Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Mol Cell ; 82(11): 2132-2147.e6, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35447083

ABSTRACT

Mouse pericentromeric DNA is composed of tandem major satellite repeats, which are heterochromatinized and cluster together to form chromocenters. These clusters are refractory to DNA repair through homologous recombination (HR). The mechanisms by which pericentromeric heterochromatin imposes a barrier on HR and the implications of repeat clustering are unknown. Here, we compare the spatial recruitment of HR factors upon double-stranded DNA breaks (DSBs) induced in human and mouse pericentromeric heterochromatin, which differ in their capacity to form clusters. We show that while DSBs increase the accessibility of human pericentromeric heterochromatin by disrupting HP1α dimerization, mouse pericentromeric heterochromatin repeat clustering imposes a physical barrier that requires many layers of de-compaction to be accessed. Our results support a model in which the 3D organization of heterochromatin dictates the spatial activation of DNA repair pathways and is key to preventing the activation of HR within clustered repeats and the onset of chromosomal translocations.


Subject(s)
Heterochromatin , Translocation, Genetic , Animals , Cluster Analysis , DNA Breaks, Double-Stranded , Heterochromatin/genetics , Homologous Recombination/genetics , Mice
2.
Mol Cell ; 81(12): 2596-2610.e7, 2021 06 17.
Article in English | MEDLINE | ID: mdl-33961796

ABSTRACT

p53-binding protein 1 (53BP1) regulates both the DNA damage response and p53 signaling. Although 53BP1's function is well established in DNA double-strand break repair, how its role in p53 signaling is modulated remains poorly understood. Here, we identify the scaffolding protein AHNAK as a G1 phase-enriched interactor of 53BP1. We demonstrate that AHNAK binds to the 53BP1 oligomerization domain and controls its multimerization potential. Loss of AHNAK results in hyper-accumulation of 53BP1 on chromatin and enhanced phase separation, culminating in an elevated p53 response, compromising cell survival in cancer cells but leading to senescence in non-transformed cells. Cancer transcriptome analyses indicate that AHNAK-53BP1 cooperation contributes to the suppression of p53 target gene networks in tumors and that loss of AHNAK sensitizes cells to combinatorial cancer treatments. These findings highlight AHNAK as a rheostat of 53BP1 function, which surveys cell proliferation by preventing an excessive p53 response.


Subject(s)
Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism , Cell Line, Tumor , Chromatin/metabolism , DNA/genetics , DNA Breaks, Double-Stranded , DNA Repair , G1 Phase/physiology , Histones/metabolism , Humans , MCF-7 Cells , Membrane Proteins/genetics , Membrane Proteins/physiology , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Signal Transduction/physiology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/physiology
3.
Nature ; 600(7890): 748-753, 2021 12.
Article in English | MEDLINE | ID: mdl-34853474

ABSTRACT

Centromeric integrity is key for proper chromosome segregation during cell division1. Centromeres have unique chromatin features that are essential for centromere maintenance2. Although they are intrinsically fragile and represent hotspots for chromosomal rearrangements3, little is known about how centromere integrity in response to DNA damage is preserved. DNA repair by homologous recombination requires the presence of the sister chromatid and is suppressed in the G1 phase of the cell cycle4. Here we demonstrate that DNA breaks that occur at centromeres in G1 recruit the homologous recombination machinery, despite the absence of a sister chromatid. Mechanistically, we show that the centromere-specific histone H3 variant CENP-A and its chaperone HJURP, together with dimethylation of lysine 4 in histone 3 (H3K4me2), enable a succession of events leading to the licensing of homologous recombination in G1. H3K4me2 promotes DNA-end resection by allowing DNA damage-induced centromeric transcription and increased formation of DNA-RNA hybrids. CENP-A and HJURP interact with the deubiquitinase USP11, enabling formation of the RAD51-BRCA1-BRCA2 complex5 and rendering the centromeres accessible to RAD51 recruitment and homologous recombination in G1. Finally, we show that inhibition of homologous recombination in G1 leads to centromeric instability and chromosomal translocations. Our results support a model in which licensing of homologous recombination at centromeric breaks occurs throughout the cell cycle to prevent the activation of mutagenic DNA repair pathways and preserve centromeric integrity.


Subject(s)
Chromosomal Proteins, Non-Histone , DNA Repair , DNA-Binding Proteins , Centromere/genetics , Centromere/metabolism , Centromere Protein A , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Segregation , DNA , DNA-Binding Proteins/metabolism , Histones/metabolism , Homologous Recombination
4.
Nature ; 600(7888): 329-333, 2021 12.
Article in English | MEDLINE | ID: mdl-34819671

ABSTRACT

Efficient humoral responses rely on DNA damage, mutagenesis and error-prone DNA repair. Diversification of B cell receptors through somatic hypermutation and class-switch recombination are initiated by cytidine deamination in DNA mediated by activation-induced cytidine deaminase (AID)1 and by the subsequent excision of the resulting uracils by uracil DNA glycosylase (UNG) and by mismatch repair proteins1-3. Although uracils arising in DNA are accurately repaired1-4, how these pathways are co-opted to generate mutations and double-strand DNA breaks in the context of somatic hypermutation and class-switch recombination is unknown1-3. Here we performed a genome-wide CRISPR-Cas9 knockout screen for genes involved in class-switch recombination and identified FAM72A, a protein that interacts with the nuclear isoform of UNG (UNG2)5 and is overexpressed in several cancers5. We show that the FAM72A-UNG2 interaction controls the levels of UNG2 and that class-switch recombination is defective in Fam72a-/- B cells due to the upregulation of UNG2. Moreover, we show that somatic hypermutation is reduced in Fam72a-/- B cells and that its pattern is skewed upon upregulation of UNG2. Our results are consistent with a model in which FAM72A interacts with UNG2 to control its physiological level by triggering its degradation, regulating the level of uracil excision and thus the balance between error-prone and error-free DNA repair. Our findings have potential implications for tumorigenesis, as reduced levels of UNG2 mediated by overexpression of Fam72a would shift the balance towards mutagenic DNA repair, rendering cells more prone to acquire mutations.


Subject(s)
B-Lymphocytes , DNA Mismatch Repair , Immunoglobulin Class Switching , Immunoglobulin Switch Region , Mutation , Somatic Hypermutation, Immunoglobulin , Animals , Female , Male , Mice , B-Lymphocytes/metabolism , CRISPR-Cas Systems/genetics , Genome/genetics , Immunoglobulin Class Switching/genetics , Immunoglobulin Switch Region/genetics , Somatic Hypermutation, Immunoglobulin/genetics , Up-Regulation , Uracil/metabolism
5.
Cell ; 143(1): 122-33, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20887897

ABSTRACT

Activation-induced cytidine deaminase (AID) initiates antibody gene diversification by creating U:G mismatches. However, AID is not specific for antibody genes; Off-target lesions can activate oncogenes or cause chromosome translocations. Despite its importance in these transactions little is known about how AID finds its targets. We performed an shRNA screen to identify factors required for class switch recombination (CSR) of antibody loci. We found that Spt5, a factor associated with stalled RNA polymerase II (Pol II) and single stranded DNA (ssDNA), is required for CSR. Spt5 interacts with AID, it facilitates association between AID and Pol II, and AID recruitment to its Ig and non-Ig targets. ChIP-seq experiments reveal that Spt5 colocalizes with AID and stalled Pol II. Further, Spt5 accumulation at sites of Pol II stalling is predictive of AID-induced mutation. We propose that AID is targeted to sites of Pol II stalling in part via its association with Spt5.


Subject(s)
B-Lymphocytes/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Cytidine Deaminase/metabolism , Immunoglobulin Class Switching , RNA Polymerase II/metabolism , Transcriptional Elongation Factors/metabolism , Animals , Cell Line , Cell Line, Tumor , Fibroblasts/metabolism , Humans , Immunoglobulins/genetics , Mice
6.
Eur J Immunol ; 53(7): e2350373, 2023 07.
Article in English | MEDLINE | ID: mdl-37143384

ABSTRACT

During immune responses, B cells engaging a cognate antigen are recruited to GCs in secondary lymphoid organs where they will diversify their BCR to generate highly specific and adapted humoral responses. They do so, by inducing the expression of activation-induced cytidine deaminase (AID), which initiates somatic hypermutation (SHM) and class switch recombination (CSR). AID deaminates cytosines in ss DNA, generating U:G mismatches that are processed to induce ds DNA break intermediates during CSR that result in the expression of a different antibody isotype. Interestingly, hypoxia regions have been reported in GCs and suggesting that hypoxia could modulate the humoral response. Furthermore, hypoxia inducible transcription factor (HIF) can bind to the AID promoter and induce AID expression in a non-B-cell setting, suggesting that it might be involved in the transcriptional induction of AID in B cells, hence, regulating SHM and CSR. We, thus, hypothesized that HIF could regulate the efficiency of CSR. Here, we show that the inactivation of both the HIF-1α and HIF-1ß subunits of the HIF transcription factor in murine CH12 B cells results in defective CSR and that this is due to the suboptimal induction of AID expression.


Subject(s)
Cytidine Deaminase , Gene Expression Regulation , Animals , Mice , B-Lymphocytes , Cytidine Deaminase/metabolism , Immunoglobulin Class Switching , Immunoglobulin Isotypes/metabolism , Somatic Hypermutation, Immunoglobulin , Transcription Factors/genetics
7.
Mol Cell ; 63(2): 293-305, 2016 07 21.
Article in English | MEDLINE | ID: mdl-27397684

ABSTRACT

Repetitive DNA is packaged into heterochromatin to maintain its integrity. We use CRISPR/Cas9 to induce DSBs in different mammalian heterochromatin structures. We demonstrate that in pericentric heterochromatin, DSBs are positionally stable in G1 and recruit NHEJ factors. In S/G2, DSBs are resected and relocate to the periphery of heterochromatin, where they are retained by RAD51. This is independent of chromatin relaxation but requires end resection and RAD51 exclusion from the core. DSBs that fail to relocate are engaged by NHEJ or SSA proteins. We propose that the spatial disconnection between end resection and RAD51 binding prevents the activation of mutagenic pathways and illegitimate recombination. Interestingly, in centromeric heterochromatin, DSBs recruit both NHEJ and HR proteins throughout the cell cycle. Our results highlight striking differences in the recruitment of DNA repair factors between pericentric and centromeric heterochromatin and suggest a model in which the commitment to specific DNA repair pathways regulates DSB position.


Subject(s)
Centromere/metabolism , Chromatin Assembly and Disassembly , DNA Breaks, Double-Stranded , DNA Repair , Heterochromatin/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , CRISPR-Cas Systems , Centromere/chemistry , Centromere/genetics , DNA End-Joining Repair , G2 Phase , Heterochromatin/chemistry , Heterochromatin/genetics , Histones/genetics , Histones/metabolism , Ku Autoantigen/genetics , Ku Autoantigen/metabolism , Mice , NIH 3T3 Cells , RNA Interference , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Rad52 DNA Repair and Recombination Protein/genetics , Rad52 DNA Repair and Recombination Protein/metabolism , Recombinational DNA Repair , S Phase , Time Factors , Transfection
8.
J Cell Sci ; 134(10)2021 05 15.
Article in English | MEDLINE | ID: mdl-34037234

ABSTRACT

Many cellular processes, ranging from cell division to differentiation, are controlled by nuclear pore complexes (NPCs). However, studying the contributions of individual NPC subunits to these processes in vertebrates has long been impeded by their complexity and the lack of efficient genetic tools. Here, we use genome editing in mouse embryonic stem cells (mESCs) to characterize the role of NPC structural components, focusing on the short arm of the Y-complex that comprises Nup85, Seh1 and Nup43. We show that Seh1 and Nup43, although dispensable in pluripotent mESCs, are required for their normal cell growth rates, their viability upon differentiation and for the maintenance of proper NPC density. mESCs with an N-terminally truncated Nup85 mutation (in which interaction with Seh1 is greatly impaired) feature a similar reduction of NPC density. However, their proliferation and differentiation are unaltered, indicating that it is the integrity of the Y-complex, rather than the number of NPCs, that is critical to ensure these processes.


Subject(s)
Mouse Embryonic Stem Cells , Nuclear Pore , Animals , Cell Differentiation/genetics , Gene Editing , Mice , Nuclear Pore/genetics , Nuclear Pore Complex Proteins/genetics
9.
Cell ; 135(6): 1028-38, 2008 Dec 12.
Article in English | MEDLINE | ID: mdl-19070574

ABSTRACT

Chromosomal translocation requires formation of paired double-strand DNA breaks (DSBs) on heterologous chromosomes. One of the most well characterized oncogenic translocations juxtaposes c-myc and the immunoglobulin heavy-chain locus (IgH) and is found in Burkitt's lymphomas in humans and plasmacytomas in mice. DNA breaks in IgH leading to c-myc/IgH translocations are created by activation-induced cytidine deaminase (AID) during antibody class switch recombination or somatic hypermutation. However, the source of DNA breaks at c-myc is not known. Here, we provide evidence for the c-myc promoter region being required in targeting AID-mediated DNA damage to produce DSBs in c-myc that lead to c-myc/IgH translocations in primary B lymphocytes. Thus, in addition to producing somatic mutations and DNA breaks in antibody genes, AID is also responsible for the DNA lesions in oncogenes that are required for their translocation.


Subject(s)
Cytidine Deaminase/metabolism , Genes, Immunoglobulin Heavy Chain , Genes, myc , Translocation, Genetic , Animals , B-Lymphocytes/metabolism , Burkitt Lymphoma/genetics , Burkitt Lymphoma/metabolism , DNA Breaks, Double-Stranded , Embryonic Stem Cells , Humans , Integrases/metabolism , Mice , Mice, Inbred C57BL , Plasmacytoma/genetics , Plasmacytoma/metabolism
10.
Genes Dev ; 28(22): 2450-63, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25366693

ABSTRACT

Faithful DNA repair is essential to avoid chromosomal rearrangements and promote genome integrity. Nuclear organization has emerged as a key parameter in the formation of chromosomal translocations, yet little is known as to whether DNA repair can efficiently occur throughout the nucleus and whether it is affected by the location of the lesion. Here, we induce DNA double-strand breaks (DSBs) at different nuclear compartments and follow their fate. We demonstrate that DSBs induced at the nuclear membrane (but not at nuclear pores or nuclear interior) fail to rapidly activate the DNA damage response (DDR) and repair by homologous recombination (HR). Real-time and superresolution imaging reveal that DNA DSBs within lamina-associated domains do not migrate to more permissive environments for HR, like the nuclear pores or the nuclear interior, but instead are repaired in situ by alternative end-joining. Our results are consistent with a model in which nuclear position dictates the choice of DNA repair pathway, thus revealing a new level of regulation in DSB repair controlled by spatial organization of DNA within the nucleus.


Subject(s)
Cell Nucleus/metabolism , DNA Breaks, Double-Stranded , DNA Repair , Cell Line, Tumor , Chromatin/genetics , HeLa Cells , Homologous Recombination/genetics , Humans , Nuclear Envelope/metabolism , Nuclear Lamina/metabolism
11.
Biol Cell ; 112(1): 22-37, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31789463

ABSTRACT

BACKGROUND: The centrosome regulates cell spatial organisation by controlling the architecture of the microtubule (MT) cytoskeleton. Conversely, the position of the centrosome within the cell depends on cytoskeletal networks it helps organizing. In mammalian cells, centrosome positioning involves a population of MT stably anchored at centrioles, the core components of the centrosome. An MT-anchoring complex containing the proteins ninein and Cep170 is enriched at subdistal appendages (SAP) that decorate the older centriole (called mother centriole) and at centriole proximal ends. Here, we studied the role played at the centrosome by hVFL3/CCDC61, the human ortholog of proteins required for anchoring distinct sets of cytoskeletal fibres to centrioles in unicellular eukaryotes. RESULTS: We show that hVFL3 co-localises at SAP and at centriole proximal ends with components of the MT-anchoring complex, and physically interacts with Cep170. Depletion of hVFL3 increased the distance between mother and daughter centrioles without affecting the assembly of a filamentous linker that tethers the centrioles and contains the proteins rootletin and C-Nap1. When the linker was disrupted by inactivating C-Nap1, hVFL3-depletion exacerbated centriole splitting, a phenotype also observed following depletion of other SAP components. This supported that hVFL3 is required for SAP function, which we further established by showing that centrosome positioning is perturbed in hVFL3-depleted interphase cells. Finally, we found that hVFL3 is an MT-binding protein. CONCLUSIONS AND SIGNIFICANCE: Together, our results support that hVFL3 is required for anchoring MT at SAP during interphase and ensuring proper centrosome cohesion and positioning. The role of the VFL3 family of proteins thus appears to have been conserved in evolution despite the great variation in the shape of centriole appendages in different eukaryotic species.


Subject(s)
Carrier Proteins/metabolism , Centrioles , Centrosome , Tubulin/metabolism , Animals , CRISPR-Cas Systems , Carrier Proteins/genetics , Cell Line , Centrioles/metabolism , Centrioles/ultrastructure , Centrosome/metabolism , Centrosome/ultrastructure , Cilia/ultrastructure , Cytoskeletal Proteins/metabolism , Cytoskeleton/ultrastructure , Humans , Microscopy, Electron , Microtubules/metabolism , Microtubules/ultrastructure , RNA, Small Interfering
12.
Eur J Immunol ; 48(4): 720-723, 2018 04.
Article in English | MEDLINE | ID: mdl-29293266

ABSTRACT

The Mediator complex is known to orchestrate transcription. Here we show that B cell conditional deficient mice for the Med1 subunit display robust somatic hypermutation. Nevertheless, the mutation frequency at A residues is decreased and the expected A/T ratio is abolished, implicating Mediator in the second phase of somatic hypermutation.


Subject(s)
B-Lymphocytes/cytology , Mediator Complex Subunit 1/deficiency , Mediator Complex Subunit 1/genetics , Somatic Hypermutation, Immunoglobulin/genetics , Animals , B-Lymphocytes/immunology , Germinal Center/cytology , Germinal Center/immunology , Mice , Mice, Transgenic
13.
Mol Cell ; 41(1): 33-45, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21211721

ABSTRACT

PARP-3 is a member of the ADP-ribosyl transferase superfamily of unknown function. We show that PARP-3 is stimulated by DNA double-strand breaks (DSBs) in vitro and functions in the same pathway as the poly (ADP-ribose)-binding protein APLF to accelerate chromosomal DNA DSB repair. We implicate PARP-3 in the accumulation of APLF at DSBs and demonstrate that APLF promotes the retention of XRCC4/DNA ligase IV complex in chromatin, suggesting that PARP-3 and APLF accelerate DNA ligation during nonhomologous end-joining (NHEJ). Consistent with this, we show that class switch recombination in Aplf(-/-) B cells is biased toward microhomology-mediated end-joining, a pathway that operates in the absence of XRCC4/DNA ligase IV, and that the requirement for PARP-3 and APLF for NHEJ is circumvented by overexpression of XRCC4/DNA ligase IV. These data identify molecular roles for PARP-3 and APLF in chromosomal DNA double-strand break repair reactions.


Subject(s)
Carrier Proteins/physiology , Cell Cycle Proteins/physiology , Phosphoproteins/physiology , Poly(ADP-ribose) Polymerases/physiology , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , DNA Breaks, Double-Stranded , DNA Repair/physiology , DNA-(Apurinic or Apyrimidinic Site) Lyase , Gene Deletion , Humans , Mice , Phosphoproteins/genetics , Phosphoproteins/metabolism , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Poly-ADP-Ribose Binding Proteins , Recombinant Fusion Proteins/physiology
14.
Eur J Immunol ; 47(4): 665-676, 2017 04.
Article in English | MEDLINE | ID: mdl-28105679

ABSTRACT

To mount highly specific and adapted immune responses, B lymphocytes assemble and diversify their antibody repertoire through mechanisms involving the formation of programmed DNA damage. Immunoglobulin class switch recombination (CSR) is triggered by DNA lesions induced by activation-induced cytidine deaminase, which are processed to double-stranded DNA break (DSB) intermediates. These DSBs activate the cellular DNA damage response and enroll numerous DNA repair factors, involving poly(ADP-ribose) polymerases Parp1, Parp2, and Parp3 to promote appropriate DNA repair and efficient long-range recombination. The macroParp Parp9, which is overexpressed in certain lymphomas, has been recently implicated in DSB repair, acting together with Parp1. Here, we examine the contribution of Parp9 to the resolution of physiological DSBs incurred during V(D)J recombination and CSR by generating Parp9-/- mice. We find that Parp9-deficient mice are viable, fertile, and do not show any overt phenotype. Moreover, we find that Parp9 is dispensable for B-cell development. Finally, we show that CSR and DNA end-joining are robust in the absence of Parp9, indicating that Parp9 is not essential in vivo to achieve physiological DSB repair, or that strong compensatory mechanisms exist.


Subject(s)
B-Lymphocytes/physiology , DNA End-Joining Repair , Immunoglobulin Class Switching , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Adaptive Immunity , Animals , Cells, Cultured , DNA Breaks, Double-Stranded , DNA Damage , DNA Repair , Immunoglobulins/genetics , Immunoglobulins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly(ADP-ribose) Polymerases/genetics
15.
PLoS Genet ; 11(5): e1005240, 2015 May.
Article in English | MEDLINE | ID: mdl-26000965

ABSTRACT

To generate highly specific and adapted immune responses, B cells diversify their antibody repertoire through mechanisms involving the generation of programmed DNA damage. Somatic hypermutation (SHM) and class switch recombination (CSR) are initiated by the recruitment of activation-induced cytidine deaminase (AID) to immunoglobulin loci and by the subsequent generation of DNA lesions, which are differentially processed to mutations during SHM or to double-stranded DNA break intermediates during CSR. The latter activate the DNA damage response and mobilize multiple DNA repair factors, including Parp1 and Parp2, to promote DNA repair and long-range recombination. We examined the contribution of Parp3 in CSR and SHM. We find that deficiency in Parp3 results in enhanced CSR, while SHM remains unaffected. Mechanistically, this is due to increased occupancy of AID at the donor (Sµ) switch region. We also find evidence of increased levels of DNA damage at switch region junctions and a bias towards alternative end joining in the absence of Parp3. We propose that Parp3 plays a CSR-specific role by controlling AID levels at switch regions during CSR.


Subject(s)
Gene Expression Regulation , Immunoglobulin Class Switching/genetics , Poly(ADP-ribose) Polymerases/metabolism , Animals , B-Lymphocytes/metabolism , Base Sequence , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , DNA Breaks, Double-Stranded , DNA Repair , Genetic Loci , Immunoglobulin G/blood , Immunoglobulin M/blood , Immunoglobulin Switch Region/genetics , Mice , Mice, Knockout , Molecular Sequence Data , Poly(ADP-ribose) Polymerases/genetics , Recombination, Genetic , Somatic Hypermutation, Immunoglobulin/genetics
16.
Immunity ; 28(5): 630-8, 2008 May.
Article in English | MEDLINE | ID: mdl-18455451

ABSTRACT

MicroRNAs (miRNAs) are small noncoding RNAs that regulate vast networks of genes that share miRNA target sequences. To examine the physiologic effects of an individual miRNA-mRNA interaction in vivo, we generated mice that carry a mutation in the putative microRNA-155 (miR-155) binding site in the 3'-untranslated region of activation-induced cytidine deaminase (AID), designated Aicda(155) mice. AID is required for immunoglobulin gene diversification in B lymphocytes, but it also promotes chromosomal translocations. Aicda(155) caused an increase in steady-state Aicda mRNA and protein amounts by increasing the half-life of the mRNA, resulting in a high degree of Myc-Igh translocations. A similar but more pronounced translocation phenotype was also found in miR-155-deficient mice. Our experiments indicate that miR-155 can act as a tumor suppressor by reducing potentially oncogenic translocations generated by AID.


Subject(s)
B-Lymphocytes/enzymology , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Genes, myc , Immunoglobulin Heavy Chains/genetics , MicroRNAs/metabolism , Translocation, Genetic , 3' Untranslated Regions , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Genes, Immunoglobulin , Immunoglobulin Class Switching , Lipopolysaccharides/immunology , Mice , Mice, Mutant Strains , MicroRNAs/genetics , Mutation , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Somatic Hypermutation, Immunoglobulin
17.
Exp Cell Res ; 329(1): 18-25, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25017100

ABSTRACT

Poly(ADP-ribosyl)ation (PARylation) is a post-translational modification of proteins catalysed by Poly(ADP-ribose) polymerases (PARP). A wealth of recent advances in the biochemical and functional characterization of the DNA-dependent PARP family members have highlighted their key contribution in the DNA damage response network, the best characterized being the role of PARP1 and PARP2 in the resolution of single-strand breaks as part of the BER/SSBR process. How PARylation contributes to the repair of double-strand breaks is less well defined but has become recently the subject of significant research in the field. The aim of this review is to provide an overview of the current knowledge concerning the role of the DNA-activated PARP1, PARP2 and PARP3 in cellular response to double-strand breaks (DSB). In addition, we outline the biological significance of these properties in response to programmed DNA lesions formed during physiological processes such as antibody repertoire assembly and diversification.


Subject(s)
DNA Damage/genetics , DNA Repair , DNA/genetics , Poly(ADP-ribose) Polymerases/metabolism , Animals , Humans
18.
bioRxiv ; 2024 May 07.
Article in English | MEDLINE | ID: mdl-38352321

ABSTRACT

Transcript buffering entails the reciprocal modulation of mRNA synthesis and degradation rates to maintain stable RNA levels under varying cellular conditions. Current research supports a global, non-sequence-specific connection between mRNA synthesis and degradation, but the underlying mechanisms are still unclear. In this study, we investigated changes in RNA metabolism following acute depletion of TIP60/KAT5, the acetyltransferase subunit of the NuA4 transcriptional coactivator complex, in mouse embryonic stem cells. By combining RNA sequencing of nuclear, cytoplasmic, and newly synthesised transcript fractions with biophysical modelling, we demonstrate that TIP60 predominantly enhances transcription of numerous genes, while a smaller set of genes undergoes TIP60-dependent transcriptional repression. Surprisingly, transcription changes caused by TIP60 depletion were offset by corresponding changes in RNA nuclear export and cytoplasmic stability, indicating gene-specific buffering mechanisms. Similarly, disruption of the unrelated ATAC coactivator complex also resulted in gene-specific transcript buffering. These findings reveal that transcript buffering functions at a gene-specific level and suggest that cells dynamically adjust RNA splicing, export, and degradation in response to individual RNA synthesis alterations, thereby sustaining cellular homeostasis.

19.
J Exp Med ; 204(9): 2225-32, 2007 Sep 03.
Article in English | MEDLINE | ID: mdl-17724134

ABSTRACT

Chromosome translocations between oncogenes and the region spanning the immunoglobulin (Ig) heavy chain (IgH) variable (V), diversity (D), and joining (J) gene segments (Ig V-J(H) region) are found in several mature B cell lymphomas in humans and mice. The breakpoints are frequently adjacent to the recombination signal sequences targeted by recombination activating genes 1 and 2 during antigen receptor assembly in pre-B cells, suggesting that these translocations might be the result of aberrant V(D)J recombination. However, in mature B cells undergoing activation-induced cytidine deaminase (AID)-dependent somatic hypermutation (SHM), duplications or deletions that would necessitate a double-strand break make up 6% of all the Ig V-J(H) region-associated somatic mutations. Furthermore, DNA breaks can be detected at this locus in B cells undergoing SHM. To determine whether SHM might induce c-myc to Ig V-J(H) translocations, we searched for such events in both interleukin (IL) 6 transgenic (IL-6 tg) and AID(-/-) IL-6 tg mice. Here, we report that AID is required for c-myc to Ig V-J(H) translocations induced by IL-6.


Subject(s)
Chromosomes, Mammalian/genetics , Cytidine Deaminase/metabolism , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , Proto-Oncogene Proteins c-myc/genetics , Translocation, Genetic , Animals , Chromosome Breakage , Cytidine Deaminase/deficiency , Female , Humans , Interleukin-6/deficiency , Male , Mice , Mice, Transgenic , Somatic Hypermutation, Immunoglobulin/genetics
20.
Nat Cell Biol ; 7(7): 675-85, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15965469

ABSTRACT

Nijmegen breakage syndrome (NBS) is a chromosomal fragility disorder that shares clinical and cellular features with ataxia telangiectasia. Here we demonstrate that Nbs1-null B cells are defective in the activation of ataxia-telangiectasia-mutated (Atm) in response to ionizing radiation, whereas ataxia-telangiectasia- and Rad3-related (Atr)-dependent signalling and Atm activation in response to ultraviolet light, inhibitors of DNA replication, or hypotonic stress are intact. Expression of the main human NBS allele rescues the lethality of Nbs1-/- mice, but leads to immunodeficiency, cancer predisposition, a defect in meiotic progression in females and cell-cycle checkpoint defects that are associated with a partial reduction in Atm activity. The Mre11 interaction domain of Nbs1 is essential for viability, whereas the Forkhead-associated (FHA) domain is required for T-cell and oocyte development and efficient DNA damage signalling. Reconstitution of Nbs1 knockout mice with various mutant isoforms demonstrates the biological impact of impaired Nbs1 function at the cellular and organismal level.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , DNA-Binding Proteins/metabolism , Disease Models, Animal , Nuclear Proteins/physiology , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , ATP-Binding Cassette Transporters/metabolism , Acid Anhydride Hydrolases , Animals , Ataxia Telangiectasia Mutated Proteins , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Binding Sites/genetics , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Chromosome Aberrations , Chromosome Breakage , Chromosome Disorders/genetics , Chromosome Disorders/metabolism , Chromosome Disorders/pathology , DNA Damage/genetics , DNA Damage/physiology , DNA Repair Enzymes , DNA Replication/genetics , Female , Genetic Predisposition to Disease/genetics , Gonads/abnormalities , Humans , Immunoglobulin Class Switching/genetics , Lymphoma, Non-Hodgkin/etiology , Lymphoma, Non-Hodgkin/genetics , MRE11 Homologue Protein , Male , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation , Syndrome , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL