Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nat Med ; 15(5): 553-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19377486

ABSTRACT

H2A histone family member X (H2AX, encoded by H2AFX) and its C-terminal phosphorylation (gamma-H2AX) participates in the DNA damage response and mediates DNA repair. Hypoxia is a physiological stress that induces a replication-associated DNA damage response. Moreover, hypoxia is the major driving force for neovascularization, as the hypoxia-mediated induction of vascular growth factors triggers endothelial cell proliferation. Here we studied the role of the hypoxia-induced DNA damage response in endothelial cell function and in hypoxia-driven neovascularization in vivo. Hypoxia induced replication-associated generation of gamma-H2AX in endothelial cells in vitro and in mice. Both in cultured cells and in mice, endothelial cell proliferation under hypoxic conditions was reduced by H2AX deficiency. Whereas developmental angiogenesis was not affected in H2afx(-/-) mice, hypoxia-induced neovascularization during pathologic proliferative retinopathy, in response to hind limb ischemia or during tumor angiogenesis was substantially lower in H2afx(-/-) mice. Moreover, endothelial-specific H2afx deletion resulted in reduced hypoxia-driven retina neovascularization and tumor neovascularization. Our findings establish that H2AX, and hence activation of the DNA repair response, is needed for endothelial cells to maintain their proliferation under hypoxic conditions and is crucial for hypoxia-driven neovascularization.


Subject(s)
Endothelium, Vascular/physiopathology , Histones/deficiency , Histones/genetics , Neovascularization, Pathologic/genetics , Retinal Vessels/physiopathology , Animals , DNA Damage , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Gene Deletion , Hindlimb , Humans , Hydroxyurea/pharmacology , Hypoxia/genetics , Hypoxia/physiopathology , Ischemia/genetics , Ischemia/physiopathology , Mice , Mice, Knockout , Neovascularization, Pathologic/prevention & control , Phosphorylation , Umbilical Veins/drug effects , Umbilical Veins/physiology , Umbilical Veins/physiopathology
2.
Methods Mol Biol ; 523: 125-40, 2009.
Article in English | MEDLINE | ID: mdl-19381917

ABSTRACT

The recognition and repair of DNA lesions occurs within a chromatin environment. Genetically tagging fluorescent proteins to DNA damage response proteins has provided spatial and temporal details concerning the establishment of biochemical subnuclear regions geared toward metabolizing genomic lesions. A specific marker for chromatin regions containing DNA breaks is required to study the initial dynamic structural changes in chromatin when DNA breaks occur. Here we present the experimental protocols used to investigate the dynamics of chromatin structure immediately after the simultaneous photoactivation of PAGFP-tagged core histone H2B and introduction of DNA breaks using UVA laser microirradiation on a laser scanning confocal microscope.


Subject(s)
Chromatin/metabolism , DNA Breaks/radiation effects , Fibroblasts/cytology , Fibroblasts/radiation effects , Lasers , Microscopy, Confocal/methods , Optical Phenomena , Animals , Calibration , Cell Survival/radiation effects , Cells, Cultured , Fibroblasts/metabolism , Histones/metabolism , Mice , Phosphorylation/radiation effects , Staining and Labeling , Transfection , Ultraviolet Rays
3.
Genes Dev ; 22(15): 2048-61, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18676810

ABSTRACT

H4K20 methylation is a broad chromatin modification that has been linked with diverse epigenetic functions. Several enzymes target H4K20 methylation, consistent with distinct mono-, di-, and trimethylation states controlling different biological outputs. To analyze the roles of H4K20 methylation states, we generated conditional null alleles for the two Suv4-20h histone methyltransferase (HMTase) genes in the mouse. Suv4-20h-double-null (dn) mice are perinatally lethal and have lost nearly all H4K20me3 and H4K20me2 states. The genome-wide transition to an H4K20me1 state results in increased sensitivity to damaging stress, since Suv4-20h-dn chromatin is less efficient for DNA double-strand break (DSB) repair and prone to chromosomal aberrations. Notably, Suv4-20h-dn B cells are defective in immunoglobulin class-switch recombination, and Suv4-20h-dn deficiency impairs the stem cell pool of lymphoid progenitors. Thus, conversion to an H4K20me1 state results in compromised chromatin that is insufficient to protect genome integrity and to process a DNA-rearranging differentiation program in the mouse.


Subject(s)
Chromatin/metabolism , Gene Rearrangement , Genome , Histone-Lysine N-Methyltransferase/metabolism , Alleles , Animals , Chromatin/chemistry , Chromatin/genetics , Chromosome Painting , Crosses, Genetic , Heterozygote , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Methylation , Mice , Mice, Knockout , Protein Methyltransferases , Repressor Proteins/genetics , Repressor Proteins/metabolism
4.
Cell ; 130(1): 63-75, 2007 Jul 13.
Article in English | MEDLINE | ID: mdl-17599403

ABSTRACT

DNA double-strand breaks (DSBs) induce a signal transmitted by the ataxia-telangiectasia mutated (ATM) kinase, which suppresses illegitimate joining of DSBs and activates cell-cycle checkpoints. Here we show that a significant fraction of mature ATM-deficient lymphocytes contain telomere-deleted ends produced by failed end joining during V(D)J recombination. These RAG-1/2 endonuclease-dependent, terminally deleted chromosomes persist in peripheral lymphocytes for at least 2 weeks in vivo and are stable over several generations in vitro. Restoration of ATM kinase activity in mature lymphocytes that have transiently lost ATM function leads to loss of cells with terminally deleted chromosomes. Thus, maintenance of genomic stability in lymphocytes requires faithful end joining as well a checkpoint that prevents the long-term persistence and transmission of DSBs. Silencing this checkpoint permits DNA ends produced by V(D)J recombination in a lymphoid precursor to serve as substrates for translocations with chromosomes subsequently damaged by other means in mature cells.


Subject(s)
B-Lymphocytes/physiology , Cell Cycle Proteins/metabolism , Chromosome Breakage , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Recombination, Genetic , T-Lymphocytes/physiology , Tumor Suppressor Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , B-Lymphocytes/radiation effects , Cell Cycle Proteins/genetics , DNA Damage , DNA-Binding Proteins/genetics , Genes, cdc , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunoglobulins/genetics , Immunoglobulins/metabolism , Interleukin-4/immunology , Lipopolysaccharides/immunology , Mice , Mice, Knockout , Models, Genetic , Protein Serine-Threonine Kinases/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Translocation, Genetic , Tumor Suppressor Proteins/genetics
5.
J Exp Med ; 204(5): 1003-11, 2007 May 14.
Article in English | MEDLINE | ID: mdl-17485521

ABSTRACT

The chromosomal instability syndromes Nijmegen breakage syndrome (NBS) and ataxia telangiectasia (AT) share many overlapping phenotypes, including cancer predisposition, radiation sensitivity, cell-cycle checkpoint defects, immunodeficiency, and gonadal dysfunction. The NBS protein Nbs1 is not only a downstream target of AT mutated (ATM) kinase but also acts upstream, promoting optimal ATM activation, ATM recruitment to breaks, and ATM accessibility to substrates. By reconstituting Nbs1 knockout mice with bacterial artificial chromosomes, we have assessed the contribution of distinct regions of Nbs1 to the ATM-dependent DNA damage response. We find that T cell and oocyte development, as well as DNA damage-induced G2/M and S phase checkpoint arrest and radiation survival are dependent on the N-terminal forkhead-associated domain, but not on the principal residues phosphorylated by ATM (S278 and S343) or on the evolutionarily conserved C-terminal region of Nbs1. However, the C-terminal region regulates irradiation-induced apoptosis. These studies provide insight into the complex interplay between Nbs1 and ATM in the DNA damage response.


Subject(s)
Apoptosis/physiology , Cell Cycle Proteins/metabolism , Chromosomal Instability/physiology , DNA Damage/physiology , Nuclear Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , Cell Cycle Proteins/genetics , Chromosomes, Artificial, Bacterial , DNA Primers , DNA-Binding Proteins/metabolism , Fluorescent Antibody Technique , Humans , Immunoprecipitation , In Situ Nick-End Labeling , Mice , Mice, Knockout , Nuclear Proteins/genetics , Oocytes/growth & development , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , T-Lymphocytes/physiology , Tumor Suppressor Proteins/metabolism
6.
Nature ; 443(7108): 222-5, 2006 Sep 14.
Article in English | MEDLINE | ID: mdl-16906133

ABSTRACT

The ATM (ataxia telangiectasia mutated) protein kinase is activated under physiological and pathological conditions that induce DNA double-strand breaks (DSBs). Loss of ATM or failure of its activation in humans and mice lead to defective cellular responses to DSBs, such as cell cycle checkpoints, radiation sensitivity, immune dysfunction, infertility and cancer predisposition. A widely used biological marker to identify the active form of ATM is the autophosphorylation of ATM at a single, conserved serine residue (Ser 1981 in humans; Ser 1987 in mouse). Here we show that Atm-dependent responses are functional at the organismal and cellular level in mice that express a mutant form of Atm (mutation of Ser to Ala at position 1987) as their sole Atm species. Moreover, the mutant protein does not exhibit dominant-negative interfering activity when expressed physiologically or overexpressed in the context of Atm heterozygous mice. These results suggest an alternative mode for stimulation of Atm by DSBs in which Atm autophosphorylation at Ser 1987, like trans-phosphorylation of downstream substrates, is a consequence rather than a cause of Atm activation.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Phosphoserine/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , DNA Damage , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Genomic Instability , Lymphocytes/cytology , Lymphocytes/metabolism , Mice , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Transgenes/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
7.
Cell Cycle ; 5(17): 1910-2, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16929171

ABSTRACT

The cellular response to DNA breaks consists of a complex signaling network that coordinates the initial recognition of the lesion with the induction of cell cycle checkpoints and DNA repair. With DNA wrapped around histone proteins and packaged into higher order levels of chromatin structure, the detection of a single DNA break (DSB) in the genome is the molecular equivalent of finding a needle in a haystack. A recent study from our laboratory used high-resolution electron microscropy and live cell imaging to demonstrate that chromatin undergoes a marked reorganization in response to a DSB. In an energy dependent manner, chromatin rapidly decondenses to a more open configuration in the regions surrounding the lesion. We propose that this ATP dependent chromatin-remodeling event facilitates the subsequent recognition and processing of damaged DNA. While the chromatin surrounding the lesion remodels to a more open configuration, the DNA break itself remains relatively immobile over time, consistent with the idea that DNA damage response proteins migrate to positionally stable sites of damaged DNA. The lack of significant movement of chromatin regions containing DSBs has implications for the process by which chromosomal translocations form.


Subject(s)
Chromatin/chemistry , DNA Damage , Models, Genetic , Animals , Chromatin/metabolism
8.
J Cell Biol ; 172(6): 823-34, 2006 Mar 13.
Article in English | MEDLINE | ID: mdl-16520385

ABSTRACT

The repair of DNA double-strand breaks (DSBs) is facilitated by the phosphorylation of H2AX, which organizes DNA damage signaling and chromatin remodeling complexes in the vicinity of the lesion. The disruption of DNA integrity induces an alteration of chromatin architecture that has been proposed to activate the DNA damage transducing kinase ataxia telangiectasia mutated. However, little is known about the physical properties of damaged chromatin. In this study, we use a photoactivatable version of GFP-tagged histone H2B to examine the mobility and structure of chromatin containing DSBs in living cells. We find that chromatin containing DSBs exhibits limited mobility but undergoes an energy-dependent local expansion immediately after DNA damage. The localized expansion observed in real time corresponds to a 30-40% reduction in the density of chromatin fibers in the vicinity of DSBs, as measured by energy-filtering transmission electron microscopy. The observed opening of chromatin occurs independently of H2AX and ATM. We propose that localized adenosine triphosphate-dependent decondensation of chromatin at DSBs establishes an accessible subnuclear environment that facilitates DNA damage signaling and repair.


Subject(s)
Adenosine Triphosphate/metabolism , Chromatin/genetics , DNA Damage/genetics , DNA Repair/genetics , DNA/genetics , Animals , Cells, Cultured , Chromatin/chemistry , Chromatin/ultrastructure , Chromosome Positioning/genetics , DNA/ultrastructure , Energy Metabolism/genetics , Female , Fibroblasts , Green Fluorescent Proteins , HeLa Cells , Histones/genetics , Histones/metabolism , Humans , Male , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Signal Transduction/genetics
9.
Mol Cell ; 21(2): 187-200, 2006 Jan 20.
Article in English | MEDLINE | ID: mdl-16427009

ABSTRACT

MDC1 functions in checkpoint activation and DNA repair following DNA damage. To address the physiological role of MDC1, we disrupted the MDC1 gene in mice. MDC1-/- mice recapitulated many phenotypes of H2AX-/- mice, including growth retardation, male infertility, immune defects, chromosome instability, DNA repair defects, and radiation sensitivity. At the molecular level, H2AX, MDC1, and ATM form a positive feedback loop, with MDC1 directly mediating the interaction between H2AX and ATM. MDC1 binds phosphorylated H2AX through its BRCT domain and ATM through its FHA domain. Through these interactions, MDC1 accumulates activated ATM flanking the sites of DNA damage, facilitating further ATM-dependent phosphorylation of H2AX and the amplification of DNA damage signals. In the absence of MDC1, many downstream ATM signaling events are defective. These results suggest that MDC1, as a signal amplifier of the ATM pathway, is vital in controlling proper DNA damage response and maintaining genomic stability.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Ataxia Telangiectasia Mutated Proteins , DNA Repair , Female , Genomic Instability , Infertility, Male/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Signal Transduction , Trans-Activators
10.
Proc Natl Acad Sci U S A ; 102(30): 10730-5, 2005 Jul 26.
Article in English | MEDLINE | ID: mdl-16027367

ABSTRACT

Cells adapted to high NaCl have many DNA breaks both in cell culture and in the renal inner medulla in vivo; yet they survive, function, and even proliferate. Here, we show that Ku86 is important for maintaining chromosomal integrity despite the continued presence of DNA breaks. The Ku heterodimer is part of DNA-dependent PK (DNA-PK), a complex that contributes by nonhomologous end joining to repair of double-strand breaks. We demonstrate that cells deficient in Ku86, but not cells deficient in DNA-PKcs (the catalytic subunit of DNA-PK), are hypersensitive to high NaCl as manifested by profound inhibition of proliferation, aberrant mitosis, and increased chromosomal fragmentation. Lower eukaryotes, including the soil nematode Caenorhabditis elegans, lack a DNA-PKcs homologue but are able to adapt to high NaCl. We show that cells of C. elegans adapted to high NaCl have many DNA breaks, similar to the mammalian cells adapted to high NaCl. Ku86 mutant C. elegans as well as C. elegans fed with cku86 dsRNA also display hypersensitivity to high NaCl, characterized by a reduced number of progeny and prolonged generation time in high NaCl. We propose that Ku86 ameliorates the effects of high NaCl-induced DNA breaks in adapted cells by supporting alignment of the broken ends of the DNA and thus maintaining integrity of the fragmented chromatin.


Subject(s)
Adaptation, Physiological , Antigens, Nuclear/metabolism , Chromatin/physiology , DNA Damage , DNA Repair/physiology , DNA-Binding Proteins/metabolism , Sodium Chloride/toxicity , Animals , Antigens, Nuclear/genetics , Antigens, Nuclear/physiology , Blotting, Western , Caenorhabditis elegans , Cell Cycle/drug effects , Cell Line , Chromatin/drug effects , Chromatin/genetics , Cricetinae , Cricetulus , Cytogenetic Analysis , DNA Repair/genetics , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Humans , Ku Autoantigen , Mice , Nuclear Proteins/metabolism , RNA Interference , Reproduction/drug effects
11.
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
12.
Development ; 131(20): 5185-95, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15459109

ABSTRACT

The nerve growth factor (NGF) receptor TrkA is widely expressed in non-neural tissues suggesting pleiotropic functions outside the nervous system. Based on pharmacological and immuno-depletion experiments, it has been hypothesized that NGF plays an important role in the normal development and function of the immune system. However, attempts to unravel these functions by conventional gene targeting in mice have been hampered by the early postnatal lethality caused by null mutations. We have developed a novel 'reverse conditional' gene targeting strategy by which TrkA function is restored specifically in the nervous system. Mice lacking TrkA in non-neuronal tissues are viable and appear grossly normal. All major immune system cell populations are present in normal numbers and distributions. However, mutant mice have elevated serum levels of certain immunoglobulin classes and accumulate B1 cells with aging. These data, confirmed in a classical reconstitution model using embryonic fetal liver from TrkA-null mice, demonstrate that endogenous NGF modulates B cell development through TrkA in vivo. Furthermore, they demonstrate that many of the dramatic effects previously reported by pharmacological or immuno-depletion approaches do not reflect physiological developmental roles of TrkA in the immune system.


Subject(s)
B-Lymphocytes/metabolism , Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Animals , B-Lymphocytes/immunology , Immune System/embryology , Immunoglobulins/blood , Immunologic Memory/genetics , Immunologic Memory/immunology , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mice, Knockout , Nerve Growth Factor/deficiency , Nerve Growth Factor/genetics , Receptor, trkA/deficiency , Receptor, trkA/genetics , Receptor, trkA/immunology
13.
J Cell Biol ; 166(6): 801-13, 2004 Sep 13.
Article in English | MEDLINE | ID: mdl-15364958

ABSTRACT

Bloom's syndrome is a rare autosomal recessive genetic disorder characterized by chromosomal aberrations, genetic instability, and cancer predisposition, all of which may be the result of abnormal signal transduction during DNA damage recognition. Here, we show that BLM is an intermediate responder to stalled DNA replication forks. BLM colocalized and physically interacted with the DNA damage response proteins 53BP1 and H2AX. Although BLM facilitated physical interaction between p53 and 53BP1, 53BP1 was required for efficient accumulation of both BLM and p53 at the sites of stalled replication. The accumulation of BLM/53BP1 foci and the physical interaction between them was independent of gamma-H2AX. The active Chk1 kinase was essential for both the accurate focal colocalization of 53BP1 with BLM and the consequent stabilization of BLM. Once the ATR/Chk1- and 53BP1-mediated signal from replicational stress is received, BLM functions in multiple downstream repair processes, thereby fulfilling its role as a caretaker tumor suppressor.


Subject(s)
Bloom Syndrome/enzymology , DNA Helicases/metabolism , Intracellular Signaling Peptides and Proteins , Protein Kinases/metabolism , S Phase , Blotting, Western , Bromodeoxyuridine/metabolism , Carrier Proteins , Cell Line , Checkpoint Kinase 1 , DNA Damage , DNA Helicases/genetics , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Hydroxyurea/pharmacology , Kinetics , Microscopy, Confocal , Phosphoproteins , Phosphorylation , Precipitin Tests , RNA, Small Interfering , Tumor Suppressor p53-Binding Protein 1
15.
Biochem Cell Biol ; 81(3): 123-9, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12897845

ABSTRACT

Phosphorylated H2AX (gamma-H2AX) is essential to the efficient recognition and (or) repair of DNA double strand breaks (DSBs), and many molecules, often thousands, of H2AX become rapidly phosphorylated at the site of each nascent DSB. An antibody to gamma-H2AX reveals that this highly amplified process generates nuclear foci. The phosphorylation site is a serine four residues from the C-terminus which has been evolutionarily conserved in organisms from giardia intestinalis to humans. Mice and yeast lacking the conserved serine residue demonstrate a variety of defects in DNA DSB processing. H2AX Delta/Delta mice are smaller, sensitive to ionizing radiation, defective in class switch recombination and spermatogenesis while cells from the mice demonstrate substantially increased numbers of genomic defects. gamma-H2AX foci formation is a sensitive biological dosimeter and presents new and exciting opportunities to understand important biological processes, human diseases, and individual variations in radiation sensitivity. These potentialities demonstrate the importance of understanding the parameters and functions of gamma-H2AX formation.


Subject(s)
DNA Damage , Histones/metabolism , Amino Acid Sequence , Animals , Chromatin/genetics , Chromatin/metabolism , Chromatin/radiation effects , Chromosomes/genetics , Chromosomes/metabolism , Chromosomes/radiation effects , DNA Repair , Histones/genetics , Humans , Mice , Molecular Sequence Data , Molecular Structure , Phosphorylation/radiation effects , Sequence Homology, Amino Acid , Time Factors
16.
Cell ; 114(3): 371-383, 2003 Aug 08.
Article in English | MEDLINE | ID: mdl-12914701

ABSTRACT

Histone H2AX becomes phosphorylated in chromatin domains flanking sites of DNA double-strand breakage associated with gamma-irradiation, meiotic recombination, DNA replication, and antigen receptor rearrangements. Here, we show that loss of a single H2AX allele compromises genomic integrity and enhances the susceptibility to cancer in the absence of p53. In comparison with heterozygotes, tumors arise earlier in the H2AX homozygous null background, and H2AX(-/-) p53(-/-) lymphomas harbor an increased frequency of clonal nonreciprocal translocations and amplifications. These include complex rearrangements that juxtapose the c-myc oncogene to antigen receptor loci. Restoration of the H2AX null allele with wild-type H2AX restores genomic stability and radiation resistance, but this effect is abolished by substitution of the conserved serine phosphorylation sites in H2AX with alanine or glutamic acid residues. Our results establish H2AX as genomic caretaker that requires the function of both gene alleles for optimal protection against tumorigenesis.


Subject(s)
Chromosome Aberrations , DNA/metabolism , Disease Susceptibility , Histones/metabolism , Animals , B-Lymphocytes/physiology , Cells, Cultured , DNA/genetics , DNA/radiation effects , DNA Damage , DNA Repair , Fibroblasts/cytology , Fibroblasts/physiology , Fibroblasts/radiation effects , Gamma Rays , Histones/genetics , Karyotyping , Lymphoma/genetics , Lymphoma/metabolism , Lymphoma/pathology , Mice , Mice, Knockout , Phosphorylation , Survival Rate , T-Lymphocytes/cytology , T-Lymphocytes/physiology , Tumor Suppressor Protein p53/genetics
17.
Nat Cell Biol ; 5(7): 675-9, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12792649

ABSTRACT

Histone H2AX is rapidly phosphorylated in the chromatin micro-environment surrounding a DNA double-strand break (DSB). Although H2AX deficiency is not detrimental to life, H2AX is required for the accumulation of numerous essential proteins into irradiation induced foci (IRIF). However, the relationship between IRIF formation, H2AX phosphorylation (gamma-H2AX) and the detection of DNA damage is unclear. Here, we show that the migration of repair and signalling proteins to DSBs is not abrogated in H2AX(-/-) cells, or in H2AX-deficient cells that have been reconstituted with H2AX mutants that eliminate phosphorylation. Despite their initial recruitment to DSBs, numerous factors, including Nbs1, 53BP1 and Brca1, subsequently fail to form IRIF. We propose that gamma-H2AX does not constitute the primary signal required for the redistribution of repair complexes to damaged chromatin, but may function to concentrate proteins in the vicinity of DNA lesions. The differential requirements for factor recruitment to DSBs and sequestration into IRIF may explain why essential regulatory pathways controlling the ability of cells to respond to DNA damage are not abolished in the absence of H2AX.


Subject(s)
Cell Nucleus/genetics , Chromatin/genetics , DNA Damage/genetics , Eukaryotic Cells/metabolism , Histones/deficiency , Animals , Cell Line , DNA Damage/radiation effects , DNA Repair/genetics , DNA-Binding Proteins/genetics , Histones/genetics , Humans , Mice , Nuclear Proteins/genetics , Phosphorylation , Signal Transduction/genetics
18.
Dev Cell ; 4(4): 497-508, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12689589

ABSTRACT

During meiotic prophase in male mammals, the X and Y chromosomes condense to form a macrochromatin body, termed the sex, or XY, body, within which X- and Y-linked genes are transcriptionally repressed. The molecular basis and biological function of both sex body formation and meiotic sex chromosome inactivation (MSCI) are unknown. A phosphorylated form of H2AX, a histone H2A variant implicated in DNA repair, accumulates in the sex body in a manner independent of meiotic recombination-associated double-strand breaks. Here we show that the X and Y chromosomes of histone H2AX-deficient spermatocytes fail to condense to form a sex body, do not initiate MSCI, and exhibit severe defects in meiotic pairing. Moreover, other sex body proteins, including macroH2A1.2 and XMR, do not preferentially localize with the sex chromosomes in the absence of H2AX. Thus, H2AX is required for the chromatin remodeling and associated silencing in male meiosis.


Subject(s)
Chromatin/metabolism , Gene Silencing/physiology , Histones/deficiency , Meiosis/genetics , Sex Chromosomes/metabolism , Spermatocytes/metabolism , Testis/metabolism , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/genetics , Chromosome Pairing/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histones/genetics , Male , Mice , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Structure, Tertiary/genetics , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Rad51 Recombinase , Sex Chromatin/genetics , Sex Chromatin/metabolism , Sex Chromosome Aberrations , Sex Chromosomes/genetics , Spermatocytes/pathology , Testis/abnormalities , Testis/pathology
19.
J Biol Chem ; 278(22): 20303-12, 2003 May 30.
Article in English | MEDLINE | ID: mdl-12660252

ABSTRACT

DNA double-strand breaks originating from diverse causes in eukaryotic cells are accompanied by the formation of phosphorylated H2AX (gammaH2AX) foci. Here we show that gammaH2AX formation is also a cellular response to topoisomerase I cleavage complexes known to induce DNA double-strand breaks during replication. In HCT116 human carcinoma cells exposed to the topoisomerase I inhibitor camptothecin, the resulting gammaH2AX formation can be prevented with the phosphatidylinositol 3-OH kinase-related kinase inhibitor wortmannin; however, in contrast to ionizing radiation, only camptothecin-induced gammaH2AX formation can be prevented with the DNA replication inhibitor aphidicolin and enhanced with the checkpoint abrogator 7-hydroxystaurosporine. This gammaH2AX formation is suppressed in ATR (ataxia telangiectasia and Rad3-related) deficient cells and markedly decreased in DNA-dependent protein kinase-deficient cells but is not abrogated in ataxia telangiectasia cells, indicating that ATR and DNA-dependent protein kinase are the kinases primarily involved in gammaH2AX formation at the sites of replication-mediated DNA double-strand breaks. Mre11- and Nbs1-deficient cells are still able to form gammaH2AX. However, H2AX-/- mouse embryonic fibroblasts exposed to camptothecin fail to form Mre11, Rad50, and Nbs1 foci and are hypersensitive to camptothecin. These results demonstrate a conserved gammaH2AX response for double-strand breaks induced by replication fork collision. gammaH2AX foci are required for recruiting repair and checkpoint protein complexes to the replication break sites.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage , DNA Replication , DNA Topoisomerases, Type I/metabolism , DNA-Binding Proteins/metabolism , Histones/metabolism , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Animals , Blotting, Western , Camptothecin/pharmacology , DNA Repair Enzymes , Enzyme Inhibitors/pharmacology , Humans , Hydrolysis , MRE11 Homologue Protein , Mice , Mice, Knockout , Microscopy, Confocal , Phosphorylation , Topoisomerase I Inhibitors , Tumor Cells, Cultured
20.
Nat Cell Biol ; 4(12): 993-7, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12447390

ABSTRACT

Activation of the ataxia telangiectasia mutated (ATM) kinase triggers diverse cellular responses to ionizing radiation (IR), including the initiation of cell cycle checkpoints. Histone H2AX, p53 binding-protein 1 (53BP1) and Chk2 are targets of ATM-mediated phosphorylation, but little is known about their roles in signalling the presence of DNA damage. Here, we show that mice lacking either H2AX or 53BP1, but not Chk2, manifest a G2-M checkpoint defect close to that observed in ATM(-/-) cells after exposure to low, but not high, doses of IR. Moreover, H2AX regulates the ability of 53BP1 to efficiently accumulate into IR-induced foci. We propose that at threshold levels of DNA damage, H2AX-mediated concentration of 53BP1 at double-strand breaks is essential for the amplification of signals that might otherwise be insufficient to prevent entry of damaged cells into mitosis.


Subject(s)
Carrier Proteins/genetics , DNA Damage/genetics , G2 Phase/genetics , Histones/genetics , Intracellular Signaling Peptides and Proteins , Mitosis/genetics , Phosphoproteins , Protein Serine-Threonine Kinases , Animals , Cell Line , Checkpoint Kinase 2 , Chromosomal Proteins, Non-Histone , DNA-Binding Proteins , G2 Phase/physiology , Gene Expression Regulation , Mice , Mitosis/physiology , Phosphorylation , Protein Kinases/genetics , Tumor Suppressor p53-Binding Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...