Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 487
Filtrar
1.
PLoS Comput Biol ; 17(6): e1009077, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34161317

RESUMEN

The vertebrate hindbrain is segmented into rhombomeres (r) initially defined by distinct domains of gene expression. Previous studies have shown that noise-induced gene regulation and cell sorting are critical for the sharpening of rhombomere boundaries, which start out rough in the forming neural plate (NP) and sharpen over time. However, the mechanisms controlling simultaneous formation of multiple rhombomeres and accuracy in their sizes are unclear. We have developed a stochastic multiscale cell-based model that explicitly incorporates dynamic morphogenetic changes (i.e. convergent-extension of the NP), multiple morphogens, and gene regulatory networks to investigate the formation of rhombomeres and their corresponding boundaries in the zebrafish hindbrain. During pattern initiation, the short-range signal, fibroblast growth factor (FGF), works together with the longer-range morphogen, retinoic acid (RA), to specify all of these boundaries and maintain accurately sized segments with sharp boundaries. At later stages of patterning, we show a nonlinear change in the shape of rhombomeres with rapid left-right narrowing of the NP followed by slower dynamics. Rapid initial convergence improves boundary sharpness and segment size by regulating cell sorting and cell fate both independently and coordinately. Overall, multiple morphogens and tissue dynamics synergize to regulate the sizes and boundaries of multiple segments during development.


Asunto(s)
Tipificación del Cuerpo/fisiología , Modelos Biológicos , Pez Cebra/embriología , Animales , Tipificación del Cuerpo/genética , Biología Computacional , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Regulación del Desarrollo de la Expresión Génica , Sustancias de Crecimiento/fisiología , Rombencéfalo/citología , Rombencéfalo/embriología , Transducción de Señal , Procesos Estocásticos , Tretinoina/fisiología , Pez Cebra/genética
2.
Nat Struct Mol Biol ; 28(6): 521-532, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34045724

RESUMEN

Totipotent cells hold enormous potential for regenerative medicine. Thus, the development of cellular models recapitulating totipotent-like features is of paramount importance. Cells resembling the totipotent cells of early embryos arise spontaneously in mouse embryonic stem (ES) cell cultures. Such '2-cell-like-cells' (2CLCs) recapitulate 2-cell-stage features and display expanded cell potential. Here, we used 2CLCs to perform a small-molecule screen to identify new pathways regulating the 2-cell-stage program. We identified retinoids as robust inducers of 2CLCs and the retinoic acid (RA)-signaling pathway as a key component of the regulatory circuitry of totipotent cells in embryos. Using single-cell RNA-seq, we reveal the transcriptional dynamics of 2CLC reprogramming and show that ES cells undergo distinct cellular trajectories in response to RA. Importantly, endogenous RA activity in early embryos is essential for zygotic genome activation and developmental progression. Overall, our data shed light on the gene regulatory networks controlling cellular plasticity and the totipotency program.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Células Madre Totipotentes/citología , Tretinoina/fisiología , Acitretina/farmacología , Animales , Masa Celular Interna del Blastocisto/citología , Diferenciación Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Femenino , Redes Reguladoras de Genes/genética , Genes Reporteros , Isotretinoína/farmacología , Masculino , Ratones/embriología , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Piperazinas/farmacología , Pirazoles/farmacología , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/farmacología , RNA-Seq , Receptores de Ácido Retinoico/antagonistas & inhibidores , Receptores de Ácido Retinoico/fisiología , Transducción de Señal/efectos de los fármacos , Células Madre Totipotentes/efectos de los fármacos , Transcripción Genética , Tretinoina/antagonistas & inhibidores , Tretinoina/farmacología , Receptor de Ácido Retinoico gamma
3.
Dev Biol ; 473: 119-129, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33607112

RESUMEN

Retinoic acid (RA) signaling is required to restrict heart size through limiting the posterior boundary of the vertebrate cardiac progenitor field within the anterior lateral plate mesoderm (ALPM). However, we still do not fully understand how different cardiac progenitor populations that contribute to the developing heart, including earlier-differentiating first heart field (FHF), later-differentiating second heart field (SHF), and neural crest-derived progenitors, are each affected in RA-deficient embryos. Here, we quantified the number of cardiac progenitors and differentiating cardiomyocytes (CMs) in RA-deficient zebrafish embryos. While Nkx2.5+ cells were increased overall in the nascent hearts of RA-deficient embryos, unexpectedly, we found that the major effect within this population was a significant expansion in the number of differentiating FHF CMs. In contrast to the expansion of the FHF, there was a progressive decrease in SHF progenitors at the arterial pole as the heart tube elongated. Temporal differentiation assays and immunostaining in RA-deficient embryos showed that the outflow tracts (OFTs) of the hearts were significantly smaller, containing fewer differentiated SHF-derived ventricular CMs and a complete absence of SHF-derived smooth muscle at later stages. At the venous pole of the heart, pacemaker cells of the sinoatrial node also failed to differentiate in RA-deficient embryos. Interestingly, genetic lineage tracing showed that the number of neural-crest derived CMs was not altered within the enlarged hearts of RA-deficient zebrafish embryos. Altogether, our data show that the enlarged hearts in RA-deficient zebrafish embryos are comprised of an expansion in earlier differentiating FHF-derived CMs coupled with a progressive depletion of the SHF, suggesting RA signaling determines the relative ratios of earlier- and later-differentiation cardiac progenitors within an expanded cardiac progenitor pool.


Asunto(s)
Corazón/embriología , Mesodermo/metabolismo , Tretinoina/metabolismo , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Regulación del Desarrollo de la Expresión Génica/genética , Corazón/fisiología , Ventrículos Cardíacos/metabolismo , Mesodermo/fisiología , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Organogénesis/genética , Transducción de Señal/genética , Factores de Transcripción/metabolismo , Tretinoina/fisiología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
4.
Dev Dyn ; 250(7): 1036-1050, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33452727

RESUMEN

BACKGROUND: Pharyngeal arches (PA) are sequentially generated in an anterior-to-posterior order. Ripply3 is essential for posterior PA development in mouse embryos and its expression is sequentially activated in ectoderm and endoderm prior to formation of each PA. Since the PA phenotype of Ripply3 knockout (KO) mice is similar to that of retinoic acid (RA) signal-deficient embryos, we investigated the relationship between RA signaling and Ripply3 in mouse embryos. RESULTS: In BMS493 (pan-RAR antagonist) treated embryos, which are defective in third and fourth PA development, Ripply3 expression is decreased in the region posterior to PA2 at E9.0. This expression remains and its distribution is expanded posteriorly at E9.5. Conversely, high dose RA exposure does not apparently change its expression at E9.0 and 9.5. Knockout of retinaldehyde dehydrogenase 2 (Raldh2), which causes more severe PA defect, attenuates sequential Ripply3 expression at PA1 and reduces its expression level. EGFP reporter expression driven by a 6 kb Ripply3 promoter fragment recapitulates the endogenous Ripply3 mRNA expression during PA development in wild-type, but its distribution is expanded posteriorly in BMS493-treated and Raldh2 KO embryos. CONCLUSION: Spatio-temporal regulation of Ripply3 expression by RA signaling is indispensable for the posterior PA development in mouse.


Asunto(s)
Región Branquial/embriología , Proteínas Represoras/genética , Tretinoina/metabolismo , Animales , Benzoatos/farmacología , Región Branquial/efectos de los fármacos , Región Branquial/metabolismo , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Ratones Transgénicos , Morfogénesis/efectos de los fármacos , Morfogénesis/genética , Embarazo , Proteínas Represoras/metabolismo , Receptor alfa de Ácido Retinoico/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Estilbenos/farmacología , Tretinoina/farmacología , Tretinoina/fisiología
5.
PLoS Biol ; 18(11): e3000902, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33201874

RESUMEN

Coordinated development of muscles, tendons, and their attachment sites ensures emergence of functional musculoskeletal units that are adapted to diverse anatomical demands among different species. How these different tissues are patterned and functionally assembled during embryogenesis is poorly understood. Here, we investigated the morphogenesis of extraocular muscles (EOMs), an evolutionary conserved cranial muscle group that is crucial for the coordinated movement of the eyeballs and for visual acuity. By means of lineage analysis, we redefined the cellular origins of periocular connective tissues interacting with the EOMs, which do not arise exclusively from neural crest mesenchyme as previously thought. Using 3D imaging approaches, we established an integrative blueprint for the EOM functional unit. By doing so, we identified a developmental time window in which individual EOMs emerge from a unique muscle anlage and establish insertions in the sclera, which sets these muscles apart from classical muscle-to-bone type of insertions. Further, we demonstrate that the eyeballs are a source of diffusible all-trans retinoic acid (ATRA) that allow their targeting by the EOMs in a temporal and dose-dependent manner. Using genetically modified mice and inhibitor treatments, we find that endogenous local variations in the concentration of retinoids contribute to the establishment of tendon condensations and attachment sites that precede the initiation of muscle patterning. Collectively, our results highlight how global and site-specific programs are deployed for the assembly of muscle functional units with precise definition of muscle shapes and topographical wiring of their tendon attachments.


Asunto(s)
Músculos Oculomotores/embriología , Músculos Oculomotores/crecimiento & desarrollo , Tretinoina/metabolismo , Animales , Tejido Conectivo/fisiología , Desarrollo Embrionario , Ojo , Imagenología Tridimensional/métodos , Ratones/embriología , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Morfogénesis , Transducción de Señal , Tendones/fisiología , Tretinoina/fisiología
6.
Mech Dev ; 164: 103644, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32911082

RESUMEN

The molecular regulators that determine the precise position of the vertebrate limb along the anterio-posterior axis have not been identified. One model suggests that a combination of hox genes in the lateral plate mesoderm (LPM) promotes formation of the limb field, however redundancy among duplicated paralogs has made this model difficult to confirm. In this study, we identify an optimal window during mid-gastrulation stages when transient mis-regulation of retinoic acid signaling or the caudal related transcription factor, Cdx4, both known regulators of hox genes, can alter the position of the pectoral fin field. We show that increased levels of either RA or Cdx4 during mid-gastrulation are sufficient to rostrally shift the position of the pectoral fin field at the expense of surrounding gene expression in the anterior lateral plate mesoderm (aLPM). Alternatively, embryos deficient for both Cdx4 and Cdx1a (Cdx-deficient) form pectoral fins that are shifted towards the posterior and reveal an additional effect on size of the pectoral fin buds. Prior to formation of the pectoral fin buds, the fin field in Cdx-deficient embryos is visibly expanded into the posterior LPM (pLPM) region at the expense of surrounding gene expression. The effects on gene expression immediately post-gastrulation and during somitogenesis support a model where RA and Cdx4 act in parallel to regulate the position of the pectoral fin. Our transient method is a potentially useful model for studying the mechanisms of limb positioning along the AP axis.


Asunto(s)
Aletas de Animales/crecimiento & desarrollo , Gastrulación , Factores de Transcripción/genética , Tretinoina/fisiología , Proteínas de Pez Cebra/genética , Pez Cebra/embriología , Animales , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Genes Homeobox , Mesodermo , Pez Cebra/genética
7.
Br J Cancer ; 122(8): 1231-1241, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32147670

RESUMEN

BACKGROUND: Genome-wide association studies (GWASs) have enriched the fields of genomics and drug development. Adrenocortical carcinoma (ACC) is a rare cancer with a bimodal age distribution and inadequate treatment options. Paediatric ACC is frequently associated with TP53 mutations, with particularly high incidence in Southern Brazil due to the TP53 p.R337H (R337H) germline mutation. The heterogeneous risk among carriers suggests other genetic modifiers could exist. METHODS: We analysed clinical, genotype and gene expression data derived from paediatric ACC, R337H carriers, and adult ACC patients. We restricted our analyses to single nucleotide polymorphisms (SNPs) previously identified in GWASs to associate with disease or human traits. RESULTS: A SNP, rs971074, in the alcohol dehydrogenase 7 gene significantly and reproducibly associated with allelic differences in ACC age-of-onset in both cohorts. Patients homozygous for the minor allele were diagnosed up to 16 years earlier. This SNP resides in a gene involved in the retinoic acid (RA) pathway and patients with differing levels of RA pathway gene expression in their tumours associate with differential ACC progression. CONCLUSIONS: These results identify a novel genetic component to ACC development that resides in the retinoic acid pathway, thereby informing strategies to develop management, preventive and therapeutic treatments for ACC.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/genética , Carcinoma Corticosuprarrenal/genética , Genes p53 , Polimorfismo de Nucleótido Simple , Tretinoina/fisiología , Adolescente , Neoplasias de la Corteza Suprarrenal/epidemiología , Carcinoma Corticosuprarrenal/epidemiología , Factores de Edad , Edad de Inicio , Alcohol Deshidrogenasa/genética , Niño , Preescolar , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Incidencia , Lactante , Masculino
8.
Science ; 367(6482)2020 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-32054698

RESUMEN

Sex determination of germ cells is vital to creating the sexual dichotomy of germ cell development, thereby ensuring sexual reproduction. However, the underlying mechanisms remain unclear. Here, we show that ZGLP1, a conserved transcriptional regulator with GATA-like zinc fingers, determines the oogenic fate in mice. ZGLP1 acts downstream of bone morphogenetic protein, but not retinoic acid (RA), and is essential for the oogenic program and meiotic entry. ZGLP1 overexpression induces differentiation of in vitro primordial germ cell-like cells (PGCLCs) into fetal oocytes by activating the oogenic programs repressed by Polycomb activities, whereas RA signaling contributes to oogenic program maturation and PGC program repression. Our findings elucidate the mechanism for mammalian oogenic fate determination, providing a foundation for promoting in vitro gametogenesis and reproductive medicine.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Oocitos/fisiología , Oogénesis/genética , Proteínas Represoras/fisiología , Diferenciación Sexual/genética , Factores de Transcripción/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Femenino , Feto/citología , Masculino , Meiosis/genética , Ratones , Ratones Noqueados , Oocitos/citología , Proteínas del Grupo Polycomb/metabolismo , Proteínas Represoras/genética , Procesos de Determinación del Sexo , Transducción de Señal , Factores de Transcripción/genética , Transcriptoma , Tretinoina/fisiología
9.
Cell Death Differ ; 27(1): 363-378, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31197235

RESUMEN

To discover novel therapeutic targets for triple-negative breast cancer (TNBC) and cancer stem cells (CSCs), we screened long non-coding RNAs (lncRNAs) most enriched in TNBCs for high expression in CSCs defined by high Aldefluor activity and associated with worse patient outcomes. This led to the identification of non-coding RNA in the aldehyde dehydrogenase 1 A pathway (NRAD1), also known as LINC00284. Targeting NRAD1 in TNBC tumors using antisense oligonucleotides reduced cell survival, tumor growth, and the number of cells with CSC characteristics. Expression of NRAD1 is regulated by an enzyme that causes Aldefluor activity in CSCs, aldehyde dehydrogenase 1A3 (ALDH1A3) and its product retinoic acid. Cellular fractionation revealed that NRAD1 is primarily nuclear localized, which suggested a potential function in gene regulation. This was confirmed by transcriptome profiling and chromatin isolation by RNA purification, followed by sequencing (ChIRP-seq), which demonstrated that NRAD1 has enriched chromatin interactions among the genes it regulates. Gene Ontology enrichment analysis revealed that NRAD1 regulates expression of genes involved in differentiation and catabolic processes. NRAD1 also contributes to gene expression changes induced by ALDH1A3; thereby, the induction of NRAD1 is a novel mechanism through which ALDH1A3 regulates gene expression. Together, these data identify lncRNA NRAD1 as a downstream effector of ALDH1A3, and a target for TNBCs and CSCs, with functions in cell survival and regulation of gene expression.


Asunto(s)
Aldehído Oxidorreductasas/metabolismo , Regulación Neoplásica de la Expresión Génica , Células Madre Neoplásicas/metabolismo , ARN Largo no Codificante/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Animales , Línea Celular Tumoral , Núcleo Celular/genética , Femenino , Humanos , Ratones SCID , ARN Largo no Codificante/antagonistas & inhibidores , Tretinoina/fisiología , Neoplasias de la Mama Triple Negativas/mortalidad , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/terapia
10.
Neurobiol Aging ; 85: 1-10, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31689598

RESUMEN

Dietary micronutrients constitute a major environmental factor influencing aging processes. Vitamin A (vit. A) is the precursor of retinoic acid, a bioactive molecule that controls the expression of several genes involved in brain function. Evidence suggests a reduction of vit. A bioavailability with aging, but its impact on neuronal network is poorly understood. We investigated the mechanisms linking memory impairments with specific alterations of retinoic acid metabolism in the hippocampus. We compared young (10 weeks) and aged (16 months) rats, supplemented or not with dietary vit. A (20 IU retinol/g) for 4 weeks. Our study reveals that aging induced dysregulation of gene expression involved in vit. A and retinoic acid metabolism in the liver. Furthermore, vit. A supplementation restored the integrity of the hippocampal neuronal morphology altered by aging. Importantly, we found a high correlation between hippocampal levels of retinoic acid and memory performance. The present work establishes the link between collapse of retinoid metabolism and age-related cognitive decline, highlighting the role of vit. A in maintaining memory through aging.


Asunto(s)
Envejecimiento , Hipocampo/metabolismo , Trastornos de la Memoria/etiología , Memoria , Tretinoina/metabolismo , Animales , Expresión Génica/efectos de los fármacos , Ratas Wistar , Tretinoina/farmacología , Tretinoina/fisiología
11.
eNeuro ; 7(1)2020.
Artículo en Inglés | MEDLINE | ID: mdl-31879367

RESUMEN

Myelination of the CNS relies on the production and differentiation of oligodendrocyte (OL) precursor cells (OPCs) into mature OLs. During the first month of postnatal life, OPCs that populate the corpus callosum (CC) arise from neural stem cells (NSCs) in the subcallosal subventricular zone (SVZ), and then differentiate to generate myelinating OLs. However, the signals that regulate these processes are not fully understood. In this study, we show that endogenous expression of the retinoic acid (RA)-synthesizing enzyme retinaldehyde dehydrogenase 2 (RALDH2) is required for OPC generation and differentiation in the postnatal subcortical white matter. In male and female pups, conditional deletion of Raldh2 reduced OPC numbers and differentiation. Moreover, decreased OPC numbers coincided with reductions in NSC survival and expression of the sonic hedgehog (SHH) signaling effector protein Gli1 in the SVZ. Additionally, GFAP expression in the CC was decreased, and cortical neuron numbers were altered. Our work suggests a role for endogenous RALDH2-dependent RA synthesis in OPC production and differentiation in the CC, as well as in the development of other cell types derived from NSCs in the embryonic ventricular zone (VZ) and SVZ, as well as the postnatal subcallosal SVZ.


Asunto(s)
Cuerpo Calloso , Células Precursoras de Oligodendrocitos , Tretinoina , Aldehído Oxidorreductasas , Animales , Diferenciación Celular , Femenino , Masculino , Ratones , Oligodendroglía , Tretinoina/fisiología
12.
Nutrients ; 11(12)2019 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-31771174

RESUMEN

Maternal and perinatal undernutrition affects the lung development of litters and it may produce long-lasting alterations in respiratory health. This can be demonstrated using animal models and epidemiological studies. During pregnancy, maternal diet controls lung development by direct and indirect mechanisms. For sure, food intake and caloric restriction directly influence the whole body maturation and the lung. In addition, the maternal food intake during pregnancy controls mother, placenta, and fetal endocrine systems that regulate nutrient uptake and distribution to the fetus and pulmonary tissue development. There are several hormones involved in metabolic regulations, which may play an essential role in lung development during pregnancy. This review focuses on the effect of metabolic hormones in lung development and in how undernutrition alters the hormonal environment during pregnancy to disrupt normal lung maturation. We explore the role of GLP-1, ghrelin, and leptin, and also retinoids and cholecalciferol as hormones synthetized from diet precursors. Finally, we also address how metabolic hormones altered during pregnancy may affect lung pathophysiology in the adulthood.


Asunto(s)
Retardo del Crecimiento Fetal/fisiopatología , Hormonas/fisiología , Pulmón/crecimiento & desarrollo , Pulmón/patología , Desnutrición/fisiopatología , Fenómenos Fisiologicos Nutricionales Maternos , Animales , Colecalciferol/fisiología , Femenino , Desarrollo Fetal , Ghrelina/fisiología , Péptido 1 Similar al Glucagón/fisiología , Humanos , Leptina/fisiología , Embarazo , Retinoides/fisiología , Tretinoina/fisiología
13.
Development ; 146(21)2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31597660

RESUMEN

Retinoic acid (RA) is crucial for mammalian spermatogonia differentiation, and stimulates Stra8 expression, a gene required for meiosis. Certain fish species, including zebrafish, have lost the stra8 gene. While RA still seems important for spermatogenesis in fish, it is not known which stage(s) respond to RA or whether its effects are integrated into the endocrine regulation of spermatogenesis. In zebrafish, RA promoted spermatogonia differentiation, supported androgen-stimulated meiosis, and reduced spermatocyte and spermatid apoptosis. Follicle-stimulating hormone (Fsh) stimulated RA production. Expressing a dominant-negative RA receptor variant in germ cells clearly disturbed spermatogenesis but meiosis and spermiogenesis still took place, although sperm quality was low in 6-month-old adults. This condition also activated Leydig cells. Three months later, spermatogenesis apparently had recovered, but doubling of testis weight demonstrated hypertrophy, apoptosis/DNA damage among spermatids was high and sperm quality remained low. We conclude that RA signaling is important for zebrafish spermatogenesis but is not of crucial relevance. As Fsh stimulates androgen and RA production, germ cell-mediated, RA-dependent reduction of Leydig cell activity may form a hitherto unknown intratesticular negative-feedback loop.


Asunto(s)
Andrógenos/fisiología , Sistema Endocrino/fisiología , Hormona Folículo Estimulante/fisiología , Transducción de Señal , Espermatogénesis , Tretinoina/fisiología , Animales , Busulfano/química , Diferenciación Celular/genética , Retroalimentación Fisiológica , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Retinoides/fisiología , Espermátides/fisiología , Espermatocitos/fisiología , Espermatogonias/fisiología , Testículo/fisiología , Transgenes , Pez Cebra
14.
Development ; 146(13)2019 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-31273085

RESUMEN

Retinoic acid (RA), a metabolite of retinol (vitamin A), functions as a ligand for nuclear RA receptors (RARs) that regulate development of chordate animals. RA-RARs can activate or repress transcription of key developmental genes. Genetic studies in mouse and zebrafish embryos that are deficient in RA-generating enzymes or RARs have been instrumental in identifying RA functions, revealing that RA signaling regulates development of many organs and tissues, including the body axis, spinal cord, forelimbs, heart, eye and reproductive tract. An understanding of the normal functions of RA signaling during development will guide efforts for use of RA as a therapeutic agent to improve human health. Here, we provide an overview of RA signaling and highlight its key functions during development.


Asunto(s)
Genes del Desarrollo , Receptores de Ácido Retinoico/fisiología , Tretinoina/farmacología , Tretinoina/fisiología , Animales , Embrión de Mamíferos , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Genes del Desarrollo/efectos de los fármacos , Genes del Desarrollo/genética , Humanos , Ratones , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Tretinoina/metabolismo , Pez Cebra
15.
Proc Biol Sci ; 286(1904): 20190401, 2019 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-31185860

RESUMEN

The diversity of teeth patterns in actinopterygians is impressive with tooth rows in many locations in the oral and pharyngeal regions. The first-formed tooth has been hypothesized to serve as an initiator controlling the formation of the subsequent teeth. In zebrafish, the existence of the first tooth (named 4 V1) is puzzling as its replacement is induced before the opening of the mouth. Functionally, it has been shown that 4 V1 formation requires fibroblast growth factor (FGF) and retinoic acid (RA) signalling. Here, we show that the ablation of 4 V1 prevents the development of the dental row demonstrating its dependency over it. If endogenous levels of FGF and RA are restored after 4 V1 ablation, embryonic dentition starts again by de novo formation of a first tooth, followed by the dental row. Similarly, induction of anterior ectopic teeth induces subsequent tooth formation, demonstrating that the initiator tooth is necessary and sufficient for dental row formation, probably via FGF ligands released by 4 V1 to induce the formation of subsequent teeth. Our results show that by modifying the formation of the initiator tooth it is possible to control the formation of a dental row. This could help to explain the diversity of tooth patterns observed in actinopterygians and more broadly, how diverse traits evolved through molecular fine-tuning.


Asunto(s)
Dentición , Pez Cebra/crecimiento & desarrollo , Animales , Tipificación del Cuerpo , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/farmacología , Factores de Crecimiento de Fibroblastos/fisiología , Transducción de Señal , Diente/anatomía & histología , Diente/efectos de los fármacos , Diente/crecimiento & desarrollo , Tretinoina/metabolismo , Tretinoina/farmacología , Tretinoina/fisiología , Pez Cebra/anatomía & histología , Pez Cebra/embriología
16.
Exp Dermatol ; 28(8): 976-980, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31125456

RESUMEN

Carbonic anhydrases (CAs) are ubiquitously expressed enzymes and catalyse an important physiological reaction of interconverting the hydration of carbon dioxide to bicarbonates, which is crucial for maintaining acid/base equilibrium in certain tissues. While 15 different isoforms of CAs are present in various cell types in human tissues, their expression pattern in the epidermis remains to be investigated. Here, we report the expression of 5 CA isoforms (CAII, CAIX, CAXI, CAXII and CAXIII) in human primary keratinocytes. Further, we demonstrate that the expression of CAII and CAIX in these cells is significantly up-regulated by the biologically active metabolites of vitamin A (all-trans retinoic acid) and vitamin D (1α,25-dihydroxyvitamin D3 ), respectively. Taken together, apart from providing new information on the expression of CAs in the skin, our results highlight a previously undisclosed connection between vitamin A and CAII expression and vitamin D and CAIX expression.


Asunto(s)
Calcitriol/fisiología , Anhidrasas Carbónicas/metabolismo , Queratinocitos/enzimología , Tretinoina/fisiología , Línea Celular , Humanos
17.
Reprod Toxicol ; 86: 56-61, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30940504

RESUMEN

In female mice, the Müllerian duct develops into the oviduct, uterus and vagina. The fate of epithelia is determined by factors secreted from the mesenchyme. Retinoic acid (RA) and its receptors are present in the mesenchyme of cranial Müllerian duct. RA induces Müllerian duct to uterine epithelial differentiation whereas inhibition of RA receptors induces vaginal epithelial differentiation. Thus, RA signaling in the Müllerian duct is required to promote differentiation of the mesenchyme into the future uterus. Perinatal estrogen exposure induces various abnormalities in Müllerian duct-derived organs. These include a cranial shift of the border among oviduct, uterus and vagina as well as precancerous lesions suppressed by co-treatment with RA and estrogen. Since RA synthesis enzymes and receptors are expressed both in the epithelium and stroma after birth, RA signaling may act in the epithelia to maintain adult epithelial homeostasis and to prevent irreversible lesions induced by perinatal estrogen exposure.


Asunto(s)
Conductos Paramesonéfricos/crecimiento & desarrollo , Tretinoina/fisiología , Útero/crecimiento & desarrollo , Animales , Estrógenos/metabolismo , Femenino , Humanos , Transducción de Señal
18.
Cell Rep ; 26(13): 3522-3536.e5, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30917309

RESUMEN

In humans, many cases of congenital insensitivity to pain (CIP) are caused by mutations of components of the NGF/TrkA signaling pathway, which is required for survival and specification of nociceptors and plays a major role in pain processing. Mutations in PRDM12 have been identified in CIP patients that indicate a putative role for this transcriptional regulator in pain sensing. Here, we show that Prdm12 expression is restricted to developing and adult nociceptors and that its genetic ablation compromises their viability and maturation. Mechanistically, we find that Prdm12 is required for the initiation and maintenance of the expression of TrkA by acting as a modulator of Neurogenin1/2 transcription factor activity, in frogs, mice, and humans. Altogether, our results identify Prdm12 as an evolutionarily conserved key regulator of nociceptor specification and as an actionable target for new pain therapeutics.


Asunto(s)
Proteínas Portadoras/fisiología , Proteínas del Tejido Nervioso/fisiología , Neurogénesis/fisiología , Nociceptores/citología , Animales , Apoptosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas Portadoras/genética , Línea Celular , Evolución Molecular , Femenino , Ganglios Sensoriales/citología , Técnicas de Inactivación de Genes , Células Madre Embrionarias Humanas , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Cresta Neural/citología , Nociceptores/metabolismo , Receptor trkA/metabolismo , Tretinoina/fisiología , Xenopus laevis
19.
Development ; 146(6)2019 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-30824552

RESUMEN

Neonatal germ cell development provides the foundation of spermatogenesis. However, a systematic understanding of this process is still limited. To resolve cellular and molecular heterogeneity in this process, we profiled single cell transcriptomes of undifferentiated germ cells from neonatal mouse testes and employed unbiased clustering and pseudotime ordering analysis to assign cells to distinct cell states in the developmental continuum. We defined the unique transcriptional programs underlying migratory capacity, resting cellular states and apoptosis regulation in transitional gonocytes. We also identified a subpopulation of primitive spermatogonia marked by CD87 (plasminogen activator, urokinase receptor), which exhibited a higher level of self-renewal gene expression and migration potential. We further revealed a differentiation-primed state within the undifferentiated compartment, in which elevated Oct4 expression correlates with lower expression of self-renewal pathway factors, higher Rarg expression, and enhanced retinoic acid responsiveness. Lastly, a knockdown experiment revealed the role of Oct4 in the regulation of gene expression related to the MAPK pathway and cell adhesion, which may contribute to stem cell differentiation. Our study thus provides novel insights into cellular and molecular regulation during early germ cell development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Análisis de Secuencia de ARN , Espermatogonias/citología , Animales , Animales Recién Nacidos , Apoptosis , Adhesión Celular , Diferenciación Celular , Perfilación de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Microscopía Fluorescente , Factor 3 de Transcripción de Unión a Octámeros/fisiología , Receptores de Ácido Retinoico/fisiología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/fisiología , Espermatogénesis/genética , Transcriptoma , Tretinoina/fisiología , Receptor de Ácido Retinoico gamma
20.
BMC Ophthalmol ; 19(1): 23, 2019 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-30658598

RESUMEN

BACKGROUND: By investigating that (i) all-trans retinoic acid (ATRA) affects human retinal pigment epithelium (RPE) in expressing and secreting transforming growth factor (TGF)-ß2 and (ii) U73122 (phospholipase C inhibitor) and SQ22536 (adenylyl cyclase inhibitor) regulate the ATRA-induced secretion of TGF-ß2 in human RPE, we sought to interpret the signaling pathway of ATRA in promoting the development of myopia. METHODS: The RPE cell line (D407) was treated with (i) ATRA (10 µM), (ii) U73122 (5-40 µM) and ATRA (10 µM), or (iii) SQ22536 (5-40 µM) and ATRA (10 µM). The control group was no-treated. After stimulated at 2, 4, 8, 16, 24, and 48 h, The expression and secretion of TGF-ß2 was detected. RESULTS: TGF-ß2 in the cytoplasm was time-dependent increased by ATRA (p < 0.001). A time-dependent increase in the TGF-ß2 protein of the supernatant was induced by ATRA (p < 0.001). U73122 (in the range of 5 to 40 µM) could suppress the secretion of TGF-ß2 induced by ATRA (p < 0.001), and 40 µM U73122 could completely inhibit the up-regulated effect of 10 µM ATRA. However, SQ22536 (in the range of 5 to 40 µM) had no impact on the secretion of TGF-ß2 induced by ATRA (p > 0.05). CONCLUSIONS: In RPE cells, ATRA stimulates the secretion of TGF-ß2 via the phospholipase C signaling pathway but not the adenylyl cyclase signaling pathway. U73122 may inhibit the promotion of ATRA in the development of myopia.


Asunto(s)
Adenilil Ciclasas/fisiología , Miopía/fisiopatología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Factor de Crecimiento Transformador beta2/metabolismo , Tretinoina/fisiología , Fosfolipasas de Tipo C/fisiología , Células Cultivadas , Citoplasma/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Epitelio Pigmentado de la Retina/metabolismo , Transducción de Señal/fisiología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...