Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 52(6): 3069-3087, 2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38321933

RESUMO

Coordinating epigenomic inheritance and cell cycle progression is essential for organogenesis. UHRF1 connects these functions during development by facilitating maintenance of DNA methylation and cell cycle progression. Here, we provide evidence resolving the paradoxical phenotype of uhrf1 mutant zebrafish embryos which have activation of pro-proliferative genes and increased number of hepatocytes in S-phase, but the liver fails to grow. We uncover decreased Cdkn2a/b and persistent Cdk4/6 activation as the mechanism driving uhrf1 mutant hepatocytes into S-phase. This induces replication stress, DNA damage and Atr activation. Palbociclib treatment of uhrf1 mutants prevented aberrant S-phase entry, reduced DNA damage, and rescued most cellular and developmental phenotypes, but it did not rescue DNA hypomethylation, transposon expression or the interferon response. Inhibiting Atr reduced DNA replication and increased liver size in uhrf1 mutants, suggesting that Atr activation leads to dormant origin firing and prevents hepatocyte proliferation. Cdkn2a/b was downregulated pro-proliferative genes were also induced in a Cdk4/6 dependent fashion in the liver of dnmt1 mutants, suggesting DNA hypomethylation as a mechanism of Cdk4/6 activation during development. This shows that the developmental defects caused by DNA hypomethylation are attributed to persistent Cdk4/6 activation, DNA replication stress, dormant origin firing and cell cycle inhibition.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia , Quinase 4 Dependente de Ciclina , Quinase 6 Dependente de Ciclina , Metilação de DNA , Fígado , Peixe-Zebra , Animais , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Ciclo Celular/genética , Pontos de Checagem do Ciclo Celular/genética , Divisão Celular/genética , Quinase 4 Dependente de Ciclina/genética , Quinase 4 Dependente de Ciclina/metabolismo , Quinase 6 Dependente de Ciclina/genética , Quinase 6 Dependente de Ciclina/metabolismo , DNA/metabolismo , Replicação do DNA/genética , Embrião não Mamífero , Fígado/crescimento & desenvolvimento , Fígado/metabolismo , Fase S , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Ativação Enzimática/genética
2.
Bioessays ; 45(1): e2200036, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36403219

RESUMO

All animals have evolved solutions to manage their genomes, enabling the efficient organization of meters of DNA strands in the nucleus and allowing for nuanced regulation of gene expression while keeping transposable elements suppressed. Epigenetic modifications are central to accomplishing all these. Recent advances in sequencing technologies and the development of techniques that profile epigenetic marks and chromatin accessibility using reagents that can be used in any species has catapulted epigenomic studies in diverse animal species, shedding light on the multitude of epigenomic mechanisms utilized across the evolutionary tree. Now, comparative epigenomics is a rapidly growing field that is uncovering mechanistic aspects of epigenetic modifications and chromatin organization in non-model invertebrates, ranging from octopus to sponges. This review puts recent discoveries in the epigenetics of non-model invertebrates in historical context, and describes new insight into the patterning and functions of DNA methylation and other highly conserved epigenetic modifications.


Assuntos
Epigênese Genética , Epigenômica , Animais , Metilação de DNA , Evolução Biológica , Elementos de DNA Transponíveis
3.
Trends Genet ; 36(8): 587-597, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32487496

RESUMO

A wealth of studies over several decades has revealed an epigenetic prepattern that determines the competence of cellular differentiation in the developing liver. More recently, studies focused on the impact of epigenetic factors during liver regeneration suggest that an epigenetic code in the quiescent liver may establish its regenerative potential. We review work on the pioneer factors and other chromatin remodelers that impact the gene expression patterns instructing hepatocyte and biliary cell specification and differentiation, along with the requirement of epigenetic regulatory factors for hepatic outgrowth. We then explore recent studies involving the role of epigenetic regulators, Arid1a and Uhrf1, in efficient activation of proregenerative genes during liver regeneration, thus highlighting the epigenetic mechanisms of liver disease and tumor development.


Assuntos
Montagem e Desmontagem da Cromatina , Epigênese Genética , Hepatopatias/genética , Hepatopatias/patologia , Regeneração Hepática , Fígado/citologia , Animais , Humanos , Fígado/metabolismo
4.
BMC Biol ; 20(1): 202, 2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-36104784

RESUMO

BACKGROUND: Epigenetic regulatory mechanisms are divergent across the animal kingdom, yet these mechanisms are not well studied in non-model organisms. Unique features of cephalopods make them attractive for investigating behavioral, sensory, developmental, and regenerative processes, and recent studies have elucidated novel features of genome organization and gene and transposon regulation in these animals. However, it is not known how epigenetics regulates these interesting cephalopod features. We combined bioinformatic and molecular analysis of Octopus bimaculoides to investigate the presence and pattern of DNA methylation and examined the presence of DNA methylation and 3 histone post-translational modifications across tissues of three cephalopod species. RESULTS: We report a dynamic expression profile of the genes encoding conserved epigenetic regulators, including DNA methylation maintenance factors in octopus tissues. Levels of 5-methyl-cytosine in multiple tissues of octopus, squid, and bobtail squid were lower compared to vertebrates. Whole genome bisulfite sequencing of two regions of the brain and reduced representation bisulfite sequencing from a hatchling of O. bimaculoides revealed that less than 10% of CpGs are methylated in all samples, with a distinct pattern of 5-methyl-cytosine genome distribution characterized by enrichment in the bodies of a subset of 14,000 genes and absence from transposons. Hypermethylated genes have distinct functions and, strikingly, many showed similar expression levels across tissues while hypomethylated genes were silenced or expressed at low levels. Histone marks H3K27me3, H3K9me3, and H3K4me3 were detected at different levels across tissues of all species. CONCLUSIONS: Our results show that the DNA methylation and histone modification epigenetic machinery is conserved in cephalopods, and that, in octopus, 5-methyl-cytosine does not decorate transposable elements, but is enriched on the gene bodies of highly expressed genes and could cooperate with the histone code to regulate tissue-specific gene expression.


Assuntos
Cefalópodes , Animais , Cefalópodes/genética , Cefalópodes/metabolismo , Citosina/metabolismo , Elementos de DNA Transponíveis/genética , Epigênese Genética , Sulfitos
5.
Genomics ; 114(1): 107-124, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34863900

RESUMO

Restoring homeostasis following proteostatic stress hinges on a stress-specific transcriptional signature. How these signatures are regulated is unknown. We use functional genomics to uncover how activating transcription factor 6 (ATF6), a central factor in the unfolded protein response, regulates its target genes in response to toxicant induced and physiological stress in the liver. We identified 652 conserved putative ATF6 targets (CPATs), which functioned in metabolism, development and proteostasis. Strikingly, Atf6 activation in the zebrafish liver by transgenic nAtf6 overexpression, ethanol and arsenic exposure resulted in a distinct CPAT signature for each; with only 34 CPATs differentially expressed in all conditions. In contrast, during liver regeneration in mice resulted in a dynamic differential expression pattern of 53% of CPATs. These CPATs were distinguished by residing in open chromatin, H3K4me3 occupancy and the absence of H3K27me3 on their promoters. This suggests that a permissive epigenetic landscape allows stress-specific Atf6 target gene expression.


Assuntos
Fator 6 Ativador da Transcrição , Epigênese Genética , Peixe-Zebra , Fator 6 Ativador da Transcrição/genética , Fator 6 Ativador da Transcrição/metabolismo , Animais , Fígado/metabolismo , Camundongos , Resposta a Proteínas não Dobradas , Peixe-Zebra/genética
6.
Genome Res ; 28(9): 1415-1425, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30061115

RESUMO

With the emergence of zebrafish as an important model organism, a concerted effort has been made to study its transcriptome. This effort is limited, however, by gaps in zebrafish annotation, which are especially pronounced concerning transcripts dynamically expressed during zygotic genome activation (ZGA). To date, short-read sequencing has been the principal technology for zebrafish transcriptome annotation. In part because these sequence reads are too short for assembly methods to resolve the full complexity of the transcriptome, the current annotation is rudimentary. By providing direct observation of full-length transcripts, recently refined long-read sequencing platforms can dramatically improve annotation coverage and accuracy. Here, we leveraged the SMRT platform to study the transcriptome of zebrafish embryos before and after ZGA. Our analysis revealed additional novelty and complexity in the zebrafish transcriptome, identifying 2539 high-confidence novel transcripts that originated from previously unannotated loci and 1835 high-confidence new isoforms in previously annotated genes. We validated these findings using a suite of computational approaches including structural prediction, sequence homology, and functional conservation analyses, as well as by confirmatory transcript quantification with short-read sequencing data. Our analyses provided insight into new homologs and paralogs of functionally important proteins and noncoding RNAs, isoform switching occurrences, and different classes of novel splicing events. Several novel isoforms representing distinct splicing events were validated through PCR experiments, including the discovery and validation of a novel 8-kb transcript spanning multiple mir-430 elements, an important driver of early development. Our study provides a significantly improved zebrafish transcriptome annotation resource.


Assuntos
Anotação de Sequência Molecular , Transcriptoma , Peixe-Zebra/genética , Animais , Análise de Sequência de RNA/métodos , Análise de Sequência de RNA/normas , Homologia de Sequência do Ácido Nucleico
7.
J Am Chem Soc ; 142(44): 18782-18794, 2020 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-33090806

RESUMO

Nanoscale imine-linked covalent organic frameworks (nCOFs) were first loaded with the anticancer drug Doxorubicin (Dox), coated with magnetic iron oxide nanoparticles (γ-Fe2O3 NPs), and stabilized with a shell of poly(l-lysine) cationic polymer (PLL) for simultaneous synergistic thermo-chemotherapy treatment and MRI imaging. The pH responsivity of the resulting nanoagents (γ-SD/PLL) allowed the release of the drug selectively within the acidic microenvironment of late endosomes and lysosomes of cancer cells (pH 5.4) and not in physiological conditions (pH 7.4). γ-SD/PLL could efficiently generate high heat (48 °C) upon exposure to an alternating magnetic field due to the nCOF porous structure that facilitates the heat conduction, making γ-SD/PLL excellent heat mediators in an aqueous solution. The drug-loaded magnetic nCOF composites were cytotoxic due to the synergistic toxicity of Dox and the effects of hyperthermia in vitro on glioblastoma U251-MG cells and in vivo on zebrafish embryos, but they were not significantly toxic to noncancerous cells (HEK293). To the best of our knowledge, this is the first report of multimodal MRI probe and chemo-thermotherapeutic magnetic nCOF composites.


Assuntos
Compostos Férricos/química , Iminas/química , Nanopartículas de Magnetita/química , Nanopartículas/química , Animais , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/química , Doxorrubicina/metabolismo , Doxorrubicina/farmacologia , Portadores de Fármacos/química , Embrião não Mamífero/efeitos dos fármacos , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Hipertermia Induzida , Imageamento por Ressonância Magnética , Polilisina/química , Porosidade , Temperatura , Peixe-Zebra/crescimento & desenvolvimento
8.
Development ; 144(16): 2925-2939, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28698226

RESUMO

Complex cytoplasmic nucleotide-sensing mechanisms can recognize foreign DNA based on a lack of methylation and initiate an immune response to clear the infection. Zebrafish embryos with global DNA hypomethylation caused by mutations in the ubiquitin-like with PHD and ring finger domains 1 (uhrf1) or DNA methyltransferase 1 (dnmt1) genes exhibit a robust interferon induction characteristic of the first line of defense against viral infection. We found that this interferon induction occurred in non-immune cells and examined whether intracellular viral sensing pathways in these cells were the trigger. RNA-seq analysis of uhrf1 and dnmt1 mutants revealed widespread induction of Class I retrotransposons and activation of cytoplasmic DNA viral sensors. Attenuating Sting, phosphorylated Tbk1 and, importantly, blocking reverse transcriptase activity suppressed the expression of interferon genes in uhrf1 mutants. Thus, activation of transposons in cells with global DNA hypomethylation mimics a viral infection by activating cytoplasmic DNA sensors. This suggests that antiviral pathways serve as surveillance of cells that have derepressed intragenomic parasites due to DNA hypomethylation.


Assuntos
Metilação de DNA/fisiologia , Retroelementos/genética , Peixe-Zebra/embriologia , Animais , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Transativadores/genética , Transativadores/metabolismo , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
9.
Toxicol Appl Pharmacol ; 409: 115307, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33147493

RESUMO

Inorganic arsenic (iAs) is one of the most endemic toxicants worldwide and oxidative stress is a key cellular pathway underlying iAs toxicity. Other cellular stress response pathways, such as the unfolded protein response (UPR), are also impacted by iAs exposure, however it is not known how these pathways intersect to cause disease. We optimized the use of zebrafish larvae to identify the relationship between these cellular stress response pathways and arsenic toxicity. We found that the window of iAs susceptibility during zebrafish development corresponds with the development of the liver, and that even a 24-h exposure can cause lethality if administered to mature larvae, but not to early embryos. Acute exposure of larvae to iAs generates reactive oxygen species (ROS), an antioxidant response, endoplasmic reticulum (ER) stress and UPR activation in the liver. An in vivo assay using transgenic larvae expressing a GFP-tagged secreted glycoprotein in hepatocytes (Tg(fabp10a:Gc-EGFP)) revealed acute iAs exposure selectively decreased expression of Gc-EGFP, indicating that iAs impairs secretory protein folding in the liver. The transcriptional output of UPR activation is preceded by ROS production and activation of genes involved in the oxidative stress response. These studies implicate redox imbalance as the mechanism of iAs-induced ER stress and suggest that crosstalk between these pathways underlie iAs-induced hepatic toxicity.


Assuntos
Arsênio/toxicidade , Fígado/efeitos dos fármacos , Oxirredução/efeitos dos fármacos , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Animais , Animais Geneticamente Modificados , Antioxidantes/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Larva/efeitos dos fármacos , Larva/metabolismo , Fígado/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Peixe-Zebra
10.
Chemistry ; 26(23): 5270-5279, 2020 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-32077541

RESUMO

Triphenylphosphine (TPP) surface-functionalized and F-108 Pluronic-stabilized gold nanoparticles (F-108@TPP-AuNPs) have been synthesized through a one-step approach, leading to well-defined (9.6±1.6 nm) and water-soluble nanoparticles by microwave heating an aqueous solution of TPP-AuI Cl in the presence of a Pluronic polymer under basic conditions. TPP release was negligible under physiological conditions, but enhanced significantly at an acidic pH (5.4) mimicking that of a cancer cell. Laser irradiation (532 nm) raised the temperature of an aqueous solution of F-108@TPP-AuNPs to 51.7 °C within 5 min, confirming efficient light-to-heat conversion capabilities without significant photodegradation. TEM confirmed intracellular localization of F-108@TPP-AuNPs in the cytosol, endosomes and lysosomes of HeLa cells. F-108@TPP-AuNPs were well tolerated by HeLa cells and zebrafish embryos at ambient temperatures and became toxic upon heat activation, suggesting synergistic interactions between heat and cytotoxic action by TPP.


Assuntos
Antineoplásicos/farmacologia , Ouro/química , Nanopartículas Metálicas/química , Compostos Organofosforados/química , Antineoplásicos/química , Células HeLa , Humanos , Fototerapia , Polímeros/química , Temperatura
11.
Development ; 142(3): 510-21, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25564650

RESUMO

UHRF1 (ubiquitin-like, containing PHD and RING finger domains, 1) recruits DNMT1 to hemimethylated DNA during replication and is essential for maintaining DNA methylation. uhrf1 mutant zebrafish have global DNA hypomethylation and display embryonic defects, including a small liver, and they die as larvae. We make the surprising finding that, despite their reduced organ size, uhrf1 mutants express high levels of genes controlling S-phase and have many more cells undergoing DNA replication, as measured by BrdU incorporation. In contrast to wild-type hepatocytes, which are continually dividing during hepatic outgrowth and thus dilute the BrdU label, uhrf1 mutant hepatocytes retain BrdU throughout outgrowth, reflecting cell cycle arrest. Pulse-chase-pulse experiments with BrdU and EdU, and DNA content analysis indicate that uhrf1 mutant cells undergo DNA re-replication and that apoptosis is the fate of many of the re-replicating and arrested hepatocytes. Importantly, the DNA re-replication phenotype and hepatic outgrowth failure are preceded by global loss of DNA methylation. Moreover, uhrf1 mutants are phenocopied by mutation of dnmt1, and Dnmt1 knockdown in uhrf1 mutants enhances their small liver phenotype. Together, these data indicate that unscheduled DNA replication and failed cell cycle progression leading to apoptosis are the mechanisms by which DNA hypomethylation prevents organ expansion in uhrf1 mutants. We propose that cell cycle arrest leading to apoptosis is a strategy that restricts propagation of epigenetically damaged cells during embryogenesis.


Assuntos
Pontos de Checagem do Ciclo Celular/fisiologia , Metilação de DNA/fisiologia , Replicação do DNA/fisiologia , Embrião não Mamífero/fisiologia , Epigênese Genética/fisiologia , Fígado/embriologia , Peixe-Zebra/embriologia , Animais , Apoptose/fisiologia , Bromodesoxiuridina , Perfilação da Expressão Gênica , Marcação In Situ das Extremidades Cortadas , Estatísticas não Paramétricas , Transativadores/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
12.
Dev Biol ; 412(1): 99-113, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26851214

RESUMO

Landmark epigenetic events underlie early embryonic development, yet how epigenetic modifiers are regulated to achieve rapid epigenome re-patterning is not known. Uhrf1 and DNA methyltransferase 1 (Dnmt1) are known to largely mediate maintenance DNA methylation and Uhrf1 is also required for both Dnmt1 localization and stability. Here, we investigate how these two key epigenetic modifiers regulate early zebrafish development and characterize the developmental consequences of disrupting their homeostatic relationship. Unlike Uhrf1 knockdown, which causes developmental arrest and death prior to gastrulation, overexpression of human UHRF1 (WT-UHRF1) caused asymmetric epiboly, inefficient gastrulation and multi-systemic defects. UHRF1 phosphorylation was previously demonstrated as essential for zebrafish embryogenesis, and we found that penetrance of the asymmetric epiboly phenotype was significantly increased in embryos injected with mRNA encoding non-phosphorylatable UHRF1 (UHRF1(S661A)). Surprisingly, both WT-UHRF1 and UHRF1(S661A) overexpression caused DNA hypomethylation. However, since other approaches that caused an equivalent degree of DNA hypomethylation did not cause the asymmetric epiboly phenotype, we conclude that bulk DNA methylation is not the primary mechanism. Instead, UHRF1(S661A) overexpression resulted in accumulation of Dnmt1 protein and the overexpression of both WT and a catalytically inactive Dnmt1 phenocopied the assymetric epiboly phenotype. Dnmt1 knockdown suppressed the phenotype caused by UHRF1(S661A) overexpression, and Uhrf1 knockdown suppressed the effect of Dnmt1 overexpression. Therefore, we conclude that the interaction between these two proteins is the mechanism underlying the gastrulation defects. This indicates that Dnmt1 stability requires UHRF1 phosphorylation and that crosstalk between the proteins is essential for the function of these two important epigenetic regulators during gastrulation.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Gástrula/metabolismo , Transativadores/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/embriologia , Animais , DNA (Citosina-5-)-Metiltransferase 1 , Fosforilação
13.
PLoS Genet ; 10(5): e1004335, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24874946

RESUMO

Fatty liver disease (FLD) is characterized by lipid accumulation in hepatocytes and is accompanied by secretory pathway dysfunction, resulting in induction of the unfolded protein response (UPR). Activating transcription factor 6 (ATF6), one of three main UPR sensors, functions to both promote FLD during acute stress and reduce FLD during chronic stress. There is little mechanistic understanding of how ATF6, or any other UPR factor, regulates hepatic lipid metabolism to cause disease. We addressed this using zebrafish genetics and biochemical analyses and demonstrate that Atf6 is necessary and sufficient for FLD. atf6 transcription is significantly upregulated in the liver of zebrafish with alcoholic FLD and morpholino-mediated atf6 depletion significantly reduced steatosis incidence caused by alcohol. Moreover, overexpression of active, nuclear Atf6 (nAtf6) in hepatocytes caused FLD in the absence of stress. mRNA-Seq and qPCR analyses of livers from five day old nAtf6 transgenic larvae revealed upregulation of genes promoting glyceroneogenesis and fatty acid elongation, including fatty acid synthase (fasn), and nAtf6 overexpression in both zebrafish larvae and human hepatoma cells increased the incorporation of 14C-acetate into lipids. Srebp transcription factors are key regulators of lipogenic enzymes, but reducing Srebp activation by scap morpholino injection neither prevented FLD in nAtf6 transgenics nor synergized with atf6 knockdown to reduce alcohol-induced FLD. In contrast, fasn morpholino injection reduced FLD in nAtf6 transgenic larvae and synergistically interacted with atf6 to reduce alcoholic FLD. Thus, our data demonstrate that Atf6 is required for alcoholic FLD and epistatically interacts with fasn to cause this disease, suggesting triglyceride biogenesis as the mechanism of UPR induced FLD.


Assuntos
Fator 6 Ativador da Transcrição/genética , Fígado Gorduroso Alcoólico/genética , Hepatócitos/metabolismo , Ativação Transcricional/genética , Fator 6 Ativador da Transcrição/metabolismo , Fator 6 Ativador da Transcrição/toxicidade , Animais , Animais Geneticamente Modificados , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Modelos Animais de Doenças , Etanol/toxicidade , Fígado Gorduroso Alcoólico/etiologia , Fígado Gorduroso Alcoólico/metabolismo , Hepatócitos/patologia , Humanos , Metabolismo dos Lipídeos/genética , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Peixe-Zebra
14.
J Cell Sci ; 127(Pt 3): 485-95, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24481493

RESUMO

Over the past decades, studies using zebrafish have significantly advanced our understanding of the cellular basis for development and human diseases. Zebrafish have rapidly developing transparent embryos that allow comprehensive imaging of embryogenesis combined with powerful genetic approaches. However, forward genetic screens in zebrafish have generated unanticipated findings that are mirrored by human genetic studies: disruption of genes implicated in basic cellular processes, such as protein secretion or cytoskeletal dynamics, causes discrete developmental or disease phenotypes. This is surprising because many processes that were assumed to be fundamental to the function and survival of all cell types appear instead to be regulated by cell-specific mechanisms. Such discoveries are facilitated by experiments in whole animals, where zebrafish provides an ideal model for visualization and manipulation of organelles and cellular processes in a live vertebrate. Here, we review well-characterized mutants and newly developed tools that underscore this notion. We focus on the secretory pathway and microtubule-based trafficking as illustrative examples of how studying cell biology in vivo using zebrafish has broadened our understanding of the role fundamental cellular processes play in embryogenesis and disease.


Assuntos
Biologia Celular , Desenvolvimento Embrionário/genética , Proteínas de Transporte Vesicular/genética , Peixe-Zebra/embriologia , Animais , Movimento Celular/genética , Humanos , Microtúbulos/genética , Microtúbulos/metabolismo , Fenótipo , Vertebrados/genética , Proteínas de Transporte Vesicular/metabolismo , Peixe-Zebra/genética
15.
Gastroenterology ; 149(6): 1361-77, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26319012

RESUMO

As the incidence of hepatobiliary diseases increases, we must improve our understanding of the molecular, cellular, and physiological factors that contribute to the pathogenesis of liver disease. Animal models help us identify disease mechanisms that might be targeted therapeutically. Zebrafish (Danio rerio) have traditionally been used to study embryonic development but are also important to the study of liver disease. Zebrafish embryos develop rapidly; all of their digestive organs are mature in larvae by 5 days of age. At this stage, they can develop hepatobiliary diseases caused by developmental defects or toxin- or ethanol-induced injury and manifest premalignant changes within weeks. Zebrafish are similar to humans in hepatic cellular composition, function, signaling, and response to injury as well as the cellular processes that mediate liver diseases. Genes are highly conserved between humans and zebrafish, making them a useful system to study the basic mechanisms of liver disease. We can perform genetic screens to identify novel genes involved in specific disease processes and chemical screens to identify pathways and compounds that act on specific processes. We review how studies of zebrafish have advanced our understanding of inherited and acquired liver diseases as well as liver cancer and regeneration.


Assuntos
Modelos Animais de Doenças , Hepatopatias , Fígado , Peixe-Zebra , Animais , Humanos , Fígado/metabolismo , Fígado/patologia , Fígado/fisiopatologia , Hepatopatias/genética , Hepatopatias/metabolismo , Hepatopatias/patologia , Hepatopatias/fisiopatologia , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/metabolismo
16.
Adv Exp Med Biol ; 916: 169-97, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27165354

RESUMO

The cancer epigenome is fundamentally different than that of normal cells. How these differences arise in and contribute to carcinogenesis is not known, and studies using model organisms such as zebrafish provide an opportunity to address these important questions. Modifications of histones and DNA comprise the complex epigenome, and these influence chromatin structure, genome stability and gene expression, all of which are fundamental to the cellular changes that cause cancer. The cancer genome atlas covers the wide spectrum of genetic changes associated with nearly every cancer type, however, this catalog is currently uni-dimensional. As the pattern of epigenetic marks and chromatin structure in cancer cells is described and overlaid on the mutational landscape, the map of the cancer genome becomes multi-dimensional and highly complex. Two major questions remain in the field: (1) how the epigenome becomes repatterned in cancer and (2) which of these changes are cancer-causing. Zebrafish provide a tractable in vivo system to monitor the epigenome during transformation and to identify epigenetic drivers of cancer. In this chapter, we review principles of cancer epigenetics and discuss recent work using zebrafish whereby epigenetic modifiers were established as cancer driver genes, thus providing novel insights into the mechanisms of epigenetic reprogramming in cancer.


Assuntos
Modelos Animais de Doenças , Epigênese Genética , Neoplasias/genética , Acetilação , Animais , Metilação de DNA , Fosforilação , Peixe-Zebra
17.
Biol Open ; 13(3)2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38446164

RESUMO

The impacts of exposure to the pervasive environmental toxicant, inorganic arsenic (iAs), on human and fish health are well characterized and several lines of evidence suggest that some impacts can manifest years after exposure cessation. Using a developmental exposure protocol whereby zebrafish embryos were exposed to 0.5 and 1.5 mM iAs from 4-120 hours post fertilization (hpf) and then removed, we investigated the sustained effects of iAs on gene expression in the liver, survival, reproductive success, and susceptibility to iAs toxicity in the subsequent generation. Persistent exposure to iAs during development had substantial effects on the hepatic transcriptome, with 23% of all expressed genes significantly changed following developmental exposure. The gsto2 gene is involved in iAs metabolism and this gene was significantly downregulated in female livers 9 months after iAs was removed. Developmental exposure to 1.5 mM iAs, but not 0.5 mM, decreased survival by over 50% at 3 months of age. Adults that were developmentally exposed to 0.5 mM iAs had reduced mating success, but their offspring had no differences in observable aspects of development or their susceptibility to iAs toxicity. This demonstrates that developmental exposure of zebrafish to iAs reduces long-term survival, reproductive success and causes sustained changes to gsto2 expression in the liver.


Assuntos
Arsênio , Peixe-Zebra , Adulto , Animais , Humanos , Feminino , Peixe-Zebra/genética , Arsênio/toxicidade , Fígado , Reprodução , Comunicação Celular
18.
Proc Natl Acad Sci U S A ; 107(19): 8689-94, 2010 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-20395551

RESUMO

Cytosine DNA methylation is a heritable epigenetic mark present in many eukaryotic organisms. Although DNA methylation likely has a conserved role in gene silencing, the levels and patterns of DNA methylation appear to vary drastically among different organisms. Here we used shotgun genomic bisulfite sequencing (BS-Seq) to compare DNA methylation in eight diverse plant and animal genomes. We found that patterns of methylation are very similar in flowering plants with methylated cytosines detected in all sequence contexts, whereas CG methylation predominates in animals. Vertebrates have methylation throughout the genome except for CpG islands. Gene body methylation is conserved with clear preference for exons in most organisms. Furthermore, genes appear to be the major target of methylation in Ciona and honey bee. Among the eight organisms, the green alga Chlamydomonas has the most unusual pattern of methylation, having non-CG methylation enriched in exons of genes rather than in repeats and transposons. In addition, the Dnmt1 cofactor Uhrf1 has a conserved function in maintaining CG methylation in both transposons and gene bodies in the mouse, Arabidopsis, and zebrafish genomes.


Assuntos
Metilação de DNA/genética , Evolução Molecular , Plantas/genética , Animais , Arabidopsis/genética , Éxons/genética , Íntrons/genética , Mutação/genética , Análise de Sequência com Séries de Oligonucleotídeos , Fases de Leitura Aberta/genética , Filogenia , Sequências Repetitivas de Ácido Nucleico/genética , Transativadores/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
19.
Dis Model Mech ; 16(2)2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36752155

RESUMO

Celebrating, educating and mobilizing the global community to achieve equality for women and girls in science is recognized worldwide each February 11 on the International Day of Women and Girls in Science. This day serves as a rousing call for people of all genders to gain unfettered access to opportunities to become successful, engaged and productive scientists. One strategy to achieve this is to develop policies to ensure that all people can pursue science education, training and careers, unimpeded by their sex, gender and gender identity. Another strategy is to assure representation of women and non-binary people at every level of the scientific workforce. Collectively, these strategies and other important efforts are shifting the global mindset to one where women and girls are recognized as vital, capable and innovative contributors to disciplines that have historically been dominated by men. These male-dominated disciplines have evolved cultural norms rooted in masculine stereotypes and the next challenge is to continue this shift away from a scientific culture in which hypercompetitive, individualistic, solo artists work alone in single-minded pursuit of a breakthrough. Instead, the reality is that contemporary biomedical scientific research is a cosmopolitan culture; one where breakthroughs depend on collaboration, where data sharing is the norm and where all are encouraged to contribute their best ideas to help solve science's most vexing and exciting problems.


Assuntos
Pesquisa Biomédica , Identidade de Gênero , Humanos , Feminino , Masculino , Recursos Humanos
20.
Cancers (Basel) ; 15(8)2023 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-37190230

RESUMO

The ataxia-telangiectasia mutated (atm) gene is activated in response to genotoxic stress and leads to activation of the tp53 tumor suppressor gene which induces either senescence or apoptosis as tumor suppressive mechanisms. Atm also serves non-canonical functions in the response to oxidative stress and chromatin reorganization. We previously reported that overexpression of the epigenetic regulator and oncogene Ubiquitin Like with PHD and Ring Finger Domains 1 (UHRF1) in zebrafish hepatocytes resulted in tp53-dependent hepatocyte senescence, a small liver and larval lethality. We investigated the role of atm on UHRF1-mediated phenotypes by generating zebrafish atm mutants. atm-/- adults were viable but had reduction in fertility. Embryos developed normally but were protected from lethality caused by etoposide or H2O2 exposure and failed to fully upregulate Tp53 targets or oxidative stress response genes in response to these treatments. In contrast to the finding that Tp53 prevents the small liver phenotype caused by UHRF1 overexpression, atm mutation and exposure to H2O2 further reduced the liver size in UHRF1 overexpressing larvae whereas treatment with the antioxidant N-acetyl cysteine suppressed this phenotype. We conclude that UHRF1 overexpression in hepatocytes causes oxidative stress, and that loss of atm further enhances this, triggering elimination of these precancerous cells, leading to a small liver.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa