Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49.814
Filtrar
1.
BMC Bioinformatics ; 25(1): 294, 2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39242990

RESUMEN

Mouse (Mus musculus) models have been heavily utilized in developmental biology research to understand mammalian embryonic development, as mice share many genetic, physiological, and developmental characteristics with humans. New explorations into the integration of temporal (stage-specific) and transcriptional (tissue-specific) data have expanded our knowledge of mouse embryo tissue-specific gene functions. To better understand the substantial impact of synonymous mutational variations in the cell-state-specific transcriptome on a tissue's codon and codon pair usage landscape, we have established a novel resource-Mouse Embryo Codon and Codon Pair Usage Tables (Mouse Embryo CoCoPUTs). This webpage not only offers codon and codon pair usage, but also GC, dinucleotide, and junction dinucleotide usage, encompassing four strains, 15 murine embryonic tissue groups, 18 Theiler stages, and 26 embryonic days. Here, we leverage Mouse Embryo CoCoPUTs and employ the use of heatmaps to depict usage changes over time and a comparison to human usage for each strain and embryonic time point, highlighting unique differences and similarities. The usage similarities found between mouse and human central nervous system data highlight the translation for projects leveraging mouse models. Data for this analysis can be directly retrieved from Mouse Embryo CoCoPUTs. This cutting-edge resource plays a crucial role in deciphering the complex interplay between usage patterns and embryonic development, offering valuable insights into variation across diverse tissues, strains, and stages. Its applications extend across multiple domains, with notable advantages for biotherapeutic development, where optimizing codon usage can enhance protein expression; one can compare strains, tissues, and mouse embryonic stages in one query. Additionally, Mouse Embryo CoCoPUTs holds great potential in the field of tissue-specific genetic engineering, providing insights for tailoring gene expression to specific tissues for targeted interventions. Furthermore, this resource may enhance our understanding of the nuanced connections between usage biases and tissue-specific gene function, contributing to the development of more accurate predictive models for genetic disorders.


Asunto(s)
Transcriptoma , Animales , Ratones , Transcriptoma/genética , Embrión de Mamíferos/metabolismo , Humanos , Desarrollo Embrionario/genética , Uso de Codones/genética
2.
Sci Rep ; 14(1): 20760, 2024 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-39237572

RESUMEN

Embryo quality assessment by optical imaging is increasing in popularity. Among available optical techniques, light sheet microscopy has emerged as a superior alternative to confocal microscopy due to its geometry, enabling faster image acquisition with reduced photodamage to the sample. However, previous assessments of photodamage induced by imaging may have failed to measure more subtle impacts. In this study, we employed DNA damage as a sensitive indicator of photodamage. We use light sheet microscopy with excitation at a wavelength of 405 nm for imaging embryo autofluorescence and compare its performance to laser scanning confocal microscopy. At an equivalent signal-to-noise ratio for images acquired with both modalities, light sheet microscopy reduced image acquisition time by ten-fold, and did not induce DNA damage when compared to non-imaged embryos. In contrast, imaging with confocal microscopy led to significantly higher levels of DNA damage within embryos and had a higher photobleaching rate. Light sheet imaging is also capable of inducing DNA damage within the embryo but requires multiple cycles of volumetric imaging. Collectively, this study confirms that light sheet microscopy is faster and safer than confocal microscopy for imaging live embryos, indicating its potential as a label-free diagnostic for embryo quality.


Asunto(s)
Daño del ADN , Embrión de Mamíferos , Microscopía Confocal , Microscopía Confocal/métodos , Animales , Ratones , Femenino , Imagen Óptica/métodos
3.
Development ; 151(17)2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39221968

RESUMEN

The lymphatic system is formed during embryonic development by the commitment of specialized lymphatic endothelial cells (LECs) and their subsequent assembly in primary lymphatic vessels. Although lymphatic cells are in continuous contact with mesenchymal cells during development and in adult tissues, the role of mesenchymal cells in lymphatic vasculature development remains poorly characterized. Here, we show that a subpopulation of mesenchymal cells expressing the transcription factor Osr1 are in close association with migrating LECs and established lymphatic vessels in mice. Lineage tracing experiments revealed that Osr1+ cells precede LEC arrival during lymphatic vasculature assembly in the back of the embryo. Using Osr1-deficient embryos and functional in vitro assays, we show that Osr1 acts in a non-cell-autonomous manner controlling proliferation and early migration of LECs to peripheral tissues. Thereby, mesenchymal Osr1+ cells control, in a bimodal manner, the production of extracellular matrix scaffold components and signal ligands crucial for lymphatic vessel formation.


Asunto(s)
Células Endoteliales , Linfangiogénesis , Vasos Linfáticos , Factores de Transcripción , Animales , Vasos Linfáticos/embriología , Vasos Linfáticos/metabolismo , Vasos Linfáticos/citología , Ratones , Linfangiogénesis/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Células Endoteliales/metabolismo , Células Endoteliales/citología , Movimiento Celular/genética , Proliferación Celular , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/citología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Mesodermo/metabolismo , Mesodermo/citología , Regulación del Desarrollo de la Expresión Génica , Linaje de la Célula
4.
Sci Adv ; 10(36): eadk2252, 2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39231227

RESUMEN

Primordial germ cells (PGCs) are the precursors of gametes and the sole mechanism by which animals transmit genetic information across generations. In the mouse embryo, the transcriptional and epigenetic regulation of PGC specification has been extensively characterized. However, the initial event that triggers the soma-germline segregation remains poorly understood. Here, we uncover a critical role for the basement membrane in regulating germline entry. We show that PGCs arise in a region of the mouse embryo that lacks contact with the basement membrane, and the addition of exogenous extracellular matrix (ECM) inhibits both PGC and PGC-like cell (PGCLC) specification in mouse embryos and stem cell models, respectively. Mechanistically, we demonstrate that the engagement of ß1 integrin with laminin blocks PGCLC specification by preventing the Wnt signaling-dependent down-regulation of the PGC transcriptional repressor, Otx2. In this way, the physical segregation of cells away from the basement membrane acts as a morphogenetic fate switch that controls the soma-germline bifurcation.


Asunto(s)
Células Germinativas , Células Madre Pluripotentes , Animales , Ratones , Células Germinativas/metabolismo , Células Germinativas/citología , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/citología , Transducción de Señal , Integrinas/metabolismo , Integrinas/genética , Membrana Basal/metabolismo , Vía de Señalización Wnt , Diferenciación Celular , Matriz Extracelular/metabolismo , Laminina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Integrina beta1/metabolismo , Integrina beta1/genética , Factores de Transcripción Otx/metabolismo , Factores de Transcripción Otx/genética , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/citología
6.
Nat Commun ; 15(1): 7860, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39251590

RESUMEN

Pluripotent mouse embryonic stem cells (ESCs) can differentiate to all germ layers and serve as an in vitro model of embryonic development. To better understand the differentiation paths traversed by ESCs committing to different lineages, we track individual differentiating ESCs by timelapse imaging followed by multiplexed high-dimensional Imaging Mass Cytometry (IMC) protein quantification. This links continuous live single-cell molecular NANOG and cellular dynamics quantification over 5-6 generations to protein expression of 37 different molecular regulators in the same single cells at the observation endpoints. Using this unique data set including kinship history and live lineage marker detection, we show that NANOG downregulation occurs generations prior to, but is not sufficient for neuroectoderm marker Sox1 upregulation. We identify a developmental cell type co-expressing both the canonical Sox1 neuroectoderm and FoxA2 endoderm markers in vitro and confirm the presence of such a population in the post-implantation embryo. RNASeq reveals cells co-expressing SOX1 and FOXA2 to have a unique cell state characterized by expression of both endoderm as well as neuroectoderm genes suggesting lineage potential towards both germ layers.


Asunto(s)
Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 3-beta del Hepatocito , Células Madre Embrionarias de Ratones , Factores de Transcripción SOXB1 , Animales , Ratones , Factor Nuclear 3-beta del Hepatocito/metabolismo , Factor Nuclear 3-beta del Hepatocito/genética , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción SOXB1/genética , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Embrionarias de Ratones/citología , Rastreo Celular/métodos , Proteína Homeótica Nanog/metabolismo , Proteína Homeótica Nanog/genética , Linaje de la Célula , Endodermo/metabolismo , Endodermo/citología , Análisis de la Célula Individual/métodos , Desarrollo Embrionario/genética , Placa Neural/metabolismo , Placa Neural/embriología , Placa Neural/citología , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/citología
8.
Sci Rep ; 14(1): 21598, 2024 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-39285243

RESUMEN

Dynamic changes in maternal‒zygotic transition (MZT) require complex regulation of zygote formation, maternal transcript decay, embryonic genome activation (EGA), and cell cycle progression. Although these changes are well described, some key regulatory factors are still elusive. Sirtuin-1 (SIRT1), an NAD+-dependent histone deacetylase, is a versatile driver of MZT via its epigenetic and nonepigenetic substrates. This study focused on the dynamics of SIRT1 in early embryos and its contribution to MZT. A conditional SIRT1-deficient knockout mouse model was used, accompanied by porcine and human embryos. Embryos across mammalian species showed the prominent localization of SIRT1 in the nucleus throughout early embryonic development. Accordingly, SIRT1 interacts with histone H4 on lysine K16 (H4K16) in both mouse and human blastocysts. While maternal SIRT1 is dispensable for MZT, at least one allele of embryonic Sirt1 is required for early embryonic development around the time of EGA. This role of SIRT1 is surprisingly mediated via a transcription-independent mode of action.


Asunto(s)
Desarrollo Embrionario , Ratones Noqueados , Sirtuina 1 , Cigoto , Sirtuina 1/metabolismo , Sirtuina 1/genética , Animales , Cigoto/metabolismo , Humanos , Ratones , Desarrollo Embrionario/genética , Femenino , Histonas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Blastocisto/metabolismo , Porcinos , Embrión de Mamíferos/metabolismo
9.
Proc Natl Acad Sci U S A ; 121(38): e2400781121, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39259589

RESUMEN

During homeostasis, the endoplasmic reticulum (ER) maintains productive transmembrane and secretory protein folding that is vital for proper cellular function. The ER-resident HSP70 chaperone, binding immunoglobulin protein (BiP), plays a pivotal role in sensing ER stress to activate the unfolded protein response (UPR). BiP function is regulated by the bifunctional enzyme filamentation induced by cyclic-AMP domain protein (FicD) that mediates AMPylation and deAMPylation of BiP in response to changes in ER stress. AMPylated BiP acts as a molecular rheostat to regulate UPR signaling, yet little is known about the molecular consequences of FicD loss. In this study, we investigate the role of FicD in mouse embryonic fibroblast (MEF) response to pharmacologically and metabolically induced ER stress. We find differential BiP AMPylation signatures when comparing robust chemical ER stress inducers to physiological glucose starvation stress and recovery. Wildtype MEFs respond to pharmacological ER stress by down-regulating BiP AMPylation. Conversely, BiP AMPylation in wildtype MEFs increases upon metabolic stress induced by glucose starvation. Deletion of FicD results in widespread gene expression changes under baseline growth conditions. In addition, FicD null MEFs exhibit dampened UPR signaling, altered cell stress recovery response, and unconstrained protein secretion. Taken together, our findings indicate that FicD is important for tampering UPR signaling, stress recovery, and the maintenance of secretory protein homeostasis.


Asunto(s)
Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico , Fibroblastos , Glucosa , Respuesta de Proteína Desplegada , Animales , Ratones , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/citología , Retículo Endoplásmico/metabolismo , Chaperón BiP del Retículo Endoplásmico/metabolismo , Fibroblastos/metabolismo , Glucosa/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico/genética , Ratones Noqueados , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/genética , Transducción de Señal
10.
Int J Mol Sci ; 25(16)2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39201474

RESUMEN

Congenital birth defects contribute significantly to preterm birth, stillbirth, perinatal death, infant mortality, and adult disability. As a first step to exploring the mechanisms underlying this major clinical challenge, we analyzed the embryonic phenotypes of lethal strains generated by random mutagenesis. In this study, we report the gross embryonic and perinatal phenotypes of 55 lethal strains randomly picked from a collection of mutants that carry piggyBac (PB) transposon inserts. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses suggested most of the analyzed mutations hit genes involved in heart and nervous development, or in Notch and Wnt signaling. Among them, 12 loci are known to be associated with human diseases. We confirmed 53 strains as embryonic or perinatal lethal, while others were subviable. Gross morphological phenotypes such as body size abnormality (29/55, 52.73%), growth or developmental delay (35/55, 63.64%), brain defects (9/55, 16.36%), vascular/heart development (31/55, 56.36%), and other structural defects (9/55, 16.36%) could be easily observed in the mutants, while three strains showed phenotypes similar to those of human patients. Furthermore, we detected body weight or body composition alterations in the heterozygotes of eight strains. One of them was the TGF-ß signaling gene Smad2. The heterozygotes showed increased energy expenditure and a lower fat-to-body weight ratio compared to wild-type mice. This study provided new insights into mammalian embryonic development and will help understand the pathology of congenital birth defects in humans. In addition, it expanded our understanding of the etiology of obesity.


Asunto(s)
Anomalías Congénitas , Fenotipo , Animales , Ratones , Anomalías Congénitas/genética , Anomalías Congénitas/patología , Femenino , Mutación , Genes Letales , Masculino , Humanos , Estudios de Asociación Genética , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología
11.
Nat Commun ; 15(1): 7390, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39191720

RESUMEN

The success of deep learning in identifying complex patterns exceeding human intuition comes at the cost of interpretability. Non-linear entanglement of image features makes deep learning a "black box" lacking human meaningful explanations for the models' decision. We present DISCOVER, a generative model designed to discover the underlying visual properties driving image-based classification models. DISCOVER learns disentangled latent representations, where each latent feature encodes a unique classification-driving visual property. This design enables "human-in-the-loop" interpretation by generating disentangled exaggerated counterfactual explanations. We apply DISCOVER to interpret classification of in vitro fertilization embryo morphology quality. We quantitatively and systematically confirm the interpretation of known embryo properties, discover properties without previous explicit measurements, and quantitatively determine and empirically verify the classification decision of specific embryo instances. We show that DISCOVER provides human-interpretable understanding of "black box" classification models, proposes hypotheses to decipher underlying biomedical mechanisms, and provides transparency for the classification of individual predictions.


Asunto(s)
Aprendizaje Profundo , Fertilización In Vitro , Humanos , Fertilización In Vitro/métodos , Procesamiento de Imagen Asistido por Computador/métodos , Embrión de Mamíferos , Femenino
12.
Commun Biol ; 7(1): 1057, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39191989

RESUMEN

High quality label-free imaging of oocytes and early embryos is essential for accurate assessment of their developmental potential, a key element of assisted reproduction procedures. To achieve this goal, we propose full-field optical coherence microscopy (FF-OCM), constructed as a compact module fully integrated with a commercial wide-field fluorescence microscope. Our system achieves optical sectioning in wide-field, high in-plane resolution of 0.5 µm, and high sensitivity to backscattered light. To demonstrate its imaging capabilities, we study live mouse oocytes and embryos at all important stages of meiotic maturation and early embryogenesis. Our system enables visualization of intracellular structures, which are not visible in common bright-field microscopy, i.e., internal structure of nuclear apparatus, cytoskeletal filaments, cellular cortex, cytoplasmic protrusions, or zona pellucida features. Additionally, we visualize and quantify intracellular dynamics like cytoplasmic stirring motion, nuclear envelope fluctuations and nucleolus mobility. Altogether, we demonstrate that FF-OCM is a powerful tool for research in developmental biology that also holds great potential for non-invasive time-lapse monitoring of oocyte and embryo quality in assisted reproduction.


Asunto(s)
Oocitos , Animales , Oocitos/citología , Ratones , Femenino , Embrión de Mamíferos/citología , Desarrollo Embrionario , Tomografía de Coherencia Óptica/métodos
13.
Nat Commun ; 15(1): 7310, 2024 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-39181896

RESUMEN

In mammals, global passive demethylation contributes to epigenetic reprogramming during early embryonic development. At this stage, the majority of DNA-methyltransferase 1 (DNMT1) protein is excluded from nucleus, which is considered the primary cause. However, whether the remaining nuclear activity of DNMT1 is regulated by additional mechanisms is unclear. Here, we report that nuclear DNMT1 abundance is finetuned through proteasomal degradation in mouse zygotes. We identify a maternal factor, Pramel15, which targets DNMT1 for degradation via Cullin-RING E3 ligases. Loss of Pramel15 elevates DNMT1 levels in the zygote pronuclei, impairs zygotic DNA demethylation, and causes a stochastic gain of DNA methylation in early embryos. Thus, Pramel15 can modulate the residual level of DNMT1 in the nucleus during zygotic DNA replication, thereby ensuring efficient DNA methylation reprogramming in early embryos.


Asunto(s)
Núcleo Celular , ADN (Citosina-5-)-Metiltransferasa 1 , Desmetilación del ADN , Cigoto , Animales , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/genética , Cigoto/metabolismo , Ratones , Núcleo Celular/metabolismo , Femenino , Metilación de ADN , Proteolisis , Desarrollo Embrionario/genética , Masculino , Embrión de Mamíferos/metabolismo , Ratones Noqueados , Regulación del Desarrollo de la Expresión Génica , Replicación del ADN
14.
Sci Rep ; 14(1): 19757, 2024 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-39187532

RESUMEN

Despite its high cost, the success rate for in vitro fertilization (IVF) remains < 33% in humans, driving the need for new techniques to improve embryo culture outcomes. The well-of-the-well (WOW) culture system is a platform for in-vitro mammalian embryo culture that has been shown to enhance the developmental competence of embryos and clinical pregnancy rates in humans. However, discovery and testing of the best design for optimal embryo culture quality is hindered by the lack of a method to flexibly produce WOW dishes of various designs. Here, we present a low-cost and simple method to fabricate WOW dishes with microwells of arbitrary shapes and dimensions. We use a low-cost 3D printing service to fabricate a poly(dimethylsiloxane) (PDMS)-based WOW insert that can be paired with a standard in vitro fertilization (IVF) dish to create WOW dishes with new microwell shapes, including pyramidal and hemispherical designs. We validate the fabrication quality of the WOW inserts and demonstrate the utility of the assembled WOW dishes for observation and grading of mouse embryo quality. Moreover, our results indicate that WOW dishes with hemispherical microwells result in better culture outcomes than traditional flat-bottomed IVF dishes and those with other microwell shapes, including the semi-elliptical microwells used in commercial WOW dishes. The proposed fabrication strategy thus provides a way to rapidly fabricate and test new WOW dishes that may bolster IVF success rates.


Asunto(s)
Técnicas de Cultivo de Embriones , Fertilización In Vitro , Impresión Tridimensional , Técnicas de Cultivo de Embriones/métodos , Técnicas de Cultivo de Embriones/instrumentación , Animales , Ratones , Fertilización In Vitro/métodos , Femenino , Embrión de Mamíferos , Desarrollo Embrionario , Humanos , Dimetilpolisiloxanos/química
15.
Int J Mol Sci ; 25(15)2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39125691

RESUMEN

Cell immortalization, a hallmark of cancer development, is a process that cells can undergo on their path to carcinogenesis. Spontaneously immortalized mouse embryonic fibroblasts (MEFs) have been used for decades; however, changes in the global transcriptome during this process have been poorly described. In our research, we characterized the poly-A RNA transcriptome changes after spontaneous immortalization. To this end, differentially expressed genes (DEGs) were screened using DESeq2 and characterized by gene ontology enrichment analysis and protein-protein interaction (PPI) network analysis to identify the potential hub genes. In our study, we identified changes in the expression of genes involved in proliferation regulation, cell adhesion, immune response and transcriptional regulation in immortalized MEFs. In addition, we performed a comparative analysis with previously reported MEF immortalization data, where we propose a predicted gene regulatory network model in immortalized MEFs based on the altered expression of Mapk11, Cdh1, Chl1, Zic1, Hoxd10 and the novel hub genes Il6 and Itgb2.


Asunto(s)
Fibroblastos , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Transcriptoma , Animales , Ratones , Fibroblastos/metabolismo , Mapas de Interacción de Proteínas/genética , Embrión de Mamíferos/metabolismo , Ontología de Genes
16.
Int J Mol Sci ; 25(15)2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-39125754

RESUMEN

The Dlk1-Dio3 domain is important for normal embryonic growth and development. The heart is the earliest developing and functioning organ of the embryo. In this study, we constructed a transcriptional termination model by inserting termination sequences and clarified that the lack of long non-coding RNA (lncRNA) expression in the Dlk1-Dio3 domain caused the death of maternal insertion mutant (MKI) and homozygous mutant (HOMO) mice starting from E13.5. Parental insertion mutants (PKI) can be born and grow normally. Macroscopically, dying MKI and HOMO embryos showed phenomena such as embryonic edema and reduced heart rate. Hematoxylin and eosin (H.E.) staining showed thinning of the myocardium in MKI and HOMO embryos. In situ hybridization (IHC) and quantitative reverse-transcription polymerase chain reaction (qRT-PCR) showed downregulation of lncGtl2, Rian, and Mirg expression in MKI and HOMO hearts. The results of single-cell RNA sequencing (scRNA-Seq) analysis indicated that the lack of lncRNA expression in the Dlk1-Dio3 domain led to reduced proliferation of epicardial cells and may be an important cause of cardiac dysplasia. In conclusion, this study demonstrates that Dlk1-Dio3 domain lncRNAs play an integral role in ventricular development.


Asunto(s)
Proteínas de Unión al Calcio , Regulación del Desarrollo de la Expresión Génica , Corazón , Yoduro Peroxidasa , ARN Largo no Codificante , Animales , ARN Largo no Codificante/genética , Ratones , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Corazón/embriología , Corazón/crecimiento & desarrollo , Yoduro Peroxidasa/genética , Yoduro Peroxidasa/metabolismo , Femenino , Desarrollo Embrionario/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proliferación Celular/genética , Embrión de Mamíferos/metabolismo , Proteínas Nucleares
17.
Methods Mol Biol ; 2818: 133-145, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39126471

RESUMEN

Oogenesis is the central process required to produce viable oocytes in female mammals. It is initiated during embryonic development, and it involves the specification of primordial germ cells (PGCs) and progresses through the activation of the meiotic program, reaching a crucial phase in prophase I before pausing at diplotene around the time of birth. The significance of meiosis, particularly the prophase I stage, cannot be overstated, as it plays a pivotal role in ensuring the formation of healthy gametes, a prerequisite for successful reproduction. While research has explored meiosis across various organisms, understanding how environmental factors, including radiation, drugs, endocrine disruptors, reproductive age, or diet, influence this complex developmental process remains incomplete. In this chapter, we describe an ex vivo culture method to investigate meiotic prophase I and beyond and the disruption of oogenesis by external factors. Using this methodology, it is possible to evaluate the effects of individual xenobiotics by administering chemicals at specific points during oogenesis. This culture technique was optimized to study the effects of two selected endocrine disruptors (vinclozolin and MEHP), demonstrating that vinclozolin exposure delayed meiotic differentiation and MEHP exposure reduced follicle size. This approach also opens avenues for future applications, involving the exploration of established or novel pharmaceutical substances and their influence on essential events during prophase I, such as homologous recombination and chromosome segregation. These processes collectively dictate the ultimate fitness of oocytes, with potential implications for factors relevant to the reproductive age and fertility.


Asunto(s)
Meiosis , Ovario , Animales , Femenino , Ratones , Ovario/citología , Meiosis/efectos de los fármacos , Oogénesis/efectos de los fármacos , Oocitos/citología , Oocitos/efectos de los fármacos , Profase Meiótica I/efectos de los fármacos , Disruptores Endocrinos/farmacología , Oxazoles/farmacología , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos
18.
Gene ; 929: 148838, 2024 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-39127412

RESUMEN

Single-tube nested PCR (STnPCR) is a technique that improves nested PCR, reducing potential contamination and false-positive results, enhancing the amplification sensitivity. Despite being commonly used for the detection of microorganisms, STnPCR can be a valuable tool for bovine genotyping, encompassing essential targets as ROSA26 and TSPY, pivotal in the fields of animal reproduction, genetic improvement, and transgenic research. The objective of this study was to improve and innovate STnPCR for gene detection in cattle. We aimed to detect the ROSA26 and TSPY genes using low-concentration DNA samples, including single cells, small cell groups (one to five cells), in vitro-produced embryos, and bovine tissue samples. Moreover, we refined STnPCR for gene detection in up to single cells by conducting sensitivity testing with different concentration ratios of internal and external primers. Successful amplification of the ROSA26 and TSPY genes was achieved across all tested primer concentrations, even in single cells, with more consistent results observed at lower primer concentrations. Additionally, simultaneous gene amplification was achieved through STnPCR multiplexing, representing the first study of multiplex STnPCR in cattle. These outcomes not only confirm its effectiveness in detecting genetic markers for animal genetic improvement and transgenic elements but also pave the way for its widespread adoption in reproductive studies in bovines.


Asunto(s)
Técnicas de Genotipaje , Reacción en Cadena de la Polimerasa , Animales , Bovinos/genética , Reacción en Cadena de la Polimerasa/métodos , Técnicas de Genotipaje/métodos , Embrión de Mamíferos , Análisis de la Célula Individual/métodos , Genotipo
19.
Sci Rep ; 14(1): 19014, 2024 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-39152124

RESUMEN

We previously identified a unique genetic feature of Autism Spectrum Disorder (ASD) in human patients and established mouse models, a low to very low level of six microRNAs, miR-19a-3p, miR-361-5p, miR-3613-3p, miR-150-5p, miR-126-3p and miR-499a-5p. We attempted to interfere experimentally in mice with two of them, miR19a-3p and miR499a-5p by microinjecting into zygote pronuclei either the complementary sequence or an excess of the microRNA. Both resulted in low levels in the tissues and sperm of the targeted microRNAs and their pri and pre precursors. This method stably modify predetermined levels of miRNAs and identify miRNA alterations that cause changes in autistic behavior and predispose the individual to an inherited disease. Excess miRNA results in single-stranded miRNA variations in both free and DNA-bound RNA (R-loop) fractions in mouse models thus appearing to affect their own transcription. Analysis of miRNAs fractions in human patients blood samples confirm low level of six microRNAs also in R-loop fractions.


Asunto(s)
MicroARNs , Fenotipo , Animales , MicroARNs/genética , MicroARNs/metabolismo , Ratones , Humanos , Masculino , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Femenino , Modelos Animales de Enfermedad , Embrión de Mamíferos/metabolismo , Adulto
20.
Mol Cell ; 84(16): 3128-3140.e4, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39096898

RESUMEN

The IscB proteins, as the ancestors of Cas9 endonuclease, hold great promise due to their small size and potential for diverse genome editing. However, their activity in mammalian cells is unsatisfactory. By introducing three residual substitutions in IscB, we observed an average 7.5-fold increase in activity. Through fusing a sequence-non-specific DNA-binding protein domain, the eIscB-D variant achieved higher editing efficiency, with a maximum of 91.3%. Moreover, engineered ωRNA was generated with a 20% reduction in length and slightly increased efficiency. The engineered eIscB-D/eωRNA system showed an average 20.2-fold increase in activity compared with the original IscB. Furthermore, we successfully adapted eIscB-D for highly efficient cytosine and adenine base editing. Notably, eIscB-D is highly active in mouse cell lines and embryos, enabling the efficient generation of disease models through mRNA/ωRNA injection. Our study suggests that these miniature genome-editing tools have great potential for diverse applications.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Animales , Edición Génica/métodos , Ratones , Humanos , Embrión de Mamíferos/metabolismo , Células HEK293 , Ingeniería de Proteínas/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA