Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Cell ; 166(3): 538-554, 2016 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-27471964

RESUMEN

Most complex trait-associated variants are located in non-coding regulatory regions of the genome, where they have been shown to disrupt transcription factor (TF)-DNA binding motifs. Variable TF-DNA interactions are therefore increasingly considered as key drivers of phenotypic variation. However, recent genome-wide studies revealed that the majority of variable TF-DNA binding events are not driven by sequence alterations in the motif of the studied TF. This observation implies that the molecular mechanisms underlying TF-DNA binding variation and, by extrapolation, inter-individual phenotypic variation are more complex than originally anticipated. Here, we summarize the findings that led to this important paradigm shift and review proposed mechanisms for local, proximal, or distal genetic variation-driven variable TF-DNA binding. In addition, we discuss the biomedical implications of these findings for our ability to dissect the molecular role(s) of non-coding genetic variants in complex traits, including disease susceptibility.


Asunto(s)
ADN/metabolismo , Variación Genética , Factores de Transcripción/metabolismo , Animales , Humanos , Unión Proteica/genética
2.
EMBO J ; 41(18): e108206, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35996853

RESUMEN

Adipose stem and precursor cells (ASPCs) give rise to adipocytes and determine the composition and plasticity of adipose tissue. Recently, several studies have demonstrated that ASPCs partition into at least three distinct cell subpopulations, including the enigmatic CD142+ cells. An outstanding challenge is to functionally characterise this population, as discrepant properties, from adipogenic to non- and anti-adipogenic, have been reported for these cells. To resolve these phenotypic ambiguities, we characterised mammalian subcutaneous CD142+ ASPCs across various experimental conditions, demonstrating that CD142+ ASPCs exhibit high molecular and phenotypic robustness. Specifically, we find these cells to be firmly non- and anti-adipogenic both in vitro and in vivo, with their inhibitory signals also impacting adipogenic human cells. However, these CD142+ ASPC-specific properties exhibit surprising temporal phenotypic alterations, and emerge only in an age-dependent manner. Finally, using multi-omic and functional assays, we show that the inhibitory nature of these adipogenesis-regulatory CD142+ ASPCs (Aregs) is driven by specifically expressed secretory factors that cooperate with the retinoic acid signalling pathway to transform the adipogenic state of CD142- ASPCs into a non-adipogenic, Areg-like state.


Asunto(s)
Adipogénesis , Tretinoina , Adipocitos/metabolismo , Tejido Adiposo , Anfirregulina/metabolismo , Animales , Diferenciación Celular , Humanos , Mamíferos , Transducción de Señal , Tretinoina/farmacología
3.
Biomed Microdevices ; 26(2): 18, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38416278

RESUMEN

High-throughput transcriptomics is of increasing fundamental biological and clinical interest. The generation of molecular data from large collections of samples, such as biobanks and drug libraries, is boosting the development of new biomarkers and treatments. Focusing on gene expression, the transcriptomic market exploits the benefits of next-generation sequencing (NGS), leveraging RNA sequencing (RNA-seq) as standard for measuring genome-wide gene expression in biological samples. The cumbersome sample preparation, including RNA extraction, conversion to cDNA and amplification, prevents high-throughput translation of RNA-seq technologies. Bulk RNA barcoding and sequencing (BRB-seq) addresses this limitation by enabling sample preparation in multi-well plate format. Sample multiplexing combined with early pooling into a single tube reduces reagents consumption and manual steps. Enabling simultaneous pooling of all samples from the multi-well plate into one tube, our technology relies on smart labware: a pooling lid comprising fluidic features and small pins to transport the liquid, adapted to standard 96-well plates. Operated with standard fluidic tubes and pump, the system enables over 90% recovery of liquid in a single step in less than a minute. Large scale manufacturing of the lid is demonstrated with the transition from a milled polycarbonate/steel prototype into an injection molded polystyrene lid. The pooling lid demonstrated its value in supporting high-throughput barcode-based sequencing by pooling 96 different DNA barcodes directly from a standard 96-well plate, followed by processing within the single sample pool. This new pooling technology shows great potential to address medium throughput needs in the BRB-seq workflow, thereby addressing the challenge of large-scale and cost-efficient sample preparation for RNA-seq.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , ARN , Heces
4.
PLoS Biol ; 19(3): e3001158, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33780434

RESUMEN

Since its emergence in December 2019, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has spread globally and become a major public health burden. Despite its close phylogenetic relationship to SARS-CoV, SARS-CoV-2 exhibits increased human-to-human transmission dynamics, likely due to efficient early replication in the upper respiratory epithelium of infected individuals. Since different temperatures encountered in the human upper and lower respiratory tract (33°C and 37°C, respectively) have been shown to affect the replication kinetics of several respiratory viruses, as well as host innate immune response dynamics, we investigated the impact of temperature on SARS-CoV-2 and SARS-CoV infection using the primary human airway epithelial cell culture model. SARS-CoV-2, in contrast to SARS-CoV, replicated to higher titers when infections were performed at 33°C rather than 37°C. Although both viruses were highly sensitive to type I and type III interferon pretreatment, a detailed time-resolved transcriptome analysis revealed temperature-dependent interferon and pro-inflammatory responses induced by SARS-CoV-2 that were inversely proportional to its replication efficiency at 33°C or 37°C. These data provide crucial insight on pivotal virus-host interaction dynamics and are in line with characteristic clinical features of SARS-CoV-2 and SARS-CoV, as well as their respective transmission efficiencies.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Regulación Viral de la Expresión Génica/genética , SARS-CoV-2/genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Animales , Antivirales/farmacología , Células Cultivadas , Chlorocebus aethiops , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/virología , Regulación Viral de la Expresión Génica/efectos de los fármacos , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Humanos , Interferones/farmacología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Especificidad de la Especie , Temperatura , Células Vero , Replicación Viral/efectos de los fármacos , Replicación Viral/genética
5.
Nature ; 559(7712): 103-108, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29925944

RESUMEN

Adipocyte development and differentiation have an important role in the aetiology of obesity and its co-morbidities1,2. Although multiple studies have investigated the adipogenic stem and precursor cells that give rise to mature adipocytes3-14, our understanding of their in vivo origin and properties is incomplete2,15,16. This is partially due to the highly heterogeneous and unstructured nature of adipose tissue depots17, which has proven difficult to molecularly dissect using classical approaches such as fluorescence-activated cell sorting and Cre-lox lines based on candidate marker genes16,18. Here, using the resolving power of single-cell transcriptomics19 in a mouse model, we reveal distinct subpopulations of adipose stem and precursor cells in the stromal vascular fraction of subcutaneous adipose tissue. We identify one of these subpopulations as CD142+ adipogenesis-regulatory cells, which can suppress adipocyte formation in vivo and in vitro in a paracrine manner. We show that adipogenesis-regulatory cells are refractory to adipogenesis and that they are functionally conserved in humans. Our findings point to a potentially critical role for adipogenesis-regulatory cells in modulating adipose tissue plasticity, which is linked to metabolic control, differential insulin sensitivity and type 2 diabetes.


Asunto(s)
Adipogénesis , Células del Estroma/citología , Grasa Subcutánea/citología , Adipocitos/citología , Adipocitos/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Resistencia a la Insulina , Masculino , Ratones , Comunicación Paracrina , Análisis de la Célula Individual , Células Madre/citología , Células Madre/metabolismo , Células del Estroma/metabolismo , Grasa Subcutánea/metabolismo , Tromboplastina/metabolismo
6.
Proc Natl Acad Sci U S A ; 117(24): 13828-13838, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32461370

RESUMEN

Despite its popularity, chromatin immunoprecipitation followed by sequencing (ChIP-seq) remains a tedious (>2 d), manually intensive, low-sensitivity and low-throughput approach. Here, we combine principles of microengineering, surface chemistry, and molecular biology to address the major limitations of standard ChIP-seq. The resulting technology, FloChIP, automates and miniaturizes ChIP in a beadless fashion while facilitating the downstream library preparation process through on-chip chromatin tagmentation. FloChIP is fast (<2 h), has a wide dynamic range (from 106 to 500 cells), is scalable and parallelized, and supports antibody- or sample-multiplexed ChIP on both histone marks and transcription factors. In addition, FloChIP's interconnected design allows for straightforward chromatin reimmunoprecipitation, which allows this technology to also act as a microfluidic sequential ChIP-seq system. Finally, we ran FloChIP for the transcription factor MEF2A in 32 distinct human lymphoblastoid cell lines, providing insights into the main factors driving collaborative DNA binding of MEF2A and into its role in B cell-specific gene regulation. Together, our results validate FloChIP as a flexible and reproducible automated solution for individual or sequential ChIP-seq.


Asunto(s)
Automatización/métodos , Secuenciación de Inmunoprecipitación de Cromatina/métodos , Histonas/metabolismo , Factores de Transcripción MEF2/metabolismo , Automatización/instrumentación , Linfocitos B/química , Linfocitos B/metabolismo , Línea Celular Tumoral , Secuenciación de Inmunoprecipitación de Cromatina/instrumentación , Histonas/química , Histonas/genética , Humanos , Factores de Transcripción MEF2/química , Factores de Transcripción MEF2/genética , Unión Proteica
7.
Nat Methods ; 14(3): 316-322, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28092692

RESUMEN

Resolving the DNA-binding specificities of transcription factors (TFs) is of critical value for understanding gene regulation. Here, we present a novel, semiautomated protein-DNA interaction characterization technology, selective microfluidics-based ligand enrichment followed by sequencing (SMiLE-seq). SMiLE-seq is neither limited by DNA bait length nor biased toward strong affinity binders; it probes the DNA-binding properties of TFs over a wide affinity range in a fast and cost-effective fashion. We validated SMiLE-seq by analyzing 58 full-length human, mouse, and Drosophila TFs from distinct structural classes. All tested TFs yielded DNA-binding models with predictive power comparable to or greater than that of other in vitro assays. De novo motif discovery on all JUN-FOS heterodimers and several nuclear receptor-TF complexes provided novel insights into partner-specific heterodimer DNA-binding preferences. We also successfully analyzed the DNA-binding properties of uncharacterized human C2H2 zinc-finger proteins and validated several using ChIP-exo.


Asunto(s)
Dedos de Zinc CYS2-HIS2/fisiología , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Factores de Transcripción/metabolismo , Animales , Sitios de Unión/genética , Biología Computacional , Drosophila/genética , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Ratones , Microfluídica/métodos , Proteínas Proto-Oncogénicas c-fos/genética , Análisis de Secuencia de ADN/métodos
8.
Cell Metab ; 36(7): 1566-1585.e9, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38729152

RESUMEN

Adipose tissue plasticity is orchestrated by molecularly and functionally diverse cells within the stromal vascular fraction (SVF). Although several mouse and human adipose SVF cellular subpopulations have by now been identified, we still lack an understanding of the cellular and functional variability of adipose stem and progenitor cell (ASPC) populations across human fat depots. To address this, we performed single-cell and bulk RNA sequencing (RNA-seq) analyses of >30 SVF/Lin- samples across four human adipose depots, revealing two ubiquitous human ASPC (hASPC) subpopulations with distinct proliferative and adipogenic properties but also depot- and BMI-dependent proportions. Furthermore, we identified an omental-specific, high IGFBP2-expressing stromal population that transitions between mesothelial and mesenchymal cell states and inhibits hASPC adipogenesis through IGFBP2 secretion. Our analyses highlight the molecular and cellular uniqueness of different adipose niches, while our discovery of an anti-adipogenic IGFBP2+ omental-specific population provides a new rationale for the biomedically relevant, limited adipogenic capacity of omental hASPCs.


Asunto(s)
Adipogénesis , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina , Epiplón , Células del Estroma , Humanos , Epiplón/metabolismo , Epiplón/citología , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Células del Estroma/metabolismo , Células del Estroma/citología , Femenino , Masculino , Persona de Mediana Edad , Tejido Adiposo/metabolismo , Tejido Adiposo/citología , Adulto , Epitelio/metabolismo , Células Madre/metabolismo , Células Madre/citología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Anciano , Animales
9.
Curr Res Immunol ; 3: 1-12, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35496820

RESUMEN

CD8 T cells have multiple functional properties that mediate acute phase and long-term immune protection. Several effector and memory CD8 T cell subsets have been described with diverse functionalities and marker profiles. In contrast to the many comprehensive mouse studies, most human studies lack samples from the acute infection phase, a major reason why current knowledge of human T cell subsets and differentiation remains incomplete, particularly with regard to the T cell heterogeneity early during the immune response. Here we analysed the human CD8 T cell response to yellow fever vaccination as the best-known model to study the human immune response to acute viral infection. We performed flow cytometry on 21 markers conventionally used in mice and in humans to describe differentiation, activation, cycling, and so-called effector functions. We found clearly distinct 'acute traits' at the peak of the response that are shared amongst all non-naïve antigen-specific subsets, including memory-differentiated cells. These acute traits were low BCL-2 and high KI67, CD38, HLA-DR, as well as increased Granzyme B and Perforin, previously attributed only to effector cells at the peak of the response. Furthermore, analysis of chromatin accessibility at the single cell level revealed that memory- and effector-differentiated cells clustered together specifically in the acute phase. Altogether, we demonstrate 'acute traits' across differentiation subsets, and point out the need to discriminate the differentiation states when studying human CD8 T cells that undergo an acute response.

10.
Nat Commun ; 13(1): 2042, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35440565

RESUMEN

Non-coding variants coordinate transcription factor (TF) binding and chromatin mark enrichment changes over regions spanning >100 kb. These molecularly coordinated regions are named "variable chromatin modules" (VCMs), providing a conceptual framework of how regulatory variation might shape complex traits. To better understand the molecular mechanisms underlying VCM formation, here, we mechanistically dissect a VCM-modulating noncoding variant that is associated with reduced chronic lymphocytic leukemia (CLL) predisposition and disease progression. This common, germline variant constitutes a 5-bp indel that controls the activity of an AXIN2 gene-linked VCM by creating a MEF2 binding site, which, upon binding, activates a super-enhancer-like regulatory element. This triggers a large change in TF binding activity and chromatin state at an enhancer cluster spanning >150 kb, coinciding with subtle, long-range chromatin compaction and robust AXIN2 up-regulation. Our results support a model in which the indel acts as an AXIN2 VCM-activating TF nucleation event, which modulates CLL pathology.


Asunto(s)
Cromatina , Leucemia Linfocítica Crónica de Células B , Cromatina/genética , Elementos de Facilitación Genéticos/genética , Regulación de la Expresión Génica , Células Germinativas/metabolismo , Humanos , Leucemia Linfocítica Crónica de Células B/genética , Factores de Transcripción/metabolismo
11.
Elife ; 102021 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-33404503

RESUMEN

Skeletal muscles are composed of gigantic cells called muscle fibers, packed with force-producing myofibrils. During development, the size of individual muscle fibers must dramatically enlarge to match with skeletal growth. How muscle growth is coordinated with growth of the contractile apparatus is not understood. Here, we use the large Drosophila flight muscles to mechanistically decipher how muscle fiber growth is controlled. We find that regulated activity of core members of the Hippo pathway is required to support flight muscle growth. Interestingly, we identify Dlg5 and Slmap as regulators of the STRIPAK phosphatase, which negatively regulates Hippo to enable post-mitotic muscle growth. Mechanistically, we show that the Hippo pathway controls timing and levels of sarcomeric gene expression during development and thus regulates the key components that physically mediate muscle growth. Since Dlg5, STRIPAK and the Hippo pathway are conserved a similar mechanism may contribute to muscle or cardiomyocyte growth in humans.


Asunto(s)
Drosophila melanogaster/fisiología , Regulación de la Expresión Génica , Vía de Señalización Hippo/fisiología , Fibras Musculares Esqueléticas/fisiología , Miofibrillas/metabolismo , Sarcómeros/genética , Animales , Drosophila melanogaster/genética
12.
Sci Rep ; 10(1): 18308, 2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-33110158

RESUMEN

Mitochondrial dysfunction was highlighted as a crucial vulnerability factor for the development of depression. However, systemic studies assessing stress-induced changes in mitochondria-associated genes in brain regions relevant to depression symptomatology remain scarce. Here, we performed a genome-wide transcriptomic study to examine mitochondrial gene expression in the prefrontal cortex (PFC) and nucleus accumbens (NAc) of mice exposed to multimodal chronic restraint stress. We identified mitochondria-associated gene pathways as most prominently affected in the PFC and with lesser significance in the NAc. A more detailed mitochondrial gene expression analysis revealed that in particular mitochondrial DNA-encoded subunits of the oxidative phosphorylation complexes were altered in the PFC. The comparison of our data with a reanalyzed transcriptome data set of chronic variable stress mice and major depression disorder subjects showed that the changes in mitochondrial DNA-encoded genes are a feature generalizing to other chronic stress-protocols as well and might have translational relevance. Finally, we provide evidence for changes in mitochondrial outputs in the PFC following chronic stress that are indicative of mitochondrial dysfunction. Collectively, our work reinforces the idea that changes in mitochondrial gene expression are key players in the prefrontal adaptations observed in individuals with high behavioral susceptibility and resilience to chronic stress.


Asunto(s)
Susceptibilidad a Enfermedades/metabolismo , Mitocondrias/metabolismo , Corteza Prefrontal/metabolismo , Estrés Psicológico/metabolismo , Animales , Perfilación de la Expresión Génica , Hormona Liberadora de Gonadotropina/análogos & derivados , Masculino , Ratones Endogámicos C57BL , Mitocondrias/genética , Núcleo Accumbens/metabolismo , Núcleo Accumbens/fisiopatología , Corteza Prefrontal/fisiopatología , Resiliencia Psicológica , Transcriptoma/fisiología
13.
Genome Biol ; 20(1): 71, 2019 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-30999927

RESUMEN

Despite its widespread use, RNA-seq is still too laborious and expensive to replace RT-qPCR as the default gene expression analysis method. We present a novel approach, BRB-seq, which uses early multiplexing to produce 3' cDNA libraries for dozens of samples, requiring just 2 hours of hands-on time. BRB-seq has a comparable performance to the standard TruSeq approach while showing greater tolerance for lower RNA quality and being up to 25 times cheaper. We anticipate that BRB-seq will transform basic laboratory practice given its capacity to generate genome-wide transcriptomic data at a similar cost as profiling four genes using RT-qPCR.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Biblioteca de Genes , Análisis de Secuencia de ARN , Secuenciación de Nucleótidos de Alto Rendimiento
14.
Sci Rep ; 7: 42130, 2017 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-28181539

RESUMEN

Brown adipocytes regulate energy expenditure via mitochondrial uncoupling, which makes them attractive therapeutic targets to tackle obesity. However, the regulatory mechanisms underlying brown adipogenesis are still poorly understood. To address this, we profiled the transcriptome and chromatin state during mouse brown fat cell differentiation, revealing extensive gene expression changes and chromatin remodeling, especially during the first day post-differentiation. To identify putatively causal regulators, we performed transcription factor binding site overrepresentation analyses in active chromatin regions and prioritized factors based on their expression correlation with the bona-fide brown adipogenic marker Ucp1 across multiple mouse and human datasets. Using loss-of-function assays, we evaluated both the phenotypic effect as well as the transcriptomic impact of several putative regulators on the differentiation process, uncovering ZFP467, HOXA4 and Nuclear Factor I A (NFIA) as novel transcriptional regulators. Of these, NFIA emerged as the regulator yielding the strongest molecular and cellular phenotypes. To examine its regulatory function, we profiled the genomic localization of NFIA, identifying it as a key early regulator of terminal brown fat cell differentiation.


Asunto(s)
Adipocitos Marrones/metabolismo , Metabolismo Energético/genética , Factores de Transcripción NFI/genética , Proteína Desacopladora 1/genética , Adipogénesis/genética , Animales , Diferenciación Celular/genética , Ensamble y Desensamble de Cromatina/genética , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica , Genómica , Proteínas de Homeodominio , Humanos , Ratones , Factores de Transcripción , Transcriptoma/genética
15.
Nat Commun ; 7: 11063, 2016 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-27026076

RESUMEN

TAF4 (TATA-binding protein-associated factor 4) and its paralogue TAF4b are components of the TFIID core module. We inactivated the murine Taf4a gene to address Taf4 function during embryogenesis. Here we show that Taf4a(-/-) embryos survive until E9.5 where primary germ layers and many embryonic structures are identified showing Taf4 is dispensable for their specification. In contrast, Taf4 is required for correct patterning of the trunk and anterior structures, ventral morphogenesis and proper heart positioning. Overlapping expression of Taf4a and Taf4b during embryogenesis suggests their redundancy at early stages. In agreement with this, Taf4a(-/-) embryonic stem cells (ESCs) are viable and comprise Taf4b-containing TFIID. Nevertheless, Taf4a(-/-) ESCs do not complete differentiation into glutamatergic neurons and cardiomyocytes in vitro due to impaired preinitiation complex formation at the promoters of critical differentiation genes. We define an essential role of a core TFIID TAF in differentiation events during mammalian embryogenesis.


Asunto(s)
Diferenciación Celular , Desarrollo Embrionario , Células Madre Embrionarias de Ratones/metabolismo , Subunidades de Proteína/metabolismo , Factor de Transcripción TFIID/metabolismo , Animales , Biomarcadores/metabolismo , Tipificación del Cuerpo/efectos de los fármacos , Tipificación del Cuerpo/genética , Anomalías Cardiovasculares/embriología , Anomalías Cardiovasculares/genética , Anomalías Cardiovasculares/patología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Supervivencia Celular/efectos de los fármacos , Pérdida del Embrión/genética , Pérdida del Embrión/patología , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Embrionario/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células Germinativas/efectos de los fármacos , Células Germinativas/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Madre Embrionarias de Ratones/efectos de los fármacos , Mutación , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Cresta Neural/efectos de los fármacos , Cresta Neural/patología , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fenotipo , Embarazo , Subunidades de Proteína/genética , Factor de Transcripción TFIID/deficiencia , Factor de Transcripción TFIID/genética , Tretinoina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA