Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 2625, 2024 Mar 23.
Article in English | MEDLINE | ID: mdl-38521763

ABSTRACT

Homology Directed Repair (HDR) enables precise genome editing, but the implementation of HDR-based therapies is hindered by limited efficiency in comparison to methods that exploit alternative DNA repair routes, such as Non-Homologous End Joining (NHEJ). In this study, we develop a functional, pooled screening platform to identify protein-based reagents that improve HDR in human hematopoietic stem and progenitor cells (HSPCs). We leverage this screening platform to explore sequence diversity at the binding interface of the NHEJ inhibitor i53 and its target, 53BP1, identifying optimized variants that enable new intermolecular bonds and robustly increase HDR. We show that these variants specifically reduce insertion-deletion outcomes without increasing off-target editing, synergize with a DNAPK inhibitor molecule, and can be applied at manufacturing scale to increase the fraction of cells bearing repaired alleles. This screening platform can enable the discovery of future gene editing reagents that improve HDR outcomes.


Subject(s)
CRISPR-Cas Systems , Recombinational DNA Repair , Humans , Gene Editing/methods , DNA Repair , DNA End-Joining Repair
2.
Nat Commun ; 12(1): 4601, 2021 07 29.
Article in English | MEDLINE | ID: mdl-34326322

ABSTRACT

Genomic sequencing of thousands of tumors has revealed many genes associated with specific types of cancer. Similarly, large scale CRISPR functional genomics efforts have mapped genes required for cancer cell proliferation or survival in hundreds of cell lines. Despite this, for specific disease subtypes, such as metastatic prostate cancer, there are likely a number of undiscovered tumor specific driver genes that may represent potential drug targets. To identify such genetic dependencies, we performed genome-scale CRISPRi screens in metastatic prostate cancer models. We then created a pipeline in which we integrated pan-cancer functional genomics data with our metastatic prostate cancer functional and clinical genomics data to identify genes that can drive aggressive prostate cancer phenotypes. Our integrative analysis of these data reveals known prostate cancer specific driver genes, such as AR and HOXB13, as well as a number of top hits that are poorly characterized. In this study we highlight the strength of an integrated clinical and functional genomics pipeline and focus on two top hit genes, KIF4A and WDR62. We demonstrate that both KIF4A and WDR62 drive aggressive prostate cancer phenotypes in vitro and in vivo in multiple models, irrespective of AR-status, and are also associated with poor patient outcome.


Subject(s)
Cell Cycle Proteins/genetics , Kinesins/genetics , Nerve Tissue Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Animals , CRISPR-Cas Systems , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , Cell Movement/physiology , Cells, Cultured , Databases, Genetic , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Kinesins/metabolism , Male , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis , Neoplasm Staging , Nerve Tissue Proteins/metabolism , Prostatic Neoplasms/metabolism , Survival Rate
3.
PLoS One ; 16(3): e0247858, 2021.
Article in English | MEDLINE | ID: mdl-33661998

ABSTRACT

Myeloproliferative neoplasms (MPNs) cause the over-production of blood cells such as erythrocytes (polycythemia vera) or platelets (essential thrombocytosis). JAK2 V617F is the most prevalent somatic mutation in many MPNs, but previous modeling of this mutation in mice relied on transgenic overexpression and resulted in diverse phenotypes that were in some cases attributed to expression level. CRISPR-Cas9 engineering offers new possibilities to model and potentially cure genetically encoded disorders via precise modification of the endogenous locus in primary cells. Here we develop "scarless" Cas9-based reagents to create and reverse the JAK2 V617F mutation in an immortalized human erythroid progenitor cell line (HUDEP-2), CD34+ adult human hematopoietic stem and progenitor cells (HSPCs), and immunophenotypic long-term hematopoietic stem cells (LT-HSCs). We find no overt in vitro increase in proliferation associated with an endogenous JAK2 V617F allele, but co-culture with wild type cells unmasks a competitive growth advantage provided by the mutation. Acquisition of the V617F allele also promotes terminal differentiation of erythroid progenitors, even in the absence of hematopoietic cytokine signaling. Taken together, these data are consistent with the gradually progressive manifestation of MPNs and reveals that endogenously acquired JAK2 V617F mutations may yield more subtle phenotypes as compared to transgenic overexpression models.


Subject(s)
Gene Editing , Myeloproliferative Disorders/genetics , CRISPR-Cas Systems , Cell Line , Coculture Techniques , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Janus Kinase 2/genetics
4.
Elife ; 82019 07 11.
Article in English | MEDLINE | ID: mdl-31294695

ABSTRACT

Overexpression of anti-apoptotic proteins MCL1 and Bcl-xL are frequently observed in many cancers. Inhibitors targeting MCL1 are in clinical development, however numerous cancer models are intrinsically resistant to this approach. To discover mechanisms underlying resistance to MCL1 inhibition, we performed multiple flow-cytometry based genome-wide CRISPR screens interrogating two drugs that directly (MCL1i) or indirectly (CDK9i) target MCL1. Remarkably, both screens identified three components (CUL5, RNF7 and UBE2F) of a cullin-RING ubiquitin ligase complex (CRL5) that resensitized cells to MCL1 inhibition. We find that levels of the BH3-only pro-apoptotic proteins Bim and Noxa are proteasomally regulated by the CRL5 complex. Accumulation of Noxa caused by depletion of CRL5 components was responsible for re-sensitization to CDK9 inhibitor, but not MCL1 inhibitor. Discovery of a novel role of CRL5 in apoptosis and resistance to multiple types of anticancer agents suggests the potential to improve combination treatments.


Subject(s)
Cullin Proteins/genetics , Cyclin-Dependent Kinase 9/genetics , Lung Neoplasms/drug therapy , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bcl-2-Like Protein 11/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Ubiquitin-Protein Ligase Complexes/genetics , Ubiquitin-Protein Ligases/genetics
5.
Genes Dev ; 30(7): 812-26, 2016 04 01.
Article in English | MEDLINE | ID: mdl-27013236

ABSTRACT

Coats plus (CP) can be caused by mutations in the CTC1 component of CST, which promotes polymerase α (polα)/primase-dependent fill-in throughout the genome and at telomeres. The cellular pathology relating to CP has not been established. We identified a homozygous POT1 S322L substitution (POT1(CP)) in two siblings with CP. POT1(CP)induced a proliferative arrest that could be bypassed by telomerase. POT1(CP)was expressed at normal levels, bound TPP1 and telomeres, and blocked ATR signaling. POT1(CP)was defective in regulating telomerase, leading to telomere elongation rather than the telomere shortening observed in other telomeropathies. POT1(CP)was also defective in the maintenance of the telomeric C strand, causing extended 3' overhangs and stochastic telomere truncations that could be healed by telomerase. Consistent with shortening of the telomeric C strand, metaphase chromosomes showed loss of telomeres synthesized by leading strand DNA synthesis. We propose that CP is caused by a defect in POT1/CST-dependent telomere fill-in. We further propose that deficiency in the fill-in step generates truncated telomeres that halt proliferation in cells lacking telomerase, whereas, in tissues expressing telomerase (e.g., bone marrow), the truncations are healed. The proposed etiology can explain why CP presents with features distinct from those associated with telomerase defects (e.g., dyskeratosis congenita).


Subject(s)
Ataxia/genetics , Brain Neoplasms/genetics , Calcinosis/genetics , Central Nervous System Cysts/genetics , Leukoencephalopathies/genetics , Muscle Spasticity/genetics , Mutation/genetics , Retinal Diseases/genetics , Seizures/genetics , Telomere Shortening/genetics , Telomere-Binding Proteins/genetics , Telomere/genetics , Telomere/pathology , Aminopeptidases/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Cells, Cultured , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Female , Humans , Metaphase , Protein Binding , Serine Proteases/metabolism , Shelterin Complex , Signal Transduction , Telomere/metabolism , Telomere Homeostasis/genetics
6.
Cell Rep ; 9(4): 1273-80, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25453752

ABSTRACT

The conserved protein Rap1 functions at telomeres in fungi, protozoa, and vertebrates. Like yeast Rap1, human Rap1 has been implicated in telomere length regulation and repression of nonhomologous end-joining (NHEJ) at telomeres. However, mouse telomeres lacking Rap1 do not succumb to NHEJ. To determine the functions of human Rap1, we generated several transcription activator-like effector nuclease (TALEN)-mediated human cell lines lacking Rap1. Loss of Rap1 did not affect the other components of shelterin, the modification of telomeric histones, the subnuclear position of telomeres, or the 3' telomeric overhang. Telomeres lacking Rap1 did not show a DNA damage response, NHEJ, or consistent changes in their length, indicating that Rap1 does not have an important function in protection or length regulation of human telomeres. As human Rap1, like its mouse and unicellular orthologs, affects gene expression, we propose that the conservation of Rap1 reflects its role in transcriptional regulation rather than a function at telomeres.


Subject(s)
Conserved Sequence , Endonucleases/metabolism , Gene Knockout Techniques , Telomere Homeostasis/genetics , Telomere-Binding Proteins/genetics , Telomere/metabolism , Trans-Activators/metabolism , Animals , Cell Proliferation , Chromatin/metabolism , Gene Expression Regulation , Humans , Mice , Shelterin Complex , Telomere-Binding Proteins/deficiency , Telomere-Binding Proteins/metabolism , Transcription, Genetic
7.
Genes Dev ; 28(22): 2477-91, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25344324

ABSTRACT

The semiconservative replication of telomeres is facilitated by the shelterin component TRF1. Without TRF1, replication forks stall in the telomeric repeats, leading to ATR kinase signaling upon S-phase progression, fragile metaphase telomeres that resemble the common fragile sites (CFSs), and the association of sister telomeres. In contrast, TRF1 does not contribute significantly to the end protection functions of shelterin. We addressed the mechanism of TRF1 action using mouse conditional knockouts of BLM, TRF1, TPP1, and Rap1 in combination with expression of TRF1 and TIN2 mutants. The data establish that TRF1 binds BLM to facilitate lagging but not leading strand telomeric DNA synthesis. As the template for lagging strand telomeric DNA synthesis is the TTAGGG repeat strand, TRF1-bound BLM is likely required to remove secondary structures formed by these sequences. In addition, the data establish that TRF1 deploys TIN2 and the TPP1/POT1 heterodimers in shelterin to prevent ATR during telomere replication and repress the accompanying sister telomere associations. Thus, TRF1 uses two distinct mechanisms to promote replication of telomeric DNA and circumvent the consequences of replication stress. These data are relevant to the expression of CFSs and provide insights into TIN2, which is compromised in dyskeratosis congenita (DC) and related disorders.


Subject(s)
DNA Replication/genetics , DNA-Binding Proteins/metabolism , Microsatellite Repeats/genetics , RecQ Helicases/metabolism , Serine Proteases/metabolism , Telomere/genetics , Telomeric Repeat Binding Protein 1/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Cells, Cultured , Enzyme Activation , Gene Knockout Techniques , Mutation , Protein Binding , RecQ Helicases/genetics , Serine Proteases/genetics , Shelterin Complex , Signal Transduction , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Telomeric Repeat Binding Protein 1/genetics
8.
Cell Rep ; 3(6): 1847-56, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23791522

ABSTRACT

The mammalian telomere-binding protein Rap1 was recently found to have additional nontelomeric functions, acting as a transcriptional cofactor and a regulator of the NF-κB pathway. Here, we assess the effect of disrupting mouse Rap1 in vivo and report on its unanticipated role in metabolic regulation and body-weight homeostasis. Rap1 inhibition causes dysregulation in hepatic as well as adipose function, leading to glucose intolerance, insulin resistance, liver steatosis, and excess fat accumulation. Furthermore, Rap1 appears to play a pivotal role in the transcriptional cascade that controls adipocyte differentiation in vitro. Using a separation-of-function allele, we show that the metabolic function of Rap1 is independent of its recruitment to TTAGGG binding elements found at telomeres and at other interstitial loci. In conclusion, our study underscores an additional function for the most conserved telomere-binding protein, forging a link between telomere biology and metabolic signaling.


Subject(s)
Body Weight/genetics , Obesity/genetics , Telomere/metabolism , rap1 GTP-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Humans , Mice , Molecular Sequence Data , Obesity/metabolism , Sequence Homology, Amino Acid , Shelterin Complex , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/genetics
9.
Cell Cycle ; 9(20): 4061-7, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20948311

ABSTRACT

Mammalian Rap1, a TRF2-interacting protein in the telomeric shelterin complex, was recently shown to repress homology-directed repair at chromosome ends. In addition, Rap1 plays a role in transcriptional regulation and NFκB signaling. Rap1 is unique among the components of shelterin in that it is conserved in budding yeast and has non-telomeric functions. Comparison of mammalian Rap1 to the Rap1 proteins of several budding yeasts and fission yeast reveal both striking similarities and notable differences. The protean nature of Rap1 is best understood by viewing it as an adaptor that can mediate a variety of protein-protein and protein-DNA interactions depending on the organism and the complex in which it is functioning.


Subject(s)
Telomere-Binding Proteins/metabolism , Telomere/metabolism , Animals , DNA Damage , DNA Repair , Humans , Shelterin Complex , Telomere-Binding Proteins/chemistry , Telomere-Binding Proteins/genetics
10.
Science ; 327(5973): 1657-61, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20339076

ABSTRACT

Shelterin is an essential telomeric protein complex that prevents DNA damage signaling and DNA repair at mammalian chromosome ends. Here we report on the role of the TRF2-interacting factor Rap1, a conserved shelterin subunit of unknown function. We removed Rap1 from mouse telomeres either through gene deletion or by replacing TRF2 with a mutant that does not bind Rap1. Rap1 was dispensable for the essential functions of TRF2--repression of ATM kinase signaling and nonhomologous end joining (NHEJ)--and mice lacking telomeric Rap1 were viable and fertile. However, Rap1 was critical for the repression of homology-directed repair (HDR), which can alter telomere length. The data reveal that HDR at telomeres can take place in the absence of DNA damage foci and underscore the functional compartmentalization within shelterin.


Subject(s)
DNA Damage , DNA Repair , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Telomere/genetics , Amino Acid Sequence , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Proliferation , Cells, Cultured , Checkpoint Kinase 2 , DNA-Binding Proteins/metabolism , Gene Deletion , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Protein Serine-Threonine Kinases/metabolism , Recombination, Genetic , Shelterin Complex , Signal Transduction , Sister Chromatid Exchange , Telomere/metabolism , Telomere-Binding Proteins/chemistry , Telomeric Repeat Binding Protein 2/genetics , Telomeric Repeat Binding Protein 2/metabolism , Tumor Suppressor Proteins/metabolism
11.
Nat Immunol ; 10(4): 420-6, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19234474

ABSTRACT

Immunoglobulin class-switch recombination (CSR) requires activation-induced cytidine deaminase (AID). Deamination of DNA by AID in transcribed switch (S) regions leads to double-stranded breaks in DNA that serve as obligatory CSR intermediates. Here we demonstrate that the catalytic and regulatory subunits of protein kinase A (PKA) were specifically recruited to S regions to promote the localized phosphorylation of AID, which led to binding of replication protein A and subsequent propagation of the CSR cascade. Accordingly, inactivation of PKA resulted in considerable disruption of CSR because of decreased AID phosphorylation and recruitment of replication protein A to S regions. We propose that PKA nucleates the formation of active AID complexes specifically on S regions to generate the high density of DNA lesions required for CSR.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/immunology , Cytidine Deaminase/immunology , Immunoglobulin Class Switching , Recombination, Genetic/immunology , Replication Protein A/immunology , Animals , Cyclic AMP-Dependent Protein Kinases/genetics , DNA Breaks, Double-Stranded , Mice , Mice, Mutant Strains , Phosphorylation , Protein Binding , Retroviridae Infections/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...