Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(25): e2409269121, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38870055

RESUMO

Sirtuin 7 (SIRT7) is a member of the mammalian family of nicotinamide adenine dinucleotide (NAD+)-dependent histone/protein deacetylases, known as sirtuins. It acts as a potent oncogene in numerous malignancies, but the molecular mechanisms employed by SIRT7 to sustain lung cancer progression remain largely uncharacterized. We demonstrate that SIRT7 exerts oncogenic functions in lung cancer cells by destabilizing the tumor suppressor alternative reading frame (ARF). SIRT7 directly interacts with ARF and prevents binding of ARF to nucleophosmin, thereby promoting proteasomal-dependent degradation of ARF. We show that SIRT7-mediated degradation of ARF increases expression of protumorigenic genes and stimulates proliferation of non-small-cell lung cancer (NSCLC) cells both in vitro and in vivo in a mouse xenograft model. Bioinformatics analysis of transcriptome data from human lung adenocarcinomas revealed a correlation between SIRT7 expression and increased activity of genes normally repressed by ARF. We propose that disruption of SIRT7-ARF signaling stabilizes ARF and thus attenuates cancer cell proliferation, offering a strategy to mitigate NSCLC progression.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Proliferação de Células , Progressão da Doença , Neoplasias Pulmonares , Sirtuínas , Humanos , Sirtuínas/metabolismo , Sirtuínas/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Animais , Camundongos , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Regulação Neoplásica da Expressão Gênica , Linhagem Celular Tumoral
2.
Nucleic Acids Res ; 51(13): 6754-6769, 2023 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-37309898

RESUMO

The Sirtuin family of NAD+-dependent enzymes plays an important role in maintaining genome stability upon stress. Several mammalian Sirtuins have been linked directly or indirectly to the regulation of DNA damage during replication through Homologous recombination (HR). The role of one of them, SIRT1, is intriguing as it seems to have a general regulatory role in the DNA damage response (DDR) that has not yet been addressed. SIRT1-deficient cells show impaired DDR reflected in a decrease in repair capacity, increased genome instability and decreased levels of γH2AX. Here we unveil a close functional antagonism between SIRT1 and the PP4 phosphatase multiprotein complex in the regulation of the DDR. Upon DNA damage, SIRT1 interacts specifically with the catalytical subunit PP4c and promotes its inhibition by deacetylating the WH1 domain of the regulatory subunits PP4R3α/ß. This in turn regulates γH2AX and RPA2 phosphorylation, two key events in the signaling of DNA damage and repair by HR. We propose a mechanism whereby during stress, SIRT1 signaling ensures a global control of DNA damage signaling through PP4.


Assuntos
Dano ao DNA , Sirtuína 1 , Animais , Humanos , Mamíferos/metabolismo , Monoéster Fosfórico Hidrolases , Fosforilação , Transdução de Sinais , Sirtuína 1/metabolismo
3.
Nucleic Acids Res ; 50(3): 1449-1464, 2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35061900

RESUMO

DNA methylation modulates telomere function. In Arabidopsis thaliana, telomeric regions have a bimodal chromatin organization with unmethylated telomeres and methylated subtelomeres. To gain insight into this organization we have generated TAIR10-Tel, a modified version of the Arabidopsis reference genome with additional sequences at most chromosome ends. TAIR10-Tel has allowed us to analyse DNA methylation at nucleotide resolution level in telomeric regions. We have analysed the wild-type strain and mutants that encode inactive versions of all currently known relevant methyltransferases involved in cytosine methylation. These analyses have revealed that subtelomeric DNA methylation extends 1 to 2 kbp from Interstitial Telomeric Sequences (ITSs) that abut or are very near to telomeres. However, DNA methylation drops at the telomeric side of the telomere-subtelomere boundaries and disappears at the inner part of telomeres. We present a comprehensive and integrative model for subtelomeric DNA methylation that should help to decipher the mechanisms that govern the epigenetic regulation of telomeres. This model involves a complex network of interactions between methyltransferases and subtelomeric DNA sequences.


Assuntos
Arabidopsis , Metilação de DNA , Arabidopsis/genética , Epigênese Genética , Metiltransferases/genética , Telômero/genética
4.
Proc Natl Acad Sci U S A ; 118(5)2021 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-33495326

RESUMO

Adaptation to different forms of environmental stress is crucial for maintaining essential cellular functions and survival. The nucleolus plays a decisive role as a signaling hub for coordinating cellular responses to various extrinsic and intrinsic cues. p53 levels are normally kept low in unstressed cells, mainly due to E3 ubiquitin ligase MDM2-mediated degradation. Under stress, nucleophosmin (NPM) relocates from the nucleolus to the nucleoplasm and binds MDM2, thereby preventing degradation of p53 and allowing cell-cycle arrest and DNA repair. Here, we demonstrate that the mammalian sirtuin SIRT7 is an essential component for the regulation of p53 stability during stress responses induced by ultraviolet (UV) irradiation. The catalytic activity of SIRT7 is substantially increased upon UV irradiation through ataxia telangiectasia mutated and Rad3 related (ATR)-mediated phosphorylation, which promotes efficient deacetylation of the SIRT7 target NPM. Deacetylation is required for stress-dependent relocation of NPM into the nucleoplasm and MDM2 binding, thereby preventing ubiquitination and degradation of p53. In the absence of SIRT7, stress-dependent stabilization of p53 is abrogated, both in vitro and in vivo, impairing cellular stress responses. The study uncovers an essential SIRT7-dependent mechanism for stabilization of the tumor suppressor p53 in response to genotoxic stress.


Assuntos
Dano ao DNA , Proteínas Nucleares/metabolismo , Sirtuínas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Raios Ultravioleta , Acetilação/efeitos da radiação , Animais , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Catálise/efeitos da radiação , Linhagem Celular Tumoral , Nucléolo Celular/metabolismo , Nucléolo Celular/efeitos da radiação , Humanos , Lisina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Nucleofosmina , Fosforilação/efeitos da radiação , Estabilidade Proteica/efeitos da radiação , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Transcrição Gênica/efeitos da radiação , Ubiquitinação/efeitos da radiação
5.
Int J Mol Sci ; 25(5)2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38474102

RESUMO

Histone deacetylase SIRT1 represses gene expression through the deacetylation of histones and transcription factors and is involved in the protective cell response to stress and aging. However, upon endoplasmic reticulum (ER) stress, SIRT1 impairs the IRE1α branch of the unfolded protein response (UPR) through the inhibition of the transcriptional activity of XBP-1 and SIRT1 deficiency is beneficial under these conditions. We hypothesized that SIRT1 deficiency may unlock the blockade of transcription factors unrelated to the UPR promoting the synthesis of chaperones and improving the stability of immature proteins or triggering the clearance of unfolded proteins. SIRT1+/+ and SIRT1-/- fibroblasts were exposed to the ER stress inducer tunicamycin and cell survival and expression of heat shock proteins were analyzed 24 h after the treatment. We observed that SIRT1 loss significantly reduced cell sensitivity to ER stress and showed that SIRT1-/- but not SIRT1+/+ cells constitutively expressed high levels of phospho-STAT3 and heat shock proteins. Hsp70 silencing in SIRT1-/- cells abolished the resistance to ER stress. Furthermore, accumulation of ubiquitinated proteins was lower in SIRT1-/- than in SIRT1+/+ cells. Our data showed that SIRT1 deficiency enabled chaperones upregulation and boosted the proteasome activity, two processes that are beneficial for coping with ER stress.


Assuntos
Proteínas de Choque Térmico , Sirtuína 1 , Proteínas de Choque Térmico/metabolismo , Regulação para Cima , Sirtuína 1/metabolismo , Endorribonucleases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Estresse do Retículo Endoplasmático , Resposta a Proteínas não Dobradas , Chaperonas Moleculares/metabolismo , Fatores de Transcrição/metabolismo
6.
Stem Cells ; 39(7): 866-881, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33621399

RESUMO

A key challenge for clinical application of induced pluripotent stem cells (iPSC) to accurately model and treat human pathologies depends on developing a method to generate genetically stable cells to reduce long-term risks of cell transplant therapy. Here, we hypothesized that CYCLIN D1 repairs DNA by highly efficient homologous recombination (HR) during reprogramming to iPSC that reduces genetic instability and threat of neoplastic growth. We adopted a synthetic mRNA transfection method using clinically compatible conditions with CYCLIN D1 plus base factors (OCT3/4, SOX2, KLF4, LIN28) and compared with methods that use C-MYC. We demonstrate that CYCLIN D1 made iPSC have (a) lower multitelomeric signal, (b) reduced double-strand DNA breaks, (c) correct nuclear localization of RAD51 protein expression, and (d) reduced single-nucleotide polymorphism (SNP) changes per chromosome, compared with the classical reprogramming method using C-MYC. CYCLIN D1 iPSC have reduced teratoma Ki67 cell growth kinetics and derived neural stem cells successfully engraft in a hostile spinal cord injury (SCI) microenvironment with efficient survival, differentiation. We demonstrate that CYCLIN D1 promotes double-stranded DNA damage repair predominantly through HR during cell reprogramming to efficiently produce iPSC. CYCLIN D1 reduces general cell stress associated with significantly lower SIRT1 gene expression and can rescue Sirt1 null mouse cell reprogramming. In conclusion, we show synthetic mRNA transfection of CYCLIN D1 repairs DNA during reprogramming resulting in significantly improved genetically stable footprint in human iPSC, enabling a new cell reprogramming method for more accurate and reliable generation of human iPSC for disease modeling and future clinical applications.


Assuntos
Células-Tronco Pluripotentes Induzidas , Animais , Diferenciação Celular , Reprogramação Celular/genética , Ciclina D1/genética , Ciclina D1/metabolismo , Reparo do DNA/genética , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
7.
Nucleic Acids Res ; 48(2): 665-681, 2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31799621

RESUMO

Sirtuins 1 and 2 (SIRT1/2) are two NAD-dependent deacetylases with major roles in inflammation. In addition to deacetylating histones and other proteins, SIRT1/2-mediated regulation is coupled with other epigenetic enzymes. Here, we investigate the links between SIRT1/2 activity and DNA methylation in macrophage differentiation due to their relevance in myeloid cells. SIRT1/2 display drastic upregulation during macrophage differentiation and their inhibition impacts the expression of many inflammation-related genes. In this context, SIRT1/2 inhibition abrogates DNA methylation gains, but does not affect demethylation. Inhibition of hypermethylation occurs at many inflammatory loci, which results in more drastic upregulation of their expression upon macrophage polarization following bacterial lipopolysaccharide (LPS) challenge. SIRT1/2-mediated gains of methylation concur with decreases in activating histone marks, and their inhibition revert these histone marks to resemble an open chromatin. Remarkably, specific inhibition of DNA methyltransferases is sufficient to upregulate inflammatory genes that are maintained in a silent state by SIRT1/2. Both SIRT1 and SIRT2 directly interact with DNMT3B, and their binding to proinflammatory genes is lost upon exposure to LPS or through pharmacological inhibition of their activity. In all, we describe a novel role for SIRT1/2 to restrict premature activation of proinflammatory genes.


Assuntos
Metilação de DNA/genética , Inflamação/genética , Sirtuína 1/genética , Sirtuína 2/genética , Acetilação , Diferenciação Celular/genética , Cromatina/genética , Regulação da Expressão Gênica/genética , Histonas/genética , Humanos , Inflamação/induzido quimicamente , Inflamação/patologia , Lipopolissacarídeos/toxicidade , Macrófagos/metabolismo , Regiões Promotoras Genéticas , Ativação Transcricional/genética
8.
Nucleic Acids Res ; 47(15): 7870-7885, 2019 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-31226208

RESUMO

Long interspersed elements-1 (LINE-1, L1) are retrotransposons that hold the capacity of self-propagation in the genome with potential mutagenic outcomes. How somatic cells restrict L1 activity and how this process becomes dysfunctional during aging and in cancer cells is poorly understood. L1s are enriched at lamin-associated domains, heterochromatic regions of the nuclear periphery. Whether this association is necessary for their repression has been elusive. Here we show that the sirtuin family member SIRT7 participates in the epigenetic transcriptional repression of L1 genome-wide in both mouse and human cells. SIRT7 depletion leads to increased L1 expression and retrotransposition. Mechanistically, we identify a novel interplay between SIRT7 and Lamin A/C in L1 repression. Our results demonstrate that SIRT7-mediated H3K18 deacetylation regulates L1 expression and promotes L1 association with elements of the nuclear lamina. The failure of such activity might contribute to the observed genome instability and compromised viability in SIRT7 knockout mice. Overall, our results reveal a novel function of SIRT7 on chromatin organization by mediating the anchoring of L1 to the nuclear envelope, and a new functional link of the nuclear lamina with transcriptional repression.


Assuntos
Genoma , Lamina Tipo A/genética , Elementos Nucleotídeos Longos e Dispersos , Sirtuínas/genética , Transcrição Gênica , Animais , Linhagem Celular , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Epigênese Genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Heterocromatina/química , Heterocromatina/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Células K562 , Lamina Tipo A/metabolismo , Fígado/citologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Miocárdio/citologia , Miocárdio/metabolismo , Lâmina Nuclear/metabolismo , Lâmina Nuclear/ultraestrutura , Sirtuínas/deficiência , Sirtuínas/metabolismo , Testículo/citologia , Testículo/metabolismo
9.
Nucleic Acids Res ; 47(10): 5016-5037, 2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-30923829

RESUMO

Histone H4 acetylation at Lysine 16 (H4K16ac) is a key epigenetic mark involved in gene regulation, DNA repair and chromatin remodeling, and though it is known to be essential for embryonic development, its role during adult life is still poorly understood. Here we show that this lysine is massively hyperacetylated in peripheral neutrophils. Genome-wide mapping of H4K16ac in terminally differentiated blood cells, along with functional experiments, supported a role for this histone post-translational modification in the regulation of cell differentiation and apoptosis in the hematopoietic system. Furthermore, in neutrophils, H4K16ac was enriched at specific DNA repeats. These DNA regions presented an accessible chromatin conformation and were associated with the cleavage sites that generate the 50 kb DNA fragments during the first stages of programmed cell death. Our results thus suggest that H4K16ac plays a dual role in myeloid cells as it not only regulates differentiation and apoptosis, but it also exhibits a non-canonical structural role in poising chromatin for cleavage at an early stage of neutrophil cell death.


Assuntos
Apoptose , Diferenciação Celular , Cromatina/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Células Mieloides/metabolismo , Acetilação , Animais , Células Cultivadas , Cromatina/genética , Epigênese Genética , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/citologia , Processamento de Proteína Pós-Traducional , Transcrição Gênica
10.
Genes Dev ; 27(6): 639-53, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23468428

RESUMO

The establishment of the epigenetic mark H4K20me1 (monomethylation of H4K20) by PR-Set7 during G2/M directly impacts S-phase progression and genome stability. However, the mechanisms involved in the regulation of this event are not well understood. Here we show that SirT2 regulates H4K20me1 deposition through the deacetylation of H4K16Ac (acetylation of H4K16) and determines the levels of H4K20me2/3 throughout the cell cycle. SirT2 binds and deacetylates PR-Set7 at K90, modulating its chromatin localization. Consistently, SirT2 depletion significantly reduces PR-Set7 chromatin levels, alters the size and number of PR-Set7 foci, and decreases the overall mitotic deposition of H4K20me1. Upon stress, the interaction between SirT2 and PR-Set7 increases along with the H4K20me1 levels, suggesting a novel mitotic checkpoint mechanism. SirT2 loss in mice induces significant defects associated with defective H4K20me1-3 levels. Accordingly, SirT2-deficient animals exhibit genomic instability and chromosomal aberrations and are prone to tumorigenesis. Our studies suggest that the dynamic cross-talk between the environment and the genome during mitosis determines the fate of the subsequent cell cycle.


Assuntos
Ciclo Celular/fisiologia , Instabilidade Genômica , Sirtuína 2/metabolismo , Acetilação , Sequência de Aminoácidos , Animais , Transformação Celular Neoplásica/genética , Cromatina/metabolismo , Dano ao DNA/genética , Técnicas de Inativação de Genes , Células HeLa , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Humanos , Pontos de Checagem da Fase M do Ciclo Celular/fisiologia , Metilação , Camundongos , Camundongos Knockout , Mitose , Ligação Proteica , Sirtuína 2/genética
11.
EMBO J ; 35(14): 1488-503, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27225932

RESUMO

Sirtuins, a family of protein deacetylases, promote cellular homeostasis by mediating communication between cells and environment. The enzymatic activity of the mammalian sirtuin SIRT7 targets acetylated lysine in the N-terminal tail of histone H3 (H3K18Ac), thus modulating chromatin structure and transcriptional competency. SIRT7 deletion is associated with reduced lifespan in mice through unknown mechanisms. Here, we show that SirT7-knockout mice suffer from partial embryonic lethality and a progeroid-like phenotype. Consistently, SIRT7-deficient cells display increased replication stress and impaired DNA repair. SIRT7 is recruited in a PARP1-dependent manner to sites of DNA damage, where it modulates H3K18Ac levels. H3K18Ac in turn affects recruitment of the damage response factor 53BP1 to DNA double-strand breaks (DSBs), thereby influencing the efficiency of non-homologous end joining (NHEJ). These results reveal a direct role for SIRT7 in DSB repair and establish a functional link between SIRT7-mediated H3K18 deacetylation and the maintenance of genome integrity.


Assuntos
Dano ao DNA , Reparo do DNA por Junção de Extremidades , DNA/metabolismo , Sirtuínas/metabolismo , Animais , Camundongos Endogâmicos C57BL , Camundongos Knockout
12.
Mol Reprod Dev ; 87(12): 1175-1187, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33184962

RESUMO

Transmission of genetic material through high-quality gametes to progeny requires accurate homologous chromosome recombination and segregation during meiosis. A failure to accomplish these processes can have major consequences in reproductive health, including infertility, and development disorders in offspring. Sirtuins, a family of NAD+ -dependent protein deacetylases and ADP-ribosyltransferases, play key roles in genome maintenance, metabolism, and aging. In recent years, Sirtuins have emerged as regulators of several reproductive processes and interventions aiming to target Sirtuin activity are of great interest in the reproductive biology field. Sirtuins are pivotal to protect germ cells against oxidative stress, a major determinant influencing ovarian aging and the quality of gametes. Sirtuins also safeguard the integrity of the genome through epigenetic programs required for regulating gene repression, DNA repair, and chromosome segregation, among others. Although these functions are relatively well characterized in many somatic tissues, how they contribute to reproductive functions is not well understood. This review summarizes our current knowledge on the role of Sirtuins in female reproductive systems and discusses the underlying molecular pathways. In addition, we highlight the importance of Sirtuins as antiaging factors in the ovary and summarize current preclinical efforts to identify treatments to extend female reproductive longevity.


Assuntos
Envelhecimento/genética , Longevidade/genética , Meiose/genética , Reprodução/genética , Sirtuínas/genética , Sirtuínas/metabolismo , Animais , Feminino , Expressão Gênica , Humanos , Masculino , Camundongos , Oogênese/genética , Ovário/metabolismo
13.
Phys Rev Lett ; 125(4): 041301, 2020 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-32794796

RESUMO

We study the gravitational collapse of axion dark matter fluctuations in the postinflationary scenario, so-called axion miniclusters, with N-body simulations. Largely confirming theoretical expectations, overdensities begin to collapse in the radiation-dominated epoch and form an early distribution of miniclusters with masses up to 10^{-12} M_{⊙}. After matter-radiation equality, ongoing mergers give rise to a steep power-law distribution of minicluster halo masses. The density profiles of well-resolved halos are Navarro-Frenk-White-like to good approximation. The fraction of axion dark matter in these bound structures is ∼0.75 at redshift z=100.

14.
Proc Natl Acad Sci U S A ; 114(40): E8352-E8361, 2017 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-28923965

RESUMO

Sirtuins (Sirt1-Sirt7) are NAD+-dependent protein deacetylases/ADP ribosyltransferases, which play decisive roles in chromatin silencing, cell cycle regulation, cellular differentiation, and metabolism. Different sirtuins control similar cellular processes, suggesting a coordinated mode of action but information about potential cross-regulatory interactions within the sirtuin family is still limited. Here, we demonstrate that Sirt1 requires autodeacetylation to efficiently deacetylate targets such as p53, H3K9, and H4K16. Sirt7 restricts Sirt1 activity by preventing Sirt1 autodeacetylation causing enhanced Sirt1 activity in Sirt7-/- mice. Increased Sirt1 activity in Sirt7-/- mice blocks PPARγ and adipocyte differentiation, thereby diminishing accumulation of white fat. Thus, reduction of Sirt1 activity restores adipogenesis in Sirt7-/- adipocytes in vitro and in vivo. We disclosed a principle controlling Sirt1 activity and uncovered an unexpected complexity in the crosstalk between two different sirtuins. We propose that antagonistic interactions between Sirt1 and Sirt7 are pivotal in controlling the signaling network required for maintenance of adipose tissue.


Assuntos
Adipogenia/fisiologia , Tecido Adiposo Branco/citologia , Tecido Adiposo/citologia , Sirtuína 1/fisiologia , Sirtuínas/fisiologia , Acetilação , Tecido Adiposo/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Catálise , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Conformação Proteica , Transdução de Sinais , Sirtuína 1/química , Sirtuínas/química
15.
Genome Res ; 26(8): 1047-56, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27405804

RESUMO

Cytosine methylation regulates the length and stability of telomeres, which can affect a wide variety of biological features, including cell differentiation, development, or illness. Although it is well established that subtelomeric regions are methylated, the presence of methylated cytosines at telomeres has remained controversial. Here, we have analyzed multiple bisulfite sequencing studies to address the methylation status of Arabidopsis thaliana telomeres. We found that the levels of estimated telomeric DNA methylation varied among studies. Interestingly, we estimated higher levels of telomeric DNA methylation in studies that produced C-rich telomeric strands with lower efficiency. However, these high methylation estimates arose due to experimental limitations of the bisulfite technique. We found a similar phenomenon for mitochondrial DNA: The levels of mitochondrial DNA methylation detected were higher in experiments with lower mitochondrial read production efficiencies. Based on experiments with high telomeric C-rich strand production efficiencies, we concluded that Arabidopsis telomeres are not methylated, which was confirmed by methylation-dependent restriction enzyme analyses. Thus, our studies indicate that telomeres are refractory to de novo DNA methylation by the RNA-directed DNA methylation machinery. This result, together with previously reported data, reveals that subtelomeric DNA methylation controls the homeostasis of telomere length.


Assuntos
Arabidopsis/genética , Metilação de DNA/genética , Homeostase do Telômero/genética , Telômero/genética , Arabidopsis/crescimento & desenvolvimento , Citosina/metabolismo , DNA Mitocondrial/genética , RNA/genética
16.
Mol Cell ; 42(2): 210-23, 2011 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-21504832

RESUMO

Sirtuins are NAD-dependent deacetylases that sense oxidative stress conditions and promote a protective cellular response. The Sirtuin SirT1 is involved in facultative heterochromatin formation through an intimate functional relationship with the H3K9me3 methyltransferase Suv39h1, a chromatin organization protein. However, SirT1 also regulates Suv39h1-dependent constitutive heterochromatin (CH) through an unknown mechanism; interestingly, SirT1 does not significantly localize in these regions. Herein, we report that SirT1 controls global levels of Suv39h1 by increasing its half-life through inhibition of Suv39h1 lysine 87 polyubiquitination by the E3-ubiquitin ligase MDM2. This in turn increases Suv39h1 turnover in CH and ensures genome integrity. Stress conditions that lead to SirT1 upregulation, such as calorie restriction, also induce higher levels of Suv39h1 in a SirT1-dependent manner in vivo. These observations reflect a direct link between oxidative stress response and Suv39h1 and support a dynamic view of heterochromatin, in which its structure adapts to cell physiology.


Assuntos
Montagem e Desmontagem da Cromatina , Instabilidade Genômica , Heterocromatina/metabolismo , Metiltransferases/metabolismo , Estresse Oxidativo , Proteínas Repressoras/metabolismo , Sirtuína 1/metabolismo , Sequência de Aminoácidos , Animais , Restrição Calórica , Estabilidade Enzimática , Células HEK293 , Meia-Vida , Células HeLa , Humanos , Lisina , Masculino , Metiltransferases/genética , Camundongos , Dados de Sequência Molecular , Mutação , Células NIH 3T3 , Poliubiquitina/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/genética , Sirtuína 1/genética , Fatores de Tempo , Transfecção , Ubiquitinação
17.
Proteomics ; 17(13-14)2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28613014

RESUMO

Sirtuins are crucial proteins involved in sensing and coordinating the response to different forms of stress, mainly through NAD+ -dependent deacetylation of proteins. For that reason, sirtuins are directly involved in many human pathologies including cancer, diabetes, cardiovascular and neurodegenerative diseases. SirT7, one of the less well-understood sirtuins, has been associated with ribosome biogenesis, gene expression, metabolism and cancer. Despite the wide range of these functions, only a handful of targets for SirT7 have so far been described. In this issue, Zhang et al. report the first proteomic screening of SirT7 substrates. Using stable isotope labeling with amino acids in cell culture (SILAC), coupled with quantitative mass spectrometry, they have identified a comprehensive list of candidates involved in a variety of functions, ranging from maintenance of chromatin architecture to gene silencing and metabolism. A selected group of these candidates has been validated by in vitro co-immunoprecipitation and deacetylation experiments. Predictive tools have identified additional candidates. The identification of these novel targets not only suggests new ways of understanding the physiological role of SirT7, but also provides new evidence to add to our existing knowledge of the global impact of sirtuins in cell homeostasis.


Assuntos
Marcação por Isótopo/métodos , Proteínas/metabolismo , Proteômica/métodos , Sirtuínas/metabolismo , Acetilação , Cromatina , Humanos , Espectrometria de Massas/métodos , Proteínas/análise
18.
J Hepatol ; 66(5): 1012-1021, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28088579

RESUMO

BACKGROUND & AIMS: Modulation of microRNA expression is a potential treatment for hepatocellular carcinoma (HCC). Therefore, the epigenetically regulated microRNA-449 family (miR-449a, miR-449b, miR-449c) was characterized with regards to its functional effects and target genes in HCC. METHODS: After transfection of miR-449a, miR-449b, and/or miR-449c, tumor-relevant functional effects were analyzed using in vitro assays and a xenograft mouse model. Binding specificities, target genes, and regulated pathways of each miRNA were identified by microarray analyses. Target genes were validated by luciferase reporter assays and expression analyses in vitro. Furthermore, target gene expression was analyzed in 61 primary human HCCs compared to normal liver tissue. RESULTS: Tumor suppressive effects, binding specificities, target genes, and regulated pathways of miR-449a and miR-449b differed from those of miR-449c. Transfection of miR-449a, miR-449b, and/or miR-449c inhibited cell proliferation and migration, induced apoptosis, and reduced tumor growth to different extents. Importantly, miR-449a, miR-449b, and, to a lesser degree, miR-449c directly targeted SOX4, which codes for a transcription factor involved in epithelial-mesenchymal transition and HCC metastasis, and thereby inhibited TGF-ß-mediated cell migration. CONCLUSIONS: This study provides detailed insights into the regulatory network of the epigenetically regulated miRNA-449 family and, for the first time, describes distinct tumor suppressive effects and target specificities of miR-449a, miR-449b, and miR-449c. Our results indicate that particularly miR-449a and miR-449b may be considered for miRNA replacement therapy to prevent HCC progression and metastasis. LAY SUMMARY: In this study, we demonstrated that the microRNA-449 family acts as a tumor suppressor in liver cancer by causing cell death and inhibiting cell migration. These effects are caused by downregulation of the oncogene SOX4, which is frequently overexpressed in liver cancer. We conclude that the microRNA-449 family may be a target for liver cancer therapy.


Assuntos
Carcinoma Hepatocelular/patologia , Movimento Celular , Genes Supressores de Tumor/fisiologia , Neoplasias Hepáticas/patologia , MicroRNAs/fisiologia , Fatores de Transcrição SOXC/genética , Fator de Crescimento Transformador beta/antagonistas & inibidores , Acetilação , Animais , Carcinoma Hepatocelular/terapia , Histonas/metabolismo , Humanos , Neoplasias Hepáticas/terapia , Camundongos , Fator de Crescimento Transformador beta/fisiologia
19.
Genes Dev ; 23(16): 1849-69, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19608767

RESUMO

Since the earliest stages of evolution, organisms have faced the challenge of sensing and adapting to environmental changes for their survival under compromising conditions such as food depletion or stress. Implicit in these responses are mechanisms developed during evolution that include the targeting of chromatin to allow or prevent expression of fundamental genes and to protect genome integrity. Among the different approaches to study these mechanisms, the analysis of the response to a moderate reduction of energy intake, also known as calorie restriction (CR), has become one of the best sources of information regarding the factors and pathways involved in metabolic adaptation from lower to higher eukaryotes. Furthermore, responses to CR are involved in life span regulation-conserved from yeast to mammals-and therefore have garnered major research interest. Herein we review current knowledge of responses to CR at the molecular level and their functional link to chromatin.


Assuntos
Restrição Calórica , Cromatina/metabolismo , Animais , Enzimas/metabolismo , Humanos , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sirtuínas/metabolismo , Fatores de Transcrição/metabolismo
20.
Nucleic Acids Res ; 40(12): 5402-14, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22406835

RESUMO

Histone H1 is an intrinsic component of chromatin, whose important contribution to chromatin structure is well-established in vitro. Little is known, however, about its functional roles in vivo. Here, we have addressed this question in Drosophila, a model system offering many advantages since it contains a single dH1 variant. For this purpose, RNAi was used to efficiently deplete dH1 in flies. Expression-profiling shows that dH1 depletion affects expression of a relatively small number of genes in a regional manner. Furthermore, depletion up-regulates inactive genes, preferentially those located in heterochromatin, while active euchromatic genes are down-regulated, suggesting that the contribution of dH1 to transcription regulation is mainly structural, organizing chromatin for proper gene-expression regulation. Up-regulated genes are remarkably enriched in transposons. In particular, R1/R2 retrotransposons, which specifically integrate in the rDNA locus, are strongly up-regulated. Actually, depletion increases expression of transposon-inserted rDNA copies, resulting in synthesis of aberrant rRNAs and enlarged nucleolus. Concomitantly, dH1-depleted cells accumulate extra-chromosomal rDNA, show increased γH2Av content, stop proliferation and activate apoptosis, indicating that depletion causes genome instability and affects proliferation. Finally, the contributions to maintenance of genome integrity and cell proliferation appear conserved in human hH1s, as their expression rescues proliferation of dH1-depleted cells.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/genética , Inativação Gênica , Instabilidade Genômica , Histonas/fisiologia , Retroelementos , Animais , Proliferação de Células , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Regulação da Expressão Gênica , Genoma de Inseto , Heterocromatina/metabolismo , Histonas/antagonistas & inibidores , Histonas/genética , Humanos , Interferência de RNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA