Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Cell Metab ; 2(1): 67-76, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16054100

ABSTRACT

The Saccharomyces cerevisiae chromatin silencing factor Sir2 suppresses genomic instability and extends replicative life span. In contrast, we find that mouse embryonic fibroblasts (MEFs) deficient for SIRT1, a mammalian Sir2 homolog, have dramatically increased resistance to replicative senescence. Extended replicative life span of SIRT1-deficient MEFs correlates with enhanced proliferative capacity under conditions of chronic, sublethal oxidative stress. In this context, SIRT1-deficient cells fail to normally upregulate either the p19(ARF) senescence regulator or its downstream target p53. However, upon acute DNA damage or oncogene expression, SIRT1-deficient cells show normal p19(ARF) induction and cell cycle arrest. Together, our findings demonstrate an unexpected SIRT1 function in promoting replicative senescence in response to chronic cellular stress and implicate p19(ARF) as a downstream effector in this pathway.


Subject(s)
Cellular Senescence , DNA Damage , Sirtuins/metabolism , Animals , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p16 , DNA Damage/drug effects , Doxorubicin/pharmacology , Fibroblasts , Genes, ras/genetics , Hydrogen Peroxide/pharmacology , Mice , Mice, Knockout , NIH 3T3 Cells , Oxidative Stress/drug effects , S Phase/drug effects , Sirtuin 1 , Sirtuins/deficiency , Sirtuins/genetics , Tumor Suppressor Protein p14ARF/metabolism
2.
J Neurosci ; 25(9): 2176-80, 2005 Mar 02.
Article in English | MEDLINE | ID: mdl-15745943

ABSTRACT

The mouse brain contains genetically distinct cells that differ with respect to chromosome number manifested as aneuploidy (Rehen et al., 2001); however, the relevance to humans is not known. Here, using double-label fluorescence in situ hybridization for the autosome chromosome 21 (chromosome 21 point probes combined with chromosome 21 "paint" probes), along with immunocytochemistry and cell sorting, we present evidence for chromosome gain and loss in the human brain. Chromosome 21 aneuploid cells constitute approximately 4% of the estimated one trillion cells in the human brain and include non-neuronal cells and postmitotic neurons identified by the neuronspecific nuclear protein marker. In comparison, human interphase lymphocytes present chromosome 21 aneuploidy rates of 0.6%. Together, these data demonstrate that human brain cells (both neurons and non-neuronal cells) can be aneuploid and that the resulting genetic mosaicism is a normal feature of the human CNS.


Subject(s)
Aneuploidy , Cerebral Cortex/cytology , Chromosomes, Human, Pair 21 , Neuroglia/metabolism , Neurons/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Cell Count/methods , Child , Child, Preschool , Chromosome Mapping , Female , Flow Cytometry/methods , Hippocampus/cytology , Humans , Immunohistochemistry/methods , In Situ Hybridization, Fluorescence/methods , Karyotyping/methods , Lymphocytes , Male , Middle Aged , Mosaicism , Phosphopyruvate Hydratase/metabolism , Postmortem Changes
3.
J Neurosci ; 24(37): 8090-6, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15371510

ABSTRACT

Aneuploid neurons populate the normal adult brain, but the cause and the consequence of chromosome abnormalities in the CNS are poorly defined. In the adult cerebral cortex of three genetic mutants, one of which is a mouse model of the human neurodegenerative disease ataxia-telangiectasia (A-T), we observed divergent levels of sex chromosome (XY) aneuploidy. Although both A-T mutated (Atm)- and transformation related protein 53 (Trp53)-dependent mechanisms are thought to clear newly postmitotic neurons with chromosome abnormalities, we found a 38% increase in the prevalence of XY aneuploidy in the adult Atm-/- cerebral cortex and a dramatic 78% decrease in Trp53-/- mutant mice. A similar 43% decrease in adult XY aneuploidy was observed in DNA repair-deficient Xrcc5-/- mutants. Additional investigation found an elevated incidence of aneuploid embryonic neural progenitor cells (NPCs) in all three mutants, but elevated apoptosis, a likely fate of embryonic NPCs with severe chromosome abnormalities, was observed only in Xrcc5-/- mutants. These data lend increasing support to the hypothesis that hereditary mutations such as ATM-deficiency, which render abnormal cells resistant to developmental clearance, can lead to late-manifesting human neurological disorders.


Subject(s)
Aneuploidy , Antigens, Nuclear/physiology , Apoptosis/physiology , Cell Cycle Proteins/physiology , Cerebral Cortex/pathology , DNA-Binding Proteins/physiology , Neurons/pathology , Protein Serine-Threonine Kinases/physiology , Sex Chromosome Aberrations , Stem Cells/pathology , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/physiology , Animals , Antigens, Nuclear/genetics , Ataxia Telangiectasia/embryology , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/pathology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Survival , Cerebral Cortex/embryology , DNA Damage , DNA Repair/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Genes, p53 , Karyotyping , Ku Autoantigen , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Sex Chromosome Aberrations/embryology , Translocation, Genetic , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
4.
J Neurosci ; 23(13): 5599-606, 2003 Jul 02.
Article in English | MEDLINE | ID: mdl-12843262

ABSTRACT

Frequent chromosomal aneuploidy has recently been discovered in normal neurons of the developing and mature murine CNS. Toward a more detailed understanding of aneuploidy and its effects on normal CNS cells, we examined the genomes of cells in the postnatal subventricular zone (SVZ), an area that harbors a large number of neural stem and progenitor cells (NPCs), which give rise to neurons and glia. Here we show that NPCs, neurons, and glia from the SVZ are frequently aneuploid. Karyotyping revealed that approximately 33% of mitotic SVZ cells lost or gained chromosomes in vivo, whereas interphase fluorescence in situ hybridization demonstrated aneuploidy in postnatal-born cells in the olfactory bulb (OB) in vivo, along with neurons, glia, and NPCs in vitro. One possible consequence of aneuploidy is altered gene expression through loss of heterozygosity (LOH). This was examined in a model of LOH: loss of transgene expression in mice hemizygous for a ubiquitously expressed enhanced green fluorescent protein (eGFP) transgene on chromosome 15. Concurrent examination of eGFP expression, transgene abundance, and chromosome 15 copy number demonstrated that a preponderance of living SVZ and OB cells not expressing eGFP lost one copy of chromosome 15; the eGFP transgene was lost in these cells as well. Although gene expression profiling revealed changes in expression levels of several genes relative to GFP-expressing controls, cells with LOH at chromosome 15 were morphologically normal and proliferated or underwent apoptosis at rates similar to those of euploid cells in vitro. These findings support the view that NPCs and postnatal-born neurons and glia can be aneuploid in vivo and functional gene expression can be permanently altered in living neural cells by chromosomal aneuploidy.


Subject(s)
Aneuploidy , Brain/metabolism , Chromosomes , Gene Expression Regulation, Developmental , Lateral Ventricles/metabolism , Animals , Brain/cytology , Brain/growth & development , Cell Division/genetics , Cell Survival/genetics , Green Fluorescent Proteins , In Situ Hybridization, Fluorescence , Karyotyping , Lateral Ventricles/cytology , Loss of Heterozygosity , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Mice , Mice, Transgenic , Neurons/cytology , Neurons/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transgenes
5.
J Neurosci ; 23(32): 10454-62, 2003 Nov 12.
Article in English | MEDLINE | ID: mdl-14614104

ABSTRACT

Recent studies based predominantly on nucleotide hybridization techniques have identified aneuploid neurons and glia in the normal brain. To substantiate these findings and address how neural aneuploidy arises, we examined individual neural progenitor cells (NPCs) undergoing mitosis. Here we report the identification of chromosomal segregation defects in normal NPCs of the mouse cerebral cortex. Immunofluorescence in fixed tissue sections revealed the presence of supernumerary centrosomes and lagging chromosomes among mitotic NPCs. The extent of aneuploidy followed the prevalence of supernumerary centrosomes within distinct cell populations. Real-time imaging of live NPCs revealed lagging chromosomes and multipolar divisions. NPCs undergoing nondisjunction were also observed, along with interphase cells that harbored micronuclei or multiple nuclei, consistent with unbalanced nuclear division. These data independently confirm the presence of aneuploid NPCs and demonstrate the occurrence of mitotic segregation defects in normal cells that can mechanistically account for aneuploidy in the CNS.


Subject(s)
Aneuploidy , Chromosome Segregation/physiology , Neurons/cytology , Stem Cells/cytology , Stem Cells/metabolism , Animals , Chromosomes/genetics , Female , Karyotyping , Metaphase/physiology , Mice , Mice, Inbred BALB C/genetics , Mitosis , Neurons/ultrastructure , Nondisjunction, Genetic
6.
Proc Natl Acad Sci U S A ; 103(19): 7378-83, 2006 May 09.
Article in English | MEDLINE | ID: mdl-16670198

ABSTRACT

Inactivation of the XRCC4 nonhomologous end-joining factor in the mouse germ line leads to embryonic lethality, in association with apoptosis of newly generated, postmitotic neurons. We now show that conditional inactivation of the XRCC4 in nestin-expressing neuronal progenitor cells, although leading to no obvious phenotype in a WT background, leads to early onset of neuronally differentiated medulloblastomas (MBs) in a p53-deficient background. A substantial proportion of the XRCC4/p53-deficient MBs have high-level N-myc gene amplification, often intrachromosomally in the context of complex translocations or other alterations of chromosome 12, on which N-myc resides, or extrachromosomally within double minutes. In addition, most XRCC4/p53-deficient MBs harbor clonal translocations of chromosome 13, which frequently involve chromosome 6 as a partner. One copy of the patched gene (Ptc), which lies on chromosome 13, was deleted in all tested XRCC4/p53-deficient MBs in the context of translocations or interstitial deletions. In addition, Cyclin D2, a chromosome 6 gene, was amplified in a subset of tumors. Notably, amplification of Myc-family or Cyclin D2 genes and deletion of Ptc also have been observed in human MBs. We therefore conclude that, in neuronal cells of mice, the nonhomologous end-joining pathway plays a critical role in suppressing genomic instability that, in a p53-deficient background, routinely contributes to genesis of MBs with recurrent chromosomal alterations.


Subject(s)
DNA-Binding Proteins/metabolism , Medulloblastoma/metabolism , Translocation, Genetic/genetics , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/metabolism , Alleles , Animals , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Down-Regulation/genetics , Gene Amplification , Intermediate Filament Proteins/metabolism , Medulloblastoma/genetics , Medulloblastoma/pathology , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Nestin , Survival Rate , Time Factors , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
7.
Nat Immunol ; 5(12): 1275-81, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15531884

ABSTRACT

Immunoglobulin H class-switch recombination (CSR) occurs between switch regions and requires transcription and activation-induced cytidine deaminase (AID). Transcription through mammalian switch regions, because of their GC-rich composition, generates stable R-loops, which provide single-stranded DNA substrates for AID. However, we show here that the Xenopus laevis switch region S(mu), which is rich in AT and not prone to form R-loops, can functionally replace a mouse switch region to mediate CSR in vivo. X. laevis S(mu)-mediated CSR occurred mostly in a region of AGCT repeats targeted by the AID-replication protein A complex when transcribed in vitro. We propose that AGCT is a primordial CSR motif that targets AID through a non-R-loop mechanism involving an AID-replication protein A complex.


Subject(s)
Conserved Sequence/genetics , Evolution, Molecular , Immunoglobulin Class Switching/genetics , Immunoglobulin Switch Region/genetics , Recombination, Genetic/genetics , Animals , Base Sequence , Cytidine Deaminase/metabolism , DNA/metabolism , DNA-Binding Proteins/metabolism , Deamination , Hybridomas/immunology , Immunoglobulin Class Switching/immunology , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed/genetics , Replication Protein A , Sequence Alignment , Spleen/immunology , Xenopus laevis/genetics
8.
Mol Cell Neurosci ; 20(2): 271-82, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12093159

ABSTRACT

Lysophosphatidic acid (LPA) is a bioactive lysophospholipid that produces process retraction and cell rounding through its cognate receptors in neuroblastoma cell lines. Although the expression profile of LPA receptors in developing brains suggests a role for LPA in central nervous system (CNS) development, how LPA influences the morphology of postmitotic CNS neurons remains to be determined. Here we have investigated the effects of exogenous LPA on the morphology of young, postmitotic neurons in primary culture. When treated with LPA, these neurons responded by not only retracting processes but also producing retraction fiber "caps" characterized by fine actin filaments emanating from a dense core. Retraction fiber caps gradually vanished due to the outward spread of regrowing membranes along the fibers, suggesting a role for caps as scaffolds for regrowth of retracted processes. Furthermore, LPA also affects neuronal migration in vitro and in vivo. Taken together, these results implicate LPA as an extracellular lipid signal affecting process outgrowth and migration of early postmitotic neurons during development.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Differentiation/physiology , Cell Movement/physiology , Cerebral Cortex/embryology , Lysophospholipids/deficiency , Neurites/metabolism , Actin Cytoskeleton/drug effects , Animals , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Size/drug effects , Cell Size/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Cytochalasin D/pharmacology , Dose-Response Relationship, Drug , Female , Fetus , Gene Expression Regulation, Developmental/physiology , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Intracellular Membranes/ultrastructure , Lysophospholipids/pharmacology , Mice , Mice, Inbred BALB C , Mice, Knockout , Microtubule-Associated Proteins/drug effects , Microtubule-Associated Proteins/metabolism , Neurites/drug effects , Neurites/ultrastructure , Polymers/metabolism , Pregnancy , Pseudopodia/drug effects , Pseudopodia/metabolism , Pseudopodia/ultrastructure , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Tubulin/drug effects , Tubulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL