Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 162
Filtrar
1.
Nat Immunol ; 15(8): 749-757, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24973821

RESUMO

Interleukin 15 (IL-15) controls both the homeostasis and the peripheral activation of natural killer (NK) cells. The molecular basis for this duality of action remains unknown. Here we found that the metabolic checkpoint kinase mTOR was activated and boosted bioenergetic metabolism after exposure of NK cells to high concentrations of IL-15, whereas low doses of IL-15 triggered only phosphorylation of the transcription factor STAT5. mTOR stimulated the growth and nutrient uptake of NK cells and positively fed back on the receptor for IL-15. This process was essential for sustaining NK cell proliferation during development and the acquisition of cytolytic potential during inflammation or viral infection. The mTORC1 inhibitor rapamycin inhibited NK cell cytotoxicity both in mice and humans; this probably contributes to the immunosuppressive activity of this drug in different clinical settings.


Assuntos
Interleucina-15/imunologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Serina-Treonina Quinases TOR/imunologia , Animais , Proliferação de Células , Células Cultivadas , Infecções por Herpesviridae/imunologia , Humanos , Imunossupressores/farmacologia , Inflamação/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Células Matadoras Naturais/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/genética , Complexos Multiproteicos/imunologia , Muromegalovirus/imunologia , Infecções por Orthomyxoviridae/imunologia , Poli I-C/imunologia , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/imunologia , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/genética
2.
EMBO Rep ; 25(3): 1650-1684, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38424230

RESUMO

Lung diseases develop when telomeres shorten beyond a critical point. We constructed a mouse model in which the catalytic subunit of telomerase (mTert), or its catalytically inactive form (mTertCI), is expressed from the p21Cdkn1a locus. Expression of either TERT or TERTCI reduces global p21 levels in the lungs of aged mice, highlighting TERT non-canonical function. However, only TERT reduces accumulation of very short telomeres, oxidative damage, endothelial cell (ECs) senescence and senile emphysema in aged mice. Single-cell analysis of the lung reveals that p21 (and hence TERT) is expressed mainly in the capillary ECs. We report that a fraction of capillary ECs marked by CD34 and endowed with proliferative capacity declines drastically with age, and this is counteracted by TERT but not TERTCI. Consistently, only TERT counteracts decline of capillary density. Natural aging effects are confirmed using the experimental model of emphysema induced by VEGFR2 inhibition and chronic hypoxia. We conclude that catalytically active TERT prevents exhaustion of the putative CD34 + EC progenitors with age, thus protecting against capillary vessel loss and pulmonary emphysema.


Assuntos
Enfisema , Rarefação Microvascular , Enfisema Pulmonar , Telomerase , Camundongos , Animais , Encurtamento do Telômero , Telomerase/genética
3.
Nature ; 587(7834): 420-425, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33177709

RESUMO

Genome introgressions drive evolution across the animal1, plant2 and fungal3 kingdoms. Introgressions initiate from archaic admixtures followed by repeated backcrossing to one parental species. However, how introgressions arise in reproductively isolated species, such as yeast4, has remained unclear. Here we identify a clonal descendant of the ancestral yeast hybrid that founded the extant Saccharomyces cerevisiae Alpechin lineage5, which carries abundant Saccharomyces paradoxus introgressions. We show that this clonal descendant, hereafter defined as a 'living ancestor', retained the ancestral genome structure of the first-generation hybrid with contiguous S. cerevisiae and S. paradoxus subgenomes. The ancestral first-generation hybrid underwent catastrophic genomic instability through more than a hundred mitotic recombination events, mainly manifesting as homozygous genome blocks generated by loss of heterozygosity. These homozygous sequence blocks rescue hybrid fertility by restoring meiotic recombination and are the direct origins of the introgressions present in the Alpechin lineage. We suggest a plausible route for introgression evolution through the reconstruction of extinct stages and propose that genome instability allows hybrids to overcome reproductive isolation and enables introgressions to emerge.


Assuntos
Evolução Molecular , Introgressão Genética/genética , Genoma Fúngico/genética , Genômica , Filogenia , Saccharomyces cerevisiae/genética , Saccharomyces/genética , Cruzamentos Genéticos , Fertilidade/genética , Aptidão Genética/genética , Instabilidade Genômica/genética , Recombinação Homóloga/genética , Perda de Heterozigosidade/genética , Meiose/genética , Mitose/genética , Reprodução Assexuada/genética , Saccharomyces/classificação , Saccharomyces/citologia , Saccharomyces cerevisiae/classificação , Saccharomyces cerevisiae/citologia
4.
Mol Cell ; 70(3): 449-461.e5, 2018 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-29727617

RESUMO

Hard-to-replicate regions of chromosomes (e.g., pericentromeres, centromeres, and telomeres) impede replication fork progression, eventually leading, in the event of replication stress, to chromosome fragility, aging, and cancer. Our knowledge of the mechanisms controlling the stability of these regions is essentially limited to telomeres, where fragility is counteracted by the shelterin proteins. Here we show that the shelterin subunit TRF2 ensures progression of the replication fork through pericentromeric heterochromatin, but not centromeric chromatin. In a process involving its N-terminal basic domain, TRF2 binds to pericentromeric Satellite III sequences during S phase, allowing the recruitment of the G-quadruplex-resolving helicase RTEL1 to facilitate fork progression. We also show that TRF2 is required for the stability of other heterochromatic regions localized throughout the genome, paving the way for future research on heterochromatic replication and its relationship with aging and cancer.


Assuntos
Replicação do DNA/genética , Genoma/genética , Heterocromatina/genética , Telômero/genética , Proteína 2 de Ligação a Repetições Teloméricas/genética , Linhagem Celular Tumoral , Centrômero/genética , Cromatina/genética , DNA Helicases/genética , Quadruplex G , Células HeLa , Humanos , Fase S/genética
5.
EMBO J ; 40(9): e108164, 2021 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-33880795

RESUMO

Cellular senescence is considered to be a major driver of aging, yet the mechanisms explaining the accumulation of senescent cells during life time remain unclear. In this issue, Lagnado et al (2021) show that neutrophils can trigger the senescence of neighboring cells by transmitting reactive oxygen species (ROS), which they normally produce to fight pathogens. The main genomic targets of the neutrophil-mediated ROS damage are telomeres, supporting an intimate interplay between telomere homeostasis and oxidative stress in senescence and consequently aging.


Assuntos
Senescência Celular , Neutrófilos , Senescência Celular/genética , Estresse Oxidativo , Espécies Reativas de Oxigênio , Telômero , Homeostase do Telômero
6.
Cell ; 142(2): 230-42, 2010 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-20655466

RESUMO

Human telomeres are protected from DNA damage by a nucleoprotein complex that includes the repeat-binding factor TRF2. Here, we report that TRF2 regulates the 5' exonuclease activity of its binding partner, Apollo, a member of the metallo-beta-lactamase family that is required for telomere integrity during S phase. TRF2 and Apollo also suppress damage to engineered interstitial telomere repeat tracts that were inserted far away from chromosome ends. Genetic data indicate that DNA topoisomerase 2alpha acts in the same pathway of telomere protection as TRF2 and Apollo. Moreover, TRF2, which binds preferentially to positively supercoiled DNA substrates, together with Apollo, negatively regulates the amount of TOP1, TOP2alpha, and TOP2beta at telomeres. Our data are consistent with a model in which TRF2 and Apollo relieve topological stress during telomere replication. Our work also suggests that cellular senescence may be caused by topological problems that occur during the replication of the inner portion of telomeres.


Assuntos
Antígenos de Neoplasias/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Replicação do DNA , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Telômero/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Senescência Celular , Dano ao DNA , Exodesoxirribonucleases , Humanos , Estrutura Terciária de Proteína
7.
Mol Cell ; 68(4): 643-644, 2017 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-29149592

RESUMO

In this issue of Molecular Cell, Kim et al. (2017) have studied the structure and organization of the shelterin protein complex protecting telomeres in Schizosaccharomyces pombe and humans and discovered an allosteric structural transition that drives the formation of the shelterin complex and participates in telomere length regulation.


Assuntos
Homeostase do Telômero/fisiologia , Proteínas de Ligação a Telômeros/metabolismo , Telômero/metabolismo , Humanos , Schizosaccharomyces , Complexo Shelterina , Telômero/genética , Proteínas de Ligação a Telômeros/genética
8.
Kidney Int ; 105(5): 980-996, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38423182

RESUMO

Collapsing focal segmental glomerulosclerosis (FSGS), also known as collapsing glomerulopathy (CG), is the most aggressive variant of FSGS and is characterized by a rapid progression to kidney failure. Understanding CG pathogenesis represents a key step for the development of targeted therapies. Previous work implicated the telomerase protein component TERT in CG pathogenesis, as transgenic TERT expression in adult mice resulted in a CG resembling that seen in human primary CG and HIV-associated nephropathy (HIVAN). Here, we used the telomerase-induced mouse model of CG (i-TERTci mice) to identify mechanisms to inhibit CG pathogenesis. Inactivation of WIP1 phosphatase, a p53 target acting in a negative feedback loop, blocked disease initiation in i-TERTci mice. Repression of disease initiation upon WIP1 deficiency was associated with senescence enhancement and required transforming growth factor-ß functions. The efficacy of a pharmacologic treatment to reduce disease severity in both i-TERTci mice and in a mouse model of HIVAN (Tg26 mice) was then assessed. Pharmacologic inhibition of WIP1 enzymatic activity in either the telomerase mice with CG or in the Tg26 mice promoted partial remission of proteinuria and ameliorated kidney histopathologic features. Histological as well as high-throughput sequencing methods further showed that selective inhibition of WIP1 does not promote kidney fibrosis or inflammation. Thus, our findings suggest that targeting WIP1 may be an effective therapeutic strategy for patients with CG.


Assuntos
Nefropatia Associada a AIDS , Glomerulosclerose Segmentar e Focal , Insuficiência Renal , Telomerase , Adulto , Humanos , Camundongos , Animais , Glomerulosclerose Segmentar e Focal/patologia , Telomerase/uso terapêutico , Nefropatia Associada a AIDS/patologia , Proteinúria , Insuficiência Renal/complicações , Modelos Animais de Doenças
9.
Mol Cell ; 61(2): 274-86, 2016 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-26774283

RESUMO

The shelterin proteins protect telomeres against activation of the DNA damage checkpoints and recombinational repair. We show here that a dimer of the shelterin subunit TRF2 wraps ∼ 90 bp of DNA through several lysine and arginine residues localized around its homodimerization domain. The expression of a wrapping-deficient TRF2 mutant, named Top-less, alters telomeric DNA topology, decreases the number of terminal loops (t-loops), and triggers the ATM checkpoint, while still protecting telomeres against non-homologous end joining (NHEJ). In Top-less cells, the protection against NHEJ is alleviated if the expression of the TRF2-interacting protein RAP1 is reduced. We conclude that a distinctive topological state of telomeric DNA, controlled by the TRF2-dependent DNA wrapping and linked to t-loop formation, inhibits both ATM activation and NHEJ. The presence of RAP1 at telomeres appears as a backup mechanism to prevent NHEJ when topology-mediated telomere protection is impaired.


Assuntos
DNA/química , Conformação de Ácido Nucleico , Telômero/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Pareamento de Bases , DNA/metabolismo , Dano ao DNA , Reparo do DNA por Junção de Extremidades , Células HeLa , Humanos , Lisina/metabolismo , Modelos Moleculares , Mutação , Estrutura Terciária de Proteína , Complexo Shelterina , Transdução de Sinais , Proteínas de Ligação a Telômeros/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/química
10.
Nucleic Acids Res ; 50(4): 2081-2095, 2022 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-35150283

RESUMO

The shelterin protein complex is required for telomere protection in various eukaryotic organisms. In mammals, the shelterin subunit TRF2 is specialized in preventing ATM activation at telomeres and chromosome end fusion in somatic cells. Here, we demonstrate that the zebrafish ortholog of TRF2 (encoded by the terfa gene) is protecting against unwanted ATM activation genome-wide. The terfa-compromised fish develop a prominent and specific embryonic neurodevelopmental failure. The heterozygous fish survive to adulthood but exhibit a premature aging phenotype. The recovery from embryonic neurodevelopmental failure requires both ATM inhibition and transcriptional complementation of neural genes. Furthermore, restoring the expression of TRF2 in glial cells rescues the embryonic neurodevelopment phenotype. These results indicate that the shelterin subunit TRF2 evolved in zebrafish as a general factor of genome maintenance and transcriptional regulation that is required for proper neurodevelopment and normal aging. These findings uncover how TRF2 links development to aging by separate functions in gene expression regulation and genome stability control.


Assuntos
Proteína 2 de Ligação a Repetições Teloméricas , Peixe-Zebra , Envelhecimento/genética , Animais , Mamíferos/genética , Complexo Shelterina , Telômero , Proteína 2 de Ligação a Repetições Teloméricas/genética , Peixe-Zebra/genética
11.
Nucleic Acids Res ; 50(13): 7493-7510, 2022 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-35819196

RESUMO

Cellular senescence triggers various types of heterochromatin remodeling that contribute to aging. However, the age-related mechanisms that lead to these epigenetic alterations remain elusive. Here, we asked how two key aging hallmarks, telomere shortening and constitutive heterochromatin loss, are mechanistically connected during senescence. We show that, at the onset of senescence, pericentromeric heterochromatin is specifically dismantled consisting of chromatin decondensation, accumulation of DNA breakages, illegitimate recombination and loss of DNA. This process is caused by telomere shortening or genotoxic stress by a sequence of events starting from TP53-dependent downregulation of the telomere protective protein TRF2. The resulting loss of TRF2 at pericentromeres triggers DNA breaks activating ATM, which in turn leads to heterochromatin decondensation by releasing KAP1 and Lamin B1, recombination and satellite DNA excision found in the cytosol associated with cGAS. This TP53-TRF2 axis activates the interferon response and the formation of chromosome rearrangements when the cells escape the senescent growth arrest. Overall, these results reveal the role of TP53 as pericentromeric disassembler and define the basic principles of how a TP53-dependent senescence inducer hierarchically leads to selective pericentromeric dismantling through the downregulation of TRF2.


Assuntos
Senescência Celular , Centrômero , Heterocromatina , Encurtamento do Telômero , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular , Cromatina , Dano ao DNA , Regulação para Baixo , Células HeLa , Humanos , Telômero/genética , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo
12.
EMBO J ; 38(11)2019 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-31000523

RESUMO

Myeloid-derived suppressor cells (MDSCs) are immature myeloid cells with strong immunosuppressive activity that promote tumor growth. In this study, we describe a mechanism by which cancer cells control MDSCs in human cancers by upregulating TRF2, a protein required for telomere stability. Specifically, we showed that the TRF2 upregulation in cancer cells has extratelomeric roles in activating the expression of a network of genes involved in the biosynthesis of heparan sulfate proteoglycan, leading to profound changes in glycocalyx length and stiffness, as revealed by atomic force microscopy. This TRF2-dependent regulation facilitated the recruitment of MDSCs, their activation via the TLR2/MyD88/IL-6/STAT3 pathway leading to the inhibition of natural killer recruitment and cytotoxicity, and ultimately tumor progression and metastasis. The clinical relevance of these findings is supported by our analysis of cancer cohorts, which showed a correlation between high TRF2 expression and MDSC infiltration, which was inversely correlated with overall patient survival.


Assuntos
Glicocálix/metabolismo , Neoplasias/imunologia , Neoplasias/patologia , Proteína 2 de Ligação a Repetições Teloméricas/fisiologia , Evasão Tumoral/fisiologia , Animais , Células Cultivadas , Feminino , Regulação Neoplásica da Expressão Gênica , Glicocálix/genética , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Células Supressoras Mieloides/metabolismo , Células Supressoras Mieloides/fisiologia , Células NIH 3T3 , Neoplasias/genética , Neoplasias/mortalidade , Telômero/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/genética , Evasão Tumoral/genética
13.
Mol Ecol ; 31(23): 6087-6099, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-34587336

RESUMO

Telomere DNA length is a complex trait controlled by both multiple loci and environmental factors. A growing number of studies are focusing on the impact of stress and stress accumulation on telomere length and the link with survival and fitness in ecological contexts. Here, we investigated the telomere changes occurring in a symbiotic coral, Stylophora pistillata, that has experienced continuous darkness over 6 months. This stress condition led to the loss of its symbionts in a similar manner to that observed during large-scale bleaching events due to climate changes and anthropogenic activities, threatening reef ecosystems worldwide. We found that continuous darkness was associated with telomere length shortening. This result, together with a phylogenetic analysis of the telomere coral proteins and a transcriptome survey of the continuous darkness condition, paves the way for future studies on the role of telomeres in the coral stress response and the importance of environmentally induced telomere shortening in endangered coral species.


Assuntos
Antozoários , Animais , Antozoários/genética , Ecossistema , Filogenia , Recifes de Corais , Simbiose/genética
14.
PLoS Biol ; 17(9): e3000483, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31545807

RESUMO

Coral reefs are the most diverse habitats in the marine realm. Their productivity, structural complexity, and biodiversity critically depend on ecosystem services provided by corals that are threatened because of climate change effects-in particular, ocean warming and acidification. The coral holobiont is composed of the coral animal host, endosymbiotic dinoflagellates, associated viruses, bacteria, and other microeukaryotes. In particular, the mandatory photosymbiosis with microalgae of the family Symbiodiniaceae and its consequences on the evolution, physiology, and stress resilience of the coral holobiont have yet to be fully elucidated. The functioning of the holobiont as a whole is largely unknown, although bacteria and viruses are presumed to play roles in metabolic interactions, immunity, and stress tolerance. In the context of climate change and anthropogenic threats on coral reef ecosystems, the Tara Pacific project aims to provide a baseline of the "-omics" complexity of the coral holobiont and its ecosystem across the Pacific Ocean and for various oceanographically distinct defined areas. Inspired by the previous Tara Oceans expeditions, the Tara Pacific expedition (2016-2018) has applied a pan-ecosystemic approach on coral reefs throughout the Pacific Ocean, drawing an east-west transect from Panama to Papua New Guinea and a south-north transect from Australia to Japan, sampling corals throughout 32 island systems with local replicates. Tara Pacific has developed and applied state-of-the-art technologies in very-high-throughput genetic sequencing and molecular analysis to reveal the entire microbial and chemical diversity as well as functional traits associated with coral holobionts, together with various measures on environmental forcing. This ambitious project aims at revealing a massive amount of novel biodiversity, shedding light on the complex links between genomes, transcriptomes, metabolomes, organisms, and ecosystem functions in coral reefs and providing a reference of the biological state of modern coral reefs in the Anthropocene.


Assuntos
Antozoários/microbiologia , Recifes de Corais , Expedições , Microbiota , Animais , Metabolômica , Metagenômica , Oceano Pacífico , Simbiose
15.
EMBO Rep ; 21(4): e49076, 2020 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-32096305

RESUMO

Repressor/activator protein 1 (RAP1) is a highly evolutionarily conserved protein found at telomeres. Although yeast Rap1 is a key telomere capping protein preventing non-homologous end joining (NHEJ) and consequently telomere fusions, its role at mammalian telomeres in vivo is still controversial. Here, we demonstrate that RAP1 is required to protect telomeres in replicative senescent human cells. Downregulation of RAP1 in these cells, but not in young or dividing pre-senescent cells, leads to telomere uncapping and fusions. The anti-fusion effect of RAP1 was further explored in a HeLa cell line where RAP1 expression was depleted through an inducible CRISPR/Cas9 strategy. Depletion of RAP1 in these cells gives rise to telomere fusions only when telomerase is inhibited. We further show that the fusions triggered by RAP1 loss are dependent upon DNA ligase IV. We conclude that human RAP1 is specifically involved in protecting critically short telomeres. This has important implications for the functions of telomeres in senescent cells.


Assuntos
Telômero , Fator de Transcrição AP-1 , Animais , Senescência Celular/genética , Dano ao DNA , Células HeLa , Humanos , Telômero/genética , Proteínas de Ligação a Telômeros/genética
16.
Nucleic Acids Res ; 47(7): 3365-3382, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-30698737

RESUMO

The telomeric protein TRF2 is overexpressed in several human malignancies and contributes to tumorigenesis even though the molecular mechanism is not completely understood. By using a high-throughput approach based on the multiplexed Luminex X-MAP technology, we demonstrated that TRF2 dramatically affects VEGF-A level in the secretome of cancer cells, promoting endothelial cell-differentiation and angiogenesis. The pro-angiogenic effect of TRF2 is independent from its role in telomere capping. Instead, TRF2 binding to a distal regulatory element promotes the expression of SULF2, an endoglucosamine-6-sulfatase that impairs the VEGF-A association to the plasma membrane by inducing post-synthetic modification of heparan sulfate proteoglycans (HSPGs). Finally, we addressed the clinical relevance of our findings showing that TRF2/SULF2 expression is a worse prognostic biomarker in colorectal cancer (CRC) patients.


Assuntos
Neoplasias do Colo/metabolismo , Sulfotransferases/genética , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Microambiente Tumoral , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/irrigação sanguínea , Neoplasias do Colo/patologia , Proteoglicanas de Heparan Sulfato/química , Proteoglicanas de Heparan Sulfato/metabolismo , Heparina/metabolismo , Humanos , Masculino , Camundongos , Camundongos Nus , Metástase Neoplásica , Neovascularização Patológica , Sulfatases , Sulfotransferases/biossíntese , Proteína 2 de Ligação a Repetições Teloméricas/deficiência , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Nat Rev Genet ; 15(7): 491-503, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24913665

RESUMO

Telomeres protect chromosome ends from degradation and inappropriate DNA damage response activation through their association with specific factors. Interestingly, these telomeric factors are able to localize outside telomeric regions, where they can regulate the transcription of genes involved in metabolism, immunity and differentiation. These findings delineate a signalling pathway by which telomeric changes control the ability of their associated factors to regulate transcription. This mechanism is expected to enable a greater diversity of cellular responses that are adapted to specific cell types and telomeric changes, and may therefore represent a pivotal aspect of development, ageing and telomere-mediated diseases.


Assuntos
Reparo do DNA , Transdução de Sinais/genética , Telômero/química , Transcrição Gênica , Apoptose , Senescência Celular , Dano ao DNA , Regulação da Expressão Gênica , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Complexo Shelterina , Telomerase/genética , Telomerase/metabolismo , Telômero/metabolismo , Proteínas de Ligação a Telômeros/genética , Proteínas de Ligação a Telômeros/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/genética , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
19.
Nat Rev Mol Cell Biol ; 8(10): 825-38, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17885666

RESUMO

The replication of the ends of linear chromosomes, or telomeres, poses unique problems, which must be solved to maintain genome integrity and to allow cell division to occur. Here, we describe and compare the timing and specific mechanisms that are required to initiate, control and coordinate synthesis of the leading and lagging strands at telomeres in yeasts, ciliates and mammals. Overall, it emerges that telomere replication relies on a strong synergy between the conventional replication machinery, telomere protection systems, DNA-damage-response pathways and chromosomal organization.


Assuntos
Telomerase/genética , Telômero/genética , Animais , Período de Replicação do DNA/genética , Humanos , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética
20.
Nucleic Acids Res ; 45(4): 1820-1834, 2017 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-27923994

RESUMO

Telomere repeat binding factor 2 (TRF2) has been increasingly recognized to be involved in telomere maintenance and DNA damage response. Here, we show that TRF2 directly binds SIRT6 in a DNA independent manner and that this interaction is increased upon replication stress. Knockdown of SIRT6 up-regulates TRF2 protein levels and counteracts its down-regulation during DNA damage response, leading to cell survival. Moreover, we report that SIRT6 deactetylates in vivo the TRFH domain of TRF2, which in turn, is ubiquitylated in vivo activating the ubiquitin-dependent proteolysis. Notably, overexpression of the TRF2cT mutant failed to be stabilized by SIRT6 depletion, demonstrating that the TRFH domain is required for its post-transcriptional modification. Finally, we report an inverse correlation between SIRT6 and TRF2 protein expression levels in a cohort of colon rectal cancer patients. Taken together our findings describe TRF2 as a novel SIRT6 substrate and demonstrate that acetylation of TRF2 plays a crucial role in the regulation of TRF2 protein stability, thus providing a new route for modulating its expression level during oncogenesis and damage response.


Assuntos
Dano ao DNA , Sirtuínas/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Acetilação , Antineoplásicos Fitogênicos/farmacologia , Camptotecina/farmacologia , Linhagem Celular , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Humanos , Imuno-Histoquímica , Modelos Moleculares , Poli(ADP-Ribose) Polimerases/metabolismo , Ligação Proteica , Conformação Proteica , Estabilidade Proteica , Proteólise/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo , Sirtuínas/química , Especificidade por Substrato , Proteína 2 de Ligação a Repetições Teloméricas/química , Proteína 2 de Ligação a Repetições Teloméricas/genética , Ubiquitinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA