Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Nat Commun ; 11(1): 4826, 2020 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-32958757

RESUMO

DNA replication initiates from multiple genomic locations called replication origins. In metazoa, DNA sequence elements involved in origin specification remain elusive. Here, we examine pluripotent, primary, differentiating, and immortalized human cells, and demonstrate that a class of origins, termed core origins, is shared by different cell types and host ~80% of all DNA replication initiation events in any cell population. We detect a shared G-rich DNA sequence signature that coincides with most core origins in both human and mouse genomes. Transcription and G-rich elements can independently associate with replication origin activity. Computational algorithms show that core origins can be predicted, based solely on DNA sequence patterns but not on consensus motifs. Our results demonstrate that, despite an attributed stochasticity, core origins are chosen from a limited pool of genomic regions. Immortalization through oncogenic gene expression, but not normal cellular differentiation, results in increased stochastic firing from heterochromatin and decreased origin density at TAD borders.


Assuntos
DNA/biossíntese , DNA/química , Origem de Replicação/genética , Animais , Composição de Bases , Sequência de Bases , Carcinogênese , Diferenciação Celular , Células Cultivadas , Replicação do DNA/genética , Genoma Humano/genética , Heterocromatina/genética , Humanos , Camundongos , Motivos de Nucleotídeos , Transcrição Gênica
3.
Nucleic Acids Res ; 48(4): 1886-1904, 2020 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-31853544

RESUMO

Imbalance in the level of the pyrimidine degradation products dihydrouracil and dihydrothymine is associated with cellular transformation and cancer progression. Dihydropyrimidines are degraded by dihydropyrimidinase (DHP), a zinc metalloenzyme that is upregulated in solid tumors but not in the corresponding normal tissues. How dihydropyrimidine metabolites affect cellular phenotypes remains elusive. Here we show that the accumulation of dihydropyrimidines induces the formation of DNA-protein crosslinks (DPCs) and causes DNA replication and transcriptional stress. We used Xenopus egg extracts to recapitulate DNA replication invitro. We found that dihydropyrimidines interfere directly with the replication of both plasmid and chromosomal DNA. Furthermore, we show that the plant flavonoid dihydromyricetin inhibits human DHP activity. Cellular exposure to dihydromyricetin triggered DPCs-dependent DNA replication stress in cancer cells. This study defines dihydropyrimidines as potentially cytotoxic metabolites that may offer an opportunity for therapeutic-targeting of DHP activity in solid tumors.


Assuntos
Amidoidrolases/genética , Transformação Celular Neoplásica/genética , Replicação do DNA/genética , Transcrição Gênica , Animais , Antineoplásicos/uso terapêutico , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Uracila/análogos & derivados , Uracila/metabolismo , Xenopus laevis/genética , Xenopus laevis/crescimento & desenvolvimento
4.
Cell Rep ; 28(11): 2851-2865.e4, 2019 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-31509747

RESUMO

Hematopoiesis is particularly sensitive to DNA damage. Myeloid tumor incidence increases in patients with DNA repair defects and after chemotherapy. It is not known why hematopoietic cells are highly vulnerable to DNA damage. Addressing this question is complicated by the paucity of mouse models of hematopoietic malignancies due to defective DNA repair. We show that DNA repair-deficient Mcm8- and Mcm9-knockout mice develop myeloid tumors, phenocopying prevalent myelodysplastic syndromes. We demonstrate that these tumors are preceded by a lifelong DNA damage burden in bone marrow and that they acquire proliferative capacity by suppressing signaling of the tumor suppressor and cell cycle controller RB, as often seen in patients. Finally, we found that absence of MCM9 and the tumor suppressor Tp53 switches tumorigenesis to lymphoid tumors without precedent myeloid malignancy. Our results demonstrate that MCM8/9 deficiency drives myeloid tumor development and establishes a DNA damage burdened mouse model for hematopoietic malignancies.


Assuntos
Diferenciação Celular/genética , Dano ao DNA/genética , Regulação Leucêmica da Expressão Gênica/genética , Neoplasias Hematológicas/metabolismo , Proteínas de Manutenção de Minicromossomo/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Envelhecimento/genética , Envelhecimento/metabolismo , Envelhecimento/fisiologia , Animais , Apoptose/genética , Medula Óssea/metabolismo , Medula Óssea/patologia , Proliferação de Células/genética , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/patologia , Camundongos , Camundongos Knockout , Proteínas de Manutenção de Minicromossomo/genética , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais/genética , Esplenomegalia/genética , Esplenomegalia/metabolismo , Proteína Supressora de Tumor p53/genética
5.
Mol Cell ; 59(5): 831-9, 2015 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-26300262

RESUMO

DNA mismatch repair (MMR) is an evolutionarily conserved process that corrects DNA polymerase errors during replication to maintain genomic integrity. In E. coli, the DNA helicase UvrD is implicated in MMR, yet an analogous helicase activity has not been identified in eukaryotes. Here, we show that mammalian MCM9, a protein involved in replication and homologous recombination, forms a complex with MMR initiation proteins (MSH2, MSH3, MLH1, PMS1, and the clamp loader RFC) and is essential for MMR. Mcm9-/- cells display microsatellite instability and MMR deficiency. The MCM9 complex has a helicase activity that is required for efficient MMR since wild-type but not helicase-dead MCM9 restores MMR activity in Mcm9-/- cells. Moreover, MCM9 loading onto chromatin is MSH2-dependent, and in turn MCM9 stimulates the recruitment of MLH1 to chromatin. Our results reveal a role for MCM9 and its helicase activity in mammalian MMR.


Assuntos
Reparo de Erro de Pareamento de DNA/fisiologia , Proteínas de Manutenção de Minicromossomo/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Bases , Cromatina/genética , Cromatina/metabolismo , DNA/genética , DNA/metabolismo , DNA Helicases/química , DNA Helicases/genética , DNA Helicases/metabolismo , Reparo de Erro de Pareamento de DNA/genética , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Técnicas de Inativação de Genes , Células HeLa , Humanos , Instabilidade de Microssatélites , Proteínas de Manutenção de Minicromossomo/deficiência , Proteínas de Manutenção de Minicromossomo/genética , Proteína 1 Homóloga a MutL , Proteína 2 Homóloga a MutS/química , Proteína 2 Homóloga a MutS/genética , Proteína 2 Homóloga a MutS/metabolismo , Proteína 3 Homóloga a MutS , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo
6.
Nat Commun ; 4: 2065, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23817338

RESUMO

Cdt1 is required for loading the replicative DNA helicase MCM2/7, a process known as DNA replication licensing. Here we show that 129 mouse strains express a Cdt1 mutated allele with enhanced licensing activity. The mutation, named Δ(6)PEST, involves a six-amino acid deletion within a previously uncharacterized PEST-like domain. Cdt1 Δ(6)PEST and more extensive deletions exhibit increased re-replication and transformation activities that are independent of the Geminin and E3 ligase pathways. This PEST domain negatively regulates cell cycle-dependent chromatin recruitment of Cdt1 in G2/M phases of the cell cycle. Mass spectrometry analysis indicates that Cdt1 is phosphorylated at sites within the deleted PEST domain during mitosis. This study reveals a conserved new regulatory Cdt1 domain crucial for proper DNA licensing activity and suggests a mechanism by which the presence of Cdt1 in G2/M phases does not lead to premature origin licensing. These results also question the usage of 129 mouse strains for knockout analyses.


Assuntos
Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Replicação do DNA , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Mutação/genética , Alelos , Sequência de Aminoácidos , Animais , Sequência de Bases , Carcinogênese , Extratos Celulares , Linhagem Celular , Cromatina/metabolismo , Geminina/metabolismo , Humanos , Camundongos , Camundongos da Linhagem 129 , Mitose , Modelos Biológicos , Dados de Sequência Molecular , Células NIH 3T3 , Fosforilação , Estrutura Terciária de Proteína , Deleção de Sequência , Ubiquitina-Proteína Ligases/metabolismo
7.
Mol Cell ; 47(4): 523-34, 2012 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-22771120

RESUMO

We generated knockout mice for MCM8 and MCM9 and show that deficiency for these genes impairs homologous recombination (HR)-mediated DNA repair during gametogenesis and somatic cells cycles. MCM8(-/-) mice are sterile because spermatocytes are blocked in meiotic prophase I, and females have only arrested primary follicles and frequently develop ovarian tumors. MCM9(-/-) females also are sterile as ovaries are completely devoid of oocytes. In contrast, MCM9(-/-) testes produce spermatozoa, albeit in much reduced quantity. Mcm8(-/-) and Mcm9(-/-) embryonic fibroblasts show growth defects and chromosomal damage and cannot overcome a transient inhibition of replication fork progression. In these cells, chromatin recruitment of HR factors like Rad51 and RPA is impaired and HR strongly reduced. We further demonstrate that MCM8 and MCM9 form a complex and that they coregulate their stability. Our work uncovers essential functions of MCM8 and MCM9 in HR-mediated DSB repair during gametogenesis, replication fork maintenance, and DNA repair.


Assuntos
Proteínas de Ciclo Celular/deficiência , Proteínas de Ligação a DNA/deficiência , Gametogênese/genética , Instabilidade Genômica , Recombinação Homóloga/genética , Animais , Proteínas de Ciclo Celular/genética , Cromatina/genética , Reparo do DNA , Replicação do DNA/genética , Proteínas de Ligação a DNA/genética , Feminino , Fibroblastos/metabolismo , Células Germinativas/metabolismo , Masculino , Prófase Meiótica I/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Manutenção de Minicromossomo , Ovário/metabolismo , Espermatócitos/metabolismo
8.
Sci Rep ; 2: 279, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22359734

RESUMO

Terminal differentiation is the process by which cycling cells stop proliferating to start new specific functions. It involves dramatic changes in chromatin organization as well as gene expression. In the present report we used cell flow cytometry and genome wide DNA combing to investigate DNA replication during murine erythroleukemia-induced terminal cell differentiation. The results obtained indicated that the rate of replication fork movement slows down and the inter-origin distance becomes shorter during the precommitment and commitment periods before cells stop proliferating and accumulate in G1. We propose this is a general feature caused by the progressive heterochromatinization that characterizes terminal cell differentiation.


Assuntos
Diferenciação Celular , Proliferação de Células , Replicação do DNA , Animais , Sequência de Bases , Linhagem Celular Tumoral , Primers do DNA , Citometria de Fluxo , Técnica Indireta de Fluorescência para Anticorpo , Camundongos
9.
Proc Natl Acad Sci U S A ; 108(42): 17331-6, 2011 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-21908712

RESUMO

Transfer of somatic cell nuclei to enucleated eggs and ectopic expression of specific transcription factors are two different reprogramming strategies used to generate pluripotent cells from differentiated cells. However, these methods are poorly efficient, and other unknown factors might be required to increase their success rate. Here we show that Xenopus egg extracts at the metaphase stage (M phase) have a strong reprogramming activity on mouse embryonic fibroblasts (MEFs). First, they reset replication properties of MEF nuclei toward a replication profile characteristic of early development, and they erase several epigenetic marks, such as trimethylation of H3K9, H3K4, and H4K20. Second, when MEFs are reversibly permeabilized in the presence of M-phase Xenopus egg extracts, they show a transient increase in cell proliferation, form colonies, and start to express specific pluripotency markers. Finally, transient exposure of MEF nuclei to M-phase Xenopus egg extracts increases the success of nuclear transfer to enucleated mouse oocytes and strongly synergizes with the production of pluripotent stem cells by ectopic expression of transcription factors. The mitotic stage of the egg extract is crucial, because none of these effects is detected when using interphasic Xenopus egg extracts. Our data demonstrate that mitosis is essential to make mammalian somatic nuclei prone to reprogramming and that, surprisingly, the heterologous Xenopus system has features that are conserved enough to remodel mammalian nuclei.


Assuntos
Desdiferenciação Celular/fisiologia , Oócitos/metabolismo , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Desdiferenciação Celular/genética , Células Cultivadas , Montagem e Desmontagem da Cromatina/genética , Primers do DNA/genética , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Técnicas In Vitro , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Mitose , Técnicas de Transferência Nuclear , Oócitos/citologia , Xenopus
10.
EMBO Rep ; 7(8): 812-6, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16845368

RESUMO

In multicellular organisms, changes in the DNA replication programme could act to integrate differentiation with cell division in various developmental and transcriptional contexts. Here, we have addressed the use of DNA replication origins during differentiation in the HoxB domain-a cluster of nine genes developmentally regulated in a collinear manner. In undifferentiated mouse P19 cells, we detected several DNA replication origins in the 100 kb HoxB locus, indicating a relaxed origin use when the locus is transcriptionally silent. By contrast, in retinoic-acid-induced differentiated cells, when HoxB transcription is activated, a general silencing of DNA replication origins occurs in the locus except one located downstream of Hoxb1, at the 3' boundary of the HoxB domain. Silencing of the replication origins is associated with histone hyperacetylation, whereas the active Hoxb1 origin persists as a hypoacetylated island. These findings provide direct evidence for the differentiated use of origins in HoxB genes, and we suggest that this regulation might contribute to the regulated expression of HoxB genes during development.


Assuntos
Replicação do DNA/genética , Regulação da Expressão Gênica/genética , Proteínas de Homeodomínio/genética , Acetilação , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Regulação da Expressão Gênica/efeitos dos fármacos , Histonas/genética , Histonas/metabolismo , Proteínas de Homeodomínio/metabolismo , Modelos Genéticos , Complexo de Reconhecimento de Origem/genética , Complexo de Reconhecimento de Origem/metabolismo , Origem de Replicação/genética , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética , Tretinoína/farmacologia
11.
Cell ; 123(5): 787-801, 2005 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-16325575

RESUMO

Animal cloning by nuclear-transfer experiments frequently fails due to the inability of transplanted nuclei to support normal embryonic development. We show here that the formation of mitotic chromosomes in the egg context is crucial for adapting differentiated nuclei for early development. Differentiated erythrocyte nuclei replicate inefficiently in Xenopus eggs but do so as rapidly as sperm nuclei if a prior single mitosis is permitted. This mitotic remodeling involves a topoisomerase II-dependent shortening of chromatin loop domains and an increased recruitment of replication initiation factors onto chromatin, leading to a short interorigin spacing characteristic of early developmental stages. It also occurs within each early embryonic cell cycle and dominantly regulates initiation of DNA replication for the subsequent S phase. These results indicate that mitotic conditioning is crucial to reset the chromatin structure of differentiated adult donor cells for embryonic DNA replication and suggest that it is an important step in nuclear cloning.


Assuntos
Cromossomos/química , Clonagem de Organismos , Replicação do DNA , Mitose/fisiologia , Replicon , Xenopus laevis/embriologia , Acetilação , Animais , Extratos Celulares/química , Núcleo Celular/fisiologia , Núcleo Celular/ultraestrutura , Cromossomos/metabolismo , DNA Topoisomerases Tipo II/metabolismo , Eritrócitos/citologia , Eritrócitos/metabolismo , Histonas/metabolismo , Humanos , Masculino , Técnicas de Transferência Nuclear , Conformação de Ácido Nucleico , Origem de Replicação , Espermatozoides/citologia , Espermatozoides/metabolismo , Xenopus laevis/fisiologia
12.
Cell ; 120(3): 315-28, 2005 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-15707891

RESUMO

MCM2-7 proteins are replication factors required to initiate DNA synthesis and are currently the best candidates for replicative helicases. We show that the MCM2-7-related protein MCM8 is required to efficiently replicate chromosomal DNA in Xenopus egg extracts. MCM8 does not associate with the soluble MCM2-7 complex and binds chromatin upon initiation of DNA synthesis. MCM8 depletion does not affect replication licensing or MCM3 loading but slows down DNA synthesis and reduces chromatin recruitment of RPA34 and DNA polymerase-alpha. Recombinant MCM8 displays both DNA helicase and ATPase activities in vitro. Reconstitution experiments show that ATP binding in MCM8 is required to rescue DNA synthesis in MCM8-depleted extracts. MCM8 colocalizes with replication foci and RPA34 on chromatin. We suggest that MCM8 functions in the elongation step of DNA replication as a helicase that facilitates the recruitment of RPA34 and stimulates the processivity of DNA polymerases at replication foci.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Cromossomos/fisiologia , DNA Helicases/metabolismo , Replicação do DNA/fisiologia , Mitose/genética , Proteínas de Xenopus/metabolismo , Adenosina Trifosfatases/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/isolamento & purificação , DNA/biossíntese , DNA Helicases/genética , DNA Helicases/isolamento & purificação , DNA Polimerase I/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo/fisiologia , Feminino , Componente 2 do Complexo de Manutenção de Minicromossomo , Proteínas de Manutenção de Minicromossomo , Dados de Sequência Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oócitos , Coelhos , Xenopus , Proteínas de Xenopus/genética , Proteínas de Xenopus/isolamento & purificação
14.
Gene ; 332: 129-38, 2004 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-15145062

RESUMO

Only a very few origins have been mapped in different multicellular organisms, and they do not share detectable consensus sequence elements. Moreover, it is not clear if origins are localized at similar positions in the corresponding locus in genomes of different organisms. Here, we have mapped DNA replication origins in the c-myc locus both in Xenopus and mouse, allowing a comparison of the corresponding sites in three different animal species (Xenopus, mouse, human). An origin of DNA replication is present in the three homologous c-myc loci. In Xenopus, a main DNA replication origin was located 3 kilobases (kb) upstream of the active c-myc promoter, whereas, in mouse, we detected an origin 1 kb upstream of the promoter, as previously mapped in human c-myc. We also identified a nuclear matrix attachment region in both Xenopus and mouse, which is localized to two different regions of the c-myc promoter region. However, in both cases, the nuclear matrix attachment sites are close to the DNA replication origin mapped in the locus. These data suggest that global features of chromatin organization in different organisms may contribute to DNA replication origin localization.


Assuntos
Replicação do DNA/genética , Matriz Nuclear/genética , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-myc/genética , Animais , Sítios de Ligação/genética , Linhagem Celular Tumoral , DNA/biossíntese , DNA/genética , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário , Camundongos , Xenopus
15.
EMBO J ; 22(14): 3737-48, 2003 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12853488

RESUMO

To study the relationship between DNA replication and transcription in vivo, we investigated Hox gene activation in two vertebrate systems: the embryogenesis of Xenopus and the retinoic acid-induced differentiation of pluripotent mouse P19 cells. We show that the first cell cycles following the mid- blastula transition in Xenopus are necessary and sufficient for HoxB activation, whereas later cell cycles are necessary for the correct expression pattern. In P19 cells, HoxB expression requires proliferation, and the entire locus is activated within one cell cycle. Using synchronous cultures, we found that activation of HoxB genes is colinear within a single cell cycle, occurs during S phase and requires S phase. The HoxB locus replicates early, whereas replication is still required for maximal expression later in S phase. Thus, induction of HoxB genes occurs in a DNA replication-dependent manner and requires only one cell cycle. We propose that S-phase remodelling licenses the locus for transcriptional regulation.


Assuntos
Replicação do DNA , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Homeodomínio/metabolismo , Animais , Ciclo Celular , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Genes Homeobox , Hidroxiureia/farmacologia , Camundongos , Inibidores da Síntese de Ácido Nucleico/farmacologia , Óvulo/efeitos dos fármacos , Fase S , Ativação Transcricional , Tretinoína/farmacologia , Células Tumorais Cultivadas , Xenopus/embriologia , Xenopus/genética
16.
J Struct Biol ; 140(1-3): 57-66, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12490154

RESUMO

We have characterized Xenopus ISWI, a catalytic subunit of a family of chromatin-remodeling complexes. We show that ISWI is expressed constitutively during development but poorly expressed in adult tissues except oocytes which contain a large store of maternal protein. We further analyzed its localization both in vivo and in vitro in Xenopus cell cycle extracts and identified that ISWI binds to chromatin at the G1-S period. However, its association to chromatin does not require ongoing DNA replication. Immunodepletion of ISWI has no effect on either sperm chromatin decondensation or the kinetics and efficiency of DNA replication. Nucleosome assembly also occurs in ISWI-depleted extracts, but nucleosome spacing is disturbed. From these results, we conclude that ISWI is not necessary for sperm chromatin decondensation and the accelerated rates of DNA replication that characterize early development.


Assuntos
Adenosina Trifosfatases/biossíntese , Adenosina Trifosfatases/química , Cromatina/metabolismo , Fatores de Transcrição/biossíntese , Fatores de Transcrição/química , Sequência de Aminoácidos , Animais , Northern Blotting , Ciclo Celular , Centrifugação com Gradiente de Concentração , Clonagem Molecular , DNA Complementar/metabolismo , Fase G1 , Immunoblotting , Microscopia de Fluorescência , Dados de Sequência Molecular , Nucleossomos/metabolismo , Ligação Proteica , Fase S , Homologia de Sequência de Aminoácidos , Sacarose/farmacologia , Fatores de Tempo , Distribuição Tecidual , Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA