Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
2.
Nat Biotechnol ; 42(3): 458-469, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37127662

ABSTRACT

Inefficient knock-in of transgene cargos limits the potential of cell-based medicines. In this study, we used a CRISPR nuclease that targets a site within an exon of an essential gene and designed a cargo template so that correct knock-in would retain essential gene function while also integrating the transgene(s) of interest. Cells with non-productive insertions and deletions would undergo negative selection. This technology, called SLEEK (SeLection by Essential-gene Exon Knock-in), achieved knock-in efficiencies of more than 90% in clinically relevant cell types without impacting long-term viability or expansion. SLEEK knock-in rates in T cells are more efficient than state-of-the-art TRAC knock-in with AAV6 and surpass more than 90% efficiency even with non-viral DNA cargos. As a clinical application, natural killer cells generated from induced pluripotent stem cells containing SLEEK knock-in of CD16 and mbIL-15 show substantially improved tumor killing and persistence in vivo.


Subject(s)
CRISPR-Cas Systems , Gene Editing , CRISPR-Cas Systems/genetics , Gene Knock-In Techniques , Transgenes/genetics
3.
Cell Rep ; 40(10): 111312, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36070702

ABSTRACT

Down syndrome (DS), driven by an extra copy of chromosome 21 (HSA21), and fragile X syndrome (FXS), driven by loss of the RNA-binding protein FMRP, are two common genetic causes of intellectual disability and autism. Based upon the number of DS-implicated transcripts bound by FMRP, we hypothesize that DS and FXS may share underlying mechanisms. Comparing DS and FXS human pluripotent stem cell (hPSC) and glutamatergic neuron models, we identify increased protein expression of select targets and overlapping transcriptional perturbations. Moreover, acute upregulation of endogenous FMRP in DS patient cells using CRISPRa is sufficient to significantly reduce expression levels of candidate proteins and reverse 40% of global transcriptional perturbations. These results pinpoint specific molecular perturbations shared between DS and FXS that can be leveraged as a strategy for target prioritization; they also provide evidence for the functional relevance of previous associations between FMRP targets and disease-implicated genes.


Subject(s)
Down Syndrome , Fragile X Syndrome , Pluripotent Stem Cells , Down Syndrome/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Humans , Neurons/metabolism , Pluripotent Stem Cells/metabolism
4.
Stem Cell Reports ; 16(9): 2138-2148, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34416176

ABSTRACT

Human pluripotent stem cells (hPSCs) have proven to be valuable tools for both drug discovery and the development of cell-based therapies. However, the long non-coding RNA XIST, which is essential for the establishment and maintenance of X chromosome inactivation, is repressed during culture, thereby causing erosion of dosage compensation in female hPSCs. Here, we report that the de novo DNA methyltransferases DNMT3A/3B are necessary for XIST repression in female hPSCs. We found that the deletion of both genes, but not the individual genes, inhibited XIST silencing, maintained the heterochromatin mark of H3K27me3, and did not cause global overdosage in X-linked genes. Meanwhile, DNMT3A/3B deletion after XIST repression failed to restore X chromosome inactivation. Our findings revealed that de novo DNA methyltransferases are primary factors responsible for initiating erosion of dosage compensation in female hPSCs, and XIST silencing is stably maintained in a de novo DNA-methylation-independent manner.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A/genetics , Gene Expression Regulation , Gene Silencing , Pluripotent Stem Cells/metabolism , RNA, Long Noncoding/genetics , Chromatin Assembly and Disassembly , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA Methyltransferase 3A/metabolism , Dosage Compensation, Genetic , Epigenesis, Genetic , Gene Expression Profiling , Genes, X-Linked , Genetic Background , Heterochromatin/genetics , Heterochromatin/metabolism , Humans , Models, Biological , Pluripotent Stem Cells/cytology , DNA Methyltransferase 3B
5.
DNA Repair (Amst) ; 106: 103176, 2021 10.
Article in English | MEDLINE | ID: mdl-34365116

ABSTRACT

DNA damage can be cytotoxic and mutagenic, and it is directly linked to aging, cancer, and other diseases. To counteract the deleterious effects of DNA damage, cells have evolved highly conserved DNA repair pathways. Many commonly used DNA repair assays are relatively low throughput and are limited to analysis of one protein or one pathway. Here, we have explored the capacity of the CometChip platform for parallel analysis of multiple DNA repair activities. Taking advantage of the versatility of the traditional comet assay and leveraging micropatterning techniques, the CometChip platform offers increased throughput and sensitivity compared to the traditional comet assay. By exposing cells to DNA damaging agents that create substrates of Base Excision Repair, Nucleotide Excision Repair, and Non-Homologous End Joining, we show that the CometChip is an effective method for assessing repair deficiencies in all three pathways. With these applications of the CometChip platform, we expand the utility of the comet assay for precise, high-throughput, parallel analysis of multiple DNA repair activities.


Subject(s)
Comet Assay/methods , DNA Damage , DNA Repair , High-Throughput Screening Assays/methods , Cell Line , Cell Line, Tumor , DNA/drug effects , DNA/metabolism , DNA/radiation effects , DNA End-Joining Repair , Humans , Mutagens/toxicity
6.
Free Radic Biol Med ; 174: 89-99, 2021 10.
Article in English | MEDLINE | ID: mdl-34324980

ABSTRACT

Although DNA repair is known to impact susceptibility to cancer and other diseases, relatively few population studies have been performed to evaluate DNA repair kinetics in people due to the difficulty of assessing DNA repair in a high-throughput manner. Here we use the CometChip, a high-throughput comet assay, to explore inter-individual variation in repair of oxidative damage to DNA, a known risk factor for aging, cancer and other diseases. DNA repair capacity after H2O2-induced DNA oxidation damage was quantified in peripheral blood mononuclear cells (PBMCs). For 10 individuals, blood was drawn at several times over the course of 4-6 weeks. In addition, blood was drawn once from each of 56 individuals. DNA damage levels were quantified prior to exposure to H2O2 and at 0, 15, 30, 60, and 120-min post exposure. We found that there is significant variability in DNA repair efficiency among individuals. When subdivided into quartiles by DNA repair efficiency, we found that the average t1/2 is 81 min for the slowest group and 24 min for the fastest group. This work shows that the CometChip can be used to uncover significant differences in repair kinetics among people, pointing to its utility in future epidemiological and clinical studies.


Subject(s)
Hydrogen Peroxide , Leukocytes, Mononuclear , Comet Assay , DNA Damage , DNA Repair , Humans , Individuality , Kinetics , Lymphocytes , Oxidative Stress/genetics
7.
Sci Rep ; 10(1): 635, 2020 01 20.
Article in English | MEDLINE | ID: mdl-31959800

ABSTRACT

CRISPR-Cas9-mediated gene interference (CRISPRi) and activation (CRISPRa) approaches hold promise for functional gene studies and genome-wide screens in human pluripotent stem cells (hPSCs). However, in contrast to CRISPR-Cas9 nuclease approaches, the efficiency of CRISPRi/a depends on continued expression of the dead Cas9 (dCas9) effector and guide RNA (gRNA), which can vary substantially depending on transgene design and delivery. Here, we design and generate new fluorescently labeled piggyBac (PB) vectors to deliver uniform and sustained expression of multiplexed gRNAs. In addition, we generate hPSC lines harboring AAVS1-integrated, inducible and fluorescent dCas9-KRAB and dCas9-VPR transgenes to allow for accurate quantification and tracking of cells that express both the dCas9 effectors and gRNAs. We then employ these systems to target the TCF4 gene in hPSCs and assess expression levels of the dCas9 effectors, individual gRNAs and targeted gene. We also assess the performance of our PB system for single gRNA delivery, confirming its utility for library format applications. Collectively, our results provide proof-of-principle application of a stable, multiplexed PB gRNA delivery system that can be widely exploited to further enable genome engineering studies in hPSCs. Paired with diverse CRISPR tools including our dual fluorescence CRISPRi/a cell lines, this system can facilitate functional dissection of individual genes and pathways as well as larger-scale screens for studies of development and disease.


Subject(s)
CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , DNA Transposable Elements , Gene Transfer Techniques , Genetic Vectors , Pluripotent Stem Cells , RNA, Guide, Kinetoplastida , Basic Helix-Loop-Helix Transcription Factors , Cell Line , Drosophila Proteins , Humans , Transgenes
8.
PLoS One ; 14(2): e0208341, 2019.
Article in English | MEDLINE | ID: mdl-30811507

ABSTRACT

The DNA repair protein O6-methylguanine DNA methyltransferase (MGMT) strongly influences the effectiveness of cancer treatment with chemotherapeutic alkylating agents, and MGMT status in cancer cells could potentially contribute to tailored therapies for individual patients. However, the promoter methylation and immunohistochemical assays presently used for measuring MGMT in clinical samples are indirect, cumbersome and sometimes do not accurately report MGMT activity. Here we directly compare the accuracy of 6 analytical methods, including two fluorescent reporter assays, against the in vitro MGMT activity assay that is considered the gold standard for measuring MGMT DNA repair capacity. We discuss the relative advantages of each method. Our data indicate that two recently developed fluorescence-based assays measure MGMT activity accurately and efficiently, and could provide a functional dimension to clinical efforts to identify patients who are likely to benefit from alkylating chemotherapy.


Subject(s)
Biological Assay/methods , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Fluorescent Dyes/administration & dosage , Tumor Suppressor Proteins/genetics , Antineoplastic Agents, Alkylating/pharmacology , Cell Line , DNA Methylation/drug effects , DNA Repair/genetics , Humans , Promoter Regions, Genetic/genetics
9.
Proc Natl Acad Sci U S A ; 114(48): E10379-E10388, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29122935

ABSTRACT

The integrity of our DNA is challenged with at least 100,000 lesions per cell on a daily basis. Failure to repair DNA damage efficiently can lead to cancer, immunodeficiency, and neurodegenerative disease. Base excision repair (BER) recognizes and repairs minimally helix-distorting DNA base lesions induced by both endogenous and exogenous DNA damaging agents. Levels of BER-initiating DNA glycosylases can vary between individuals, suggesting that quantitating and understanding interindividual differences in DNA repair capacity (DRC) may enable us to predict and prevent disease in a personalized manner. However, population studies of BER capacity have been limited because most methods used to measure BER activity are cumbersome, time consuming and, for the most part, only allow for the analysis of one DNA glycosylase at a time. We have developed a fluorescence-based multiplex flow-cytometric host cell reactivation assay wherein the activity of several enzymes [four BER-initiating DNA glycosylases and the downstream processing apurinic/apyrimidinic endonuclease 1 (APE1)] can be tested simultaneously, at single-cell resolution, in vivo. Taking advantage of the transcriptional properties of several DNA lesions, we have engineered specific fluorescent reporter plasmids for quantitative measurements of 8-oxoguanine DNA glycosylase, alkyl-adenine DNA glycosylase, MutY DNA glycosylase, uracil DNA glycosylase, and APE1 activity. We have used these reporters to measure differences in BER capacity across a panel of cell lines collected from healthy individuals, and to generate mathematical models that predict cellular sensitivity to methylmethane sulfonate, H2O2, and 5-FU from DRC. Moreover, we demonstrate the suitability of these reporters to measure differences in DRC in multiple pathways using primary lymphocytes from two individuals.


Subject(s)
Biological Variation, Population/physiology , DNA Damage/physiology , DNA Glycosylases/metabolism , DNA Repair/physiology , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Cell Line , DNA/genetics , DNA/metabolism , DNA Damage/drug effects , DNA Glycosylases/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Flow Cytometry/methods , Fluorouracil/toxicity , Gene Knockdown Techniques , Healthy Volunteers , Humans , Hydrogen Peroxide/toxicity , Methyl Methanesulfonate/toxicity , Models, Biological , Mutagenesis/drug effects , Mutagenesis/physiology , Mutagens/toxicity , Precision Medicine/methods , Primary Cell Culture , RNA, Small Interfering/metabolism , Single-Cell Analysis/methods , T-Lymphocytes
10.
Stem Cell Reports ; 9(4): 1315-1327, 2017 10 10.
Article in English | MEDLINE | ID: mdl-29020615

ABSTRACT

Scaling of CRISPR-Cas9 technology in human pluripotent stem cells (hPSCs) represents an important step for modeling complex disease and developing drug screens in human cells. However, variables affecting the scaling efficiency of gene editing in hPSCs remain poorly understood. Here, we report a standardized CRISPR-Cas9 approach, with robust benchmarking at each step, to successfully target and genotype a set of psychiatric disease-implicated genes in hPSCs and provide a resource of edited hPSC lines for six of these genes. We found that transcriptional state and nucleosome positioning around targeted loci was not correlated with editing efficiency. However, editing frequencies varied between different hPSC lines and correlated with genomic stability, underscoring the need for careful cell line selection and unbiased assessments of genomic integrity. Together, our step-by-step quantification and in-depth analyses provide an experimental roadmap for scaling Cas9-mediated editing in hPSCs to study psychiatric disease, with broader applicability for other polygenic diseases.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Biomarkers , Cell Differentiation/genetics , Cell Line , Gene Expression , Gene Targeting , Genes, Reporter , Genomic Instability , Humans , INDEL Mutation , Mental Disorders/etiology , Mental Disorders/metabolism , Mental Disorders/psychology , Neurons/cytology , Neurons/metabolism , Workflow
11.
Cancer Res ; 77(1): 198-206, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27793847

ABSTRACT

Cancer cells can resist the effects of DNA-damaging therapeutic agents via utilization of DNA repair pathways, suggesting that DNA repair capacity (DRC) measurements in cancer cells could be used to identify patients most likely to respond to treatment. However, the limitations of available technologies have so far precluded adoption of this approach in the clinic. We recently developed fluorescence-based multiplexed host cell reactivation (FM-HCR) assays to measure DRC in multiple pathways. Here we apply a mathematical model that uses DRC in multiple pathways to predict cellular resistance to killing by DNA-damaging agents. This model, developed using FM-HCR and drug sensitivity measurements in 24 human lymphoblastoid cell lines, was applied to a panel of 12 patient-derived xenograft (PDX) models of glioblastoma to predict glioblastoma response to treatment with the chemotherapeutic DNA-damaging agent temozolomide. This work showed that, in addition to changes in O6-methylguanine DNA methyltransferase (MGMT) activity, small changes in mismatch repair (MMR), nucleotide excision repair (NER), and homologous recombination (HR) capacity contributed to acquired temozolomide resistance in PDX models and led to reduced relative survival prolongation following temozolomide treatment of orthotopic mouse models in vivo Our data indicate that measuring the combined status of MMR, HR, NER, and MGMT provided a more robust prediction of temozolomide resistance than assessments of MGMT activity alone. Cancer Res; 77(1); 198-206. ©2016 AACR.


Subject(s)
Brain Neoplasms/genetics , DNA Repair/physiology , Drug Resistance, Neoplasm/genetics , Glioblastoma/genetics , Models, Theoretical , Animals , Antineoplastic Agents/pharmacology , Area Under Curve , Cell Line, Tumor , DNA Repair/drug effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Humans , Mice , ROC Curve , Temozolomide , Xenograft Model Antitumor Assays
12.
Cancer Res ; 75(15): 3127-38, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26025730

ABSTRACT

Glioblastoma (GBM) is often treated with the cytotoxic drug temozolomide, but the disease inevitably recurs in a drug-resistant form after initial treatment. Here, we report that in GBM cells, even a modest decrease in the mismatch repair (MMR) components MSH2 and MSH6 have profound effects on temozolomide sensitivity. RNAi-mediated attenuation of MSH2 and MSH6 showed that such modest decreases provided an unexpectedly strong mechanism of temozolomide resistance. In a mouse xenograft model of human GBM, small changes in MSH2 were sufficient to suppress temozolomide-induced tumor regression. Using The Cancer Genome Atlas to analyze mRNA expression patterns in tumors from temozolomide-treated GBM patients, we found that MSH2 transcripts in primary GBM could predict patient responses to initial temozolomide therapy. In recurrent disease, the absence of microsatellite instability (the standard marker for MMR deficiency) suggests a lack of involvement of MMR in the resistant phenotype of recurrent disease. However, more recent studies reveal that decreased MMR protein levels occur often in recurrent GBM. In accordance with our findings, these reported decreases may constitute a mechanism by which GBM evades temozolomide sensitivity while maintaining microsatellite stability. Overall, our results highlight the powerful effects of MSH2 attenuation as a potent mediator of temozolomide resistance and argue that MMR activity offers a predictive marker for initial therapeutic response to temozolomide treatment.


Subject(s)
Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , MutS Homolog 2 Protein/metabolism , Animals , Antineoplastic Agents, Alkylating/pharmacology , Carmustine/pharmacology , Cell Line, Tumor/drug effects , Cell Line, Tumor/radiation effects , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dacarbazine/pharmacology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Gene Knockdown Techniques , Genes, p53 , Glioblastoma/metabolism , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Mice, Inbred C57BL , MutS Homolog 2 Protein/genetics , Radiation, Ionizing , Survival Analysis , Temozolomide , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
13.
Proc Natl Acad Sci U S A ; 111(18): E1823-32, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24757057

ABSTRACT

The capacity to repair different types of DNA damage varies among individuals, making them more or less susceptible to the detrimental health consequences of damage exposures. Current methods for measuring DNA repair capacity (DRC) are relatively labor intensive, often indirect, and usually limited to a single repair pathway. Here, we describe a fluorescence-based multiplex flow-cytometric host cell reactivation assay (FM-HCR) that measures the ability of human cells to repair plasmid reporters, each bearing a different type of DNA damage or different doses of the same type of DNA damage. FM-HCR simultaneously measures repair capacity in any four of the following pathways: nucleotide excision repair, mismatch repair, base excision repair, nonhomologous end joining, homologous recombination, and methylguanine methyltransferase. We show that FM-HCR can measure interindividual DRC differences in a panel of 24 cell lines derived from genetically diverse, apparently healthy individuals, and we show that FM-HCR may be used to identify inhibitors or enhancers of DRC. We further develop a next-generation sequencing-based HCR assay (HCR-Seq) that detects rare transcriptional mutagenesis events due to lesion bypass by RNA polymerase, providing an added dimension to DRC measurements. FM-HCR and HCR-Seq provide powerful tools for exploring relationships among global DRC, disease susceptibility, and optimal treatment.


Subject(s)
DNA Damage , DNA Repair , Genetic Techniques , Cell Line , DNA End-Joining Repair , DNA Mismatch Repair , Flow Cytometry , Genes, Reporter , Guanine/analogs & derivatives , Guanine/metabolism , High-Throughput Nucleotide Sequencing , Humans , Mutagenesis , Plasmids/genetics , Sequence Analysis, RNA , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...