Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Invest Ophthalmol Vis Sci ; 64(2): 6, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36734965

RESUMO

Purpose: During lens fiber cell differentiation, organelles are removed in an ordered manner to ensure lens clarity. A critical step in this process is removal of the cell nucleus, but the mechanisms by which this occurs are unclear. In this study, we investigate the role of a cyclin-dependent kinase 1 (CDK1) regulatory loop in controlling lens fiber cell denucleation (LFCD). Methods: We examined lens differentiation histologically in two different vertebrate models. An embryonic chick lens culture system was used to test the role of CDK1, cell division cycle 25 (CDC25), WEE1, and PP2A in LFCD. Additionally, we used three mouse models that express high levels of the CDK inhibitor p27 to test whether increased p27 levels affect LFCD. Results: Using chick lens organ cultures, small-molecule inhibitors of CDK1 and CDC25 inhibit LFCD, while inhibiting the CDK1 inhibitory kinase WEE1 potentiates LFCD. Additionally, treatment with an inhibitor of PP2A, which indirectly inhibits CDK1 activity, also increased LFCD. Three different mouse models that express increased levels of p27 through different mechanisms show impaired LFCD. Conclusions: Here we define a conserved nonmitotic role for CDK1 and its upstream regulators in controlling LFCD. We find that CDK1 functionally interacts with WEE1, a nuclear kinase that inhibits CDK1 activity, and CDC25 activating phosphatases in cells where CDK1 activity must be exquisitely regulated to allow for LFCD. We also provide genetic evidence in multiple in vivo models that p27, a CDK1 inhibitor, inhibits lens growth and LFCD.


Assuntos
Proteína Quinase CDC2 , Mitose , Camundongos , Animais , Proteína Quinase CDC2/genética , Proteína Quinase CDC2/metabolismo , Ciclo Celular , Fosforilação , Proteínas de Ciclo Celular/genética , Diferenciação Celular
2.
JCI Insight ; 4(23)2019 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-31622281

RESUMO

Mice homozygous for a hypomorphic allele of DNA replication factor minichromosome maintenance protein 2 (designated Mcm2cre/cre) develop precursor T cell lymphoblastic leukemia/lymphoma (pre-T LBL) with 4-32 small interstitial deletions per tumor. Mice that express a NUP98-HOXD13 (NHD13) transgene develop multiple types of leukemia, including myeloid and T and B lymphocyte. All Mcm2cre/cre NHD13+ mice develop pre-T LBL, and 26% develop an unrelated, concurrent B cell precursor acute lymphoblastic leukemia (BCP-ALL). Copy number alteration (CNA) analysis demonstrated that pre-T LBLs were characterized by homozygous deletions of Pten and Tcf3 and partial deletions of Notch1 leading to Notch1 activation. In contrast, BCP-ALLs were characterized by recurrent deletions involving Pax5 and Ptpn1 and copy number gain of Abl1 and Nup214 resulting in a Nup214-Abl1 fusion. We present a model in which Mcm2 deficiency leads to replicative stress, DNA double strand breaks (DSBs), and resultant CNAs due to errors in DNA DSB repair. CNAs that involve critical oncogenic pathways are then selected in vivo as malignant lymphoblasts because of a fitness advantage. Some CNAs, such as those involving Abl1 and Notch1, represent attractive targets for therapy.


Assuntos
Carcinogênese/genética , Carcinogênese/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Fenótipo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Carcinogênese/patologia , Linhagem Celular Tumoral , Variações do Número de Cópias de DNA , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Humanos , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Endogâmicos C57BL , Componente 2 do Complexo de Manutenção de Minicromossomo/genética , Componente 2 do Complexo de Manutenção de Minicromossomo/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Fator de Transcrição PAX5/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Baço/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcriptoma
3.
Trends Cancer ; 5(5): 308-324, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31174843

RESUMO

Recent studies highlight the importance of the RB1 tumor suppressor as a target for cancer therapy. Canonically, RB1 regulates cell cycle progression and represents the downstream target for cyclin-dependent kinase (CDK) 4/6 inhibitors that are in clinical use. However, newly discovered features of the RB1 pathway suggest new therapeutic strategies to counter resistance and improve precision medicine. These therapeutic strategies include deepening cell cycle exit with CDK4/6 inhibitor combinations, selectively targeting tumors that have lost RB1, and expanding therapeutic index by mitigating therapy-associated adverse effects. In addition, RB1 impacts immunological features of tumors and the microenvironment that can enhance sensitivity to immunotherapy. Lastly, RB1 specifies epigenetically determined cell lineage states that are disrupted during therapy resistance and could be re-installed through the direct use of epigenetic therapies. Thus, new opportunities are emerging to improve cancer therapy by exploiting the RB1 pathway.


Assuntos
Ciclo Celular/genética , Proteínas de Ligação a Retinoblastoma/genética , Ubiquitina-Proteína Ligases/genética , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Biomarcadores , Ciclo Celular/efeitos dos fármacos , Suscetibilidade a Doenças , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Heterogeneidade Genética , Humanos , Imunoterapia , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Neoplasias/etiologia , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas de Ligação a Retinoblastoma/metabolismo , Transdução de Sinais/efeitos dos fármacos , Evasão Tumoral/genética , Ubiquitina-Proteína Ligases/metabolismo
4.
Cell ; 174(5): 1127-1142.e19, 2018 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-30078706

RESUMO

Replication origins, fragile sites, and rDNA have been implicated as sources of chromosomal instability. However, the defining genomic features of replication origins and fragile sites are among the least understood elements of eukaryote genomes. Here, we map sites of replication initiation and breakage in primary cells at high resolution. We find that replication initiates between transcribed genes within nucleosome-depleted structures established by long asymmetrical poly(dA:dT) tracts flanking the initiation site. Paradoxically, long (>20 bp) (dA:dT) tracts are also preferential sites of polar replication fork stalling and collapse within early-replicating fragile sites (ERFSs) and late-replicating common fragile sites (CFSs) and at the rDNA replication fork barrier. Poly(dA:dT) sequences are fragile because long single-strand poly(dA) stretches at the replication fork are unprotected by the replication protein A (RPA). We propose that the evolutionary expansion of poly(dA:dT) tracts in eukaryotic genomes promotes replication initiation, but at the cost of chromosome fragility.


Assuntos
Replicação do DNA , DNA Ribossômico/química , Nucleossomos/metabolismo , Poli dA-dT/química , Origem de Replicação , Motivos de Aminoácidos , Animais , Linhagem Celular , Imunoprecipitação da Cromatina , Instabilidade Cromossômica , Sítios Frágeis do Cromossomo , Fragilidade Cromossômica , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Saccharomyces cerevisiae , Schizosaccharomyces , Sítio de Iniciação de Transcrição , Transcrição Gênica
5.
PLoS Genet ; 13(9): e1007006, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28915237

RESUMO

Ribosomal RNAs (rRNAs) in budding yeast are encoded by ~100-200 repeats of a 9.1kb sequence arranged in tandem on chromosome XII, the ribosomal DNA (rDNA) locus. Copy number of rDNA repeat units in eukaryotic cells is maintained far in excess of the requirement for ribosome biogenesis. Despite the importance of the repeats for both ribosomal and non-ribosomal functions, it is currently not known how "normal" copy number is determined or maintained. To identify essential genes involved in the maintenance of rDNA copy number, we developed a droplet digital PCR based assay to measure rDNA copy number in yeast and used it to screen a yeast conditional temperature-sensitive mutant collection of essential genes. Our screen revealed that low rDNA copy number is associated with compromised DNA replication. Further, subculturing yeast under two separate conditions of DNA replication stress selected for a contraction of the rDNA array independent of the replication fork blocking protein, Fob1. Interestingly, cells with a contracted array grew better than their counterparts with normal copy number under conditions of DNA replication stress. Our data indicate that DNA replication stresses select for a smaller rDNA array. We speculate that this liberates scarce replication factors for use by the rest of the genome, which in turn helps cells complete DNA replication and continue to propagate. Interestingly, tumors from mini chromosome maintenance 2 (MCM2)-deficient mice also show a loss of rDNA repeats. Our data suggest that a reduction in rDNA copy number may indicate a history of DNA replication stress, and that rDNA array size could serve as a diagnostic marker for replication stress. Taken together, these data begin to suggest the selective pressures that combine to yield a "normal" rDNA copy number.


Assuntos
Variações do Número de Cópias de DNA/genética , Replicação do DNA/genética , DNA Ribossômico/genética , Recombinação Genética , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ligação a DNA/genética , Camundongos , Componente 2 do Complexo de Manutenção de Minicromossomo/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Temperatura
6.
PLoS Genet ; 13(1): e1006547, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28045896

RESUMO

Insufficient licensing of DNA replication origins has been shown to result in genome instability, stem cell deficiency, and cancers. However, it is unclear whether the DNA damage resulting from deficient replication licensing occurs generally or if specific sites are preferentially affected. To map locations of ongoing DNA damage in vivo, the DNAs present in red blood cell micronuclei were sequenced. Many micronuclei are the product of DNA breaks that leave acentromeric remnants that failed to segregate during mitosis and should reflect the locations of breaks. To validate the approach we show that micronuclear sequences identify known common fragile sites under conditions that induce breaks at these locations (hydroxyurea). In MCM2 deficient mice a different set of preferred breakage sites is identified that includes the tumor suppressor gene Tcf3, which is known to contribute to T-lymphocytic leukemias that arise in these mice, and the 45S rRNA gene repeats.


Assuntos
Replicação do DNA , Instabilidade Genômica , Testes para Micronúcleos/métodos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Dano ao DNA , Eritrócitos/patologia , Camundongos , Micronúcleos com Defeito Cromossômico , Componente 2 do Complexo de Manutenção de Minicromossomo/deficiência , Componente 2 do Complexo de Manutenção de Minicromossomo/genética
7.
Genome Res ; 25(4): 558-69, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25762552

RESUMO

Minichromosome maintenance (MCM) proteins are loaded onto chromatin during G1-phase and define potential locations of DNA replication initiation. MCM protein deficiency results in genome instability and high rates of cancer in mouse models. Here we develop a method of nascent strand capture and release and show that MCM2 deficiency reduces DNA replication initiation in gene-rich regions of the genome. DNA structural properties are shown to correlate with sequence motifs associated with replication origins and with locations that are preferentially affected by MCM2 deficiency. Reduced nascent strand density correlates with sites of recurrent focal CNVs in tumors arising in MCM2-deficient mice, consistent with a direct relationship between sites of reduced DNA replication initiation and genetic damage. Between 10% and 90% of human tumors, depending on type, carry heterozygous loss or mutation of one or more MCM2-7 genes, which is expected to compromise DNA replication origin licensing and result in elevated rates of genome damage at a subset of gene-rich locations.


Assuntos
Replicação do DNA/genética , Componente 2 do Complexo de Manutenção de Minicromossomo/genética , Neoplasias/genética , Origem de Replicação/genética , Animais , Sequência de Bases , Linhagem Celular , Cromatina/genética , Variações do Número de Cópias de DNA/genética , Fase G1/genética , Instabilidade Genômica/genética , Humanos , Camundongos , Modelos Animais , Análise de Sequência de DNA
8.
J Biol Methods ; 2(4)2015.
Artigo em Inglês | MEDLINE | ID: mdl-26949711

RESUMO

Nascent strand capture and release (NSCR) is a method for isolation of short nascent strands to identify origins of DNA replication. The protocol provided involves isolation of total DNA, denaturation, size fractionation on a sucrose gradient, 5'-biotinylation of the appropriate size nucleic acids, binding to a streptavidin coated magnetic beads, intensive washing, and specific release of only the RNA-containing chimeric nascent strand DNA using ribonuclease I (RNase I). The method has been applied to mammalian cells derived from proliferative tissues and cell culture but could be used for any system where DNA replication is primed by a small RNA resulting in chimeric RNA-DNA molecules.

9.
Proc Natl Acad Sci U S A ; 111(6): E672-81, 2014 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-24469795

RESUMO

Using complete genome analysis, we sequenced five bladder tumors accrued from patients with muscle-invasive transitional cell carcinoma of the urinary bladder (TCC-UB) and identified a spectrum of genomic aberrations. In three tumors, complex genotype changes were noted. All three had tumor protein p53 mutations and a relatively large number of single-nucleotide variants (SNVs; average of 11.2 per megabase), structural variants (SVs; average of 46), or both. This group was best characterized by chromothripsis and the presence of subclonal populations of neoplastic cells or intratumoral mutational heterogeneity. Here, we provide evidence that the process of chromothripsis in TCC-UB is mediated by nonhomologous end-joining using kilobase, rather than megabase, fragments of DNA, which we refer to as "stitchers," to repair this process. We postulate that a potential unifying theme among tumors with the more complex genotype group is a defective replication-licensing complex. A second group (two bladder tumors) had no chromothripsis, and a simpler genotype, WT tumor protein p53, had relatively few SNVs (average of 5.9 per megabase) and only a single SV. There was no evidence of a subclonal population of neoplastic cells. In this group, we used a preclinical model of bladder carcinoma cell lines to study a unique SV (translocation and amplification) of the gene glutamate receptor ionotropic N-methyl D-aspertate as a potential new therapeutic target in bladder cancer.


Assuntos
Cromossomos Humanos , Heterogeneidade Genética , Genoma Humano , Neoplasias da Bexiga Urinária/genética , Humanos , Hibridização in Situ Fluorescente , Componente 4 do Complexo de Manutenção de Minicromossomo/genética , Mutação , Canal de Sódio Disparado por Voltagem NAV1.6/genética , Oncogenes , Polimorfismo de Nucleotídeo Único , Receptores de N-Metil-D-Aspartato/genética , Proteína Supressora de Tumor p53/genética
10.
Nat Commun ; 4: 2626, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24149709

RESUMO

Insufficient cell proliferation has been suggested as a potential cause of age-related tissue dysgenesis in mammals. However, genetic manipulation of cell cycle regulators in the germ lines of mice results in changes in animal size but not progeroid phenotypes. Here we increase levels of the cyclin-dependent kinase inhibitor Cdkn1b (p27kip1) in adult mice through doxycycline-inducible expression and show this results in reduced cell proliferation in multiple tissues. The mice undergo changes resembling ageing even in the absence of an elevated DNA damage response or evidence of senescent cells, suggesting an altered balance between genetic and tissue ageing. In contrast, suppressing cell proliferation by doxycycline treatment of neonates retards growth, but the onset of degenerative changes is delayed during the period of reduced body mass. These results support the hypothesis that many of the most recognizable features of mammalian ageing can result from an imbalance between cell production and the mass of tissue that must be maintained.


Assuntos
Envelhecimento/genética , Inibidor de Quinase Dependente de Ciclina p27/genética , Genoma , Envelhecimento/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Contagem de Células , Proliferação de Células/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Doxiciclina/farmacologia , Expressão Gênica , Camundongos , Camundongos Transgênicos , Células-Tronco/citologia , Células-Tronco/metabolismo
11.
Nucleic Acids Res ; 40(11): 4914-24, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22362746

RESUMO

The MiniChromosome Maintenance 2-7 (MCM2-7) complex provides essential replicative helicase function. Insufficient MCMs impair the cell cycle and cause genomic instability (GIN), leading to cancer and developmental defects in mice. Remarkably, depletion or mutation of one Mcm can decrease all Mcm levels. Here, we use mice and cells bearing a GIN-causing hypomophic allele of Mcm4 (Chaos3), in conjunction with disruption alleles of other Mcms, to reveal two new mechanisms that regulate MCM protein levels and pre-RC formation. First, the Mcm4(Chaos3) allele, which disrupts MCM4:MCM6 interaction, triggers a Dicer1 and Drosha-dependent ≈ 40% reduction in Mcm2-7 mRNAs. The decreases in Mcm mRNAs coincide with up-regulation of the miR-34 family of microRNAs, which is known to be Trp53-regulated and target Mcms. Second, MCM3 acts as a negative regulator of the MCM2-7 helicase in vivo by complexing with MCM5 in a manner dependent upon a nuclear-export signal-like domain, blocking the recruitment of MCMs onto chromatin. Therefore, the stoichiometry of MCM components and their localization is controlled post-transcriptionally at both the mRNA and protein levels. Alterations to these pathways cause significant defects in cell growth reflected by disease phenotypes in mice.


Assuntos
Proteínas de Ciclo Celular/metabolismo , DNA Helicases/genética , Proteínas de Ligação a DNA/metabolismo , Animais , Ciclo Celular , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Células Cultivadas , Cromatina/metabolismo , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Regulação para Baixo , Homeostase , Leucina/análise , Camundongos , MicroRNAs/metabolismo , Componente 3 do Complexo de Manutenção de Minicromossomo , Componente 4 do Complexo de Manutenção de Minicromossomo , Componente 6 do Complexo de Manutenção de Minicromossomo , Mutação , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenótipo , Domínios e Motivos de Interação entre Proteínas , Interferência de RNA , RNA Mensageiro/metabolismo
12.
Stem Cells ; 28(7): 1250-9, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20503265

RESUMO

Stem cell quiescence has been hypothesized to suppress the rate at which genetic mutations accumulate within tissues by reducing the number of divisions a cell undergoes. However, recent studies have suggested that stem cells in the small intestine are rapidly dividing. This observation raises the issue of whether replication related errors are an important contributor to the accumulation of genetic damage and, if so, how genomic integrity is maintained within the small intestine. Here, reporter-marked small intestinal epithelial cells, resulting from mini-chromosome maintenance protein 2 (Mcm2) gene driven Cre-mediated recombination, are shown to be retained at the +1 position within the crypt and to contribute to the intestinal epithelia over long periods. Additionally, we show that the rate of cycling of +1 position Mcm2-expressing stem cells is heterogeneous with cycling times ranging between 1 and 4 days. Further, this heterogeneity depends on the p53 signaling pathway and could provide the basis for retention and expansion, through niche succession and crypt fission, of genetically intact stem cells. This somatic selection process would require active cellular replication.


Assuntos
Ciclo Celular , Genoma , Intestino Delgado/citologia , Intestino Delgado/metabolismo , Animais , Camundongos , Camundongos Knockout , Componente 2 do Complexo de Manutenção de Minicromossomo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/metabolismo
13.
Nucleic Acids Res ; 35(21): e141, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17986461

RESUMO

Yeast two-hybrid (Y2H) has been successfully used for genome-wide screens to identify protein-protein interactions for several model organisms. Nonetheless, the logistics of pair-wise screening has resulted in a cumbersome and incomplete application of this method to complex genomes. Here, we develop a modification of Y2H that eliminates the requirement for pair-wise screening. This is accomplished by incorporating lox sequences into Y2H vectors such that cDNAs encoding interacting partners become physically linked in the presence of Cre recombinase in vivo. Once linked, DNA from complex pools of clones can be processed without losing the identity of the interacting partners. Short linked sequence tags from each pair of interacting partner (binary interaction Tags or BI-Tags) are then recovered and sequenced. To validate the approach, comparisons between interactions found using traditional Y2H and the BI-Tag method were made, which demonstrate that the BI-Tag technology accurately represents the complexity of the interaction partners found in the screens. The technology described here sufficiently improves the throughput of the Y2H approach to make feasible the generation of near comprehensive interaction maps for complex organisms.


Assuntos
Integrases/metabolismo , Sitios de Sequências Rotuladas , Técnicas do Sistema de Duplo-Híbrido , Animais , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Biblioteca Gênica , Vetores Genéticos , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/genética , Recombinação Genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Leveduras/genética
14.
Stem Cells ; 25(12): 3121-32, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17717065

RESUMO

Mcm2 is a component of the DNA replication licensing complex that marks DNA replication origins during G1 of the cell cycle for use in the subsequent S-phase. It is expressed in stem/progenitor cells in a variety of regenerative tissues in mammals. Here, we have used the Mcm2 gene to develop a transgenic mouse in which somatic stem/progenitor cells can be genetically modified in the adult. In these mice, a tamoxifen-inducible form of Cre recombinase is integrated 3' to the Mcm2 coding sequence and expressed via an internal ribosome entry site (IRES). Heterozygous Mcm2(IRES-CreERT2/wild-type (wt)) mice are phenotypically indistinguishable from wild-type at least through 1 year of age. In bigenic Mcm2(IRES-CreERT2/wt); Z/EG reporter mice, tamoxifen-dependent enhanced green fluorescence protein expression is inducible in a wide variety of somatic stem cells and their progeny. However, in Mcm2(IRES-CreERT2/IRES-CreERT2) homozygous embryos or mouse embryonic fibroblasts, Mcm2 is reduced to approximately one-third of wild-type levels. Despite the fact that these mice develop normally and are asymptomatic as young adults, life span is greatly reduced, with most surviving to only approximately 10-12 weeks of age. They demonstrate severe deficiencies in the proliferative cell compartments of a variety of tissues, including the subventricular zone of the brain, muscle, and intestinal crypts. However, the immediate cause of death in most of these animals is cancer, where the majority develop lymphomas. These studies directly demonstrate that deficiencies in the function of the core DNA replication machinery that are compatible with development and survival nonetheless result in a chronic phenotype leading to stem cell deficiency in multiple tissues and cancer. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Proteínas de Ciclo Celular/genética , Regulação Neoplásica da Expressão Gênica , Linfoma/genética , Linfoma/patologia , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Células-Tronco/metabolismo , Células-Tronco/patologia , Animais , Sequência de Bases , Contagem de Células , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/biossíntese , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Integrases/genética , Linfoma/metabolismo , Linfoma/mortalidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Componente 2 do Complexo de Manutenção de Minicromossomo , Dados de Sequência Molecular , Mutagênese Insercional , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/biossíntese , Ribossomos/genética , Ribossomos/metabolismo , Células-Tronco/fisiologia , Análise de Sobrevida
15.
Stem Cells ; 25(1): 132-8, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17008428

RESUMO

Previous studies have demonstrated expression of the minichromosome maintenance protein Mcm2 in cells that remain competent to divide, including stem/progenitor cells of the subventricular zone (SVZ) within the brain. Here, a transgenic mouse line in which the Mcm2 gene drives expression of enhanced green fluorescent protein (EGFP) was constructed by insertion of an internal ribosomal entry site (IRES)-EGFP cassette into the last exon of the gene, 3' to the stop codon. In these mice, expression of EGFP is observed in the SVZ and several other tissues with high proliferative activity, including the spleen, intestine, hair follicles, and bone marrow. These observations suggest that EGFP fluorescence in this mouse line provides an index of the proliferative capacity of different tissues. Immunohistological analysis demonstrates a direct concordance between expression of EGFP and Mcm2, consistent with a transcriptional level downregulation of Mcm2 expression in postmitotic cells. To test the utility of EGFP expression for recovery of live cells retaining the capacity to divide, EGFP-expressing and -nonexpressing cells from bone marrow and brain were isolated from an adult Mcm2(IRES-EGFP) mouse by fluorescence-activated cell sorting and assayed for clonal growth. The EGFP-positive fraction contained the entire clonogenic population of the bone marrow and greater than 90% of neurosphere-forming cells from the brain. Brain-derived clonogenic cells were shown to remain competent to differentiate towards all three neural lineages. These studies demonstrate that the Mcm2(IRES-EGFP) transgenic line constructed here can be used for recovery of proliferation competent cells from different tissue types.


Assuntos
Proteínas de Fluorescência Verde/genética , Proteínas Nucleares/genética , Células-Tronco/fisiologia , Animais , Blastocisto/citologia , Blastocisto/fisiologia , Éxons , Genes Reporter , Genoma , Proteínas de Fluorescência Verde/análise , Camundongos , Camundongos Transgênicos , Componente 2 do Complexo de Manutenção de Minicromossomo , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Transfecção
16.
Melanoma Res ; 15(5): 363-73, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16179863

RESUMO

The objective of this study was to determine which transcription factors regulate the expression of the Pax3 gene in the mouse B16 F1 melanoma cell line. The results showed that the -14 kilobase pair (kbp) Pax3 promoter, but not the -1.6 kbp Pax3 promoter, promoted Pax3 gene expression in B16 cells. Comparison of the sequence of the -14 kbp human Pax3 promoter with mouse Pax3 promoters indicated that homology sequences were located between -6.9 and -5.8 kbp, and also that the 1.1 kbp fragment (between -6.9 and -5.8 kbp), linked -1.6 kbp proximal to the Pax3 promoter [plasmid PGPax3PIV (N6.9/5.8) delta SST Lacz], could mimic the functions of plasmid PGPax3 -14(N-1.6) Lacz. Mutations of the core binding elements of either Pax3 site I or II or both sites I and II reduced significantly the beta-galactosidase (beta-gal) activity in the cells. However, mutations of the core binding sequences of site A or B increased significantly the beta-gal activity in the cells. Biochemistry analysis demonstrated that POU transcription factors (Oct-1 and Brn-2) bind to the specific binding elements of both sites I and II to stimulate Pax3 gene expression, whereas the TALE homeodomain-containing proteins (Pbx and Prep1) bind with the core binding sequences of sites A and B to repress the expression of the Pax3 gene in B16 cells.


Assuntos
Regulação Neoplásica da Expressão Gênica/fisiologia , Melanoma Experimental/genética , Fatores de Transcrição Box Pareados/biossíntese , Fatores de Transcrição/fisiologia , Animais , Linhagem Celular Tumoral , Dimerização , Ensaio de Desvio de Mobilidade Eletroforética , Proteínas de Homeodomínio/metabolismo , Melanoma Experimental/metabolismo , Camundongos , Fator 1 de Transcrição de Octâmero/metabolismo , Fator de Transcrição PAX3 , Fatores do Domínio POU/metabolismo , Fatores de Transcrição Box Pareados/genética , Regiões Promotoras Genéticas , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção
17.
Aging Cell ; 3(6): 391-7, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15569356

RESUMO

Genomic instability within somatic stem cells may lead to the accumulation of mutations and contribute to cancer or other age-related phenotypes. However, determining the frequency of mutations that differ among individual stem cells is difficult from whole tissue samples because each event is diluted in the total population of both stem cells and differentiated tissue. Here the ability to expand neural stem/progenitor cells clonally permitted measurement of genomic alterations derived from a single initial cell. C57Bl/6 x DBA/2 hybrid mice were used and PCR analysis with strain-specific primers was performed to detect loss of heterozygosity on nine different chromosomes for each neurosphere. The frequency with which changes occurred in neurospheres derived from 2-month- and 2-year-old mice was compared. In 15 neurospheres derived from young animals both parental chromosomes were present for all nine chromosome pairs. In contrast, 16/17 neurospheres from old animals demonstrated loss of heterozygosity (LOH) on one or more chromosomes and seven exhibited a complete deletion of at least one chromosomal region. For chromosomes 9 and 19 there is a significant bias in the allele that is lost where in each case the C57Bl/6 allele is retained in 6/6 neurospheres exhibiting LOH. These data suggest that aging leads to a substantial mutational load within the neural stem cell compartment which can be expected to affect the normal function of these cells. Furthermore, the retention of specific alleles for chromosomes 9 and 19 suggests that a subset of mutational events lead to an allele-specific survival advantage within the neural stem cell compartment.


Assuntos
Senescência Celular/fisiologia , Neurônios/fisiologia , Células-Tronco/fisiologia , Alelos , Animais , Senescência Celular/genética , Cromossomos/genética , Perda de Heterozigosidade/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Mutação , Neurônios/citologia , Fenótipo , Reação em Cadeia da Polimerase/métodos , Seleção Genética
18.
Gene Expr Patterns ; 4(6): 671-85, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15465489

RESUMO

Pax3 is a paired-homeodomain class transcription factor that serves a role in dorsal-ventral and medial-lateral patterning during vertebrate embryogenesis. Its expression is localized to dorsal domains within the developing neural tube and lateral domains within the developing somite. Additionally, modulation of its expression occurs along the rostral-caudal axis. Previous studies [Development 124 (1997) 617] have localized sequence elements required for expression of Pax3 in the neural tube and neural crest to a 1.6 kbp promoter fragment. In the present study, four discrete DNA elements within the 1.6 kbp promoter fragment are shown by electrophoretic mobility shift assays (EMSA) to exhibit sequence specific interactions with proteins present in nuclear extracts from P19 EC cells induced to express Pax3 by treatment with retinoic acid (RA). Proteins interacting at each of these elements are identified based on biochemical purification using DNA affinity chromatography or a candidate approach. These identifications were confirmed by the ability of specific antibodies to super-shift DNA-protein complexes in EMSA. Two of the four DNA sequence elements are shown to interact with the neural specific Pou-domain class III transcription factors Brn1 and Brn2. The remaining sites contain either consensus binding elements for heterodimers of Pbx and an anterior set of Hox family members, from paralogous groups 1-5, or monomeric Meis and are shown to interact with members of the Pbx and Meis families. Ectopic expression of Brn2 plus HoxA1 but not either factor alone, is sufficient to induce efficient expression from the endogenous Pax3 promoter in P19 EC stem cells under conditions where they would not otherwise express Pax3. Finally, in transgenic mice, mutation of either of the Pou-domain protein binding sites results in reduced expression throughout the neural tube while mutation of the Pbx/Hox binding site results in loss of expression in the anterior domain in which Hox family members from paralogous groups 1-5 are expressed. These observations demonstrate that binding elements for both neural and anterior-posterior position specific transcription factors mediate domains of Pax3 expression.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/biossíntese , Neuropeptídeos/biossíntese , Transativadores/biossíntese , Fatores de Transcrição/biossíntese , Animais , Sítios de Ligação , Linhagem Celular , Núcleo Celular/metabolismo , DNA/metabolismo , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/metabolismo , Hibridização In Situ , Técnicas In Vitro , Camundongos , Camundongos Transgênicos , Mutação , Proteínas do Tecido Nervoso , Crista Neural/embriologia , Neuropeptídeos/química , Fator de Transcrição PAX3 , Fatores do Domínio POU , Fatores de Transcrição Box Pareados , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Estrutura Terciária de Proteína , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Fatores de Tempo , Transativadores/química , Fatores de Transcrição/metabolismo , Transfecção , Tretinoína/farmacologia , beta-Galactosidase/metabolismo
19.
Bioorg Med Chem Lett ; 14(15): 3875-9, 2004 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-15225688

RESUMO

Structure-based-design studies, with the crystal structure of the HOXB1-PBX1/DNA transcription factor complex, were used to identify 1,4-disubstituted naphthalenes as potential antagonists. An initial library of 32 analogs was synthesized, two of which were found to be more potent than the reported activity for a 12 amino acid peptide antagonist. Antagonists were also identified of the related BRN1/DNA and BRN2/DNA transcription factor complexes indicating that a 1,4-disubstituted naphthalene may be a privileged scaffold for preparing screening libraries targeting this family of transcription factor complexes.


Assuntos
Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Homeodomínio/antagonistas & inibidores , Naftalenos/farmacologia , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Fatores de Transcrição/antagonistas & inibidores , Cristalografia por Raios X , DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/química , Proteínas de Homeodomínio/química , Humanos , Cinética , Modelos Moleculares , Naftalenos/síntese química , Naftalenos/química , Fator de Transcrição 1 de Leucemia de Células Pré-B , Proteínas Proto-Oncogênicas/química
20.
J Neurosci ; 24(7): 1726-33, 2004 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-14973255

RESUMO

The mammalian brain contains neural stem cells (NSCs) that allow continued neurogenesis throughout the life of the animal. However, neurogenesis is known to decline during aging and, to the extent that neurogenesis is required for normal CNS function, this may contribute to neurodegenerative disease. Decreased neurogenesis could result from loss of NSCs or dysfunction at some later step, and distinguishing these possibilities is important for understanding the cause of the decline. However, because of the inability to distinguish NSCs from their rapidly dividing progeny in situ, it has not been possible to quantitatively assess the NSC populations in young and old animals. In this report we show that the G1 phase-specific expression of the replication factor Mcm2 is a useful marker for detecting slowly cycling putative NSCs in situ and confirm the identity of these cells using both cytosine beta-D-arabinofuranoside (Ara-C) treatment and a double nucleoside analog-labeling technique. The ability to distinguish NSCs from proliferative progenitors has allowed characterization of the expression of several markers including Nestin, Musashi, and GFAP in these different cell types. Furthermore, comparison of the NSC populations in the subventricular zones of young (2-4 months) and old (24-26 months) mice demonstrates an approximately twofold reduction in the older mice. A similar twofold reduction is also observed in the number of neurospheres recovered in culture from old relative to young animals. The reduction in the neural stem cell population documented here is sufficient to account for the reduced level of neurogenesis in old animals.


Assuntos
Envelhecimento/metabolismo , Desoxiuridina/análogos & derivados , Ventrículos Laterais/citologia , Proteínas do Tecido Nervoso , Neurônios/citologia , Células-Tronco/citologia , Administração Oral , Animais , Antígenos de Diferenciação/biossíntese , Bromodesoxiuridina/farmacocinética , Contagem de Células , Ciclo Celular/fisiologia , Divisão Celular , Células Cultivadas , Citarabina/farmacologia , Desoxiuridina/administração & dosagem , Desoxiuridina/metabolismo , Desoxiuridina/farmacocinética , Proteína Glial Fibrilar Ácida/biossíntese , Idoxuridina/administração & dosagem , Idoxuridina/metabolismo , Idoxuridina/farmacocinética , Proteínas de Filamentos Intermediários/biossíntese , Masculino , Camundongos , Componente 2 do Complexo de Manutenção de Minicromossomo , Nestina , Neurônios/metabolismo , Proteínas Nucleares/biossíntese , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...