Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
DNA Repair (Amst) ; 140: 103702, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38878564

RESUMO

The maintenance of genome integrity by DNA damage response machineries is key to protect cells against pathological development. In cell nuclei, these genome maintenance machineries operate in the context of chromatin, where the DNA wraps around histone proteins. Here, we review recent findings illustrating how the chromatin substrate modulates genome maintenance mechanisms, focusing on the regulatory role of histone variants and post-translational modifications. In particular, we discuss how the pre-existing chromatin landscape impacts DNA damage formation and guides DNA repair pathway choice, and how DNA damage-induced chromatin alterations control DNA damage signaling and repair, and DNA damage segregation through cell divisions. We also highlight that pathological alterations of histone proteins may trigger genome instability by impairing chromosome segregation and DNA repair, thus defining new oncogenic mechanisms and opening up therapeutic options.


Assuntos
Cromatina , Dano ao DNA , Reparo do DNA , Instabilidade Genômica , Histonas , Processamento de Proteína Pós-Traducional , Humanos , Cromatina/metabolismo , Histonas/metabolismo , Animais , Segregação de Cromossomos
2.
Nucleic Acids Res ; 52(5): 2372-2388, 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38214234

RESUMO

Pediatric high-grade gliomas (pHGG) are devastating and incurable brain tumors with recurrent mutations in histone H3.3. These mutations promote oncogenesis by dysregulating gene expression through alterations of histone modifications. We identify aberrant DNA repair as an independent mechanism, which fosters genome instability in H3.3 mutant pHGG, and opens new therapeutic options. The two most frequent H3.3 mutations in pHGG, K27M and G34R, drive aberrant repair of replication-associated damage by non-homologous end joining (NHEJ). Aberrant NHEJ is mediated by the DNA repair enzyme polynucleotide kinase 3'-phosphatase (PNKP), which shows increased association with mutant H3.3 at damaged replication forks. PNKP sustains the proliferation of cells bearing H3.3 mutations, thus conferring a molecular vulnerability, specific to mutant cells, with potential for therapeutic targeting.


Assuntos
Neoplasias Encefálicas , Glioma , Histonas , Criança , Humanos , Neoplasias Encefálicas/patologia , Reparo do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Glioma/patologia , Histonas/genética , Histonas/metabolismo , Mutação , Fosfotransferases (Aceptor do Grupo Álcool)/genética
3.
Nat Commun ; 12(1): 3835, 2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34158510

RESUMO

Transcription restart after a genotoxic challenge is a fundamental yet poorly understood process. Here, we dissect the interplay between transcription and chromatin restoration after DNA damage by focusing on the human histone chaperone complex HIRA, which is required for transcription recovery post UV. We demonstrate that HIRA is recruited to UV-damaged chromatin via the ubiquitin-dependent segregase VCP to deposit new H3.3 histones. However, this local activity of HIRA is dispensable for transcription recovery. Instead, we reveal a genome-wide function of HIRA in transcription restart that is independent of new H3.3 and not restricted to UV-damaged loci. HIRA coordinates with ASF1B to control transcription restart by two independent pathways: by stabilising the associated subunit UBN2 and by reducing the expression of the transcription repressor ATF3. Thus, HIRA primes UV-damaged chromatin for transcription restart at least in part by relieving transcription inhibition rather than by depositing new H3.3 as an activating bookmark.


Assuntos
Proteínas de Ciclo Celular/genética , Dano ao DNA , Chaperonas de Histonas/genética , Transdução de Sinais/genética , Fatores de Transcrição/genética , Transcrição Gênica , Fator 3 Ativador da Transcrição/genética , Fator 3 Ativador da Transcrição/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Cromatina/genética , Cromatina/metabolismo , Cromatina/efeitos da radiação , Reparo do DNA , Células HeLa , Chaperonas de Histonas/metabolismo , Histonas/metabolismo , Humanos , Fatores de Transcrição/metabolismo , Raios Ultravioleta , Proteína com Valosina/genética , Proteína com Valosina/metabolismo
4.
Nat Commun ; 12(1): 3127, 2021 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-34035299

RESUMO

Cornelia de Lange syndrome is a multisystem developmental disorder typically caused by mutations in the gene encoding the cohesin loader NIPBL. The associated phenotype is generally assumed to be the consequence of aberrant transcriptional regulation. Recently, we identified a missense mutation in BRD4 associated with a Cornelia de Lange-like syndrome that reduces BRD4 binding to acetylated histones. Here we show that, although this mutation reduces BRD4-occupancy at enhancers it does not affect transcription of the pluripotency network in mouse embryonic stem cells. Rather, it delays the cell cycle, increases DNA damage signalling, and perturbs regulation of DNA repair in mutant cells. This uncovers a role for BRD4 in DNA repair pathway choice. Furthermore, we find evidence of a similar increase in DNA damage signalling in cells derived from NIPBL-deficient individuals, suggesting that defective DNA damage signalling and repair is also a feature of typical Cornelia de Lange syndrome.


Assuntos
Dano ao DNA , Reparo do DNA , Síndrome de Cornélia de Lange/genética , Mutação , Animais , Proteínas de Ciclo Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Predisposição Genética para Doença/genética , Humanos , Camundongos , RNA-Seq/métodos , Transdução de Sinais/genética , Fatores de Transcrição/genética
5.
Nat Commun ; 12(1): 2428, 2021 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-33893291

RESUMO

Heterochromatin is a critical chromatin compartment, whose integrity governs genome stability and cell fate transitions. How heterochromatin features, including higher-order chromatin folding and histone modifications associated with transcriptional silencing, are maintained following a genotoxic stress challenge is unknown. Here, we establish a system for targeting UV damage to pericentric heterochromatin in mammalian cells and for tracking the heterochromatin response to UV in real time. We uncover profound heterochromatin compaction changes during repair, orchestrated by the UV damage sensor DDB2, which stimulates linker histone displacement from chromatin. Despite massive heterochromatin unfolding, heterochromatin-specific histone modifications and transcriptional silencing are maintained. We unveil a central role for the methyltransferase SETDB1 in the maintenance of heterochromatic histone marks after UV. SETDB1 coordinates histone methylation with new histone deposition in damaged heterochromatin, thus protecting cells from genome instability. Our data shed light on fundamental molecular mechanisms safeguarding higher-order chromatin integrity following DNA damage.


Assuntos
Dano ao DNA , Reparo do DNA , DNA/genética , Heterocromatina/genética , Animais , Linhagem Celular Tumoral , Montagem e Desmontagem da Cromatina/genética , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Heterocromatina/efeitos da radiação , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Humanos , Células MCF-7 , Metilação , Camundongos , Células NIH 3T3 , Raios Ultravioleta
6.
Cells ; 9(11)2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-33167489

RESUMO

Chromatin integrity is key for cell homeostasis and for preventing pathological development. Alterations in core chromatin components, histone proteins, recently came into the spotlight through the discovery of their driving role in cancer. Building on these findings, in this review, we discuss how histone variants and their associated chaperones safeguard genome stability and protect against tumorigenesis. Accumulating evidence supports the contribution of histone variants and their chaperones to the maintenance of chromosomal integrity and to various steps of the DNA damage response, including damaged chromatin dynamics, DNA damage repair, and damage-dependent transcription regulation. We present our current knowledge on these topics and review recent advances in deciphering how alterations in histone variant sequence, expression, and deposition into chromatin fuel oncogenic transformation by impacting cell proliferation and cell fate transitions. We also highlight open questions and upcoming challenges in this rapidly growing field.


Assuntos
Instabilidade Genômica , Histonas/metabolismo , Animais , Centrômero/metabolismo , Cromossomos/genética , Dano ao DNA , Humanos , Isoformas de Proteínas/metabolismo
7.
Mol Cell ; 72(5): 888-901.e7, 2018 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-30344095

RESUMO

Safeguarding cell function and identity following a genotoxic stress challenge entails a tight coordination of DNA damage signaling and repair with chromatin maintenance. How this coordination is achieved and with what impact on chromatin integrity remains elusive. Here, we address these questions by investigating the mechanisms governing the distribution in mammalian chromatin of the histone variant H2A.X, a central player in damage signaling. We reveal that H2A.X is deposited de novo at sites of DNA damage in a repair-coupled manner, whereas the H2A.Z variant is evicted, thus reshaping the chromatin landscape at repair sites. Our mechanistic studies further identify the histone chaperone FACT (facilitates chromatin transcription) as responsible for the deposition of newly synthesized H2A.X. Functionally, we demonstrate that FACT potentiates H2A.X-dependent signaling of DNA damage. We propose that new H2A.X deposition in chromatin reflects DNA damage experience and may help tailor DNA damage signaling to repair progression.


Assuntos
Reparo do DNA , Proteínas de Ligação a DNA/genética , DNA/genética , Proteínas de Grupo de Alta Mobilidade/genética , Histonas/genética , Fatores de Elongação da Transcrição/genética , Alfa-Amanitina/farmacologia , Animais , Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Linhagem Celular Tumoral , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , DNA/metabolismo , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Proteínas de Grupo de Alta Mobilidade/metabolismo , Histonas/metabolismo , Humanos , Camundongos , Morfolinas/farmacologia , Células NIH 3T3 , Nucleossomos/química , Nucleossomos/efeitos dos fármacos , Nucleossomos/metabolismo , Venenos/farmacologia , Pirimidinas/farmacologia , Pironas/farmacologia , Transdução de Sinais , Fatores de Elongação da Transcrição/metabolismo
8.
Methods Mol Biol ; 1832: 243-253, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30073531

RESUMO

In eukaryotic cell nuclei, all DNA transactions, including DNA damage repair, take place on a chromatin substrate, the integrity of which is central to gene expression programs and cell identity. However, substantial chromatin rearrangements accompany the repair response, culminating in the deposition of new histones. How the original epigenetic information conveyed by chromatin may be preserved in this context is a burning question. Elucidating the fate of parental histones, which characterize the pre-damage chromatin state, is a key step forward in deciphering the mechanisms that safeguard epigenome stability. Here, we present an in vivo approach for tracking parental histone H3 variant dynamics in real time after UVC laser-induced damage in human cells.


Assuntos
Cromatina/efeitos da radiação , Dano ao DNA , Histonas/metabolismo , Imageamento Tridimensional/métodos , Raios Ultravioleta , Linhagem Celular Tumoral , Humanos , Isoformas de Proteínas/metabolismo , Coloração e Rotulagem
9.
Mol Cell ; 64(1): 65-78, 2016 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-27642047

RESUMO

Chromatin integrity is critical for cell function and identity but is challenged by DNA damage. To understand how chromatin architecture and the information that it conveys are preserved or altered following genotoxic stress, we established a system for real-time tracking of parental histones, which characterize the pre-damage chromatin state. Focusing on histone H3 dynamics after local UVC irradiation in human cells, we demonstrate that parental histones rapidly redistribute around damaged regions by a dual mechanism combining chromatin opening and histone mobilization on chromatin. Importantly, parental histones almost entirely recover and mix with new histones in repairing chromatin. Our data further define a close coordination of parental histone dynamics with DNA repair progression through the damage sensor DDB2 (DNA damage-binding protein 2). We speculate that this mechanism may contribute to maintaining a memory of the original chromatin landscape and may help preserve epigenome stability in response to DNA damage.


Assuntos
Cromatina/efeitos da radiação , Reparo do DNA , Imunofluorescência/métodos , Histonas/genética , Osteoblastos/efeitos da radiação , Linhagem Celular Tumoral , Cromatina/química , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina , Dano ao DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Instabilidade Genômica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Histonas/antagonistas & inibidores , Histonas/metabolismo , Humanos , Osteoblastos/citologia , Osteoblastos/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Raios Ultravioleta
10.
Cell ; 155(1): 94-106, 2013 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-24074863

RESUMO

Understanding how to recover fully functional and transcriptionally active chromatin when its integrity has been challenged by genotoxic stress is a critical issue. Here, by investigating how chromatin dynamics regulate transcriptional activity in response to DNA damage in human cells, we identify a pathway involving the histone chaperone histone regulator A (HIRA) to promote transcription restart after UVC damage. Our mechanistic studies reveal that HIRA accumulates at sites of UVC irradiation upon detection of DNA damage prior to repair and deposits newly synthesized H3.3 histones. This local action of HIRA depends on ubiquitylation events associated with damage recognition. Furthermore, we demonstrate that the early and transient function of HIRA in response to DNA damage primes chromatin for later reactivation of transcription. We propose that HIRA-dependent histone deposition serves as a chromatin bookmarking system to facilitate transcription recovery after genotoxic stress.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Cromatina , Dano ao DNA/efeitos da radiação , Chaperonas de Histonas/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Linhagem Celular Tumoral , Reparo do DNA , Células HeLa , Histonas/metabolismo , Humanos , Ubiquitinação , Raios Ultravioleta
12.
J Cell Biol ; 193(1): 97-108, 2011 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-21444690

RESUMO

Chromosomal deletions and rearrangements in tumors are often associated with common fragile sites, which are specific genomic loci prone to gaps and breaks in metaphase chromosomes. Common fragile sites appear to arise through incomplete DNA replication because they are induced after partial replication inhibition by agents such as aphidicolin. Here, we show that in G1 cells, large nuclear bodies arise that contain p53 binding protein 1 (53BP1), phosphorylated H2AX (γH2AX), and mediator of DNA damage checkpoint 1 (MDC1), as well as components of previously characterized OPT (Oct-1, PTF, transcription) domains. Notably, we find that incubating cells with low aphidicolin doses increases the incidence and number of 53BP1-OPT domains in G1 cells, and by chromatin immunoprecipitation and massively parallel sequencing analysis of γH2AX, we demonstrate that OPT domains are enriched at common fragile sites. These findings invoke a model wherein incomplete DNA synthesis during S phase leads to a DNA damage response and formation of 53BP1-OPT domains in the subsequent G1.


Assuntos
Replicação do DNA/genética , Fase G1/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Fator 1 de Transcrição de Octâmero/genética , Fatores de Transcrição/genética , Transcrição Gênica/genética , Células Cultivadas , Humanos , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
13.
Histopathology ; 57(5): 716-24, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21083601

RESUMO

AIMS: Chromatin assembly factor-1 (CAF-1), whose function is critical for maintaining chromatin stability during DNA replication and repair, has been identified as a proliferation marker in breast cancer. The aim was to investigate CAF-1 as a proliferation marker in a wide variety of solid tumours, and to assess its potential value in predicting clinical outcome. METHODS AND RESULTS: Using immunocytochemistry on paraffin-embedded tissue sections, the CAF-1 labelling index was compared with known proliferation markers Ki-67 and minichromosome maintenance (MCM), and its association with clinicopathological data and patients' outcome analysed. CAF-1 expression showed a strong positive correlation with Ki-67, used routinely to detect proliferating cells, while it generally displayed weaker correlations with MCM markers, known to label cells with replicative potential. CAF-1 expression was associated significantly with histological grade in breast, cervical, endometrial and renal cell carcinomas, and with disease stage in endometrial and renal carcinomas. Furthermore, high expression of CAF-1 was an independent predictor of adverse clinical outcome in renal, endometrial and cervical carcinomas. CONCLUSIONS: CAF-1 is a proliferation marker in various malignant tumours with prognostic value in renal, endometrial and cervical carcinomas, which supports the value of CAF-1 as a clinical marker of cancer progression.


Assuntos
Biomarcadores Tumorais/metabolismo , Fator 1 de Modelagem da Cromatina/metabolismo , Idoso , Proliferação de Células , Fator 1 de Modelagem da Cromatina/genética , Replicação do DNA , Feminino , Humanos , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Masculino , Pessoa de Meia-Idade , Proteína 1 de Manutenção de Minicromossomo/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Prognóstico
14.
EMBO J ; 29(18): 3130-9, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20693977

RESUMO

The chromatin remodelling factor chromodomain helicase DNA-binding protein 4 (CHD4) is a catalytic subunit of the NuRD transcriptional repressor complex. Here, we reveal novel functions for CHD4 in the DNA-damage response (DDR) and cell-cycle control. We show that CHD4 mediates rapid poly(ADP-ribose)-dependent recruitment of the NuRD complex to DNA-damage sites, and we identify CHD4 as a phosphorylation target for the apical DDR kinase ataxia-telangiectasia mutated. Functionally, we show that CHD4 promotes repair of DNA double-strand breaks and cell survival after DNA damage. In addition, we show that CHD4 acts as an important regulator of the G1/S cell-cycle transition by controlling p53 deacetylation. These results provide new insights into how the chromatin remodelling complex NuRD contributes to maintaining genome stability.


Assuntos
Autoantígenos/metabolismo , Ciclo Celular/fisiologia , Montagem e Desmontagem da Cromatina , Dano ao DNA , DNA Helicases/metabolismo , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Autoantígenos/genética , Western Blotting , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , DNA Helicases/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Imunofluorescência , Histonas/fisiologia , Humanos , Imunoprecipitação , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/genética , Camundongos , Camundongos Knockout , Fosforilação , Poli Adenosina Difosfato Ribose/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Supressora de Tumor p53/fisiologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
15.
Nat Struct Mol Biol ; 17(9): 1144-51, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20802485

RESUMO

DNA double-strand break (DSB) repair occurs within chromatin and can be modulated by chromatin-modifying enzymes. Here we identify the related human histone deacetylases HDAC1 and HDAC2 as two participants in the DNA-damage response. We show that acetylation of histone H3 Lys56 (H3K56) was regulated by HDAC1 and HDAC2 and that HDAC1 and HDAC2 were rapidly recruited to DNA-damage sites to promote hypoacetylation of H3K56. Furthermore, HDAC1- and 2-depleted cells were hypersensitive to DNA-damaging agents and showed sustained DNA-damage signaling, phenotypes that reflect defective DSB repair, particularly by nonhomologous end-joining (NHEJ). Collectively, these results show that HDAC1 and HDAC2 function in the DNA-damage response by promoting DSB repair and thus provide important insights into the radio-sensitizing effects of HDAC inhibitors that are being developed as cancer therapies.


Assuntos
Quebras de DNA de Cadeia Dupla , Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/metabolismo , Linhagem Celular Tumoral , Reparo do DNA , Histona Desacetilase 1/genética , Histona Desacetilase 2/genética , Histonas/metabolismo , Humanos , RNA Interferente Pequeno
16.
Science ; 325(5945): 1240-3, 2009 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-19661379

RESUMO

Posttranslational modifications play key roles in regulating chromatin plasticity. Although various chromatin-remodeling enzymes have been described that respond to specific histone modifications, little is known about the role of poly[adenosine 5'-diphosphate (ADP)-ribose] in chromatin remodeling. Here, we identify a chromatin-remodeling enzyme, ALC1 (Amplified in Liver Cancer 1, also known as CHD1L), that interacts with poly(ADP-ribose) and catalyzes PARP1-stimulated nucleosome sliding. Our results define ALC1 as a DNA damage-response protein whose role in this process is sustained by its association with known DNA repair factors and its rapid poly(ADP-ribose)-dependent recruitment to DNA damage sites. Furthermore, we show that depletion or overexpression of ALC1 results in sensitivity to DNA-damaging agents. Collectively, these results provide new insights into the mechanisms by which poly(ADP-ribose) regulates DNA repair.


Assuntos
Montagem e Desmontagem da Cromatina , Cromatina/metabolismo , DNA Helicases/metabolismo , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Poli Adenosina Difosfato Ribose/metabolismo , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Linhagem Celular , Dano ao DNA , DNA Helicases/química , DNA Helicases/genética , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Humanos , Peróxido de Hidrogênio/farmacologia , Imunoprecipitação , Cinética , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Nucleossomos/metabolismo , Fleomicinas/farmacologia , Poli(ADP-Ribose) Polimerase-1 , Inibidores de Poli(ADP-Ribose) Polimerases , Poli(ADP-Ribose) Polimerases/metabolismo , Estrutura Terciária de Proteína , Radiação Ionizante , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
17.
Cancer Lett ; 220(1): 1-9, 2005 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-15737682

RESUMO

The structural organization of DNA into chromatin is of key importance to regulate genome function and stability. Maintenance of such an organization is thus crucial to preserve cellular identity. At each cell cycle, during S phase, this is achieved by duplication of chromatin structure in tight coordination with DNA replication. Such a coordinate process requires histone synthesis and their deposition onto DNA by chromatin assembly factors to be efficiently coupled to DNA synthesis. In this review, we highlight the intimate relationship between these chromatin-related events and DNA replication and we show how it is possible to take advantage of this coupling in order to identify cells with high replicative potential such as tumor cells. On the basis of recent data, we discuss the potential use of chromatin-associated factors as new proliferation markers of interest for cancer diagnosis and prognosis.


Assuntos
Montagem e Desmontagem da Cromatina , Replicação do DNA , Histonas/biossíntese , Biomarcadores , Ciclo Celular , Proliferação de Células , Fator 1 de Modelagem da Cromatina , Proteínas Cromossômicas não Histona/análise , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/metabolismo , Chaperonas Moleculares/metabolismo , Neoplasias/diagnóstico
18.
Cancer Res ; 64(7): 2371-81, 2004 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15059888

RESUMO

Histone synthesis and chromatin assembly are mainly associated with DNA replication and are thus intimately involved in cell cycle regulation. The expression of key components involved in these events in human cells was studied in relation to cell-proliferative status. Among several chromatin assembly factors, chromatin assembly factor (CAF)-1 stood out as the most discriminating marker of the proliferative state. We show, using both immunofluorescence and Western blot analysis, that the expression of both CAF-1 large subunits, p150 and p60, is massively down-regulated during quiescence in several cell lines. Upon exit from the quiescent state, the CAF-1 subunits are re-expressed early, before DNA replication. The amounts of either total or chromatin-associated pools of CAF-1 proteins correlate directly with cell proliferation. Regulation of CAF-1 expression is partly controlled at the RNA level, as shown by quantitative reverse transcription-PCR and Northern blot experiments. Biological material from benign and malignant human breast tumors analyzed by immunocytochemistry and immunohistochemistry exhibits a strong positive correlation between CAF-1 p60 expression and the following proliferation markers: S-phase fraction (r = 0.84, P < 0.0001); Ki-67 (r = 0.94, P < 0.0001); and proliferating cell nuclear antigen (r = 0.95, P = 0.0001). We discuss the advantages of using CAF-1 to assess cell proliferation. High CAF-1 p60 levels are also shown to be associated with various prognostic factors. Our data highlight the precise association of CAF-1 expression with the proliferative state and validate the use of this factor as a useful proliferation marker and prognostic indicator in malignant and benign breast lesions.


Assuntos
Biomarcadores Tumorais/fisiologia , Neoplasias da Mama/patologia , Proteínas Cromossômicas não Histona/fisiologia , Proteínas de Ligação a DNA/fisiologia , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Divisão Celular/fisiologia , Linhagem Celular Tumoral , Cromatina/metabolismo , Fator 1 de Modelagem da Cromatina , Proteínas Cromossômicas não Histona/biossíntese , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Células HeLa , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Fase de Repouso do Ciclo Celular/fisiologia , Fase S/fisiologia , Fatores de Transcrição
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA