportion of hepatocytes stably transduced increased 4- to 8-fold from <5%, and activity levels increased correspondingly, with markedly increased survival and stable urinary orotate levels in the OTC-deficient Spfash mouse following elimination of residual endogenous murine OTC. CONCLUSIONS: The present study demonstrates the first in vivo utility of a hybrid rAAV/SB100X transposon system to achieve stable long-term therapeutic gene expression following delivery to the highly proliferative newborn mouse liver. These results have relevance to the treatment of genetic metabolic liver diseases with neonatal onset.


Subject(s)
Animals, Newborn , DNA Transposable Elements , Dependovirus , Gene Transfer Techniques , Genetic Vectors , Hepatocytes , Liver , Transduction, Genetic , Animals , Dependovirus/genetics , DNA Transposable Elements/genetics , Liver/metabolism , Mice , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Hepatocytes/metabolism , Factor IX/genetics , Ornithine Carbamoyltransferase/genetics , Ornithine Carbamoyltransferase/metabolism , Transposases/genetics , Transposases/metabolism , Humans , Transgenes , Genetic Therapy/methods , Mice, Inbred C57BL
4.
Genes Dev ; 24(17): 1939-50, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20810650

ABSTRACT

In response to DNA damage, cells activate a complex signal transduction network called the DNA damage response (DDR). To enhance our current understanding of the DDR network, we performed a genome-wide RNAi screen to identify genes required for resistance to ionizing radiation (IR). Along with a number of known DDR genes, we discovered a large set of novel genes whose depletion leads to cellular sensitivity to IR. Here we describe TTI1 (Tel two-interacting protein 1) and TTI2 as highly conserved regulators of the DDR in mammals. TTI1 and TTI2 protect cells from spontaneous DNA damage, and are required for the establishment of the intra-S and G2/M checkpoints. TTI1 and TTI2 exist in multiple complexes, including a 2-MDa complex with TEL2 (telomere maintenance 2), called the Triple T complex, and phosphoinositide-3-kinase-related protein kinases (PIKKs) such as ataxia telangiectasia-mutated (ATM). The components of the TTT complex are mutually dependent on each other, and act as critical regulators of PIKK abundance and checkpoint signaling.


Subject(s)
Carrier Proteins , Cell Cycle Proteins/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-ets , Signal Transduction , Tumor Suppressor Proteins/metabolism , 1-Phosphatidylinositol 4-Kinase/metabolism , Ataxia Telangiectasia Mutated Proteins , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle/genetics , Cell Line , Cell Line, Tumor , DNA Damage/radiation effects , Genes, cdc , Genome-Wide Association Study , Humans , Infrared Rays , Intracellular Signaling Peptides and Proteins , Molecular Chaperones , Protein Stability , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , RNA Interference
7.
EMBO Mol Med ; 13(3): e13545, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33475257

ABSTRACT

Precise correction of the CD40LG gene in T cells and hematopoietic stem/progenitor cells (HSPC) holds promise for treating X-linked hyper-IgM Syndrome (HIGM1), but its actual therapeutic potential remains elusive. Here, we developed a one-size-fits-all editing strategy for effective T-cell correction, selection, and depletion and investigated the therapeutic potential of T-cell and HSPC therapies in the HIGM1 mouse model. Edited patients' derived CD4 T cells restored physiologically regulated CD40L expression and contact-dependent B-cell helper function. Adoptive transfer of wild-type T cells into conditioned HIGM1 mice rescued antigen-specific IgG responses and protected mice from a disease-relevant pathogen. We then obtained ~ 25% CD40LG editing in long-term repopulating human HSPC. Transplanting such proportion of wild-type HSPC in HIGM1 mice rescued immune functions similarly to T-cell therapy. Overall, our findings suggest that autologous edited T cells can provide immediate and substantial benefits to HIGM1 patients and position T-cell ahead of HSPC gene therapy because of easier translation, lower safety concerns and potentially comparable clinical benefits.


Subject(s)
Hyper-IgM Immunodeficiency Syndrome, Type 1 , Hyper-IgM Immunodeficiency Syndrome , Animals , Gene Editing , Hematopoietic Stem Cells , Humans , Mice , T-Lymphocytes
8.
CRISPR J ; 3(3): 177-187, 2020 06.
Article in English | MEDLINE | ID: mdl-32584143

ABSTRACT

Multiplexed genome editing with DNA endonucleases has broad application, including for cellular therapies, but chromosomal translocations, natural byproducts of inducing simultaneous genomic breaks, have not been explored in detail. Here we apply various CRISPR-Cas nucleases to edit the T cell receptor alpha and beta 2 microglobulin genes in human primary T cells and comprehensively evaluate the frequency and stability of the resulting translocations. A thorough translocation frequency analysis using three orthogonal methods (droplet digital PCR, unidirectional sequencing, and metaphase fluorescence in situ hybridization) yielded comparable results and an overall translocation rate of ∼7% between two simultaneous CRISPR-Cas9 induced edits. In addition, we show that chromosomal translocations can be reduced when using different nuclease combinations, or by the presence of a homologous single stranded oligo donor for multiplexed genome editing. Importantly, the two different approaches for translocation reduction are compatible with cell therapy applications.


Subject(s)
CRISPR-Cas Systems , Gene Editing/methods , T-Lymphocytes , Translocation, Genetic , CD4-Positive T-Lymphocytes , CRISPR-Associated Protein 9/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , DNA/genetics , Endonucleases/genetics , Genome, Human , Humans , In Situ Hybridization, Fluorescence , Multifactorial Inheritance , RNA, Guide, Kinetoplastida , Streptococcus pyogenes
9.
Cancer Metab ; 8: 1, 2020.
Article in English | MEDLINE | ID: mdl-31908776

ABSTRACT

BACKGROUND: Metabolic programs in cancer cells are influenced by genotype and the tissue of origin. We have previously shown that central carbon metabolism is rewired in pancreatic ductal adenocarcinoma (PDA) to support proliferation through a glutamate oxaloacetate transaminase 1 (GOT1)-dependent pathway. METHODS: We utilized a doxycycline-inducible shRNA-mediated strategy to knockdown GOT1 in PDA and colorectal cancer (CRC) cell lines and tumor models of similar genotype. These cells were analyzed for the ability to form colonies and tumors to test if tissue type impacted GOT1 dependence. Additionally, the ability of GOT1 to impact the response to chemo- and radiotherapy was assessed. Mechanistically, the associated specimens were examined using a combination of steady-state and stable isotope tracing metabolomics strategies and computational modeling. Statistics were calculated using GraphPad Prism 7. One-way ANOVA was performed for experiments comparing multiple groups with one changing variable. Student's t test (unpaired, two-tailed) was performed when comparing two groups to each other. Metabolomics data comparing three PDA and three CRC cell lines were analyzed by performing Student's t test (unpaired, two-tailed) between all PDA metabolites and CRC metabolites. RESULTS: While PDA exhibits profound growth inhibition upon GOT1 knockdown, we found CRC to be insensitive. In PDA, but not CRC, GOT1 inhibition disrupted glycolysis, nucleotide metabolism, and redox homeostasis. These insights were leveraged in PDA, where we demonstrate that radiotherapy potently enhanced the effect of GOT1 inhibition on tumor growth. CONCLUSIONS: Taken together, these results illustrate the role of tissue type in dictating metabolic dependencies and provide new insights for targeting metabolism to treat PDA.

10.
Nat Commun ; 8: 13905, 2017 01 09.
Article in English | MEDLINE | ID: mdl-28067217

ABSTRACT

The CRISPR-Cas9 system provides a versatile toolkit for genome engineering that can introduce various DNA lesions at specific genomic locations. However, a better understanding of the nature of these lesions and the repair pathways engaged is critical to realizing the full potential of this technology. Here we characterize the different lesions arising from each Cas9 variant and the resulting repair pathway engagement. We demonstrate that the presence and polarity of the overhang structure is a critical determinant of double-strand break repair pathway choice. Similarly, single nicks deriving from different Cas9 variants differentially activate repair: D10A but not N863A-induced nicks are repaired by homologous recombination. Finally, we demonstrate that homologous recombination is required for repairing lesions using double-stranded, but not single-stranded DNA as a template. This detailed characterization of repair pathway choice in response to CRISPR-Cas9 enables a more deterministic approach for designing research and therapeutic genome engineering strategies.


Subject(s)
BRCA2 Protein/genetics , CRISPR-Cas Systems , DNA/genetics , Gene Editing/methods , Genome, Human , Rad51 Recombinase/genetics , Recombinational DNA Repair , BRCA2 Protein/antagonists & inhibitors , BRCA2 Protein/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , CRISPR-Associated Protein 9 , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats , DNA/metabolism , DNA Breaks, Double-Stranded , Endonucleases/genetics , Endonucleases/metabolism , HEK293 Cells , Humans , K562 Cells , Osteoblasts/cytology , Osteoblasts/metabolism , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rad51 Recombinase/antagonists & inhibitors , Rad51 Recombinase/metabolism
11.
Methods Enzymol ; 409: 442-62, 2006.
Article in English | MEDLINE | ID: mdl-16793417

ABSTRACT

Replication of the eukaryotic genome is a difficult task, as cells must coordinate chromosome replication with chromatin remodeling, DNA recombination, DNA repair, transcription, cell cycle progression, and sister chromatid cohesion. Yet, DNA replication is a potentially genotoxic process, particularly when replication forks encounter a bulge in the template: forks under these conditions may stall and restart or even break down leading to fork collapse. It is now clear that fork collapse stimulates chromosomal rearrangements and therefore represents a potential source of DNA damage. Hence, the comprehension of the mechanisms that preserve replication fork integrity or that promote fork collapse are extremely relevant for the understanding of the cellular processes controlling genome stability. Here we describe some experimental approaches that can be used to physically visualize the quality of replication forks in the yeast S. cerevisiae and to distinguish between stalled and collapsed forks.


Subject(s)
DNA Replication , Saccharomyces cerevisiae/genetics , Blotting, Southern , Cell Division , Chromatography, Gel , DNA, Fungal/genetics , Electrophoresis, Agar Gel , Electrophoresis, Gel, Two-Dimensional , Microscopy, Electron , Saccharomyces cerevisiae/cytology
12.
Oncogene ; 23(6): 1206-13, 2004 Feb 12.
Article in English | MEDLINE | ID: mdl-14647447

ABSTRACT

The replication checkpoint controls the integrity of replicating chromosomes by stabilizing stalled forks, thus preventing the accumulation of abnormal replication and recombination intermediates that contribute to genome instability. Checkpoint-defective cells are susceptible to rearrangements at chromosome fragile sites when replication pauses, and certain human cancer prone diseases suffer checkpoint abnormalities. It is unclear as to how the checkpoint stabilizes stalled forks and how cells sense replication blocks. We have analysed the checkpoint contribution in controlling replisome-fork association when replication pauses. We show that in yeast wild-type cells, stalled forks exhibit stable replisome complexes and the checkpoint sensors Ddc1 and Ddc2, thus activating Rad53 checkpoint kinase. Ddc1/Ddc2 recruitment on stalled forks and Rad53 activation are influenced by the single-strand-binding protein replication factor A (RFA). rad53 forks exhibit a defective association with DNA polymerases alpha, epsilon and delta. Further, in rad53 mutants, stalled forks progressively generate abnormal structures that turn into checkpoint signals by accumulating RFA, Ddc1 and Ddc2. We suggest that, following replication blocks, checkpoint activation mediated by RFA-ssDNA filaments stabilizes stalled forks by controlling replisome-fork association, thus preventing unscheduled recruitment of recombination enzymes that could otherwise cause the pathological processing of the forks.


Subject(s)
Cell Cycle Proteins , Chromosome Aberrations , DNA Replication/genetics , Protein Serine-Threonine Kinases/genetics , Saccharomyces cerevisiae/genetics , Checkpoint Kinase 2 , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Gene Rearrangement , Genetic Predisposition to Disease/genetics , Humans , Neoplasms/genetics , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction/genetics
13.
Science ; 332(6035): 1313-7, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21659603

ABSTRACT

The DNA damage response (DDR) is brought about by a protein kinase cascade that orchestrates DNA repair through transcriptional and posttranslational mechanisms. Cell cycle arrest is a hallmark of the DDR. We screened for cells that lacked damage-induced cell cycle arrest and uncovered a critical role for Fanconi anemia and homologous recombination proteins in ATR (ataxia telangiectasia and Rad3-related) signaling. Three DDR candidates, the RNA processing protein INTS7, the circadian transcription factor CLOCK, and a previously uncharacterized protein RHINO, were recruited to sites of DNA damage. RHINO independently bound the Rad9-Rad1-Hus1 complex (9-1-1) and the ATR activator TopBP1. RHINO was recruited to sites of DNA damage by the 9-1-1 complex to promote Chk1 activation. We suggest that RHINO functions together with the 9-1-1 complex and TopBP1 to fully activate ATR.


Subject(s)
Carrier Proteins/physiology , Cell Cycle Proteins/metabolism , Chemokines/physiology , DNA Repair , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Ataxia Telangiectasia Mutated Proteins , Carrier Proteins/metabolism , Cell Cycle/genetics , Cell Line, Tumor , Chemokines/genetics , Chemokines, CXC , DNA Damage , DNA-Binding Proteins/metabolism , Exonucleases/metabolism , Humans , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism
14.
Mol Cell ; 17(1): 153-9, 2005 Jan 07.
Article in English | MEDLINE | ID: mdl-15629726

ABSTRACT

The replication checkpoint coordinates the cell cycle with DNA replication and recombination, preventing genome instability and cancer. The budding yeast Rad53 checkpoint kinase stabilizes stalled forks and replisome-fork complexes, thus preventing the accumulation of ss-DNA regions and reversed forks at collapsed forks. We searched for factors involved in the processing of stalled forks in HU-treated rad53 cells. Using the neutral-neutral two-dimensional electrophoresis technique (2D gel) and psoralen crosslinking combined with electron microscopy (EM), we found that the Exo1 exonuclease is recruited to stalled forks and, in rad53 mutants, counteracts reversed fork accumulation by generating ss-DNA intermediates. Hence, Exo1-mediated fork processing resembles the action of E. coli RecJ nuclease at damaged forks. Fork stability and replication restart are influenced by both DNA polymerase-fork association and Exo1-mediated processing. We suggest that Exo1 counteracts fork reversal by resecting newly synthesized chains and resolving the sister chromatid junctions that cause regression of collapsed forks.


Subject(s)
DNA Replication , Exodeoxyribonucleases/metabolism , Saccharomyces cerevisiae/metabolism , Cell Cycle , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Checkpoint Kinase 2 , DNA Replication/drug effects , DNA, Fungal/biosynthesis , DNA, Fungal/ultrastructure , DNA, Single-Stranded/metabolism , Exodeoxyribonucleases/genetics , Genes, Fungal , Hydroxyurea/pharmacology , Microscopy, Electron , Mutation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
15.
Genes Dev ; 19(3): 339-50, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15687257

ABSTRACT

S-phase cells overcome chromosome lesions through replication-coupled recombination processes that seem to be assisted by recombination-dependent DNA structures and/or replication-related sister chromatid junctions. RecQ helicases, including yeast Sgs1 and human BLM, have been implicated in both replication and recombination and protect genome integrity by preventing unscheduled mitotic recombination events. We have studied the RecQ helicase-mediated mechanisms controlling genome stability by analyzing replication forks encountering a damaged template in sgs1 cells. We show that, in sgs1 mutants, recombination-dependent cruciform structures accumulate at damaged forks. Their accumulation requires Rad51 protein, is counteracted by Srs2 DNA helicase, and does not prevent fork movement. Sgs1, but not Srs2, promotes resolution of these recombination intermediates. A functional Rad53 checkpoint kinase that is known to protect the integrity of the sister chromatid junctions is required for the accumulation of recombination intermediates in sgs1 mutants. Finally, top3 and top3 sgs1 mutants accumulate the same structures as sgs1 cells. We suggest that, in sgs1 cells, the unscheduled accumulation of Rad51-dependent cruciform structures at damaged forks result from defective maturation of recombination-dependent intermediates that originate from the replication-related sister chromatid junctions. Our findings might contribute to explaining some of the recombination defects of BLM cells.


Subject(s)
Adenosine Triphosphatases/deficiency , DNA Helicases/deficiency , DNA Helicases/metabolism , DNA Replication/physiology , DNA-Binding Proteins/metabolism , DNA/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , DNA Helicases/genetics , Rad51 Recombinase , RecQ Helicases , Saccharomyces cerevisiae Proteins , Yeasts/genetics , Yeasts/metabolism
16.
Mol Cell ; 12(6): 1499-510, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14690603

ABSTRACT

Cells overcome intra-S DNA damage and replication impediments by coupling chromosome replication to sister chromatid-mediated recombination and replication-bypass processes. Further, molecular junctions between replicated molecules have been suggested to assist sister chromatid cohesion until anaphase. Using two-dimensional gel electrophoresis, we have identified, in yeast cells, replication-dependent X-shaped molecules that appear during origin activation, branch migrate, and distribute along the replicon through a mechanism influenced by the rate of fork progression. Their formation is independent of Rad51- and Rad52-mediated homologous recombination events and is not affected by DNA damage or replication blocks. Further, in hydroxyurea-treated rad53 mutants, altered in the replication checkpoint, the branched molecules progressively degenerate and likely contribute to generate pathological structures. We suggest that cells couple sister chromatid tethering with replication initiation by generating specialized joint molecules resembling hemicatenanes: this process might prime cohesion and assist sister chromatid-mediated recombination and replication events.


Subject(s)
Cell Cycle Proteins , Chromatids/metabolism , DNA Nucleotidyltransferases/metabolism , DNA Replication , DNA-Binding Proteins/metabolism , Replication Origin , Checkpoint Kinase 2 , DNA Damage , DNA, Fungal/genetics , DNA, Fungal/metabolism , Nucleic Acid Conformation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rad51 Recombinase , Rad52 DNA Repair and Recombination Protein , Recombination, Genetic , Replicon , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL