Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Neural Dev ; 19(1): 13, 2024 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-39049046

RESUMO

The cell-adhesion molecule NEPH1 is required for maintaining the structural integrity and function of the glomerulus in the kidneys. In the nervous system of Drosophila and C. elegans, it is involved in synaptogenesis and axon branching, which are essential for establishing functional circuits. In the mammalian nervous system, the expression regulation and function of Neph1 has barely been explored. In this study, we provide a spatiotemporal characterization of Neph1 expression in mouse dorsal root ganglia (DRGs) and spinal cord. After the neurogenic phase, Neph1 is broadly expressed in the DRGs and in their putative targets at the dorsal horn of the spinal cord, comprising both GABAergic and glutamatergic neurons. Interestingly, we found that PRRXL1, a homeodomain transcription factor that is required for proper establishment of the DRG-spinal cord circuit, prevents a premature expression of Neph1 in the superficial laminae of the dorsal spinal cord at E14.5, but has no regulatory effect on the DRGs or on either structure at E16.5. By chromatin immunoprecipitation analysis of the dorsal spinal cord, we identified four PRRXL1-bound regions within the Neph1 introns, suggesting that PRRXL1 directly regulates Neph1 transcription. We also showed that Neph1 is required for branching, especially at distal neurites. Together, our work showed that Prrxl1 prevents the early expression of Neph1 in the superficial dorsal horn, suggesting that Neph1 might function as a downstream effector gene for proper assembly of the DRG-spinal nociceptive circuit.


Assuntos
Gânglios Espinais , Proteínas de Homeodomínio , Neuritos , Corno Dorsal da Medula Espinal , Fatores de Transcrição , Animais , Camundongos , Corno Dorsal da Medula Espinal/metabolismo , Corno Dorsal da Medula Espinal/citologia , Neuritos/metabolismo , Neuritos/fisiologia , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Gânglios Espinais/metabolismo , Gânglios Espinais/citologia , Gânglios Espinais/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso
2.
Genesis ; 62(1): e23582, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38069547

RESUMO

Tfap2b, a pivotal transcription factor, plays critical roles within neural crest cells and their derived lineage. To unravel the intricate lineage dynamics and contribution of these Tfap2b+ cells during craniofacial development, we established a Tfap2b-CreERT2 knock-in transgenic mouse line using the CRISPR-Cas9-mediated homologous direct repair. By breeding with tdTomato reporter mice and initiating Cre activity through tamoxifen induction at distinct developmental time points, we show the Tfap2b lineage within the key neural crest-derived domains, such as the facial mesenchyme, midbrain, cerebellum, spinal cord, and limbs. Notably, the migratory neurons stemming from the dorsal root ganglia are visible subsequent to Cre activity initiated at E8.5. Intriguingly, Tfap2b+ cells, serving as the progenitors for limb development, show activity predominantly commencing at E10.5. Across the mouse craniofacial landscape, Tfap2b exhibits a widespread presence throughout the facial organs. Here we validate its role as a marker of progenitors in tooth development and have confirmed that this process initiates from E12.5. Our study not only validates the Tfap2b-CreERT2 transgenic line, but also provides a powerful tool for lineage tracing and genetic targeting of Tfap2b-expressing cells and their progenitor in a temporally and spatially regulated manner during the intricate process of development and organogenesis.


Assuntos
Sistemas CRISPR-Cas , Tamoxifeno , Camundongos , Animais , Tamoxifeno/farmacologia , Camundongos Transgênicos , Proteína Vermelha Fluorescente , Integrases/genética , Integrases/metabolismo
3.
Development ; 151(3)2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-37982461

RESUMO

Early organogenesis represents a key step in animal development, during which pluripotent cells diversify to initiate organ formation. Here, we sampled 300,000 single-cell transcriptomes from mouse embryos between E8.5 and E9.5 in 6-h intervals and combined this new dataset with our previous atlas (E6.5-E8.5) to produce a densely sampled timecourse of >400,000 cells from early gastrulation to organogenesis. Computational lineage reconstruction identified complex waves of blood and endothelial development, including a new programme for somite-derived endothelium. We also dissected the E7.5 primitive streak into four adjacent regions, performed scRNA-seq and predicted cell fates computationally. Finally, we defined developmental state/fate relationships by combining orthotopic grafting, microscopic analysis and scRNA-seq to transcriptionally determine cell fates of grafted primitive streak regions after 24 h of in vitro embryo culture. Experimentally determined fate outcomes were in good agreement with computationally predicted fates, demonstrating how classical grafting experiments can be revisited to establish high-resolution cell state/fate relationships. Such interdisciplinary approaches will benefit future studies in developmental biology and guide the in vitro production of cells for organ regeneration and repair.


Assuntos
Gastrulação , Organogênese , Camundongos , Animais , Diferenciação Celular , Organogênese/genética , Linha Primitiva , Endotélio , Embrião de Mamíferos , Mamíferos
4.
BMC Genomics ; 24(1): 663, 2023 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-37924034

RESUMO

BACKGROUND: H3K9me3 and DNA methylation co-marked CpG-rich regions (CHMs) are functionally important in mouse pre-implantation embryos, but their characteristics in other biological processes are still largely unknown. RESULTS: In this study, we performed a comprehensive analysis to characterize CHMs during 6 mouse developmental processes, identifying over 2,600 CHMs exhibiting stable co-mark of H3K9me3 and DNA methylation patterns at CpG-rich regions. We revealed the distinctive features of CHMs, including elevated H3K9me3 signals and a significant presence in euchromatin and the potential role in silencing younger long terminal repeats (LTRs), especially in some ERVK subfamilies. The results highlight the distinct nature of universal CHMs compared to CpG-rich nonCHMs in terms of location, LTR enrichment, and DNA sequence features, enhancing our understanding of CpG-rich regions' regulatory roles. CONCLUSIONS: This study characterizes the features of CHMs in multiple developmental processes and broadens our understanding of the regulatory roles of CpG-rich regions.


Assuntos
Metilação de DNA , Histonas , Animais , Camundongos , Histonas/metabolismo , DNA , Sequências Repetidas Terminais , Ilhas de CpG
5.
Genes Dev ; 37(13-14): 590-604, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37532472

RESUMO

Nucleosome positioning can alter the accessibility of DNA-binding proteins to their cognate DNA elements, and thus its precise control is essential for cell identity and function. Mammalian preimplantation embryos undergo temporal changes in gene expression and cell potency, suggesting the involvement of dynamic epigenetic control during this developmental phase. However, the dynamics of nucleosome organization during early development are poorly understood. In this study, using a low-input MNase-seq method, we show that nucleosome positioning is globally obscure in zygotes but becomes well defined during subsequent development. Down-regulation of the chromatin assembly in embryonic stem cells can partially reverse nucleosome organization into a zygote-like pattern, suggesting a possible link between the chromatin assembly pathway and fuzzy nucleosomes in zygotes. We also reveal that YY1, a zinc finger-containing transcription factor expressed upon zygotic genome activation, regulates the de novo formation of well-positioned nucleosome arrays at the regulatory elements through identifying YY1-binding sites in eight-cell embryos. The YY1-binding regions acquire H3K27ac enrichment around the eight-cell and morula stages, and YY1 depletion impairs the morula-to-blastocyst transition. Thus, our study delineates the remodeling of nucleosome organization and its underlying mechanism during early mouse development.


Assuntos
Nucleossomos , Fatores de Transcrição , Animais , Camundongos , Cromatina , Montagem e Desmontagem da Cromatina/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Mamíferos/genética , Nucleossomos/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
6.
Dev Cell ; 58(19): 1983-1995.e7, 2023 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-37499658

RESUMO

The mammalian body plan is shaped by rhythmic segmentation of mesoderm into somites, which are transient embryonic structures that form down each side of the neural tube. We have analyzed the genome-wide transcriptional and chromatin dynamics occurring within nascent somites, from early inception of somitogenesis to the latest stages of body plan establishment. We created matched gene expression and open chromatin maps for the three leading pairs of somites at six time points during mouse embryonic development. We show that the rate of somite differentiation accelerates as development progresses. We identified a conserved maturation program followed by all somites, but somites from more developed embryos concomitantly switch on differentiation programs from derivative cell lineages soon after segmentation. Integrated analysis of the somitic transcriptional and chromatin activities identified opposing regulatory modules controlling the onset of differentiation. Our results provide a powerful, high-resolution view of the molecular genetics underlying somitic development in mammals.


Assuntos
Desenvolvimento Embrionário , Somitos , Gravidez , Feminino , Camundongos , Animais , Desenvolvimento Embrionário/genética , Mesoderma , Diferenciação Celular/genética , Cromatina/genética , Mamíferos
7.
Development ; 150(10)2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-37102683

RESUMO

Signaling pathways regulate the patterns of Hox gene expression that underlie their functions in the specification of axial identity. Little is known about the properties of cis-regulatory elements and underlying transcriptional mechanisms that integrate graded signaling inputs to coordinately control Hox expression. Here, we optimized a single molecule fluorescent in situ hybridization (smFISH) technique with probes spanning introns to evaluate how three shared retinoic acid response element (RARE)-dependent enhancers in the Hoxb cluster regulate patterns of nascent transcription in vivo at the level of single cells in wild-type and mutant embryos. We predominately detect nascent transcription of only a single Hoxb gene in each cell, with no evidence for simultaneous co-transcriptional coupling of all or specific subsets of genes. Single and/or compound RARE mutations indicate that each enhancer differentially impacts global and local patterns of nascent transcription, suggesting that selectivity and competitive interactions between these enhancers is important to robustly maintain the proper levels and patterns of nascent Hoxb transcription. This implies that rapid and dynamic regulatory interactions potentiate transcription of genes through combined inputs from these enhancers in coordinating the retinoic acid response.


Assuntos
Proteínas de Homeodomínio , Tretinoína , Camundongos , Animais , Tretinoína/metabolismo , Proteínas de Homeodomínio/metabolismo , Camundongos Transgênicos , Tubo Neural/metabolismo , Hibridização in Situ Fluorescente , Elementos Facilitadores Genéticos
8.
Front Psychiatry ; 13: 998695, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36518366

RESUMO

Introduction: Autism spectrum disorder (ASD) is considered a significant behavioral problem that is characterized by impairment in social interaction and communication. It is believed that some cases of ASD originate in the intrauterine maternal environment. Therefore, we hypothesized that there might be qualitative changes in the interaction between the mother and fetus in ASD during the prenatal period, hence, we investigated the similarity patterns between maternal and fetal heart rate (HR). Methods: In this study, we first demonstrate the presence and formation of similarities between maternal and fetal RR interval (RRI) collected from typical developmental mice at different embryonic days (EDs), ED13.5, ED15.5, ED17.5, and ED18.5. The similarities were quantified by means of cross-correlation (CC) and magnitude-squared coherence (MSC) analyses. Correlation analysis between the CC coefficients and EDs and between MSC coefficients and EDs showed that the same coefficients increase with EDs, suggesting that similarities between maternal and fetal RRI are associated with typical fetal development. Next, because maternal and fetal similarities were indicative of development, a comparison analysis between the autism mouse model (injected with valproic acid (VPA)), and the control group (injected with saline) was performed for ED15.5 and ED18.5. Results: The results of the comparison showed that the CC and MSC coefficients of VPA fetuses were significantly lower than that of the control group. The lower coefficients in VPA-treated mice suggest that they could be one of the features of ASD symptoms. The findings of this study can assist in identifying potential ASD causes during the prenatal period.

9.
Dev Cell ; 57(17): 2140-2150.e5, 2022 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-36055247

RESUMO

Normal organogenesis cannot be recapitulated in vitro for mammalian organs, unlike in species including Drosophila and zebrafish. Available 3D data in the form of ex vivo images only provide discrete snapshots of the development of an organ morphology. Here, we propose a computer-based approach to recreate its continuous evolution in time and space from a set of 3D volumetric images. Our method is based on the remapping of shape data into the space of the coefficients of a spherical harmonics expansion where a smooth interpolation over time is simpler. We tested our approach on mouse limb buds and embryonic hearts. A key advantage of this method is that the resulting 4D trajectory can take advantage of all the available data while also being able to interpolate well through time intervals for which there are little or no data. This allows for a quantitative, data-driven 4D description of mouse limb morphogenesis.


Assuntos
Imageamento Tridimensional , Organogênese , Algoritmos , Animais , Imageamento Tridimensional/métodos , Mamíferos , Camundongos
10.
Dev Biol ; 483: 118-127, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34958748

RESUMO

The primordia of the post-otic mouse embryo forms largely from a bipotential cell population containing neuromesodermal progenitors (NMP) which reside in the tail bud and contribute to the elaboration of the major body axis after gastrulation. The mechanisms by which the NMP population is both maintained and subsequently directed down mesodermal and neural lineages is incompletely understood. The vertebrate transcription factor Cdx2, is essential for axial elongation and has been implicated in maintaining the NMP niche and in specification of NMP derivatives. To better understand the role of the Cdx family in axial elongation, we employed a conditional mutant allele which evokes total loss of Cdx function, and enriched for tail bud progenitors through the use of a Pax2-GFP transgenic reporter. Using this approach, we identified 349 Cdx-dependent genes by RNA sequencing (RNA-seq). From these, Gene Ontology and chromatin immunoprecipitation analysis further revealed a number of putative direct Cdx candidate target genes implicated in axial elongation, including Sp8, Isl1, Evx1, Zic3 and Nr2f1. Additional analysis of available single-cell RNA-seq data from mouse tail buds revealed the co-expression of Sp8, Isl1, Evx1 and Zic3 with Cdx2 in putative NMP cells, while Nr2f1 was excluded from this population. These findings identify a number of novel Cdx targets and provide further insight into the critical roles for Cdx in elaborating the post-otic embryo.


Assuntos
Padronização Corporal/genética , Fator de Transcrição CDX2/metabolismo , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário/genética , Proteínas de Homeodomínio/metabolismo , Transdução de Sinais/genética , Animais , Fator de Transcrição CDX2/genética , Fator I de Transcrição COUP/genética , Imunoprecipitação da Cromatina/métodos , Proteínas de Ligação a DNA/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas com Homeodomínio LIM/genética , Masculino , Mesoderma/metabolismo , Camundongos , Camundongos Transgênicos , Mutação , RNA-Seq/métodos , Células-Tronco/metabolismo , Fatores de Transcrição/genética
11.
Yi Chuan ; 43(4): 375-384, 2021 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-33972211

RESUMO

Mouse Developmental and Metabolic Phenotype Repository (MDMPR) is an open access, real-time database which dedicates to share mouse resources and phenotype data. MDMPR is supported by the National Key Research and Development Project "Establishment of Mouse Developmental and Metabolic Phenotype Repository" within the Key Project of "Developmental Programming and Its Metabolic Regulation" from the Ministry of Science and Technology of the People's Republic of China's program. In the next 5 years, MDMPR will create 500 mutant mouse models related to development and metabolism, perform standard phenotyping analysis, and establish a phenotype database. MDMPR is a combination of resources and data repository, has several sub-systems, including the ES cell database, the project management system, the breeding management system, the sperm bank management system and the phenotyping database. These systems digitalize all data and ensure their authenticity in real-time. Besides the gradual increase of data during the project, MDMPR will also integrate other resources, such as human KO ES cell database, STRING database, database of Core Transcriptional Regulatory Circuitries and Enhancer-Indel database. MDMPR is anticipated to contribute to various areas of developmental and metabolic research to investigators through more convenient accesses to the resources and data in one-stop manner, thereby accelerating the research processes and ultimately serving the medical causes of human health.


Assuntos
Fenótipo , Animais , Bases de Dados Factuais , Modelos Animais de Doenças , Camundongos
12.
Front Cell Dev Biol ; 9: 587096, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33575260

RESUMO

Programmed cell senescence is a cellular process that seems to contribute to embryo development, in addition to cell proliferation, migration, differentiation and programmed cell death, and has been observed in evolutionary distant organisms such as mammals, amphibians, birds and fish. Programmed cell senescence is a phenotype similar to stress-induced cellular senescence, characterized by the expression of the cell cycle inhibitors p21CIP1/WAF and p16INK4A, increased activity of a lysosomal enzyme with beta-galactosidase activity (coined senescence-associated beta-galactosidase) and secretion of growth factors, interleukins, chemokines, metalloproteases, etc., collectively known as a senescent-associated secretory phenotype that instructs surrounding tissue. How wide is the distribution of programmed cell senescence during mouse development and its specific mechanisms to shape the embryo are still poorly understood. Here, we investigated whether markers of programmed cell senescence are found in the developing mouse spinal cord and notochord. We found discrete areas and developmental windows with high senescence-associated beta galactosidase in both spinal cord and notochord, which was reduced in mice embryos developed ex-utero in the presence of the senolytic ABT-263. Expression of p21CIP1/WAF was documented in epithelial cells of the spinal cord and the notochord, while p16INK4A was observed in motoneurons. Treatment with the senolytic ABT-263 decreased the number of motoneurons, supporting their senescent phenotype. Our data suggest that a subpopulation of motoneurons in the developing spinal cord, as well as some notochord cells undergo programmed cell senescence.

13.
Elife ; 102021 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-33570495

RESUMO

3D imaging data necessitate 3D reference atlases for accurate quantitative interpretation. Existing computational methods to generate 3D atlases from 2D-derived atlases result in extensive artifacts, while manual curation approaches are labor-intensive. We present a computational approach for 3D atlas construction that substantially reduces artifacts by identifying anatomical boundaries in the underlying imaging data and using these to guide 3D transformation. Anatomical boundaries also allow extension of atlases to complete edge regions. Applying these methods to the eight developmental stages in the Allen Developing Mouse Brain Atlas (ADMBA) led to more comprehensive and accurate atlases. We generated imaging data from 15 whole mouse brains to validate atlas performance and observed qualitative and quantitative improvement (37% greater alignment between atlas and anatomical boundaries). We provide the pipeline as the MagellanMapper software and the eight 3D reconstructed ADMBA atlases. These resources facilitate whole-organ quantitative analysis between samples and across development.


The research community needs precise, reliable 3D atlases of organs to pinpoint where biological structures and processes are located. For instance, these maps are essential to understand where specific genes are turned on or off, or the spatial organization of various groups of cells over time. For centuries, atlases have been built by thinly 'slicing up' an organ, and then precisely representing each 2D layer. Yet this approach is imperfect: each layer may be accurate on its own, but inevitable mismatches appear between the slices when viewed in 3D or from another angle. Advances in microscopy now allow entire organs to be imaged in 3D. Comparing these images with atlases could help to detect subtle differences that indicate or underlie disease. However, this is only possible if 3D maps are accurate and do not feature mismatches between layers. To create an atlas without such artifacts, one approach consists in starting from scratch and manually redrawing the maps in 3D, a labor-intensive method that discards a large body of well-established atlases. Instead, Young et al. set out to create an automated method which could help to refine existing 'layer-based' atlases, releasing software that anyone can use to improve current maps. The package was created by harnessing eight atlases in the Allen Developing Mouse Brain Atlas, and then using the underlying anatomical images to resolve discrepancies between layers or fill out any missing areas. Known as MagellanMapper, the software was extensively tested to demonstrate the accuracy of the maps it creates, including comparison to whole-brain imaging data from 15 mouse brains. Armed with this new software, researchers can improve the accuracy of their atlases, helping them to understand the structure of organs at the level of the cell and giving them insight into a broad range of human disorders.


Assuntos
Encéfalo/anatomia & histologia , Processamento de Imagem Assistida por Computador/métodos , Imageamento Tridimensional/métodos , Imageamento por Ressonância Magnética/métodos , Animais , Encéfalo/crescimento & desenvolvimento , Feminino , Masculino , Camundongos
14.
Metabolites ; 10(11)2020 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-33228074

RESUMO

A large part of metabolomics research relies on experiments involving mouse models, which are usually 6 to 20 weeks of age. However, in this age range mice undergo dramatic developmental changes. Even small age differences may lead to different metabolomes, which in turn could increase inter-sample variability and impair the reproducibility and comparability of metabolomics results. In order to learn more about the variability of the murine plasma metabolome, we analyzed male and female C57BL/6J, C57BL/6NTac, 129S1/SvImJ, and C3HeB/FeJ mice at 6, 10, 14, and 20 weeks of age, using targeted metabolomics (BIOCRATES AbsoluteIDQ™ p150 Kit). Our analysis revealed high variability of the murine plasma metabolome during adolescence and early adulthood. A general age range with minimal variability, and thus a stable metabolome, could not be identified. Age-related metabolomic changes as well as the metabolite profiles at specific ages differed markedly between mouse strains. This observation illustrates the fact that the developmental timing in mice is strain specific. We therefore stress the importance of deliberate strain choice, as well as consistency and precise documentation of animal age, in metabolomics studies.

15.
Mech Dev ; 163: 103617, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32473204

RESUMO

During mouse embryonic development a mass of pluripotent epiblast tissue is transformed during gastrulation to generate the three definitive germ layers: endoderm, mesoderm, and ectoderm. During gastrulation, a spatiotemporally controlled sequence of events results in the generation of organ progenitors and positions them in a stereotypical fashion throughout the embryo. Key to the correct specification and differentiation of these cell fates is the establishment of an axial coordinate system along with the integration of multiple signals by individual epiblast cells to produce distinct outcomes. These signaling domains evolve as the anterior-posterior axis is established and the embryo grows in size. Gastrulation is initiated at the posteriorly positioned primitive streak, from which nascent mesoderm and endoderm progenitors ingress and begin to diversify. Advances in technology have facilitated the elaboration of landmark findings that originally described the epiblast fate map and signaling pathways required to execute those fates. Here we will discuss the current state of the field and reflect on how our understanding has shifted in recent years.


Assuntos
Padronização Corporal/genética , Diferenciação Celular/genética , Desenvolvimento Embrionário/genética , Gastrulação/genética , Especificidade de Órgãos/genética , Animais , Linhagem da Célula/genética , Ectoderma/crescimento & desenvolvimento , Endoderma/crescimento & desenvolvimento , Feminino , Gástrula/crescimento & desenvolvimento , Gastrulação/fisiologia , Camadas Germinativas/crescimento & desenvolvimento , Mesoderma/crescimento & desenvolvimento , Camundongos , Gravidez
16.
Elife ; 92020 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-32129762

RESUMO

ARL13B is a regulatory GTPase highly enriched in cilia. Complete loss of Arl13b disrupts cilia architecture, protein trafficking and Sonic hedgehog signaling. To determine whether ARL13B is required within cilia, we knocked in a cilia-excluded variant of ARL13B (V358A) and showed it retains all known biochemical function. We found that ARL13BV358A protein was expressed but could not be detected in cilia, even when retrograde ciliary transport was blocked. We showed Arl13bV358A/V358A mice are viable and fertile with normal Shh signal transduction. However, in contrast to wild type cilia, Arl13bV358A/V358A cells displayed short cilia and lacked ciliary ARL3 and INPP5E. These data indicate that ARL13B's role within cilia can be uncoupled from its function outside of cilia. Furthermore, these data imply that the cilia defects upon complete absence of ARL13B do not underlie the alterations in Shh transduction, which is unexpected given the requirement of cilia for Shh transduction.


Assuntos
Fatores de Ribosilação do ADP/genética , Fatores de Ribosilação do ADP/metabolismo , Cílios/metabolismo , Proteínas Hedgehog/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Animais , Sequência de Bases , Sistemas CRISPR-Cas , Desenvolvimento Embrionário/genética , Edição de Genes , Expressão Gênica , Camundongos , Camundongos Knockout , Mutação , Fenótipo
17.
Exp Eye Res ; 194: 108001, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32173378

RESUMO

Elastic fibres provide tissues with elasticity and flexibility. In the healthy human cornea, elastic fibres are limited to the posterior region of the peripheral stroma, but their specific functional role remains elusive. Here, we examine the physical and structural characteristics of the cornea during development in the mgΔloxPneo dominant-negative mouse model for Marfan syndrome, in which the physiological extracellular matrix of its elastic-fibre rich tissues is disrupted by the presence of a dysfunctional fibrillin-1 glycoprotein. Optical coherence tomography demonstrated a reduced corneal thickness in the mutant compared to wild type mice from embryonic day 16.5 until adulthood. X-ray scattering and electron microscopy revealed a disruption to both the elastic fibre and collagen fibril ultrastructure in the knockout mice, as well as abnormally low levels of the proteoglycan decorin. It is suggested that these alterations might be a result of increased transforming growth factor beta signalling. To conclude, this study has demonstrated corneal structure and ultrastructure to be altered when fibrillin-1 is disrupted and has provided insights into the role of fibrillin-1 in developing a functional cornea.


Assuntos
Córnea/anormalidades , Matriz Extracelular/metabolismo , Síndrome de Marfan/patologia , Animais , Córnea/metabolismo , Córnea/ultraestrutura , Modelos Animais de Doenças , Elasticidade , Feminino , Fibrilina-1/metabolismo , Síndrome de Marfan/metabolismo , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Tomografia de Coerência Óptica/métodos
18.
Curr Top Dev Biol ; 136: 429-454, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31959298

RESUMO

Gastrulation is a central process in mammalian development in which a spatiotemporally coordinated series of events driven by cross-talk between adjacent embryonic and extra-embryonic tissues results in stereotypical morphogenetic cell behaviors, massive cell proliferation and the acquisition of distinct cell identities. Gastrulation provides the blueprint of the body plan of the embryo, as well as generating extra-embryonic cell types of the embryo to make a connection with its mother. Gastrulation involves the specification of mesoderm and definitive endoderm from pluripotent epiblast, concomitant with a highly ordered elongation of tissue along the anterior-posterior (AP) axis. Interestingly, cells with an endoderm identity arise twice during mouse development. Cells with a primitive endoderm identity are specified in the preimplantation blastocyst, and which at gastrulation intercalate with the emergent definitive endoderm to form a mosaic tissue, referred to as the gut endoderm. The gut endoderm gives rise to the gut tube, which will subsequently become patterned along its AP axis into domains possessing unique visceral organ identities, such as thyroid, lung, liver and pancreas. In this way, proper endoderm development is essential for vital organismal functions, including the absorption of nutrients, gas exchange, detoxification and glucose homeostasis.


Assuntos
Embrião de Mamíferos/fisiologia , Endoderma/fisiologia , Trato Gastrointestinal/fisiologia , Gastrulação , Camadas Germinativas/fisiologia , Mesoderma/fisiologia , Morfogênese , Animais , Embrião de Mamíferos/citologia , Endoderma/citologia , Trato Gastrointestinal/citologia , Camadas Germinativas/citologia , Mesoderma/citologia , Camundongos
19.
Curr Protoc Stem Cell Biol ; 52(1): e105, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31971672

RESUMO

The complex program of mouse development entails specification of the embryonic epiblast (Epi) as well as the extra-embryonic trophectoderm (TE) and primitive endoderm (PrE). These three lineages of mouse blastocyst can be modeled in vitro using stem cells derived from primary tissues. In these cultures, cells self-renew while retaining their developmental potential if put back into a developing embryo. Indeed, embryonic stem cells (ESC), when injected into a blastocyst, readily contribute to all embryonic lineages. Similarly, trophoblast stem cells (TSCs) will give rise to all TE-derived trophoblast lineages, and extraembryonic endoderm cells (XEN) will contribute to the PrE-derived yolk sack. These model systems are a powerful tool to study early development, lineage specification, and placenta formation. Only recently reproducible and chemically defined culture systems of these cells have been described. This overview discusses such novel methods for culturing ESC/TSC/XEN, as well as their molecular signatures and developmental potential. Recent strides in expanding the developmental potential of stem cells as well as achieving models more reminiscent of their in vivo counterparts are discussed. Finally, such in vitro stem cells can self-assemble into structures resembling embryos when used in novel 3D-culture systems. This article discusses the strengths and limitations of such "synthetic embryos" in studying developmental processes. © 2020 by John Wiley & Sons, Inc.


Assuntos
Blastocisto/citologia , Técnicas de Cultura de Células/métodos , Células-Tronco Embrionárias/citologia , Modelos Biológicos , Animais , Linhagem da Célula , Desenvolvimento Embrionário , Camundongos , Trofoblastos/citologia
20.
RNA Biol ; 17(1): 150-164, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31566069

RESUMO

A sequencing-based profiling method (RiboMeth-seq) for ribose methylations was used to study methylation patterns in mouse adult tissues and during development. In contrast to previous reports based on studies of human cancer cell lines, almost all methylation sites were close to fully methylated in adult tissues. A subset of sites was differentially modified in developing tissues compared to their adult counterparts and showed clear developmental dynamics. This provides the first evidence for ribosome heterogeneity at the level of rRNA modifications during mouse development. In a prominent example, the expression levels of SNORD78 during development appeared to be regulated by alternative splicing of the Gas5 host-gene and to correlate with the methylation level of its target site at LSU-G4593. The results are discussed in the context of the specialized ribosome hypothesis.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , RNA Ribossômico/genética , RNA Ribossômico/metabolismo , Ribose/metabolismo , Processamento Alternativo , Animais , Biologia Computacional/métodos , Desenvolvimento Embrionário/genética , Perfilação da Expressão Gênica , Íntrons , Metilação , Camundongos , Especificidade de Órgãos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA