Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38585916

ABSTRACT

Long-term perturbation of de novo chromatin assembly during DNA replication has profound effects on epigenome maintenance and cell fate. The early mechanistic origin of these defects is unknown. Here, we combine acute degradation of Chromatin Assembly Factor 1 (CAF-1), a key player in de novo chromatin assembly, with single-cell genomics, quantitative proteomics, and live-microscopy to uncover these initiating mechanisms in human cells. CAF-1 loss immediately slows down DNA replication speed and renders nascent DNA hyperaccessible. A rapid cellular response, distinct from canonical DNA damage signaling, is triggered and lowers histone mRNAs. As a result, histone variants usage and their modifications are altered, limiting transcriptional fidelity and delaying chromatin maturation within a single S-phase. This multi-level response induces a cell-cycle arrest after mitosis. Our work reveals the immediate consequences of defective de novo chromatin assembly during DNA replication, explaining how at later times the epigenome and cell fate can be altered.

2.
Nucleic Acids Res ; 50(17): 9930-9947, 2022 09 23.
Article in English | MEDLINE | ID: mdl-36107780

ABSTRACT

Cells respond to double-strand breaks (DSBs) by activating DNA damage response pathways, including cell cycle arrest. We have previously shown that a single double-strand break generated via CRISPR/Cas9 is sufficient to delay cell cycle progression and compromise cell viability. However, we also found that the cellular response to DSBs can vary, independent of the number of lesions. This implies that not all DSBs are equally toxic, and raises the question if the location of a single double-strand break could influence its toxicity. To systematically investigate if DSB-location is a determinant of toxicity we performed a CRISPR/Cas9 screen targeting 6237 single sites in the human genome. Next, we developed a data-driven framework to design CRISPR/Cas9 sgRNA (crRNA) pools targeting specific chromatin features. The chromatin context was defined using ChromHMM states, Lamin-B1 DAM-iD, DNAseI hypersensitivity, and RNA-sequencing data. We computationally designed 6 distinct crRNA pools, each containing 10 crRNAs targeting the same chromatin state. We show that the toxicity of a DSB is highly similar across the different ChromHMM states. Rather, we find that the major determinants of toxicity of a sgRNA are cutting efficiency and off-target effects. Thus, chromatin features have little to no effect on the toxicity of a single CRISPR/Cas9-induced DSB.


Subject(s)
DNA Breaks, Double-Stranded , CRISPR-Cas Systems , Chromatin/genetics , DNA Repair , Humans , Lamins , RNA
3.
Nature ; 597(7877): 561-565, 2021 09.
Article in English | MEDLINE | ID: mdl-34497418

ABSTRACT

Single-cell sequencing methods have enabled in-depth analysis of the diversity of cell types and cell states in a wide range of organisms. These tools focus predominantly on sequencing the genomes1, epigenomes2 and transcriptomes3 of single cells. However, despite recent progress in detecting proteins by mass spectrometry with single-cell resolution4, it remains a major challenge to measure translation in individual cells. Here, building on existing protocols5-7, we have substantially increased the sensitivity of these assays to enable ribosome profiling in single cells. Integrated with a machine learning approach, this technology achieves single-codon resolution. We validate this method by demonstrating that limitation for a particular amino acid causes ribosome pausing at a subset of the codons encoding the amino acid. Of note, this pausing is only observed in a sub-population of cells correlating to its cell cycle state. We further expand on this phenomenon in non-limiting conditions and detect pronounced GAA pausing during mitosis. Finally, we demonstrate the applicability of this technique to rare primary enteroendocrine cells. This technology provides a first step towards determining the contribution of the translational process to the remarkable diversity between seemingly identical cells.


Subject(s)
Cell Cycle/genetics , Codon/genetics , Protein Biosynthesis , RNA-Seq/methods , Ribosomes/metabolism , Single-Cell Analysis , Amino Acids/deficiency , Amino Acids/pharmacology , Animals , Cell Cycle/drug effects , Cell Line , Female , Humans , Machine Learning , Male , Mice , Peptide Chain Elongation, Translational , Peptide Chain Initiation, Translational , Peptide Chain Termination, Translational , Protein Biosynthesis/drug effects , Reproducibility of Results , Ribosomes/drug effects
4.
Mol Cell ; 81(10): 2216-2230.e10, 2021 05 20.
Article in English | MEDLINE | ID: mdl-33848455

ABSTRACT

DNA double-strand break (DSB) repair is mediated by multiple pathways. It is thought that the local chromatin context affects the pathway choice, but the underlying principles are poorly understood. Using a multiplexed reporter assay in combination with Cas9 cutting, we systematically measure the relative activities of three DSB repair pathways as a function of chromatin context in >1,000 genomic locations. This reveals that non-homologous end-joining (NHEJ) is broadly biased toward euchromatin, while the contribution of microhomology-mediated end-joining (MMEJ) is higher in specific heterochromatin contexts. In H3K27me3-marked heterochromatin, inhibition of the H3K27 methyltransferase EZH2 reverts the balance toward NHEJ. Single-stranded template repair (SSTR), often used for precise CRISPR editing, competes with MMEJ and is moderately linked to chromatin context. These results provide insight into the impact of chromatin on DSB repair pathway balance and guidance for the design of Cas9-mediated genome editing experiments.


Subject(s)
CRISPR-Associated Protein 9/metabolism , Chromatin/metabolism , DNA Breaks, Double-Stranded , DNA Repair , Base Sequence , DNA End-Joining Repair , Euchromatin/metabolism , Gene Rearrangement , Genome, Human , Heterochromatin/metabolism , Humans , INDEL Mutation/genetics , K562 Cells , Kinetics , Protein Binding , Reproducibility of Results
5.
Cell Rep ; 34(4): 108675, 2021 01 26.
Article in English | MEDLINE | ID: mdl-33503422

ABSTRACT

DNA replication is challenged by numerous exogenous and endogenous factors that can interfere with the progression of replication forks. Substantial accumulation of single-stranded DNA during DNA replication activates the DNA replication stress checkpoint response that slows progression from S/G2 to M phase to protect genomic integrity. Whether and how mild replication stress restricts proliferation remains controversial. Here, we identify a cell cycle exit mechanism that prevents S/G2 phase arrested cells from undergoing mitosis after exposure to mild replication stress through premature activation of the anaphase promoting complex/cyclosome (APC/CCDH1). We find that replication stress causes a gradual decrease of the levels of the APC/CCDH1 inhibitor EMI1/FBXO5 through Forkhead box O (FOXO)-mediated inhibition of its transcription factor E2F1. By doing so, FOXOs limit the time during which the replication stress checkpoint is reversible and thereby play an important role in maintaining genomic stability.


Subject(s)
Cell Cycle/physiology , DNA Damage/genetics , DNA Replication/genetics , Genomic Instability/genetics , Cell Proliferation , Humans
6.
European J Org Chem ; 2020(45): 7087-7100, 2020 Dec 07.
Article in English | MEDLINE | ID: mdl-33380897

ABSTRACT

The synthesis and characterization of double porphyrin cage compounds are described. They consist of two porphyrins that are each attached to a diphenylglycoluril-based clip molecule via four ethyleneoxy spacers, and are linked together by a single alkyl chain using "click"-chemistry. Following a newly developed multistep synthesis procedure we report three of these double porphyrin cages, linked by spacers of different lengths, i.e. 3, 5, and 11 carbon atoms. The structures of the double porphyrin cages were fully characterized by NMR, which revealed that they consist of mixtures of two diastereoisomers. Their zinc derivatives are capable of forming sandwich-like complexes with the ditopic ligand 1,4-diazabicyclo[2,2,2]octane (dabco).

7.
Mol Cell ; 76(2): 346-358, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31561953

ABSTRACT

DNA double-strand breaks (DSBs) pose a constant threat to genomic integrity. Such DSBs need to be repaired to preserve homeostasis at both the cellular and organismal levels. Hence, the DNA damage response (DDR) has evolved to repair these lesions and limit their toxicity. The initiation of DNA repair depends on the activation of the DDR, and we know that the strength of DDR signaling may differentially affect cellular viability. However, we do not fully understand what determines the cytotoxicity of a DSB. Recent work has identified genomic location, (in)correct DNA repair pathway usage, and cell-cycle position as contributors to DSB-induced cytotoxicity. In this review, we discuss how these determinants affect cytotoxicity, highlight recent discoveries, and identify open questions that could help to improve our understanding about cell fate decisions after a DNA DSB.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Genomic Instability , Animals , Cell Cycle Checkpoints , Cell Death , Cell Proliferation , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly , Humans , Mitosis , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
8.
Nucleic Acids Res ; 46(19): 10132-10144, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30184135

ABSTRACT

DNA damaging agents cause a variety of lesions, of which DNA double-strand breaks (DSBs) are the most genotoxic. Unbiased approaches aimed at investigating the relationship between the number of DSBs and outcome of the DNA damage response have been challenging due to the random nature in which damage is induced by classical DNA damaging agents. Here, we describe a CRISPR/Cas9-based system that permits us to efficiently introduce DSBs at defined sites in the genome. Using this system, we show that a guide RNA targeting only a single site in the human genome can trigger a checkpoint response that is potent enough to delay cell cycle progression. Abrogation of this checkpoint leads to DNA breaks in mitosis which gives rise to aneuploid progeny.


Subject(s)
Cell Cycle Checkpoints/genetics , Cell Cycle/genetics , DNA Breaks, Double-Stranded , DNA Repair , Aneuploidy , CRISPR-Cas Systems , Cell Line , DNA Damage , Genome, Human/genetics , Humans , Mitosis/genetics
10.
Cancer Cell ; 30(4): 548-562, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27693046

ABSTRACT

Neuroblastoma is a pediatric embryonal malignancy characterized by impaired neuronal differentiation. A better understanding of neuroblastoma differentiation is essential for developing new therapeutic approaches. GDE2 (encoded by GDPD5) is a six-transmembrane-domain glycerophosphodiesterase that promotes embryonic neurogenesis. We find that high GDPD5 expression is strongly associated with favorable outcome in neuroblastoma. GDE2 induces differentiation of neuroblastoma cells, suppresses cell motility, and opposes RhoA-driven neurite retraction. GDE2 alters the Rac-RhoA activity balance and the expression of multiple differentiation-associated genes. Mechanistically, GDE2 acts by cleaving (in cis) and releasing glycosylphosphatidylinositol-anchored glypican-6, a putative co-receptor. A single point mutation in the ectodomain abolishes GDE2 function. Our results reveal GDE2 as a cell-autonomous inducer of neuroblastoma differentiation with prognostic significance and potential therapeutic value.


Subject(s)
Glypicans/metabolism , Neuroblastoma/enzymology , Neuroblastoma/pathology , Phosphoric Diester Hydrolases/metabolism , Animals , Cell Differentiation/physiology , Chickens , Glycosylphosphatidylinositols/metabolism , HEK293 Cells , Humans , Prognosis
11.
Nat Commun ; 7: 12618, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27561326

ABSTRACT

Activation of the DNA-damage response can lead to the induction of an arrest at various stages in the cell cycle. These arrests are reversible in nature, unless the damage is too excessive. Here we find that checkpoint reversibility is lost in cells that are in very late G2, but not yet fully committed to enter mitosis (antephase). We show that antephase cells exit the cell cycle and enter senescence at levels of DNA damage that induce a reversible arrest in early G2. We show that checkpoint reversibility critically depends on the presence of the APC/C inhibitor Emi1, which is degraded just before mitosis. Importantly, ablation of the cell cycle withdrawal mechanism in antephase promotes cell division in the presence of broken chromosomes. Thus, our data uncover a novel, but irreversible, DNA-damage response in antephase that is required to prevent the propagation of DNA damage during cell division.


Subject(s)
DNA Damage , G2 Phase Cell Cycle Checkpoints/genetics , Genomic Instability/genetics , Mitosis/genetics , Anaphase-Promoting Complex-Cyclosome/genetics , Anaphase-Promoting Complex-Cyclosome/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Chromosomes, Human/genetics , F-Box Proteins/genetics , F-Box Proteins/metabolism , Humans , Time-Lapse Imaging
12.
Cell Rep ; 14(11): 2519-27, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26972008

ABSTRACT

rDNA repeats constitute the most heavily transcribed region in the human genome. Tumors frequently display elevated levels of recombination in rDNA, indicating that the repeats are a liability to the genomic integrity of a cell. However, little is known about how cells deal with DNA double-stranded breaks in rDNA. Using selective endonucleases, we show that human cells are highly sensitive to breaks in 45S but not the 5S rDNA repeats. We find that homologous recombination inhibits repair of breaks in 45S rDNA, and this results in repeat loss. We identify the structural maintenance of chromosomes protein 5 (SMC5) as contributing to recombination-mediated repair of rDNA breaks. Together, our data demonstrate that SMC5-mediated recombination can lead to error-prone repair of 45S rDNA repeats, resulting in their loss and thereby reducing cellular viability.


Subject(s)
RNA, Ribosomal/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Survival/radiation effects , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone , DNA Breaks, Double-Stranded , DNA Repair , Endonucleases/metabolism , Genome, Human , Histones/metabolism , Homologous Recombination , Humans , Microsatellite Repeats/genetics , Phosphorylation , RNA, Ribosomal/genetics , RNA, Ribosomal, 5S/genetics , RNA, Ribosomal, 5S/metabolism , Radiation, Ionizing , Real-Time Polymerase Chain Reaction , Time-Lapse Imaging
13.
EMBO Rep ; 17(5): 659-70, 2016 05.
Article in English | MEDLINE | ID: mdl-26931568

ABSTRACT

In order to maintain a stable genome, cells need to detect and repair DNA damage before they complete the division cycle. To this end, cell cycle checkpoints prevent entry into the next cell cycle phase until the damage is fully repaired. Proper reentry into the cell cycle, known as checkpoint recovery, requires that a cell retains its original cell cycle state during the arrest. Here, we have identified Tousled-like kinase 2 (Tlk2) as an important regulator of recovery after DNA damage in G2. We show that Tlk2 regulates the Asf1A histone chaperone in response to DNA damage and that depletion of Asf1A also produces a recovery defect. Both Tlk2 and Asf1A are required to restore histone H3 incorporation into damaged chromatin. Failure to do so affects expression of pro-mitotic genes and compromises the cellular competence to recover from damage-induced cell cycle arrests. Our results demonstrate that Tlk2 promotes Asf1A function during the DNA damage response in G2 to allow for proper restoration of chromatin structure at the break site and subsequent recovery from the arrest.


Subject(s)
DNA Damage , G2 Phase Cell Cycle Checkpoints/genetics , Protein Kinases/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Survival/genetics , Chromatin/genetics , Chromatin/metabolism , DNA Replication , Enzyme Activation , Gene Expression Regulation , Histones/metabolism , Humans , Molecular Chaperones , Protein Kinases/genetics , RNA, Small Interfering/genetics
14.
Mol Cell ; 55(1): 59-72, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24910099

ABSTRACT

DNA damage can result in a transient cell-cycle arrest or lead to permanent cell-cycle withdrawal. Here we show that the decision to irreversibly withdraw from the cell cycle is made within a few hours following damage in G2 cells. This permanent arrest is dependent on induction of p53 and p21, resulting in the nuclear retention of Cyclin B1. This rapid response is followed by the activation of the APC/C(Cdh1) (the anaphase-promoting complex/cyclosome and its coactivator Cdh1) several hours later. Inhibition of APC/C(Cdh1) activity fails to prevent cell-cycle withdrawal, whereas preventing nuclear retention of Cyclin B1 does allow cells to remain in cycle. Importantly, transient induction of p53 in G2 cells is sufficient to induce senescence. Taken together, these results indicate that a rapid and transient pulse of p53 in G2 can drive nuclear retention of Cyclin B1 as the first irreversible step in the onset of senescence.


Subject(s)
Cellular Senescence/genetics , DNA Damage , G2 Phase , Tumor Suppressor Protein p53/physiology , Active Transport, Cell Nucleus , Cell Cycle Checkpoints , Cell Differentiation , Cyclin B1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/physiology , Tumor Suppressor Protein p53/metabolism
16.
FEMS Microbiol Lett ; 239(2): 295-9, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15476979

ABSTRACT

This study investigated the antimicrobial activities of peptides derived from the N-terminal region of human lactoferrin, and examined the contributions of individual residues to the activity of the most potent peptide. Two regions of antimicrobial activity were identified, the first corresponding to a weakly active peptide, HLP-9, comprising residues 1-9, and a second corresponding to a more potent peptide, HLP-10, comprising residues 18-26 and containing the hexapeptide motif, FQWQRN. Inhibitory studies on peptides from the first region confirm the importance of tryptophan residues in enhancing and broadening peptide activity. Inhibitory studies with glycine-substituted homologues of the more potent peptide showed that F21/G and R25/G substitutions resulted in a major reduction or complete loss of activity, while increased peptide cationicity or flexibility had little effect. Our findings demonstrate that F21 and R25 are critical determinants of potency for HLP-10, and that the second aromatic residue may act synergistically with W23 in developing and enhancing the activity of this cationic peptide.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Bacteria/drug effects , Lactoferrin/pharmacology , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides/chemistry , Humans , Lactoferrin/chemistry , Lactoferrin/genetics , Microbial Sensitivity Tests
17.
Dig Dis Sci ; 49(3): 425-33, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15139492

ABSTRACT

A strategy protecting the small intestine against deleterious side effects associated with anti-cancer therapy is arresting epithelial cell cycling temporally. Since endogenous glucagon-like peptide-2 (GLP-2) is a trophic factor specific for intestinal epithelia, the possibility of inhibiting GLP-2-mediated cell proliferation by lactoferrin, thereby protecting the small intestine against deleterious side effects of anticancer therapy, was investigated. In Caco-2 cells, GLP-2-mediated proliferation was reduced in a dose-dependent manner using lactoferrin. Furthermore, in a rat model for methotrexate-induced mucositis, lactoferrin reduced BrdU incorporation in small intestinal epithelial cells, indicating inhibition of epithelial cell proliferation in vivo. Subsequently, protection against methotrexate-induced intestinal damage was found in corresponding regions. These results show, for the first time, that lactoferrin interferes with GLP-2-induced intestinal epithelial proliferation. It may therefore be hypothesized that lactoferrin protects the intestine against anticancer therapy-induced intestinal damage, via inhibition of GLP-2-induced small intestinal epithelial cell proliferation.


Subject(s)
Antimetabolites, Antineoplastic/adverse effects , Cytoprotection/physiology , Intestinal Mucosa/drug effects , Lactoferrin/pharmacology , Methotrexate/adverse effects , Peptides/physiology , Animals , Bromodeoxyuridine/metabolism , Caco-2 Cells , Cell Division , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Epithelial Cells/physiology , Female , Glucagon-Like Peptide 2 , Glucagon-Like Peptides , Humans , Polymerase Chain Reaction , Rats , Rats, Inbred Strains
18.
Drug Deliv ; 9(1): 31-8, 2002.
Article in English | MEDLINE | ID: mdl-11839206

ABSTRACT

Lactoferrin (Lf) is a potential drug candidate for the treatment of oropharyngeal Candida infections. However, for an effective therapeutic treatment an appropriate dosage form is required. Therefore a mucoadhesive tablet for buccal application was developed. Tablets of sufficient strength could be produced on high speed tabletting machines, but they could only be obtained when the protein contained at least 7% moisture. The tablet contained sodium alginate both for its release-controlling properties as well as for its mucoadhesive properties. Furthermore, phosphate buffer was added to keep the pH of the saliva in the mouth within the range of 6.5 to 7.5. In this pH range, Lf has shown to have its highest activity against Candida growth inhibition. The tablet formulation containing Lf had the same antifungal properties as compared with Lf alone, because in most cases identical inhibitory concentrations were observed against several clinical isolates of Candida albicans and Candida glabrata. In human volunteers the tablets, containing 250 mg Lf and placed in each pouch, were able to keep the Lf concentration in the saliva at effective levels for at least 2 hr, while the pH of the saliva remained within the desired range. We concluded that the developed mucoadhesive tablet can improve the therapeutic efficacy of Lf and that it is suitable for further clinical research.


Subject(s)
Antifungal Agents/administration & dosage , Antifungal Agents/pharmacology , Candida/drug effects , Candidiasis, Oral/drug therapy , Lactoferrin/administration & dosage , Lactoferrin/pharmacology , Adhesiveness , Administration, Buccal , Adult , Candida/growth & development , Candidiasis, Oral/microbiology , Cross-Over Studies , Delayed-Action Preparations , Drug Design , Female , Humans , Hydrogen-Ion Concentration , Male , Microbial Sensitivity Tests , Mouth Mucosa , Saliva/chemistry , Tablets
SELECTION OF CITATIONS
SEARCH DETAIL
...