Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Genes Dev ; 35(1-2): 82-101, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33303642

RESUMO

The establishment and maintenance of chromatin domains shape the epigenetic memory of a cell, with the methylation of histone H3 lysine 9 (H3K9me) defining transcriptionally silent heterochromatin. We show here that the C. elegans SET-25 (SUV39/G9a) histone methyltransferase (HMT), which catalyzes H3K9me1, me2 and me3, can establish repressed chromatin domains de novo, unlike the SETDB1 homolog MET-2. Thus, SET-25 is needed to silence novel insertions of RNA or DNA transposons, and repress tissue-specific genes de novo during development. We identify two partially redundant pathways that recruit SET-25 to its targets. One pathway requires LIN-61 (L3MBTL2), which uses its four MBT domains to bind the H3K9me2 deposited by MET-2. The second pathway functions independently of MET-2 and involves the somatic Argonaute NRDE-3 and small RNAs. This pathway targets primarily highly conserved RNA and DNA transposons. These redundant SET-25 targeting pathways (MET-2-LIN-61-SET-25 and NRDE-3-SET-25) ensure repression of intact transposons and de novo insertions, while MET-2 can act alone to repress simple and satellite repeats. Removal of both pathways in the met-2;nrde-3 double mutant leads to the loss of somatic H3K9me2 and me3 and the synergistic derepression of transposons in embryos, strongly elevating embryonic lethality.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Proteínas Cromossômicas não Histona/metabolismo , Elementos de DNA Transponíveis/genética , Regulação da Expressão Gênica/genética , Heterocromatina/genética , Proteínas de Ligação a RNA/metabolismo , Animais , Proteínas de Caenorhabditis elegans/genética , Proteínas Cromossômicas não Histona/genética , Embrião não Mamífero , Inativação Gênica , Heterocromatina/metabolismo , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Metilação , Mutação , Proteínas de Ligação a RNA/genética
2.
Mol Cell ; 66(2): 247-257.e5, 2017 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-28410996

RESUMO

Recruitment of transcription factors (TFs) to repressed genes in euchromatin is essential to activate new transcriptional programs during cell differentiation. However, recruitment of all TFs, including pioneer factors, is impeded by condensed H3K27me3-containing chromatin. Single-cell and gene-specific analyses revealed that, during the first hours of induction of differentiation of mammalian embryonic stem cells (ESCs), accumulation of the repressive histone mark H3K27me3 is delayed after DNA replication, indicative of a decondensed chromatin structure in all regions of the replicating genome. This delay provides a critical "window of opportunity" for recruitment of lineage-specific TFs to DNA. Increasing the levels of post-replicative H3K27me3 or preventing S phase entry inhibited recruitment of new TFs to DNA and significantly blocked cell differentiation. These findings suggest that recruitment of lineage-specifying TFs occurs soon after replication and is facilitated by a decondensed chromatin structure. This insight may explain the developmental plasticity of stem cells and facilitate their exploitation for therapeutic purposes.


Assuntos
Diferenciação Celular , Linhagem da Célula , Montagem e Desmontagem da Cromatina , Cromatina/metabolismo , Replicação do DNA , DNA/biossíntese , Células-Tronco Embrionárias/metabolismo , Histonas/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Animais , Sítios de Ligação , Plasticidade Celular , Cromatina/química , DNA/química , DNA/genética , Metilação de DNA , Regulação da Expressão Gênica no Desenvolvimento , Histona Desmetilases/metabolismo , Histonas/química , Humanos , Metilação , Camundongos , Proteínas Nucleares/metabolismo , Conformação de Ácido Nucleico , Ligação Proteica , Relação Estrutura-Atividade , Fatores de Tempo , Fatores de Transcrição/genética
3.
Dev Biol ; 466(1-2): 90-98, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32712024

RESUMO

Spatial organization of the genome in the nucleus plays a critical role in development and regulation of transcription. A genomic region that resides at the nuclear periphery is part of the chromatin layer marked with histone H3 lysine 9 dimethyl (H3K9me2), but chromatin reorganization during cell differentiation can cause movement in and out of this nuclear compartment with patterns specific for individual cell fates. Here we describe a CRISPR-based system that allows visualization coupled with forced spatial relocalization of a target genomic locus in live cells. We demonstrate that a specified locus can be tethered to the nuclear periphery through direct binding to a dCas9-Lap2ß fusion protein at the nuclear membrane, or via targeting of a histone methyltransferase (HMT), G9a fused to dCas9, that promotes H3K9me2 labeling and localization to the nuclear periphery. The enzymatic activity of the HMT is sufficient to promote this repositioning, while disruption of the catalytic activity abolishes the localization effect. We further demonstrate that dCas9-G9a-mediated localization to the nuclear periphery is independent of nuclear actin polymerization. Our data suggest a function for epigenetic histone modifying enzymes in spatial chromatin organization and provide a system for tracking and labeling targeted genomic regions in live cells.


Assuntos
Diferenciação Celular , Cromatina/metabolismo , Epigênese Genética , Histona Metiltransferases/metabolismo , Histonas/metabolismo , Processamento de Proteína Pós-Traducional , Cromatina/genética , Células HEK293 , Histona Metiltransferases/genética , Histonas/genética , Humanos
4.
Entropy (Basel) ; 22(11)2020 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-33286995

RESUMO

As humanity grapples with the concept of autonomy for human-machine teams (A-HMTs), unresolved is the necessity for the control of autonomy that instills trust. For non-autonomous systems in states with a high degree of certainty, rational approaches exist to solve, model or control stable interactions; e.g., game theory, scale-free network theory, multi-agent systems, drone swarms. As an example, guided by artificial intelligence (AI, including machine learning, ML) or by human operators, swarms of drones have made spectacular gains in applications too numerous to list (e.g., crop management; mapping, surveillance and fire-fighting systems; weapon systems). But under states of uncertainty or where conflict exists, rational models fail, exactly where interdependence theory thrives. Large, coupled physical or information systems can also experience synergism or dysergism from interdependence. Synergistically, the best human teams are not only highly interdependent, but they also exploit interdependence to reduce uncertainty, the focus of this work-in-progress and roadmap. We have long argued that interdependence is fundamental to human autonomy in teams. But for A-HMTs, no mathematics exists to build from rational theory or social science for their design nor safe or effective operation, a severe weakness. Compared to the rational and traditional social theory, we hope to advance interdependence theory first by mapping similarities between quantum theory and our prior findings; e.g., to maintain interdependence, we previously established that boundaries reduce dysergic effects to allow teams to function (akin to blocking interference to prevent quantum decoherence). Second, we extend our prior findings with case studies to predict with interdependence theory that as uncertainty increases in non-factorable situations for humans, the duality in two-sided beliefs serves debaters who explore alternatives with tradeoffs in the search for the best path going forward. Third, applied to autonomous teams, we conclude that a machine in an A-HMT must be able to express itself to its human teammates in causal language however imperfectly.

5.
Exp Cell Res ; 336(2): 182-91, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26162853

RESUMO

Skeletal and heart muscle-specific variant of the alpha subunit of nascent polypeptide associated complex (skNAC) is exclusively found in striated muscle cells. Its function, however, is largely unknown. Previous reports could demonstrate that skNAC binds to Smyd1 (SET and MYND domain containing protein 1). The facts that (a) SET domains have histone methyltransferase activity, and (b) MYND domains are known recruiters of histone deacetylases (HDACs), implicate the skNAC-Smyd1 complex in transcriptional control. To study potential target genes, we carried out cDNA microarray analysis on differentiating C2C12 myoblasts in which expression of the skNAC gene had been knocked down. We found and confirmed a series of targets, specifically genes encoding regulators of inflammation, cellular metabolism, and cell migration. Mechanistically, as shown by Western blot, ELISA, and ChIP analysis at selected promoter regions, transcriptional control by skNAC-Smyd1 appears to be exerted at least in part by affecting a series of histone modifications, specifically H3K4 di- and trimethylation and potentially also histone acetylation. Taken together, our data suggest that the skNAC-Smyd1 complex is involved in transcriptional regulation both via the control of histone methylation and histone (de)acetylation.


Assuntos
Proteínas de Ligação a DNA/genética , Histonas/metabolismo , Chaperonas Moleculares/genética , Proteínas Musculares/genética , Fatores de Transcrição/genética , Transcrição Gênica/genética , Acetilação , Animais , Diferenciação Celular , Linhagem Celular , Movimento Celular/genética , Metabolismo Energético/genética , Regulação da Expressão Gênica , Histona Desacetilases/metabolismo , Inflamação/genética , Metilação , Camundongos , Músculo Esquelético/metabolismo , Mioblastos Cardíacos/citologia , Mioblastos Esqueléticos/citologia , Miocárdio/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas/genética , Interferência de RNA , RNA Interferente Pequeno , Succinato Desidrogenase/metabolismo , Fatores ras de Troca de Nucleotídeo Guanina/biossíntese
6.
Eur J Med Chem ; 264: 115982, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38056296

RESUMO

Histone methyltransferases (HMTs) play a critical role in gene post-translational regulation and diverse physiological processes, and are implicated in a plethora of human diseases, especially cancer. Increasing evidences demonstrate that HMTs may serve as a potential therapeutic target for cancer treatment. Thus, the development of HMTs inhibitor have been pursued with steadily increasing interest over the past decade. However, the disadvantages such as insufficient clinical efficacy, moderate selectivity, and propensity for acquired resistance have hindered the development of conventional HMT inhibitors. New technologies and methods are imperative to enhance the anticancer activity of HMT inhibitors. In this review, we first review the structure and biological functions of the several essential HMTs, such as EZH2, G9a, PRMT5, and DOT1L. The internal relationship between these HMTs and cancer is also expounded. Next, we mainly focus on the latest progress in the development of HMT modulators encompassing dual-target inhibitors, targeted protein degraders and covalent inhibitors from perspectives such as rational design, pharmacodynamics, pharmacokinetics, and clinical status. Lastly, we also discuss the challenges and future directions for HMT-based drug discovery for cancer therapy.


Assuntos
Neoplasias , Humanos , Histona Metiltransferases , Neoplasias/tratamento farmacológico , Neoplasias/genética , Descoberta de Drogas , Metiltransferases , Proteína-Arginina N-Metiltransferases
7.
PeerJ ; 7: e6396, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30755832

RESUMO

BACKGROUND: Histone lysine methyltransferases (HMTs), a category of enzymes, play essential roles in regulating transcription, cellular differentiation, and chromatin construction. The genomic landscape and clinical significance of HMTs in renal cell carcinoma (RCC) remain uncovered. METHODS: We conducted an integrative analysis of 50 HMTs in RCC and discovered the internal relations among copy number alterations (CNAs), expressive abundance, mutations, and clinical outcome. RESULTS: We confirmed 12 HMTs with the highest frequency of genetic alterations, including seven HMTs with high-level amplification, two HMTs with somatic mutation, and three HMTs with putative homozygous deletion. Patterns of copy number and expression varied among different subtypes of RCC, including clear cell renal cell carcinoma, papillary cell carcinoma, and chromophobe renal carcinoma. Kaplan-Meier survival analysis and multivariate analysis identified that CNA or mRNA expression in some HMTs were significantly associated with shorter overall patient survival. Systematic analysis identified six HMTs (ASH1L, PRDM6, NSD1, EZH2, WHSC1L1, SETD2) which were dysregulated by genetic alterations as candidate therapeutic targets. DISCUSSION: In summary, our findings strongly evidenced that genetic alteration of HMTs may play an important role in generation and development of RCC, which lays a solid foundation for the mechanism for further research in the future.

8.
Cell Rep ; 19(2): 295-306, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28402853

RESUMO

The role of chromatin structure in lineage commitment of multipotent hematopoietic progenitors (HPCs) is presently unclear. We show here that CD34+ HPCs possess a post-replicative chromatin globally devoid of the repressive histone mark H3K27me3. This H3K27-unmodified chromatin is required for recruitment of lineage-determining transcription factors (TFs) C/EBPα, PU.1, and GATA-1 to DNA just after DNA replication upon cytokine-induced myeloid or erythroid commitment. Blocking DNA replication or increasing H3K27me3 levels prevents recruitment of these TFs to DNA and suppresses cytokine-induced erythroid or myeloid differentiation. However, H3K27me3 is rapidly associated with nascent DNA in more primitive human and murine HPCs. Treatment of these cells with instructive cytokines leads to a significant delay in accumulation of H3K27me3 in nascent chromatin due to activity of the H3K27me3 demethylase UTX. Thus, HPCs utilize special mechanisms of chromatin modification for recruitment of specific TFs to DNA during early stages of lineage specification.


Assuntos
Diferenciação Celular/genética , Hematopoese/genética , Células-Tronco Hematopoéticas/citologia , Histona Desmetilases com o Domínio Jumonji/genética , Animais , Antígenos CD34/biossíntese , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Linhagem da Célula/genética , Cromatina/genética , Replicação do DNA/genética , Fator de Transcrição GATA1/genética , Humanos , Histona Desmetilases com o Domínio Jumonji/metabolismo , Camundongos , Proteínas Proto-Oncogênicas/genética , Transativadores/genética
9.
Int J Biochem Cell Biol ; 81(Pt A): 48-56, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27771440

RESUMO

Mitochondrial Targeting Sequences (MTSs) are responsible for trafficking nuclear-encoded proteins into mitochondria. Once entering the mitochondria, the MTS is recognized and cleaved off. Some MTSs are long and undergo two-step processing, as in the case of the human frataxin (FXN) protein (80aa), implicated in Friedreich's ataxia (FA). Therefore, we chose the FXN protein to examine whether nuclear-encoded mitochondrial proteins can efficiently be targeted via a heterologous MTS (hMTS) and deliver a functional protein into mitochondria. We examined three hMTSs; that of citrate synthase (cs), lipoamide deydrogenase (LAD) and C6ORF66 (ORF), as classically MTS sequences, known to be removed by one-step processing, to deliver FXN into mitochondria, in the form of fusion proteins. We demonstrate that using hMTSs for delivering FXN results in the production of 4-5-fold larger amounts of the fusion proteins, and at 4-5-fold higher concentrations. Moreover, hMTSs delivered a functional FXN protein into the mitochondria even more efficiently than the native MTSfxn, as evidenced by the rescue of FA patients' cells from oxidative stress; demonstrating a 18%-54% increase in cell survival; and a 13%-33% increase in ATP levels, as compared to the fusion protein carrying the native MTS. One fusion protein with MTScs increased aconitase activity within patients' cells, by 400-fold. The implications form our studies are of vast importance for both basic and translational research of mitochondrial proteins as any mitochondrial protein can be delivered efficiently by an hMTS. Moreover, effective targeting of functional proteins is important for restoration of mitochondrial function and treatment of related disorders.


Assuntos
Proteínas de Ligação ao Ferro/metabolismo , Mitocôndrias/metabolismo , Aconitato Hidratase/metabolismo , Ataxia de Friedreich/metabolismo , Humanos , Estresse Oxidativo , Transporte Proteico , Frataxina
10.
Epigenomics ; 8(6): 831-42, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27337224

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is often viewed to arise primarily by genetic alterations. However, today we know that many aspects of the cancer phenotype require a crosstalk among these genetic alterations with epigenetic changes. Indeed, aberrant gene expression patterns, driven by epigenetics are fixed by altered signaling from mutated oncogenes and tumor suppressors to define the PDAC phenotype. This conceptual framework may have significant mechanistic value and could offer novel possibilities for treating patients affected with PDAC. In fact, extensive investigations are leading to the development of small molecule drugs that reversibly modify the epigenome. These new 'epigenetic therapeutics' discussed herein are promising to fuel a new era of studies, by providing the medical community with new tools to treat this dismal disease.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Ductal Pancreático/tratamento farmacológico , Epigênese Genética/efeitos dos fármacos , Neoplasias Pancreáticas/tratamento farmacológico , Animais , Carcinoma Ductal Pancreático/genética , Ilhas de CpG , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Humanos , Terapia de Alvo Molecular , Neoplasias Pancreáticas/genética
11.
Cell Rep ; 14(1): 103-114, 2016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26725110

RESUMO

Trithorax proteins and long-intergenic noncoding RNAs are critical regulators of embryonic stem cell pluripotency; however, how they cooperatively regulate germ layer mesoderm specification remains elusive. We report here that HoxBlinc RNA first specifies Flk1(+) mesoderm and then promotes hematopoietic differentiation through regulation of hoxb pathways. HoxBlinc binds to the hoxb genes, recruits Setd1a/MLL1 complexes, and mediates long-range chromatin interactions to activate transcription of the hoxb genes. Depletion of HoxBlinc by shRNA-mediated knockdown or CRISPR-Cas9-mediated genetic deletion inhibits expression of hoxb genes and other factors regulating cardiac/hematopoietic differentiation. Reduced hoxb expression is accompanied by decreased recruitment of Set1/MLL1 and H3K4me3 modification, as well as by reduced chromatin loop formation. Re-expression of hoxb2-b4 genes in HoxBlinc-depleted embryoid bodies rescues Flk1(+) precursors that undergo hematopoietic differentiation. Thus, HoxBlinc plays an important role in controlling hoxb transcription networks that mediate specification of mesoderm-derived Flk1(+) precursors and differentiation of Flk1(+) cells into hematopoietic lineages.


Assuntos
Linhagem da Célula/fisiologia , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Histona-Lisina N-Metiltransferase/metabolismo , Proteínas de Homeodomínio/biossíntese , Mesoderma/embriologia , Proteína de Leucina Linfoide-Mieloide/metabolismo , RNA Longo não Codificante/biossíntese , Animais , Linhagem Celular , Embrião de Mamíferos/citologia , Histona-Lisina N-Metiltransferase/genética , Proteínas de Homeodomínio/genética , Mesoderma/citologia , Camundongos , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Proteína de Leucina Linfoide-Mieloide/genética , RNA Longo não Codificante/genética
12.
Clin Epigenetics ; 7: 127, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26692909

RESUMO

Epigenetic treatment has been approved by regulatory agencies for haematological malignancies. The success observed in cutaneous lymphomas represents a proof of principle that similar results may be obtained in solid tumours. Several agents that interfere with DNA methylation-demethylation and histones acetylation/deacetylation have been studied, and some (such as azacytidine, decitabine, valproic acid and vorinostat) are already in clinical use. The aim of this review is to provide a brief overview of the molecular events underlying the antitumour effects of epigenetic treatments and to summarise data available on clinical trials that tested the use of epigenetic agents against solid tumours. We not only list results but also try to indicate how the proper evaluation of this treatment might result in a better selection of effective agents and in a more rapid development. We divided compounds in demethylating agents and HDAC inhibitors. For each class, we report the antitumour activity and the toxic side effects. When available, we describe plasma pharmacokinetics and pharmacodynamic evaluation in tumours and in surrogate tissues (generally white blood cells). Epigenetic treatment is a reality in haematological malignancies and deserves adequate attention in solid tumours. A careful consideration of available clinical data however is required for faster drug development and possibly to re-evaluate some molecules that were perhaps discarded too early.

13.
Cancer Treat Rev ; 40(1): 153-69, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23831234

RESUMO

Genetic mutations and gross structural defects in the DNA sequence permanently alter genetic loci in ways that significantly disrupt gene function. In sharp contrast, genes modified by aberrant epigenetic modifications remain structurally intact and are subject to partial or complete reversal of modifications that restore the original (i.e. non-diseased) state. Such reversibility makes epigenetic modifications ideal targets for therapeutic intervention. The epigenome of cancer cells is extensively modified by specific hypermethylation of the promoters of tumor suppressor genes relative to the extensive hypomethylation of repetitive sequences, overall loss of acetylation, and loss of repressive marks at microsatellite/repeat regions. In this review, we discuss emerging therapies targeting specific epigenetic modifications or epigenetic modifying enzymes either alone or in combination with other treatment regimens. The limitations posed by cancer treatments elicit unintended epigenetic modifications that result in exacerbation of tumor progression are also discussed. Lastly, a brief discussion of the specificity restrictions posed by epigenetic therapies and ways to address such limitations is presented.


Assuntos
Epigênese Genética , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Animais , Metilação de DNA , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico , Histonas/metabolismo , Humanos , Neoplasias/genética , Processamento de Proteína Pós-Traducional/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA