Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.675
Filtrar
1.
Life Sci Alliance ; 7(10)2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38991729

RESUMO

Embryonic germ cells develop rapidly to establish the foundation for future developmental trajectories, and in this process, they make critical lineage choices including the configuration of their unique identity and a decision on sex. Here, we use single-cell genomics patterns for the entire embryonic germline in Drosophila melanogaster along with the somatic gonadal precursors after embryonic gonad coalescence to investigate molecular mechanisms involved in the setting up and regulation of the germline program. Profiling of the early germline chromatin landscape revealed sex- and stage-specific features. In the male germline immediately after zygotic activation, the chromatin structure underwent a brief remodeling phase during which nucleosome density was lower and deconcentrated from promoter regions. These findings echoed enrichment analysis results of our genomics data in which top candidates were factors with the ability to mediate large-scale chromatin reorganization. Together, they point to the importance of chromatin regulation in the early germline and raise the possibility of a conserved epigenetic reprogramming-like process required for proper initiation of germline development.


Assuntos
Montagem e Desmontagem da Cromatina , Cromatina , Drosophila melanogaster , Desenvolvimento Embrionário , Animais , Masculino , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Cromatina/metabolismo , Cromatina/genética , Montagem e Desmontagem da Cromatina/genética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Células Germinativas Embrionárias/metabolismo , Células Germinativas Embrionárias/citologia , Células Germinativas/metabolismo , Epigênese Genética , Feminino , Nucleossomos/metabolismo , Nucleossomos/genética , Análise de Célula Única/métodos
2.
PLoS Comput Biol ; 20(6): e1011882, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38838038

RESUMO

In embryonic development and organogenesis, cells sharing identical genetic codes acquire diverse gene expression states in a highly reproducible spatial distribution, crucial for multicellular formation and quantifiable through positional information. To understand the spontaneous growth of complexity, we constructed a one-dimensional division-decision model, simulating the growth of cells with identical genetic networks from a single cell. Our findings highlight the pivotal role of cell division in providing positional cues, escorting the system toward states rich in information. Moreover, we pinpointed lateral inhibition as a critical mechanism translating spatial contacts into gene expression. Our model demonstrates that the spatial arrangement resulting from cell division, combined with cell lineages, imparts positional information, specifying multiple cell states with increased complexity-illustrated through examples in C.elegans. This study constitutes a foundational step in comprehending developmental intricacies, paving the way for future quantitative formulations to construct synthetic multicellular patterns.


Assuntos
Redes Reguladoras de Genes , Modelos Biológicos , Animais , Redes Reguladoras de Genes/genética , Caenorhabditis elegans/genética , Caenorhabditis elegans/embriologia , Caenorhabditis elegans/crescimento & desenvolvimento , Divisão Celular/fisiologia , Divisão Celular/genética , Biologia Computacional , Desenvolvimento Embrionário/fisiologia , Desenvolvimento Embrionário/genética , Linhagem da Célula , Simulação por Computador , Regulação da Expressão Gênica no Desenvolvimento/genética
3.
Genes Dev ; 38(9-10): 436-454, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38866556

RESUMO

Genome organization can regulate gene expression and promote cell fate transitions. The differentiation of germline stem cells (GSCs) to oocytes in Drosophila involves changes in genome organization mediated by heterochromatin and the nuclear pore complex (NPC). Heterochromatin represses germ cell genes during differentiation, and NPCs anchor these silenced genes to the nuclear periphery, maintaining silencing to allow for oocyte development. Surprisingly, we found that genome organization also contributes to NPC formation, mediated by the transcription factor Stonewall (Stwl). As GSCs differentiate, Stwl accumulates at boundaries between silenced and active gene compartments. Stwl at these boundaries plays a pivotal role in transitioning germ cell genes into a silenced state and activating a group of oocyte genes and nucleoporins (Nups). The upregulation of these Nups during differentiation is crucial for NPC formation and further genome organization. Thus, cross-talk between genome architecture and NPCs is essential for successful cell fate transitions.


Assuntos
Diferenciação Celular , Proteínas de Drosophila , Genoma de Inseto , Poro Nuclear , Oogênese , Animais , Oogênese/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Diferenciação Celular/genética , Poro Nuclear/metabolismo , Poro Nuclear/genética , Genoma de Inseto/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Feminino , Drosophila melanogaster/genética , Oócitos/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Drosophila/genética , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/genética
4.
Genes Dev ; 38(9-10): 393-414, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38834239

RESUMO

The fibroblast growth factor (FGF) pathway is a conserved signaling pathway required for embryonic development. Activated FGF receptor 1 (FGFR1) drives multiple intracellular signaling cascade pathways, including ERK/MAPK and PI3K/AKT, collectively termed canonical signaling. However, unlike Fgfr1-null embryos, embryos containing hypomorphic mutations in Fgfr1 lacking the ability to activate canonical downstream signals are still able to develop to birth but exhibit severe defects in all mesodermal-derived tissues. The introduction of an additional signaling mutation further reduces the activity of Fgfr1, leading to earlier lethality, reduced somitogenesis, and more severe changes in transcriptional outputs. Genes involved in migration, ECM interaction, and phosphoinositol signaling were significantly downregulated, proteomic analysis identified changes in interactions with endocytic pathway components, and cells expressing mutant receptors show changes in endocytic trafficking. Together, we identified processes regulating early mesoderm development by mechanisms involving both canonical and noncanonical Fgfr1 pathways, including direct interaction with cell adhesion components and endocytic regulation.


Assuntos
Endocitose , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Transdução de Sinais , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Animais , Mesoderma/embriologia , Mesoderma/metabolismo , Transdução de Sinais/genética , Endocitose/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Camundongos , Desenvolvimento Embrionário/genética , Transporte Proteico , Mutação
5.
Genes Dev ; 38(9-10): 455-472, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38866557

RESUMO

Monomethylation of lysine 20 of histone H4 (H4K20me1) is catalyzed by Set8 and thought to play important roles in many aspects of genome function that are mediated by H4K20me binding proteins. We interrogated this model in a developing animal by comparing in parallel the transcriptomes of Set8 null , H4 K20R/A , and l(3)mbt mutant Drosophila melanogaster We found that the gene expression profiles of H4 K20A and H4 K20R larvae are markedly different than Set8 null larvae despite similar reductions in H4K20me1. Set8 null mutant cells have a severely disrupted transcriptome and fail to proliferate in vivo, but these phenotypes are not recapitulated by mutation of H4 K20 , indicating that the developmental defects of Set8 null animals are largely due to H4K20me1-independent effects on gene expression. Furthermore, the H4K20me1 binding protein L(3)mbt is recruited to the transcription start sites of most genes independently of H4K20me even though genes bound by L(3)mbt have high levels of H4K20me1. Moreover, both Set8 and L(3)mbt bind to purified H4K20R nucleosomes in vitro. We conclude that gene expression changes in Set8 null and H4 K20 mutants cannot be explained by loss of H4K20me1 or L(3)mbt binding to chromatin and therefore that H4K20me1 does not play a large role in gene expression.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Histona-Lisina N-Metiltransferase , Histonas , Lisina , Animais , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Histonas/metabolismo , Histonas/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Metilação , Lisina/metabolismo , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Mutação , Transcriptoma/genética , Larva/genética , Larva/metabolismo , Larva/crescimento & desenvolvimento
6.
Genes Dev ; 38(9-10): 415-435, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38866555

RESUMO

The association of genomic loci to the nuclear periphery is proposed to facilitate cell type-specific gene repression and influence cell fate decisions. However, the interplay between gene position and expression remains incompletely understood, in part because the proteins that position genomic loci at the nuclear periphery remain unidentified. Here, we used an Oligopaint-based HiDRO screen targeting ∼1000 genes to discover novel regulators of nuclear architecture in Drosophila cells. We identified the heterochromatin-associated protein Stonewall (Stwl) as a factor promoting perinuclear chromatin positioning. In female germline stem cells (GSCs), Stwl binds and positions chromatin loci, including GSC differentiation genes, at the nuclear periphery. Strikingly, Stwl-dependent perinuclear positioning is associated with transcriptional repression, highlighting a likely mechanism for Stwl's known role in GSC maintenance and ovary homeostasis. Thus, our study identifies perinuclear anchors in Drosophila and demonstrates the importance of gene repression at the nuclear periphery for cell fate.


Assuntos
Diferenciação Celular , Núcleo Celular , Cromatina , Proteínas de Drosophila , Animais , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Cromatina/metabolismo , Cromatina/genética , Núcleo Celular/metabolismo , Núcleo Celular/genética , Feminino , Diferenciação Celular/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Células-Tronco/metabolismo , Células-Tronco/citologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Drosophila/genética , Células Germinativas/metabolismo
7.
Genes Dev ; 38(7-8): 308-321, 2024 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-38719541

RESUMO

The transcription factor Oct4/Pou5f1 is a component of the regulatory circuitry governing pluripotency and is widely used to induce pluripotency from somatic cells. Here we used domain swapping and mutagenesis to study Oct4's reprogramming ability, identifying a redox-sensitive DNA binding domain, cysteine residue (Cys48), as a key determinant of reprogramming and differentiation. Oct4 Cys48 sensitizes the protein to oxidative inhibition of DNA binding activity and promotes oxidation-mediated protein ubiquitylation. Pou5f1 C48S point mutation has little effect on undifferentiated embryonic stem cells (ESCs) but upon retinoic acid (RA) treatment causes retention of Oct4 expression, deregulated gene expression, and aberrant differentiation. Pou5f1 C48S ESCs also form less differentiated teratomas and contribute poorly to adult somatic tissues. Finally, we describe Pou5f1 C48S (Janky) mice, which in the homozygous condition are severely developmentally restricted after E4.5. Rare animals bypassing this restriction appear normal at birth but are sterile. Collectively, these findings uncover a novel Oct4 redox mechanism involved in both entry into and exit from pluripotency.


Assuntos
Diferenciação Celular , Reprogramação Celular , Fator 3 de Transcrição de Octâmero , Oxirredução , Fator 3 de Transcrição de Octâmero/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Animais , Camundongos , Diferenciação Celular/genética , Reprogramação Celular/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Tretinoína/farmacologia , Tretinoína/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos
8.
Nat Neurosci ; 27(7): 1274-1284, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38773348

RESUMO

Retrotransposons are mobile DNA sequences duplicated via transcription and reverse transcription of an RNA intermediate. Cis-regulatory elements encoded by retrotransposons can also promote the transcription of adjacent genes. Somatic LINE-1 (L1) retrotransposon insertions have been detected in mammalian neurons. It is, however, unclear whether L1 sequences are mobile in only some neuronal lineages or therein promote neurodevelopmental gene expression. Here we report programmed L1 activation by SOX6, a transcription factor critical for parvalbumin (PV) interneuron development. Mouse PV interneurons permit L1 mobilization in vitro and in vivo, harbor unmethylated L1 promoters and express full-length L1 mRNAs and proteins. Using nanopore long-read sequencing, we identify unmethylated L1s proximal to PV interneuron genes, including a novel L1 promoter-driven Caps2 transcript isoform that enhances neuron morphological complexity in vitro. These data highlight the contribution made by L1 cis-regulatory elements to PV interneuron development and transcriptome diversity, uncovered due to L1 mobility in this milieu.


Assuntos
Interneurônios , Elementos Nucleotídeos Longos e Dispersos , Parvalbuminas , Animais , Interneurônios/metabolismo , Interneurônios/fisiologia , Camundongos , Elementos Nucleotídeos Longos e Dispersos/genética , Parvalbuminas/metabolismo , Retroelementos/genética , Masculino , Neurogênese/fisiologia , Neurogênese/genética , Camundongos Endogâmicos C57BL , Regulação da Expressão Gênica no Desenvolvimento/genética
9.
Dev Biol ; 512: 35-43, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38710381

RESUMO

The larval stage of the Drosophila melanogaster life cycle is characterized by rapid growth and nutrient storage that occur over three instar stages separated by molts. In the third instar, the steroid hormone ecdysone drives key developmental processes and behaviors that occur in a temporally-controlled sequence and prepare the animal to undergo metamorphosis. Accurately staging Drosophila larvae within the final third instar is critical due to the rapid developmental progress at this stage, but it is challenging because the rate of development varies widely across a population of animals even if eggs are laid within a short period of time. Moreover, many methods to stage third instar larvae are cumbersome, and inherent variability in the rate of development confounds some of these approaches. Here we demonstrate the usefulness of the Sgs3-GFP transgene, a fusion of the Salivary gland secretion 3 (Sgs3) and GFP proteins, for staging third instar larvae. Sgs3-GFP is expressed in the salivary glands in an ecdysone-dependent manner from the midpoint of the third instar, and its expression pattern changes reproducibly as larvae progress through the third instar. We show that Sgs3-GFP can easily be incorporated into experiments, that it allows collection of developmentally-equivalent individuals from a mixed population of larvae, and that its use enables precise assessment of changing levels of hormones, metabolites, and gene expression during the second half of the third instar.


Assuntos
Drosophila melanogaster , Ecdisona , Proteínas de Fluorescência Verde , Larva , Fenótipo , Glândulas Salivares , Animais , Larva/metabolismo , Larva/genética , Glândulas Salivares/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Fluorescência Verde/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Ecdisona/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Genes Reporter , Regulação da Expressão Gênica no Desenvolvimento/genética , Animais Geneticamente Modificados , Metamorfose Biológica/genética
10.
Curr Opin Genet Dev ; 86: 102192, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38604005

RESUMO

Embryonic diapause in mammals is a period of developmental pause of the embryo at the blastocyst stage. During diapause, the blastocyst has minimal cell proliferation, metabolic activity and gene expression. At reactivation, blastocyst development resumes, characterised by increases in cell number, biosynthesis and metabolism. Until recently, it has been unknown how diapause is maintained without any loss of blastocyst viability. This review focuses on recent progress in the identification of molecular pathways occurring in the blastocyst that can both cause and maintain the diapause state. A switch to lipid metabolism now appears essential to maintaining the diapause state and is induced by forkhead box protein O1. The forkhead box protein O transcription family is important for diapause in insects, nematodes and fish, but this is the first time a conclusive role has been established in mammals. Multiple epigenetic modifications are also essential to inducing and maintaining the diapause state, including both DNA and RNA methylation mechanisms. Finally, it now appears that diapause embryos, dormant stem cells and chemotherapeutic-resistant cancer cells may all share a universal system of quiescence.


Assuntos
Blastocisto , Diapausa , Desenvolvimento Embrionário , Animais , Blastocisto/metabolismo , Blastocisto/citologia , Diapausa/genética , Desenvolvimento Embrionário/genética , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Metabolismo dos Lipídeos/genética , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo
11.
Curr Opin Genet Dev ; 86: 102197, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38648722

RESUMO

Temporal control is central to deploy and coordinate genetic programs during development. At present, there is limited understanding of the molecular mechanisms that govern the duration and speed of developmental processes. Timing mechanisms may run in parallel and/or interact with each other to integrate temporal signals throughout the organism. In this piece, we consider findings on the extrinsic control of developmental tempo and discuss the intrinsic roles of cell cycle, metabolic rates, protein turnover, and post-transcriptional mechanisms in the regulation of tempo during neural development.


Assuntos
Ciclo Celular , Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Neurogênese , Animais , Diferenciação Celular/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Ciclo Celular/genética , Neurogênese/genética , Humanos , Neurônios/metabolismo , Neurônios/citologia
12.
Trends Genet ; 40(6): 480-494, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38658255

RESUMO

Embryonic neurodevelopment, particularly neural progenitor differentiation into post-mitotic neurons, has been extensively studied. While the number and composition of post-mitotic neurons remain relatively constant from birth to adulthood, the brain undergoes significant postnatal maturation marked by major property changes frequently disrupted in neural diseases. This review first summarizes recent characterizations of the functional and molecular maturation of the postnatal nervous system. We then review regulatory mechanisms controlling the precise gene expression changes crucial for the intricate sequence of maturation events, highlighting experience-dependent versus cell-intrinsic genetic timer mechanisms. Despite significant advances in understanding of the gene-environmental regulation of postnatal neuronal maturation, many aspects remain unknown. The review concludes with our perspective on exciting future research directions in the next decade.


Assuntos
Interação Gene-Ambiente , Neurogênese , Neurônios , Humanos , Neurônios/citologia , Neurônios/metabolismo , Animais , Neurogênese/genética , Diferenciação Celular/genética , Mitose/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Encéfalo/citologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/citologia
13.
Nat Genet ; 56(4): 686-696, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38467791

RESUMO

To regulate expression, enhancers must come in proximity to their target gene. However, the relationship between the timing of enhancer-promoter (E-P) proximity and activity remains unclear, with examples of uncoupled, anticorrelated and correlated interactions. To assess this, we selected 600 characterized enhancers or promoters with tissue-specific activity in Drosophila embryos and performed Capture-C in FACS-purified myogenic or neurogenic cells during specification and tissue differentiation. This enabled direct comparison between E-P proximity and activity transitioning from OFF-to-ON and ON-to-OFF states across developmental conditions. This showed remarkably similar E-P topologies between specified muscle and neuronal cells, which are uncoupled from activity. During tissue differentiation, many new distal interactions emerge where changes in E-P proximity reflect changes in activity. The mode of E-P regulation therefore appears to change as embryogenesis proceeds, from largely permissive topologies during cell-fate specification to more instructive regulation during terminal tissue differentiation, when E-P proximity is coupled to activation.


Assuntos
Elementos Facilitadores Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Animais , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Regiões Promotoras Genéticas/genética , Drosophila/genética , Diferenciação Celular/genética
15.
Curr Opin Genet Dev ; 86: 102180, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38522266

RESUMO

Genes regulating developmental processes have been identified, but the mechanisms underlying their expression with the correct timing are still under investigation. Several genes show oscillatory expression that regulates the timing of developmental processes, such as somitogenesis and neurogenesis. These oscillations are also important for other developmental processes, such as cell proliferation and differentiation. In this review, we discuss the significance of oscillatory gene expression in developmental time and other forms of regulation.


Assuntos
Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Neurogênese , Regulação da Expressão Gênica no Desenvolvimento/genética , Animais , Diferenciação Celular/genética , Neurogênese/genética , Proliferação de Células/genética , Humanos , Somitos/crescimento & desenvolvimento , Ritmo Ultradiano/genética
16.
Nat Genet ; 56(4): 697-709, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38509386

RESUMO

In mice, exit from the totipotent two-cell (2C) stage embryo requires silencing of the 2C-associated transcriptional program. However, the molecular mechanisms involved in this process remain poorly understood. Here we demonstrate that the 2C-specific transcription factor double homeobox protein (DUX) mediates an essential negative feedback loop by inducing the expression of DUXBL to promote this silencing. We show that DUXBL gains accessibility to DUX-bound regions specifically upon DUX expression. Furthermore, we determine that DUXBL interacts with TRIM24 and TRIM33, members of the TRIM superfamily involved in gene silencing, and colocalizes with them in nuclear foci upon DUX expression. Importantly, DUXBL overexpression impairs 2C-associated transcription, whereas Duxbl inactivation in mouse embryonic stem cells increases DUX-dependent induction of the 2C-transcriptional program. Consequently, DUXBL deficiency in embryos results in sustained expression of 2C-associated transcripts leading to early developmental arrest. Our study identifies DUXBL as an essential regulator of totipotency exit enabling the first divergence of cell fates.


Assuntos
Genes Homeobox , Proteínas de Homeodomínio , Células-Tronco Embrionárias Murinas , Fatores de Transcrição , Animais , Camundongos , Diferenciação Celular , Regulação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo
17.
Curr Opin Genet Dev ; 86: 102178, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38461774

RESUMO

The rate of embryonic development is a species-specific trait that depends on the properties of the intracellular environment, namely, the rate at which gene products flow through the central dogma of molecular biology. Although any given step in the production and degradation of gene products could theoretically be co-opted by evolution to modulate developmental speed, species are observed to accelerate or slow down all steps simultaneously. This suggests the rate of these molecular processes is jointly regulated by an upstream, ultimate factor. Mitochondrial metabolism was recently proposed to act as an ultimate regulator by controlling the pace of protein synthesis upstream of developmental tempo. Alternative candidates for ultimate regulators include species-specific gene expression levels of factors involved in the central dogma, as well as species-specific cell size. Overall, much work remains to be done before we can confidently identify the ultimate causes of species-specific developmental rates.


Assuntos
Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Mitocôndrias , Mitocôndrias/metabolismo , Mitocôndrias/genética , Animais , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Especificidade da Espécie , Humanos , Biossíntese de Proteínas
18.
Curr Opin Genet Dev ; 86: 102179, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38490162

RESUMO

The chronologically ordered generation of distinct cell types is essential for the establishment of neuronal diversity and the formation of neuronal circuits. Recently, single-cell transcriptomic analyses of various areas of the developing vertebrate nervous system have provided evidence for the existence of a shared temporal patterning program that partitions neurons based on the timing of neurogenesis. In this review, I summarize the findings that lead to the proposal of this shared temporal program before focusing on the developing spinal cord to discuss how temporal patterning in general and this program specifically contributes to the ordered formation of neuronal circuits.


Assuntos
Padronização Corporal , Regulação da Expressão Gênica no Desenvolvimento , Tubo Neural , Neurogênese , Medula Espinal , Vertebrados , Animais , Tubo Neural/crescimento & desenvolvimento , Neurogênese/genética , Vertebrados/crescimento & desenvolvimento , Vertebrados/genética , Vertebrados/embriologia , Padronização Corporal/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Medula Espinal/crescimento & desenvolvimento , Medula Espinal/embriologia , Neurônios/citologia , Neurônios/metabolismo , Humanos
19.
Dev Biol ; 510: 40-49, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38493946

RESUMO

The Spalt transcriptional regulators participate in a variety of cell fate decisions during multicellular development. Vertebrate Spalt proteins have been mostly associated to the organization of heterochromatic regions, but they also contribute regulatory functions through binding to A/T rich motives present in their target genes. The developmental processes in which the Drosophila spalt genes participate are well known through genetic analysis, but the mechanism by which the Spalt proteins regulate transcription are still unknown. Furthermore, despite the prominent changes in gene expression associated to mutations in the spalt genes, the specific DNA sequences they bind are unknow. Here, we analyze a DNA fragment present in the regulatory region of the knirps gene. Spalt proteins are candidate repressors of knirps expression during the formation of the venation pattern in the wing disc, and we identified a minimal conserved 30bp sequence that binds to Spalt major both in vivo and in vitro. This sequence mediates transcriptional repression in the central region of the wing blade, constituting the first confirmed case of a direct regulatory interaction between Spalt major and its target DNA in Drosophila. Interestingly, we also find similar sequences in a set of eight novel candidate Spalt target genes, pointing to a common mechanism of transcriptional repression mediated by Spalt proteins.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/metabolismo , Discos Imaginais/metabolismo , Proteínas Repressoras/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Fatores de Transcrição/metabolismo , Proteínas de Homeodomínio/metabolismo , Asas de Animais
20.
Adv Sci (Weinh) ; 11(20): e2308018, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38493496

RESUMO

Epigenetic modifiers that accumulate in oocytes, play a crucial role in steering the developmental program of cleavage embryos and initiating life. However, the identification of key maternal epigenetic regulators remains elusive. In the findings, the essential role of maternal Ep400, a chaperone for H3.3, in oocyte quality and early embryo development in mice is highlighted. Depletion of Ep400 in oocytes resulted in a decline in oocyte quality and abnormalities in fertilization. Preimplantation embryos lacking maternal Ep400 exhibited reduced major zygotic genome activation (ZGA) and experienced developmental arrest at the 2-to-4-cell stage. The study shows that EP400 forms protein complex with NFYA, occupies promoters of major ZGA genes, modulates H3.3 distribution between euchromatin and heterochromatin, promotes transcription elongation, activates the expression of genes regulating mitochondrial functions, and facilitates the expression of rate-limiting enzymes of the TCA cycle. This intricate process driven by Ep400 ensures the proper execution of the developmental program, emphasizing its critical role in maternal-to-embryonic transition.


Assuntos
Oócitos , Zigoto , Animais , Camundongos , Oócitos/metabolismo , Zigoto/metabolismo , Feminino , Desenvolvimento Embrionário/genética , Cromatina/metabolismo , Cromatina/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Epigênese Genética/genética , Proteína p300 Associada a E1A
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...