Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 141
Filtrar
1.
Nature ; 608(7924): 733-740, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35978187

RESUMO

Single-cell transcriptomics (scRNA-seq) has greatly advanced our ability to characterize cellular heterogeneity1. However, scRNA-seq requires lysing cells, which impedes further molecular or functional analyses on the same cells. Here, we established Live-seq, a single-cell transcriptome profiling approach that preserves cell viability during RNA extraction using fluidic force microscopy2,3, thus allowing to couple a cell's ground-state transcriptome to its downstream molecular or phenotypic behaviour. To benchmark Live-seq, we used cell growth, functional responses and whole-cell transcriptome read-outs to demonstrate that Live-seq can accurately stratify diverse cell types and states without inducing major cellular perturbations. As a proof of concept, we show that Live-seq can be used to directly map a cell's trajectory by sequentially profiling the transcriptomes of individual macrophages before and after lipopolysaccharide (LPS) stimulation, and of adipose stromal cells pre- and post-differentiation. In addition, we demonstrate that Live-seq can function as a transcriptomic recorder by preregistering the transcriptomes of individual macrophages that were subsequently monitored by time-lapse imaging after LPS exposure. This enabled the unsupervised, genome-wide ranking of genes on the basis of their ability to affect macrophage LPS response heterogeneity, revealing basal Nfkbia expression level and cell cycle state as important phenotypic determinants, which we experimentally validated. Thus, Live-seq can address a broad range of biological questions by transforming scRNA-seq from an end-point to a temporal analysis approach.


Assuntos
Sobrevivência Celular , Perfilação da Expressão Gênica , Macrófagos , RNA-Seq , Análise de Célula Única , Transcriptoma , Tecido Adiposo/citologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Diferenciação Celular , Perfilação da Expressão Gênica/métodos , Perfilação da Expressão Gênica/normas , Genoma/efeitos dos fármacos , Genoma/genética , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Inibidor de NF-kappaB alfa/genética , Especificidade de Órgãos , Fenótipo , RNA/genética , RNA/isolamento & purificação , RNA-Seq/métodos , RNA-Seq/normas , Reprodutibilidade dos Testes , Análise de Sequência de RNA/métodos , Análise de Sequência de RNA/normas , Análise de Célula Única/métodos , Células Estromais/citologia , Células Estromais/metabolismo , Fatores de Tempo , Transcriptoma/genética
2.
Cells ; 10(12)2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34943778

RESUMO

Retinal mitochondria are damaged in diabetes-accelerating apoptosis of capillary cells, and ultimately, leading to degenerative capillaries. Diabetes also upregulates many long noncoding RNAs (LncRNAs), including LncMALAT1 and LncNEAT1. These RNAs have more than 200 nucleotides and no open reading frame for translation. LncMALAT1 and LncNEAT1 are encoded by nuclear genome, but nuclear-encoded LncRNAs can also translocate in the mitochondria. Our aim was to investigate the role of LncMALAT1 and LncNEAT1 in mitochondrial homeostasis. Using human retinal endothelial cells, the effect of high glucose on LncMALAT1 and LncNEAT1 mitochondrial localization was examined by RNA fluorescence in situ hybridization. The role of these LncRNAs in mitochondrial membrane potential (by JC-I staining), mtDNA integrity (by extended length PCR) and in protective mtDNA nucleoids (by SYBR green staining) was examined in MALAT1- or NEAT1-siRNA transfected cells. High glucose increased LncMALAT1 and LncNEAT1 mitochondrial expression, and MALAT1-siRNA or NEAT1-siRNA ameliorated glucose-induced damage to mitochondrial membrane potential and mtDNA, and prevented decrease in mtDNA nucleoids. Thus, increased mitochondrial translocation of LncMALAT1 or LncNEAT1 in a hyperglycemic milieu plays a major role in damaging the mitochondrial structural and genomic integrity. Regulation of these LncRNAs can protect mitochondrial homeostasis, and ameliorate formation of degenerative capillaries in diabetic retinopathy.


Assuntos
Retinopatia Diabética/genética , Mitocôndrias/efeitos dos fármacos , RNA Longo não Codificante/genética , Retina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Núcleo Celular/genética , Dano ao DNA/efeitos dos fármacos , DNA Mitocondrial/efeitos dos fármacos , Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Genoma/efeitos dos fármacos , Genoma/genética , Glucose/efeitos adversos , Humanos , Hibridização in Situ Fluorescente , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/genética , Retina/efeitos dos fármacos , Retina/patologia , Transfecção
3.
STAR Protoc ; 2(3): 100799, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34527956

RESUMO

Reorganization of topologically associated domain (TAD) is considered to be a novel mechanism for cell fate transitions. Here, we present a protocol to manipulate TAD via abscisic acid (ABA)-dependent genome linking. We use this protocol to merge two adjacent TADs and evaluate the influence on cell fate transitions. The advantages are that the manipulation does not change the genome and is reversible by withdrawing ABA. The major challenge is how to select linking loci for efficient TAD reorganization. For complete details on the use and execution of this protocol, please refer to Wang et al. (2021).


Assuntos
Diferenciação Celular/genética , Técnicas Citológicas/métodos , Componentes Genômicos , Genômica/métodos , Ácido Abscísico/farmacologia , Animais , Linhagem Celular , Genoma/efeitos dos fármacos , Genoma/genética , Componentes Genômicos/efeitos dos fármacos , Componentes Genômicos/genética , Humanos , Camundongos
4.
Int J Mol Sci ; 22(11)2021 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-34071702

RESUMO

Cisplatin is a chemotherapy drug that kills cancer cells by damaging their DNA. In human cells, this damage is repaired primarily by nucleotide excision repair. While cisplatin is generally effective, many cancers exhibit initial or acquired resistance to it. Here, we studied cisplatin resistance in a defined cell line system. We conducted a comprehensive genomic characterization of the cisplatin-sensitive A2780 ovarian cancer cell line compared to A2780cis, its resistant derivative. The resistant cells acquired less damage, but had similar repair kinetics. Genome-wide mapping of nucleotide excision repair showed a shift in the resistant cells from global genome towards transcription-coupled repair. By mapping gene expression changes following cisplatin treatment, we identified 56 upregulated genes that have higher basal expression in the resistant cell line, suggesting they are primed for a cisplatin response. More than half of these genes are novel to cisplatin- or damage-response. Six out of seven primed genes tested were upregulated in response to cisplatin in additional cell lines, making them attractive candidates for future investigation. These novel candidates for cisplatin resistance could prove to be important prognostic markers or targets for tailored combined therapy in the future.


Assuntos
Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Genoma/efeitos dos fármacos , Antineoplásicos/farmacologia , Biomarcadores Tumorais , Linhagem Celular Tumoral , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo
5.
Curr Opin Chem Biol ; 62: 119-129, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34118759

RESUMO

The ENCODE and genome-wide association projects have shown that much of the genome is transcribed into RNA and much less is translated into protein. These and other functional studies suggest that the druggable transcriptome is much larger than the druggable proteome. This review highlights approaches to define druggable RNA targets and structure-activity relationships across genomic RNA. Binding compounds can be identified and optimized into structure-specific ligands by using sequence-based design with various modes of action, for example, inhibiting translation or directing pre-mRNA splicing outcomes. In addition, strategies to direct protein activity against an RNA of interest via chemically induced proximity is a burgeoning area that has been validated both in cells and in preclinical animal models, and we describe that it may allow rapid access to new avenues to affect RNA biology. These approaches and the unique modes of action suggest that more RNAs are potentially amenable to targeting than proteins.


Assuntos
Antineoplásicos/química , Genoma/efeitos dos fármacos , RNA/metabolismo , Bibliotecas de Moléculas Pequenas/química , Transcriptoma/efeitos dos fármacos , Animais , Antineoplásicos/farmacologia , Compostos Azo/farmacologia , Sequência de Bases , Desenho de Fármacos , Regulação Neoplásica da Expressão Gênica , Genoma/genética , Estudo de Associação Genômica Ampla , Genômica , Humanos , Ligantes , Modelos Animais , Pirimidinas/farmacologia , Bibliotecas de Moléculas Pequenas/metabolismo , Relação Estrutura-Atividade , Transcriptoma/genética
6.
Nucleic Acids Res ; 49(15): 8449-8461, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-33784402

RESUMO

Non-coding RNAs are regarded as promising targets for the discovery of innovative drugs due to their abundance in the genome and their involvement in many biological processes. Phytochemicals (PCs) are the primary source of ligand-based drugs due to their broad spectrum of biological activities. Since many PCs are heterocyclic and have chemical groups potentially involved in the interaction with nucleic acids, detailed interaction analysis between PCs and RNA is crucial to explore the effect of PCs on RNA functions. In this study, an integrated approach for investigating interactions between PCs and RNAs were demonstrated to verify the RNA-mediated PCs functions by using berberine (BRB) as a model PC. RNA screening of a transcriptome library followed by sequence refinement found minimal RNA motif consisting of a cytosine bulge with U-A and G-U neighbouring base pairs for interaction with BRB. NMR-based structure determination and physicochemical analyses using chemical analogues of BRB demonstrated the importance of electrostatic and stacking interactions for sequence selective interaction and RNA stabilization. The selective interaction with a relatively small RNA motif based on a chemical structure of a planer heterocyclic highlights the biological activities of various PCs mediated by the interactions with particular functional RNAs. In addition, the systematic and quantitative investigations demonstrated in this study could be useful for the development of therapeutic chemicals targeting functional RNAs, based on the PCs, in the future.


Assuntos
Berberina/farmacologia , Conformação de Ácido Nucleico , RNA não Traduzido/genética , Transcriptoma/genética , Berberina/química , Genoma/efeitos dos fármacos , Genoma/genética , Humanos , Ligantes , Motivos de Nucleotídeos/efeitos dos fármacos , Motivos de Nucleotídeos/genética , RNA não Traduzido/efeitos dos fármacos , RNA não Traduzido/ultraestrutura , Transcriptoma/efeitos dos fármacos
7.
Nat Rev Drug Discov ; 20(3): 200-216, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33510482

RESUMO

Natural products and their structural analogues have historically made a major contribution to pharmacotherapy, especially for cancer and infectious diseases. Nevertheless, natural products also present challenges for drug discovery, such as technical barriers to screening, isolation, characterization and optimization, which contributed to a decline in their pursuit by the pharmaceutical industry from the 1990s onwards. In recent years, several technological and scientific developments - including improved analytical tools, genome mining and engineering strategies, and microbial culturing advances - are addressing such challenges and opening up new opportunities. Consequently, interest in natural products as drug leads is being revitalized, particularly for tackling antimicrobial resistance. Here, we summarize recent technological developments that are enabling natural product-based drug discovery, highlight selected applications and discuss key opportunities.


Assuntos
Produtos Biológicos/química , Produtos Biológicos/farmacologia , Produtos Biológicos/uso terapêutico , Doenças Transmissíveis/tratamento farmacológico , Doenças Transmissíveis/genética , Descoberta de Drogas/métodos , Indústria Farmacêutica/métodos , Genoma/efeitos dos fármacos , Humanos , Neoplasias/tratamento farmacológico
8.
Methods Mol Biol ; 2194: 107-126, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32926364

RESUMO

The magnitude of all therapeutic responses is significantly determined by genome structure, variation, and functional interactions. This determination occurs at many levels which are discussed in the current review. Well-established examples of structural variation between individuals are known to dictate an individual's response to numerous drugs, as clearly illustrated by warfarin. The exponential rate of genomic-based interrogation is coupled with an expanding repertoire of genomic technologies and applications. This is leading to an ever more sophisticated appreciation of how structural variation, regulation of transcription and genomic structure, both individually and collectively, define cell therapeutic responses.


Assuntos
Perfilação da Expressão Gênica/métodos , Estudo de Associação Genômica Ampla/métodos , Genoma/genética , Genômica/métodos , Farmacogenética/métodos , Transcriptoma/efeitos dos fármacos , Pesquisa Translacional Biomédica/métodos , Epigenômica , Genoma/efeitos dos fármacos , Humanos , Transcriptoma/genética
9.
Toxicol Appl Pharmacol ; 409: 115319, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33160984

RESUMO

A growing body of evidence has shown that gestational exposure to environmental factors such as imbalanced diet, environmental chemicals, and stress can lead to late-onset health effects in offspring and that some of these effects are heritable by the next generation and subsequent generations. Furthermore, altered epigenetic modifications in DNA methylation, histone modifications and small RNAs in a single sperm genome have been shown to transmit disease phenotypes acquired from the environment to later generations. Recently, our group found that gestational exposure of F0 pregnant dams to an inorganic arsenic, sodium arsenite, increases the incidence of hepatic tumors in male F2 mice, and the effects are paternally transmitted to the F2. Here, we first overview the epigenetic changes involved in paternal intergenerational and transgenerational inheritance caused by exposure to environmental factors. Then, we discuss our recent studies regarding paternal inheritance of the tumor-augmenting effects in F2 mice by gestational arsenite exposure, in which we investigated alterations of DNA methylation status in F2 tumors and causative F1 sperm. We also discuss the possible targets of the F2 effects. Finally, we discuss future perspectives on the studies that are needed to fully understand the health effects of arsenic exposure.


Assuntos
Arsênio/efeitos adversos , Epigênese Genética/efeitos dos fármacos , Herança Paterna/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Animais , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/genética , Epigênese Genética/genética , Feminino , Genoma/efeitos dos fármacos , Genoma/genética , Humanos , Herança Paterna/genética , Fenótipo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética
10.
Sci Rep ; 10(1): 20654, 2020 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-33244091

RESUMO

Senegalese sole is an economically important flatfish species in aquaculture and an attractive model to decipher the molecular mechanisms governing the severe transformations occurring during metamorphosis, where retinoic acid seems to play a key role in tissue remodeling. In this study, a robust sole transcriptome was envisaged by reducing the number of assembled libraries (27 out of 111 available), fine-tuning a new automated and reproducible set of workflows for de novo assembling based on several assemblers, and removing low confidence transcripts after mapping onto a sole female genome draft. From a total of 96 resulting assemblies, two "raw" transcriptomes, one containing only Illumina reads and another with Illumina and GS-FLX reads, were selected to provide SOLSEv5.0, the most informative transcriptome with low redundancy and devoid of most single-exon transcripts. It included both Illumina and GS-FLX reads and consisted of 51,348 transcripts of which 22,684 code for 17,429 different proteins described in databases, where 9527 were predicted as complete proteins. SOLSEv5.0 was used as reference for the study of retinoic acid (RA) signalling in sole larvae using drug treatments (DEAB, a RA synthesis blocker, and TTNPB, a RA-receptor agonist) for 24 and 48 h. Differential expression and functional interpretation were facilitated by an updated version of DEGenes Hunter. Acute exposure of both drugs triggered an intense, specific and transient response at 24 h but with hardly observable differences after 48 h at least in the DEAB treatments. Activation of RA signalling by TTNPB specifically increased the expression of genes in pathways related to RA degradation, retinol storage, carotenoid metabolism, homeostatic response and visual cycle, and also modified the expression of transcripts related to morphogenesis and collagen fibril organisation. In contrast, DEAB mainly decreased genes related to retinal production, impairing phototransduction signalling in the retina. A total of 755 transcripts mainly related to lipid metabolism, lipid transport and lipid homeostasis were altered in response to both treatments, indicating non-specific drug responses associated with intestinal absorption. These results indicate that a new assembling and transcript sieving were both necessary to provide a reliable transcriptome to identify the many aspects of RA action during sole development that are of relevance for sole aquaculture.


Assuntos
Linguados/genética , Linguados/metabolismo , Larva/genética , Larva/metabolismo , Transdução de Sinais/genética , Transcriptoma/genética , Tretinoína/metabolismo , Animais , Benzoatos/farmacologia , Carotenoides/metabolismo , Colágeno/genética , Feminino , Genoma/efeitos dos fármacos , Genoma/genética , Homeostase/efeitos dos fármacos , Homeostase/genética , Larva/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Metamorfose Biológica/efeitos dos fármacos , Metamorfose Biológica/genética , Morfogênese/efeitos dos fármacos , Morfogênese/genética , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo , Retina/efeitos dos fármacos , Retina/metabolismo , Retinoides/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos
11.
Elife ; 92020 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-32956035

RESUMO

Numerous anti-cancer drugs perturb thymidylate biosynthesis and lead to genomic uracil incorporation contributing to their antiproliferative effect. Still, it is not yet characterized if uracil incorporations have any positional preference. Here, we aimed to uncover genome-wide alterations in uracil pattern upon drug treatments in human cancer cell line models derived from HCT116. We developed a straightforward U-DNA sequencing method (U-DNA-Seq) that was combined with in situ super-resolution imaging. Using a novel robust analysis pipeline, we found broad regions with elevated probability of uracil occurrence both in treated and non-treated cells. Correlation with chromatin markers and other genomic features shows that non-treated cells possess uracil in the late replicating constitutive heterochromatic regions, while drug treatment induced a shift of incorporated uracil towards segments that are normally more active/functional. Data were corroborated by colocalization studies via dSTORM microscopy. This approach can be applied to study the dynamic spatio-temporal nature of genomic uracil.


Assuntos
Antineoplásicos/farmacologia , DNA , Genoma , Uracila , DNA/análise , DNA/biossíntese , DNA/química , DNA/genética , Genoma/efeitos dos fármacos , Genoma/genética , Genômica , Células HCT116 , Humanos , Microscopia , Análise de Sequência de DNA , Uracila/análise , Uracila/biossíntese , Uracila/química
12.
Toxicon ; 187: 93-100, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32891664

RESUMO

Domoic acid (DA) is an excitatory marine neurotoxin produced by diatoms Pseudo-nitzschia spp. as a defence compound that accumulates in the food web and is associated with amnesic shellfish poisoning in humans. Although its toxicity has been well established in marine species, there is limited data on DA cytogenotoxicity in human non-target cells. Therefore, we aimed to investigate the cytogenotoxic potential of DA (0.01-10 µg/mL) in human peripheral blood cells (HPBCs) using a battery of bioassays in vitro. In addition, the influence of DA on oxidative stress parameters as a possible mechanism of action was assessed. Results revealed that DA induced dose- and time-dependent cytotoxic effects. DA significantly affected genomic instability by increasing the frequency of micronuclei and nuclear buds. Furthermore, a slight induction of primary DNA strand breaks was detected after 24 h of exposure accompanied by a significant increase in the number of abnormal size tailed nuclei. No induction of hOGG1 (human 8-oxoguanine DNA glycosylase) sensitive sites was determined upon exposure to DA. Additionally, DA induced oxidative stress by increased production of reactive oxygen species accompanied by changes in glutathione, superoxide dismutase, malondialdehyde and protein carbonyl levels. Overall, the obtained results showed adverse genotoxic effects of DA in non-target HPBCs.


Assuntos
Células Sanguíneas/efeitos dos fármacos , Genoma/efeitos dos fármacos , Ácido Caínico/análogos & derivados , Humanos , Ácido Caínico/toxicidade , Toxinas Marinhas
13.
Mutagenesis ; 35(5): 391-404, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32914844

RESUMO

DNA ligase I (LIG1) joins DNA strand breaks during DNA replication and repair transactions and contributes to genome integrity. The mutations (P529L, E566K, R641L and R771W) in LIG1 gene are described in patients with LIG1-deficiency syndrome that exhibit immunodeficiency. LIG1 senses 3'-DNA ends with a mismatch or oxidative DNA base inserted by a repair DNA polymerase. However, the ligation efficiency of the LIG1 variants for DNA polymerase-promoted mutagenesis products with 3'-DNA mismatches or 8-oxo-2'-deoxyguanosine (8-oxodG) remains undefined. Here, we report that R641L and R771W fail in the ligation of nicked DNA with 3'-8-oxodG, leading to an accumulation of 5'-AMP-DNA intermediates in vitro. Moreover, we found that the presence of all possible 12 non-canonical base pairs variously impacts the ligation efficiency by P529L and R771W depending on the architecture at the DNA end, whereas E566K exhibits no activity against all substrates tested. Our results contribute to the understanding of the substrate specificity and mismatch discrimination of LIG1 for mutagenic repair intermediates and the effect of non-synonymous mutations on ligase fidelity.


Assuntos
DNA Ligase Dependente de ATP/genética , Reparo de Erro de Pareamento de DNA/genética , Mutagênese/genética , 8-Hidroxi-2'-Desoxiguanosina/genética , Monofosfato de Adenosina/genética , Quebras de DNA de Cadeia Simples/efeitos dos fármacos , Dano ao DNA/genética , Replicação do DNA/genética , Genoma/efeitos dos fármacos , Humanos , Mutação/genética , Estresse Oxidativo/efeitos dos fármacos
14.
Aging (Albany NY) ; 12(14): 14341-14354, 2020 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-32668413

RESUMO

Inactivating mutations in the liver kinase B1 (LKB1) tumor suppressor gene underlie Peutz-Jeghers syndrome (PJS) and occur frequently in various human cancers. We previously showed that LKB1 regulates centrosome duplication via PLK1. Here, we report that LKB1 further helps to maintain genomic stability through negative regulation of survivin, a member of the chromosomal passenger complex (CPC) that mediates CPC targeting to the centromere. We found that loss of LKB1 led to accumulation of misaligned and lagging chromosomes at metaphase and anaphase and increased the appearance of multi- and micro-nucleated cells. Ectopic LKB1 expression reduced these features and improved mitotic fidelity in LKB1-deficient cells. Through pharmacological and genetic manipulations, we showed that LKB1-mediated repression of survivin is independent of AMPK, but requires p53. Consistent with the key influence of LKB1 on survivin expression, immunohistochemical analysis indicated that survivin is highly expressed in intestinal polyps from a PJS patient. Lastly, we reaffirm a potential therapeutic avenue to treat LKB1-mutated tumors by demonstrating the increased sensitivity to survivin inhibitors of LKB1-deficient cells.


Assuntos
Centrômero/efeitos dos fármacos , Genes p53/efeitos dos fármacos , Genoma/efeitos dos fármacos , Síndrome de Peutz-Jeghers/genética , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Survivina/biossíntese , Survivina/genética , Quinases Proteína-Quinases Ativadas por AMP , Linhagem Celular Tumoral , Aberrações Cromossômicas , Humanos , Pólipos Intestinais/genética , Mitose/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , RNA Interferente Pequeno/biossíntese , RNA Interferente Pequeno/genética , Ensaio Tumoral de Célula-Tronco , Regulação para Cima/genética
15.
PLoS Genet ; 16(7): e1008917, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32628663

RESUMO

Mechanisms of transcriptional control in malaria parasites are still not fully understood. The positioning patterns of G-quadruplex (G4) DNA motifs in the parasite's AT-rich genome, especially within the var gene family which encodes virulence factors, and in the vicinity of recombination hotspots, points towards a possible regulatory role of G4 in gene expression and genome stability. Here, we carried out the most comprehensive genome-wide survey, to date, of G4s in the Plasmodium falciparum genome using G4Hunter, which identifies G4 forming sequences (G4FS) considering their G-richness and G-skewness. We show an enrichment of G4FS in nucleosome-depleted regions and in the first exon of var genes, a pattern that is conserved within the closely related Laverania Plasmodium parasites. Under G4-stabilizing conditions, i.e., following treatment with pyridostatin (a high affinity G4 ligand), we show that a bona fide G4 found in the non-coding strand of var promoters modulates reporter gene expression. Furthermore, transcriptional profiling of pyridostatin-treated parasites, shows large scale perturbations, with deregulation affecting for instance the ApiAP2 family of transcription factors and genes involved in ribosome biogenesis. Overall, our study highlights G4s as important DNA secondary structures with a role in Plasmodium gene expression regulation, sub-telomeric recombination and var gene biology.


Assuntos
Quadruplex G , Malária/genética , Motivos de Nucleotídeos/genética , Plasmodium falciparum/genética , Aminoquinolinas/farmacologia , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Genoma/efeitos dos fármacos , Humanos , Malária/tratamento farmacológico , Malária/parasitologia , Ácidos Picolínicos/farmacologia , Plasmodium falciparum/patogenicidade , Regiões Promotoras Genéticas/genética , Ribossomos/efeitos dos fármacos , Ribossomos/genética
16.
Int J Mol Sci ; 21(12)2020 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-32599754

RESUMO

Throughout life, organisms are exposed to various exogenous and endogenous factors that cause DNA damages and somatic mutations provoking genomic instability. At a young age, compensatory mechanisms of genome protection are activated to prevent phenotypic and functional changes. However, the increasing stress and age-related deterioration in the functioning of these mechanisms result in damage accumulation, overcoming the functional threshold. This leads to aging and the development of age-related diseases. There are several ways to counteract these changes: 1) prevention of DNA damage through stimulation of antioxidant and detoxification systems, as well as transition metal chelation; 2) regulation of DNA methylation, chromatin structure, non-coding RNA activity and prevention of nuclear architecture alterations; 3) improving DNA damage response and repair; 4) selective removal of damaged non-functional and senescent cells. In the article, we have reviewed data about the effects of various trace elements, vitamins, polyphenols, terpenes, and other phytochemicals, as well as a number of synthetic pharmacological substances in these ways. Most of the compounds demonstrate the geroprotective potential and increase the lifespan in model organisms. However, their genome-protecting effects are non-selective and often are conditioned by hormesis. Consequently, the development of selective drugs targeting genome protection is an advanced direction.


Assuntos
Envelhecimento , Genoma/efeitos dos fármacos , Instabilidade Genômica , Preparações Farmacêuticas/administração & dosagem , Substâncias Protetoras/uso terapêutico , Animais , Humanos
17.
Bull Exp Biol Med ; 169(1): 43-47, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32488780

RESUMO

Paclitaxel in a single MTD of 40 mg/kg caused chromosome aberrations and genome changes (polyploidy) in the bone marrow cells of mice early and 3 months after the injection. The quantity of early precursors of erythropoiesis in the bone marrow decreased, as did their proliferative potential irrespective of the animal gender. Injection of paclitaxel in the MTD caused the development of bone marrow hypoplasia during the early period of observation (up to 14 days) and 3 months after injection.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Genoma/efeitos dos fármacos , Hematopoese/efeitos dos fármacos , Paclitaxel/farmacologia , Animais , Antineoplásicos/farmacologia , Células da Medula Óssea/metabolismo , Aberrações Cromossômicas/induzido quimicamente , Aberrações Cromossômicas/efeitos dos fármacos , Análise Citogenética , Eritropoese/efeitos dos fármacos , Eritropoese/genética , Feminino , Instabilidade Genômica/efeitos dos fármacos , Hematopoese/genética , Masculino , Camundongos , Camundongos Endogâmicos CBA , Testes de Mutagenicidade
18.
Nat Neurosci ; 23(6): 707-717, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32451484

RESUMO

Neuronal activation induces rapid transcription of immediate early genes (IEGs) and longer-term chromatin remodeling around secondary response genes (SRGs). Here, we use high-resolution chromosome-conformation-capture carbon-copy sequencing (5C-seq) to elucidate the extent to which long-range chromatin loops are altered during short- and long-term changes in neural activity. We find that more than 10% of loops surrounding select IEGs, SRGs, and synaptic genes are induced de novo during cortical neuron activation. IEGs Fos and Arc connect to activity-dependent enhancers via singular short-range loops that form within 20 min after stimulation, prior to peak messenger RNA levels. By contrast, the SRG Bdnf engages in both pre-existing and activity-inducible loops that form within 1-6 h. We also show that common single-nucleotide variants that are associated with autism and schizophrenia are colocalized with distinct classes of activity-dependent, looped enhancers. Our data link architectural complexity to transcriptional kinetics and reveal the rapid timescale by which higher-order chromatin architecture reconfigures during neuronal stimulation.


Assuntos
Montagem e Desmontagem da Cromatina/fisiologia , Expressão Gênica/fisiologia , Genoma/genética , Neurônios/fisiologia , Animais , Bicuculina/farmacologia , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Montagem e Desmontagem da Cromatina/genética , Proteínas do Citoesqueleto/fisiologia , Genoma/efeitos dos fármacos , Humanos , Camundongos , Proteínas do Tecido Nervoso/fisiologia , Neurônios/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/fisiologia , Tetrodotoxina/farmacologia , Fatores de Tempo
20.
J Med Chem ; 63(6): 3090-3103, 2020 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-32142285

RESUMO

Targeting G-quadruplex structures is currently viewed as a promising anticancer strategy. Searching for potent and selective G-quadruplex binders, here we describe a small series of new monohydrazone derivatives designed as analogues of a lead which was proved to stabilize G-quadruplex structures and increase R loop levels in human cancer cells. To investigate the G-quadruplex binding properties of the new molecules, in vitro biophysical studies were performed employing both telomeric and oncogene promoter G-quadruplex-forming sequences. The obtained results allowed the identification of a highly selective G-quadruplex ligand that, when studied in human cancer cells, proved to be able to stabilize both G-quadruplexes and R loops and showed a potent cell killing activity associated with the formation of micronuclei, a clear sign of genome instability.


Assuntos
Antineoplásicos/farmacologia , Dano ao DNA/efeitos dos fármacos , DNA/efeitos dos fármacos , Quadruplex G/efeitos dos fármacos , Instabilidade Genômica/efeitos dos fármacos , Hidrazonas/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/metabolismo , Linhagem Celular Tumoral , DNA/genética , DNA/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais , Genoma/efeitos dos fármacos , Humanos , Hidrazonas/síntese química , Hidrazonas/metabolismo , Ligantes , Estruturas R-Loop/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...