Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Cell ; 181(2): 236-249, 2020 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-32302568

RESUMO

Crucial transitions in cancer-including tumor initiation, local expansion, metastasis, and therapeutic resistance-involve complex interactions between cells within the dynamic tumor ecosystem. Transformative single-cell genomics technologies and spatial multiplex in situ methods now provide an opportunity to interrogate this complexity at unprecedented resolution. The Human Tumor Atlas Network (HTAN), part of the National Cancer Institute (NCI) Cancer Moonshot Initiative, will establish a clinical, experimental, computational, and organizational framework to generate informative and accessible three-dimensional atlases of cancer transitions for a diverse set of tumor types. This effort complements both ongoing efforts to map healthy organs and previous large-scale cancer genomics approaches focused on bulk sequencing at a single point in time. Generating single-cell, multiparametric, longitudinal atlases and integrating them with clinical outcomes should help identify novel predictive biomarkers and features as well as therapeutically relevant cell types, cell states, and cellular interactions across transitions. The resulting tumor atlases should have a profound impact on our understanding of cancer biology and have the potential to improve cancer detection, prevention, and therapeutic discovery for better precision-medicine treatments of cancer patients and those at risk for cancer.


Assuntos
Transformação Celular Neoplásica/metabolismo , Neoplasias/metabolismo , Microambiente Tumoral/fisiologia , Atlas como Assunto , Transformação Celular Neoplásica/patologia , Genômica/métodos , Humanos , Medicina de Precisão/métodos , Análise de Célula Única/métodos
2.
Mol Cell ; 79(5): 836-845.e7, 2020 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-32649884

RESUMO

The inactive X chromosome (Xi) is inherently susceptible to genomic aberrations. Replication stress (RS) has been proposed as an underlying cause, but the mechanisms that protect from Xi instability remain unknown. Here, we show that macroH2A1.2, an RS-protective histone variant enriched on the Xi, is required for Xi integrity and female survival. Mechanistically, macroH2A1.2 counteracts its structurally distinct and equally Xi-enriched alternative splice variant, macroH2A1.1. Comparative proteomics identified a role for macroH2A1.1 in alternative end joining (alt-EJ), which accounts for Xi anaphase defects in the absence of macroH2A1.2. Genomic instability was rescued by simultaneous depletion of macroH2A1.1 or alt-EJ factors, and mice deficient for both macroH2A1 variants harbor no overt female defects. Notably, macroH2A1 splice variant imbalance affected alt-EJ capacity also in tumor cells. Together, these findings identify macroH2A1 splicing as a modulator of genome maintenance that ensures Xi integrity and may, more broadly, predict DNA repair outcome in malignant cells.


Assuntos
Processamento Alternativo , Reparo do DNA , Epigênese Genética , Instabilidade Genômica , Histonas/fisiologia , Anáfase , Animais , Linhagem Celular , Instabilidade Cromossômica , Cromossomos Humanos X , Feminino , Histonas/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
3.
Nat Rev Genet ; 21(11): 651-670, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32764716

RESUMO

All organisms must safeguard the integrity of their DNA to avoid deleterious consequences of genome instability, which have been linked to human diseases such as autoimmune disorders, neurodegenerative diseases and cancer. Traditionally, genome maintenance has been viewed largely in terms of DNA-protein interactions. However, emerging evidence points to RNA as a key modulator of genome stability, with seemingly opposing roles in promoting chromosomal instability and protecting genome integrity. Unravelling the mechanistic and contextual basis of this duality will not only improve our understanding of the interfaces between RNA and the genome but will also provide important insights into how disrupted RNA metabolism contributes to disease origin, laying the foundation for targeted intervention.


Assuntos
Genoma Humano , Instabilidade Genômica , RNA/fisiologia , Adenosina/metabolismo , Animais , Reparo do DNA , Células Eucarióticas , Humanos , RNA Polimerase II/metabolismo , Processamento Pós-Transcricional do RNA , Retroelementos , Transcrição Gênica
4.
Mol Cell ; 69(1): 36-47.e7, 2018 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-29249653

RESUMO

Recent integrative epigenome analyses highlight the importance of functionally distinct chromatin states for accurate cell function. How these states are established and maintained is a matter of intense investigation. Here, we present evidence for DNA damage as an unexpected means to shape a protective chromatin environment at regions of recurrent replication stress (RS). Upon aberrant fork stalling, DNA damage signaling and concomitant H2AX phosphorylation coordinate the FACT-dependent deposition of macroH2A1.2, a histone variant that promotes DNA repair by homologous recombination (HR). MacroH2A1.2, in turn, facilitates the accumulation of the tumor suppressor and HR effector BRCA1 at replication forks to protect from RS-induced DNA damage. Consequently, replicating primary cells steadily accrue macroH2A1.2 at fragile regions, whereas macroH2A1.2 loss in these cells triggers DNA damage signaling-dependent senescence, a hallmark of RS. Altogether, our findings demonstrate that recurrent DNA damage contributes to the chromatin landscape to ensure the epigenomic integrity of dividing cells.


Assuntos
Carcinogênese/genética , Cromatina/genética , Dano ao DNA/genética , Reparo do DNA/genética , Replicação do DNA/genética , Histonas/genética , Recombinação Homóloga/genética , Proteína BRCA1/metabolismo , Divisão Celular/genética , Células Cultivadas , Senescência Celular/genética , Instabilidade Genômica/fisiologia , Humanos , Transdução de Sinais/genética
5.
Semin Cell Dev Biol ; 135: 59-72, 2023 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35331626

RESUMO

Histone variants represent chromatin components that diversify the structure and function of the genome. The variants of H2A, primarily H2A.X, H2A.Z and macroH2A, are well-established participants in DNA damage response (DDR) pathways, which function to protect the integrity of the genome. Through their deposition, post-translational modifications and unique protein interaction networks, these variants guard DNA from endogenous threats including replication stress and genome fragility as well as from DNA lesions inflicted by exogenous sources. A growing body of work is now providing a clearer picture on the involvement and mechanistic basis of H2A variant contribution to genome integrity. Beyond their well-documented role in gene regulation, we review here how histone H2A variants promote genome stability and how alterations in these pathways contribute to human diseases including cancer.


Assuntos
Cromatina , Histonas , Humanos , Histonas/genética , Histonas/metabolismo , Cromatina/genética , Genoma , Processamento de Proteína Pós-Traducional/genética , DNA/genética
6.
Cell ; 135(5): 907-18, 2008 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-19041753

RESUMO

Genomic instability and alterations in gene expression are hallmarks of eukaryotic aging. The yeast histone deacetylase Sir2 silences transcription and stabilizes repetitive DNA, but during aging or in response to a DNA break, the Sir complex relocalizes to sites of genomic instability, resulting in the desilencing of genes that cause sterility, a characteristic of yeast aging. Using embryonic stem cells, we show that mammalian Sir2, SIRT1, represses repetitive DNA and a functionally diverse set of genes across the mouse genome. In response to DNA damage, SIRT1 dissociates from these loci and relocalizes to DNA breaks to promote repair, resulting in transcriptional changes that parallel those in the aging mouse brain. Increased SIRT1 expression promotes survival in a mouse model of genomic instability and suppresses age-dependent transcriptional changes. Thus, DNA damage-induced redistribution of SIRT1 and other chromatin-modifying proteins may be a conserved mechanism of aging in eukaryotes.


Assuntos
Envelhecimento/genética , Cromatina/metabolismo , Instabilidade Genômica , Sirtuínas/genética , Animais , Encéfalo/metabolismo , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Células-Tronco Embrionárias , Técnicas de Inativação de Genes , Humanos , Linfoma/metabolismo , Camundongos , Dados de Sequência Molecular , Estresse Oxidativo , Sirtuína 1 , Organismos Livres de Patógenos Específicos , Neoplasias do Timo/metabolismo , Leveduras/citologia , Leveduras/metabolismo
7.
Chromosoma ; 133(1): 1-3, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38355990
8.
Nat Rev Mol Cell Biol ; 8(9): 692-702, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17700626

RESUMO

Eukaryotes come in many shapes and sizes, yet one thing that they all seem to share is a decline in vitality and health over time--a process known as ageing. If there are conserved causes of ageing, they may be traced back to common biological structures that are inherently difficult to maintain throughout life. One such structure is chromatin, the DNA-protein complex that stabilizes the genome and dictates gene expression. Studies in the budding yeast Saccharomyces cerevisiae have pointed to chromatin reorganization as a main contributor to ageing in that species, which raises the possibility that similar processes underlie ageing in more complex organisms.


Assuntos
Envelhecimento/fisiologia , Cromatina/fisiologia , Instabilidade Genômica , Animais , Expressão Gênica , Humanos , Saccharomyces cerevisiae/fisiologia
9.
Nucleic Acids Res ; 44(7): e64, 2016 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-26687720

RESUMO

DNA double-strand breaks (DSBs) and their repair can cause extensive epigenetic changes. As a result, DSBs have been proposed to promote transcriptional and, ultimately, physiological dysfunction via both cell-intrinsic and cell-non-autonomous pathways. Studying the consequences of DSBs in higher organisms has, however, been hindered by a scarcity of tools for controlled DSB induction. Here, we describe a mouse model that allows for both tissue-specific and temporally controlled DSB formation at ∼140 defined genomic loci. Using this model, we show that DSBs promote a DNA damage signaling-dependent decrease in gene expression in primary cells specifically at break-bearing genes, which is reversed upon DSB repair. Importantly, we demonstrate that restoration of gene expression can occur independently of cell cycle progression, underlining its relevance for normal tissue maintenance. Consistent with this, we observe no evidence for persistent transcriptional repression in response to a multi-day course of continuous DSB formation and repair in mouse lymphocytes in vivo Together, our findings reveal an unexpected capacity of primary cells to maintain transcriptome integrity in response to DSBs, pointing to a limited role for DNA damage as a mediator of cell-autonomous epigenetic dysfunction.


Assuntos
Quebras de DNA de Cadeia Dupla , Transcriptoma , Animais , Células Cultivadas , Endodesoxirribonucleases , Loci Gênicos , Camundongos , Camundongos Transgênicos , Transdução de Sinais
10.
EMBO Rep ; 16(11): 1520-34, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26412854

RESUMO

Long non-coding RNAs (lncRNAs) are important players in diverse biological processes. Upon DNA damage, cells activate a complex signaling cascade referred to as the DNA damage response (DDR). Using a microarray screen, we identify here a novel lncRNA, DDSR1 (DNA damage-sensitive RNA1), which is induced upon DNA damage. DDSR1 induction is triggered in an ATM-NF-κB pathway-dependent manner by several DNA double-strand break (DSB) agents. Loss of DDSR1 impairs cell proliferation and DDR signaling and reduces DNA repair capacity by homologous recombination (HR). The HR defect in the absence of DDSR1 is marked by aberrant accumulation of BRCA1 and RAP80 at DSB sites. In line with a role in regulating HR, DDSR1 interacts with BRCA1 and hnRNPUL1, an RNA-binding protein involved in DNA end resection. Our results suggest a role for the lncRNA DDSR1 in modulating DNA repair by HR.


Assuntos
Proteína BRCA1/metabolismo , Dano ao DNA , Recombinação Homóloga , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Proliferação de Células , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Regulação da Expressão Gênica , Genes BRCA1 , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Humanos , Análise em Microsséries , NF-kappa B/metabolismo , Proteínas Nucleares/metabolismo , RNA Longo não Codificante/isolamento & purificação , Transdução de Sinais , Fatores de Transcrição/metabolismo
11.
Nature ; 479(7371): 74-9, 2011 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-21964334

RESUMO

Alternative splicing of pre-messenger RNA is a key feature of transcriptome expansion in eukaryotic cells, yet its regulation is poorly understood. Spliceosome assembly occurs co-transcriptionally, raising the possibility that DNA structure may directly influence alternative splicing. Supporting such an association, recent reports have identified distinct histone methylation patterns, elevated nucleosome occupancy and enriched DNA methylation at exons relative to introns. Moreover, the rate of transcription elongation has been linked to alternative splicing. Here we provide the first evidence that a DNA-binding protein, CCCTC-binding factor (CTCF), can promote inclusion of weak upstream exons by mediating local RNA polymerase II pausing both in a mammalian model system for alternative splicing, CD45, and genome-wide. We further show that CTCF binding to CD45 exon 5 is inhibited by DNA methylation, leading to reciprocal effects on exon 5 inclusion. These findings provide a mechanistic basis for developmental regulation of splicing outcome through heritable epigenetic marks.


Assuntos
Processamento Alternativo , Metilação de DNA , RNA Polimerase II/metabolismo , Proteínas Repressoras/metabolismo , Transcrição Gênica , Animais , Fator de Ligação a CCCTC , Linhagem Celular , Células Cultivadas , Epigênese Genética , Éxons/genética , Genoma Humano/genética , Humanos , Antígenos Comuns de Leucócito/genética , Camundongos , Ligação Proteica , Sítios de Splice de RNA/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Saccharomyces cerevisiae/enzimologia , Transcrição Gênica/genética
12.
Cancer Res ; 84(6): 793-795, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38486481

RESUMO

High-grade serous ovarian carcinoma (HGSOC) is the deadliest subtype of ovarian cancer. While PARP inhibitors (PARPi) have transformed the care of advanced HGSOC, PARPi resistance poses a major limitation to their clinical utility. DNA damage checkpoint signaling via ATR kinase can counteract PARPi-induced replication stress, making ATR an attractive therapeutic target in PARPi-resistant tumors. However, ATR inhibitor (ATRi) efficacy in the clinic is low, emphasizing the need for suitable combination treatments. In this issue of Cancer Research, Huang and colleagues uncovered cytotoxic synergism between inhibition of the PI3K/AKT pathway and ATR based on high-throughput screening for ATRi drug combinations in PARPi-resistant HGSOC cells. Dual inhibition of ATR and AKT resulted in aberrant replication stress and cell death, which was attributed in part to impaired resolution of replication-stalling RNA:DNA hybrids (R loops). The authors identified the DNA/RNA helicase DHX9 as a clinically relevant candidate effector of R loop resolution in HGSOC. AKT interacted with and recruited DHX9 to R loops, where it complemented ATR in facilitating their removal. Underlining the therapeutic potential relevance of these findings, combined inhibition of ATR and AKT caused near complete tumor regression in HGSOC xenograft models, and elevated AKT/DHX9 levels correlated with poor survival in patients with HGSOC. Of note, the genotoxic consequences of dual ATRi/AKTi treatment extended beyond PARPi-resistant tumors and are likely to affect genome integrity beyond R loops. The work by Huang and colleagues thus provides compelling rationale for the exploration of combined targeting of the AKT and ATR pathways as a potentially broadly applicable treatment of advanced HGSOC. See related article by Huang et al., p. 887.


Assuntos
Neoplasias Ovarianas , Estruturas R-Loop , Humanos , Feminino , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Linhagem Celular Tumoral , Neoplasias Ovarianas/patologia , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Inibidores de Proteínas Quinases/farmacologia , DNA
14.
Biochim Biophys Acta ; 1819(7): 811-9, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22285574

RESUMO

DNA double-strand breaks (DSBs) occur in the context of a highly organized chromatin environment and are, thus, a significant threat to the epigenomic integrity of eukaryotic cells. Changes in break-proximal chromatin structure are thought to be a prerequisite for efficient DNA repair and may help protect the structural integrity of the nucleus. Unlike most bona fide DNA repair factors, chromatin influences the repair process at several levels: the existing chromatin context at the site of damage directly affects the access and kinetics of the repair machinery; DSB induced chromatin modifications influence the choice of repair factors, thereby modulating repair outcome; lastly, DNA damage can have a significant impact on chromatin beyond the site of damage. We will discuss recent findings that highlight both the complexity and importance of dynamic and tightly orchestrated chromatin reorganization to ensure efficient DSB repair and nuclear integrity. This article is part of a Special Issue entitled: Chromatin in time and space.


Assuntos
Montagem e Desmontagem da Cromatina , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Animais , Núcleo Celular/metabolismo , Senescência Celular , Replicação do DNA , Heterocromatina/genética , Heterocromatina/metabolismo , Histonas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Nucleossomos/genética , Nucleossomos/metabolismo , Processamento de Proteína Pós-Traducional , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
15.
Cancer Res ; 83(16): 2750-2762, 2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-37306706

RESUMO

ARID1A is a subunit of SWI/SNF chromatin remodeling complexes and is mutated in many types of human cancers, especially those derived from endometrial epithelium, including ovarian and uterine clear cell carcinoma (CCC) and endometrioid carcinoma (EMCA). Loss-of-function mutations in ARID1A alter epigenetic regulation of transcription, cell-cycle checkpoint control, and DNA damage repair. We report here that mammalian cells with ARID1A deficiency harbor accumulated DNA base lesions and increased abasic (AP) sites, products of glycosylase in the first step of base excision repair (BER). ARID1A mutations also delayed recruitment kinetics of BER long-patch repair effectors. Although ARID1A-deficient tumors were not sensitive to monotherapy with DNA-methylating temozolomide (TMZ), the combination of TMZ with PARP inhibitors (PARPi) potently elicited double-strand DNA breaks, replication stress, and replication fork instability in ARID1A-deficient cells. The TMZ and PARPi combination also significantly delayed in vivo growth of ovarian tumor xenografts carrying ARID1A mutations and induced apoptosis and replication stress in xenograft tumors. Together, these findings identified a synthetic lethal strategy to enhance the response of ARID1A-mutated cancers to PARP inhibition, which warrants further experimental exploration and clinical trial validation. SIGNIFICANCE: The combination of temozolomide and PARP inhibitor exploits the specific DNA damage repair status of ARID1A-inactivated ovarian cancers to suppress tumor growth.


Assuntos
Antineoplásicos , Neoplasias Ovarianas , Animais , Feminino , Humanos , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Epigênese Genética , Antineoplásicos/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Mamíferos , Proteínas de Ligação a DNA/genética , Fatores de Transcrição/genética
16.
J Exp Med ; 201(4): 603-14, 2005 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-15728238

RESUMO

The pre-T cell receptor (TCR) is expressed early during T cell development and imposes a tight selection for differentiating T cell progenitors. Pre-TCR-expressing cells are selected to survive and differentiate further, whereas pre-TCR(-) cells are "negatively" selected to die. The mechanisms of pre-TCR-mediated survival are poorly understood. Here, we describe the induction of the antiapoptotic gene BCL2A1 (A1) as a potential mechanism regulating inhibition of pre-T cell death. We characterize in detail the signaling pathway involved in A1 induction and show that A1 expression can induce pre-T cell survival by inhibiting activation of caspase-3. Moreover, we show that in vitro "knockdown" of A1 expression can compromise survival even in the presence of a functional pre-TCR. Finally, we suggest that pre-TCR-induced A1 overexpression can contribute to T cell leukemia in both mice and humans.


Assuntos
Genes bcl-2/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/fisiologia , Animais , Apoptose , Caspase 3 , Inibidores de Caspase , Linhagem Celular , Sobrevivência Celular , Regulação da Expressão Gênica , Humanos , Leucemia-Linfoma de Células T do Adulto/etiologia , Leucemia-Linfoma de Células T do Adulto/metabolismo , Glicoproteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Antígenos de Histocompatibilidade Menor , NF-kappa B/metabolismo , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Receptores de Antígenos de Linfócitos T alfa-beta , Transdução de Sinais , Linfócitos T/metabolismo , Fosfolipases Tipo C/metabolismo
17.
Front Genet ; 12: 746380, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34745220

RESUMO

Almost 25 years ago, the phosphorylation of a chromatin component, histone H2AX, was discovered as an integral part of the DNA damage response in eukaryotes. Much has been learned since then about the control of DNA repair in the context of chromatin. Recent technical and computational advances in imaging, biophysics and deep sequencing have led to unprecedented insight into nuclear organization, highlighting the impact of three-dimensional (3D) chromatin structure and nuclear topology on DNA repair. In this review, we will describe how DNA repair processes have adjusted to and in many cases adopted these organizational features to ensure accurate lesion repair. We focus on new findings that highlight the importance of chromatin context, topologically associated domains, phase separation and DNA break mobility for the establishment of repair-conducive nuclear environments. Finally, we address the consequences of aberrant 3D genome maintenance for genome instability and disease.

18.
J Exp Med ; 197(9): 1165-72, 2003 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-12719477

RESUMO

Immunoglobulin light chain (IgL) rearrangements occur more frequently at Ig kappa than at Ig lambda. Previous results suggested that the unrearranged Ig kappa locus negatively regulates Ig lambda transcription and/or rearrangement. Here, we demonstrate that expression of a VJ lambda 1-joint inserted into its physiological position in the Ig lambda locus is independent of Ig kappa rearrangements. Expression of the inserted VJ lambda 1 gene segment is developmentally controlled like that of a VJ kappa-joint inserted into the Ig kappa locus and furthermore coincides developmentally with the occurrence of Ig kappa rearrangements in wild-type mice. We conclude that developmentally controlled transcription of a gene rearrangement in the Ig lambda locus occurs in the presence of an unrearranged Ig kappa locus and is therefore not negatively regulated by the latter. Our data also indicate light chain editing in approximately 30% of lambda 1 expressing B cell progenitors.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Rearranjo Gênico , Cadeias kappa de Imunoglobulina/genética , Cadeias lambda de Imunoglobulina/genética , Animais , Sequência de Bases , Primers do DNA , Células Germinativas , Camundongos , Camundongos Transgênicos
19.
Nat Struct Mol Biol ; 26(3): 213-219, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30833786

RESUMO

The growth of telomerase-deficient cancers depends on the alternative lengthening of telomeres (ALT), a homology-directed telomere-maintenance pathway. ALT telomeres exhibit a unique chromatin environment and generally lack the nucleosome remodeler ATRX, pointing to an epigenetic basis for ALT. Recently, we identified a protective role for the ATRX-interacting macroH2A1.2 histone variant during homologous recombination and replication stress (RS). Consistent with an inherent susceptibility to RS, we show that human ALT telomeres are highly enriched for macroH2A1.2. However, in contrast to ATRX-proficient cells, ALT telomeres transiently lose macroH2A1.2 during acute RS to facilitate DNA double-strand break (DSB) formation, a process that is almost completely prevented by ectopic ATRX expression. Telomeric macroH2A1.2 is re-deposited in a DNA damage response (DDR)-dependent manner to promote homologous recombination-associated ALT pathways. Our findings thus identify the dynamic exchange of macroH2A1.2 on chromatin as an epigenetic link among ATRX loss, RS-induced DDR initiation and telomere maintenance via homologous recombination.


Assuntos
Cromatina/metabolismo , Reparo do DNA/genética , Histonas/genética , Recombinação Homóloga/genética , Homeostase do Telômero/genética , Proteína Nuclear Ligada ao X/genética , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , Células HEK293 , Células HeLa , Humanos , Telomerase/metabolismo
20.
Mol Cell Biol ; 25(10): 3896-905, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15870264

RESUMO

RNA interference (RNAi) is a naturally occurring posttranscriptional gene-silencing mechanism that has been adapted as a genetic tool for loss-of-function studies of a variety of organisms. It is more widely applicable than classical gene targeting and allows for the simultaneous inactivation of several homologous genes with a single transgene. Recently, RNAi has been used for conditional and conventional gene inactivation in mice. Unlike gene targeting, RNAi is a dynamic process, and its efficiency may vary both between cell types and throughout development. Here we demonstrate that RNAi can be used to target three separately encoded isoforms of the bcl-2 family gene bfl-1/A1 in a conditional manner in mice. The extent of gene inactivation varies between different cell types and is least efficient in mature lymphocytes. Our data suggest that RNAi is affected by factors beyond small interfering RNA-mRNA stoichiometry.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Sistema Hematopoético/metabolismo , Regiões Promotoras Genéticas/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , RNA Nuclear Pequeno/genética , Transgenes/genética , Animais , Diferenciação Celular , Células Cultivadas , Integrases/genética , Integrases/metabolismo , Camundongos , Especificidade de Órgãos , Fatores de Iniciação de Peptídeos/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Linfócitos T/citologia , Linfócitos T/metabolismo , Timo/citologia , Timo/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA