Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
bioRxiv ; 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38370781

RESUMEN

Motor neurons (MNs) are the final output of circuits driving fundamental behaviors, such as respiration and locomotion. Hox proteins are essential in generating the MN diversity required for accomplishing these functions, but the transcriptional mechanisms that enable Hox paralogs to assign distinct MN subtype identities despite their promiscuous DNA binding motif are not well understood. Here we show that Hoxa5 controls chromatin accessibility in all mouse spinal cervical MN subtypes and engages TALE co-factors to directly bind and regulate subtype-specific genes. We identify a paralog-specific interaction of Hoxa5 with the phrenic MN-specific transcription factor Scip and show that heterologous expression of Hoxa5 and Scip is sufficient to suppress limb-innervating MN identity. We also demonstrate that phrenic MN identity is stable after Hoxa5 downregulation and identify Klf proteins as potential regulators of phrenic MN maintenance. Our data identify multiple modes of Hoxa5 action that converge to induce and maintain MN identity.

3.
Front Cell Dev Biol ; 10: 806545, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35557949

RESUMEN

The skeletal system derives from multiple embryonic sources whose derivatives must develop in coordination to produce an integrated whole. In particular, interactions across the lateral somitic frontier, where derivatives of the somites and lateral plate mesoderm come into contact, are important for proper development. Many questions remain about genetic control of this coordination, and embryological information is incomplete for some structures that incorporate the frontier, including the sternum. Hox genes act in both tissues as regulators of skeletal pattern. Here, we used conditional deletion to characterize the tissue-specific contributions of Hoxa5 to skeletal patterning. We found that most aspects of the Hoxa5 skeletal phenotype are attributable to its activity in one or the other tissue, indicating largely additive roles. However, multiple roles are identified at the junction of the T1 ribs and the anterior portion of the sternum, or presternum. The embryology of the presternum has not been well described in mouse. We present a model for presternum development, and show that it arises from multiple, paired LPM-derived primordia. We show evidence that HOXA5 expression marks the embryonic precursor of a recently identified lateral presternum structure that is variably present in therians.

4.
Front Cell Dev Biol ; 9: 632303, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33732701

RESUMEN

Brown adipose tissue (BAT) plays critical thermogenic, metabolic and endocrine roles in mammals, and aberrant BAT function is associated with metabolic disorders including obesity and diabetes. The major BAT depots are clustered at the neck and forelimb levels, and arise largely within the dermomyotome of somites, from a common progenitor with skeletal muscle. However, many aspects of BAT embryonic development are not well understood. Hoxa5 patterns other tissues at the cervical and brachial levels, including skeletal, neural and respiratory structures. Here, we show that Hoxa5 also positively regulates BAT development, while negatively regulating formation of epaxial skeletal muscle. HOXA5 protein is expressed in embryonic preadipocytes and adipocytes as early as embryonic day 12.5. Hoxa5 null mutant embryos and rare, surviving adults show subtly reduced iBAT and sBAT formation, as well as aberrant marker expression, lower adipocyte density and altered lipid droplet morphology. Conversely, the epaxial muscles that arise from a common dermomyotome progenitor are expanded in Hoxa5 mutants. Conditional deletion of Hoxa5 with Myf5/Cre can reproduce both BAT and epaxial muscle phenotypes, indicating that HOXA5 is necessary within Myf5-positive cells for proper BAT and epaxial muscle development. However, recombinase-based lineage tracing shows that Hoxa5 does not act cell-autonomously to repress skeletal muscle fate. Interestingly, Hoxa5-dependent regulation of adipose-associated transcripts is conserved in lung and diaphragm, suggesting a shared molecular role for Hoxa5 in multiple tissues. Together, these findings establish a role for Hoxa5 in embryonic BAT development.

5.
Dis Model Mech ; 13(12)2020 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-33158935

RESUMEN

Pleuropulmonary blastoma (PPB) is a very rare pediatric lung disease. It can progress from abnormal epithelial cysts to an aggressive sarcoma with poor survival. PPB is difficult to diagnose as it can be confounded with other cystic lung disorders, such as congenital pulmonary airway malformation (CPAM). PPB is associated with mutations in DICER1 that perturb the microRNA (miRNA) profile in lung. How DICER1 and miRNAs act during PPB pathogenesis remains unsolved. Lung epithelial deletion of the Yin Yang1 (Yy1) gene in mice causes a phenotype mimicking the cystic form of PPB and affects the expression of key regulators of lung development. Similar changes in expression were observed in PPB but not in CPAM lung biopsies, revealing a distinctive PPB molecular signature. Deregulation of molecules promoting epithelial-mesenchymal transition (EMT) was detected in PPB specimens, suggesting that EMT might participate in tumor progression. Changes in miRNA expression also occurred in PPB lung biopsies. miR-125a-3p, a candidate to regulate YY1 expression and lung branching, was abnormally highly expressed in PPB samples. Together, these findings support the concept that reduced expression of YY1, due to the abnormal miRNA profile resulting from DICER1 mutations, contributes to PPB development via its impact on the expression of key lung developmental genes.This article has an associated First Person interview with the joint first authors of the paper.


Asunto(s)
Epitelio/patología , Eliminación de Gen , Pulmón/patología , Blastoma Pulmonar/genética , Blastoma Pulmonar/patología , Factor de Transcripción YY1/genética , Células A549 , Animales , Secuencia de Bases , Biomarcadores/metabolismo , Biopsia , Cadherinas/metabolismo , Progresión de la Enfermedad , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Factor 9 de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Modelos Biológicos , Isoformas de Proteínas/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Factor de Transcripción YY1/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
6.
Elife ; 92020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31944180

RESUMEN

The precise pattern of motor neuron (MN) activation is essential for the execution of motor actions; however, the molecular mechanisms that give rise to specific patterns of MN activity are largely unknown. Phrenic MNs integrate multiple inputs to mediate inspiratory activity during breathing and are constrained to fire in a pattern that drives efficient diaphragm contraction. We show that Hox5 transcription factors shape phrenic MN output by connecting phrenic MNs to inhibitory premotor neurons. Hox5 genes establish phrenic MN organization and dendritic topography through the regulation of phrenic-specific cell adhesion programs. In the absence of Hox5 genes, phrenic MN firing becomes asynchronous and erratic due to loss of phrenic MN inhibition. Strikingly, mice lacking Hox5 genes in MNs exhibit abnormal respiratory behavior throughout their lifetime. Our findings support a model where MN-intrinsic transcriptional programs shape the pattern of motor output by orchestrating distinct aspects of MN connectivity.


In mammals, air is moved in and out of the lungs by a sheet of muscle called the diaphragm. When this muscle contracts air gets drawn into the lungs and as the muscle relaxes this pushes air back out. Movement of the diaphragm is controlled by a group of nerve cells called motor neurons which are part of the phrenic motor column (or PMC for short) that sits within the spinal cord. The neurons within this column work together with nerve cells in the brain to coordinate the speed and duration of each breath. For the lungs to develop normally, the neurons that control how the diaphragm contracts need to start working before birth. During development, motor neurons in the PMC cluster together and connect with other nerve cells involved in breathing. But, despite their essential role, it is not yet clear how neurons in the PMC develop and join up with other nerve cells. Now, Vagnozzi et al. show that a set of genes which make the transcription factor Hox5 control the position and organization of motor neurons in the PMC. Transcription factors work as genetic switches, turning sets of genes on and off. Vagnozzi et al. showed that removing the Hox5 transcription factors from motor neurons in the PMC changed their activity and disordered their connections with other breathing-related nerve cells. Hox5 transcription factors regulate the production of proteins called cadherins which join together neighboring cells. Therefore, motor neurons lacking Hox5 were unable to make enough cadherins to securely stick together and connect with other nerve cells. Further experiments showed that removing the genes that code for Hox5 caused mice to have breathing difficulties in the first two weeks after birth. Although half of these mutant mice were eventually able to breathe normally, the other half died within a week. These breathing defects are reminiscent of the symptoms observed in sudden infant death syndrome (also known as SIDS). Abnormalities in breathing occur in many other diseases, including sleep apnea, muscular dystrophy and amyotrophic lateral sclerosis (ALS). A better understanding of how the connections between nerve cells involved in breathing are formed, and the role of Hox5 and cadherins, could lead to improved treatment options for these diseases.


Asunto(s)
Genes Homeobox , Neuronas Motoras/fisiología , Nervio Frénico/fisiología , Respiración/genética , Transcripción Genética , Animales , Ratones
8.
Mamm Genome ; 29(3-4): 281-298, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29536159

RESUMEN

The human chromosomal region 17q12-q21 is one of the best replicated genome-wide association study loci for childhood asthma. The associated SNPs span a large genomic interval that includes several protein-coding genes. Here, we tested the hypothesis that the zona pellucida-binding protein 2 (ZPBP2) gene residing in this region contributes to asthma pathogenesis using a mouse model. We tested the lung phenotypes of knock-out (KO) mice that carry a deletion of the Zpbp2 gene. The deletion attenuated airway hypersensitivity (AHR) in female, but not male, mice in the absence of allergic sensitization. Analysis of the lipid profiles of their lungs showed that female, but not male, KO mice had significantly lower levels of sphingosine-1-phosphate (S1P), very long-chain ceramides (VLCCs), and higher levels of long-chain ceramides compared to wild-type controls. Furthermore, in females, lung resistance following methacholine challenge correlated with lung S1P levels (Pearson correlation coefficient 0.57) suggesting a link between reduced AHR in KO females, Zpbp2 deletion, and S1P level regulation. In livers, spleens and blood plasma, however, VLCC, S1P, and sphingosine levels were reduced in both KO females and males. We also find that the Zpbp2 deletion was associated with gain of methylation in the adjacent DNA regions. Thus, we demonstrate that the mouse ortholog of ZPBP2 has a role in controlling AHR in female mice. Our data also suggest that Zpbp2 may act through regulation of ceramide metabolism. These findings highlight the importance of phospholipid metabolism for sexual dimorphism in AHR.


Asunto(s)
Metabolismo de los Lípidos , Pulmón/metabolismo , Proteínas de la Membrana/genética , Hipersensibilidad Respiratoria/genética , Hipersensibilidad Respiratoria/patología , Caracteres Sexuales , Animales , Asma/genética , Asma/patología , Metilación de ADN/genética , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Regulación de la Expresión Génica , Inmunoglobulina E/metabolismo , Hígado/metabolismo , Hígado/patología , Pulmón/patología , Sistema de Señalización de MAP Quinasas , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Cloruro de Metacolina , Ratones Endogámicos C57BL , Ratones Noqueados , Especificidad de Órganos , Fenotipo , Regiones Promotoras Genéticas , Esfingolípidos/metabolismo , Transcriptoma/genética
9.
Int J Dev Biol ; 62(11-12): 653-657, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30604834

RESUMEN

Forty years ago, Ed Lewis established for the first time the organization of homeotic genes along the chromosome and its importance in embryo patterning. To celebrate this seminal discovery, the International Journal of Developmental Biology decided to launch a Special Issue. It is with honor, pleasure, but also humility that we accepted the challenge of acting as guest editors for this Special Issue. We entitled the issue Hox genes: past, present and future of master regulator genes since despite four decades of amazing discoveries, numerous questions remain unanswered, which open new avenues of research. This is well-acknowledged by Robb Krumlauf and Jacqueline Deschamps in the Introductory articles. The high-level reviews and original research reports collected in this Special Issue also reflect the wide-range and important topics that are still in the spotlights including the origins of Hox genes, the regulatory events controlling their expression, the mechanisms driving the action of HOX proteins, and their multiple roles in normal development and pathogenesis.


Asunto(s)
Tipificación del Cuerpo/genética , Regulación del Desarrollo de la Expresión Génica , Genes Homeobox , Animales , Biología Evolutiva
10.
Int J Dev Biol ; 62(11-12): 665-671, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30604836

RESUMEN

For this Special Issue of The International Journal of Develomental Biology on Hox genes, the guest editors met Jacqueline Deschamps for an interview about her research career dedicated to understanding how Hox gene expression is initiated, maintained and functionally utilized in the mouse embryo. We describe here her journey through some of the main discoveries which led to our current knowledge about how Hox genes contribute to shaping the animal body plan. This journey was a human adventure also, of more than 30 years, in the light of which Jacqueline Deschamps delivers here messages to the younger generations of scientists.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Genes Homeobox , Genética , Investigación , Animales , Tipificación del Cuerpo/genética
11.
Int J Dev Biol ; 62(11-12): 785-796, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30604848

RESUMEN

HOX proteins act during development to regulate musculoskeletal morphology. HOXA5 patterns skeletal structures surrounding the cervical-thoracic transition including the vertebrae, ribs, sternum and forelimb girdle. However, the tissue types in which it acts to pattern the skeleton, and the ultimate fates of embryonic cells that activate Hoxa5 expression are unknown. A detailed characterization of HOXA5 expression by immunofluorescence was combined with Cre/LoxP genetic lineage tracing to map the fate of Hoxa5 expressing cells in axial musculoskeletal tissues and in their precursors, the somites and lateral plate mesoderm. HOXA5 protein expression is dynamic and spatially restricted in derivatives of both the lateral plate mesoderm and somites, including a subset of the lateral sclerotome, suggesting a local role in regulating early skeletal patterning. HOXA5 expression persists from somite stages through late development in differentiating skeletal and connective tissues, pointing to a continuous and direct role in skeletal patterning. In contrast, HOXA5 expression is excluded from the skeletal muscle and muscle satellite cell lineages. Furthermore, the descendants of Hoxa5-expressing cells, even after HOXA5 expression has extinguished, never contribute to these lineages. Together, these findings suggest cell autonomous roles for HOXA5 in skeletal development, as well as non-cell autonomous functions in muscle through expression in surrounding connective tissues. They also support the notion that different Hox genes display diverse tissue specificities and locations to achieve their patterning activity.


Asunto(s)
Tipificación del Cuerpo/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Sistema Musculoesquelético/embriología , Fosfoproteínas/metabolismo , Animales , Proteínas de Homeodominio/genética , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Ratones Transgénicos , Sistema Musculoesquelético/metabolismo , Organogénesis/genética , Fosfoproteínas/genética , Somitos/embriología , Somitos/metabolismo , Factores de Transcripción
12.
Int J Dev Biol ; 62(11-12): 857-864, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30604855

RESUMEN

The HOX genes are transcription factors that are expressed in coordinated spatiotemporal patterns to ensure normal development. Ectopic expression may instead lead to the development and progression of tumors. Genetic polymorphisms in the regions of four HOX gene clusters were tested for association with lung cancer in 420 cases and 3,151 controls. The effect of these variants on lung gene expression (expression quantitative trait loci, eQTL) was tested in a discovery set of 409 non-tumor lung samples and validated in two lung eQTL replication sets (n = 287 and 342). The expression levels of HOXB2 were evaluated at the mRNA and protein levels by quantitative real-time PCR and immunohistochemistry in paired tumor and non-tumor lung tissue samples. The most significant SNP associated with lung cancer in the HOXB cluster was rs10853100 located upstream of the HOXB cluster. HOXB2 was the top eQTL-regulated gene with several polymorphisms associated with its mRNA expression levels in lung tissue. This includes the lung cancer SNP rs10853100 that was significantly associated with HOXB2 expression (P=3.39E-7). In the lung eQTL discovery and replication sets, the lung cancer risk allele (T) for rs10853100 was associated with lower HOXB2 expression levels. In paired normal-tumor samples, HOXB2 mRNA and protein levels were significantly reduced in tumors when compared to non-tumor lung tissues. Genetic variants in the HOXB cluster may confer susceptibility to lung cancer by modulating the expression of HOXB2 in the lung.


Asunto(s)
Predisposición Genética a la Enfermedad , Proteínas de Homeodominio/genética , Neoplasias Pulmonares/genética , Pulmón/metabolismo , Polimorfismo de Nucleótido Simple , Factores de Transcripción/genética , Anciano , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Homeobox , Proteínas de Homeodominio/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Persona de Mediana Edad , Sitios de Carácter Cuantitativo , Factores de Transcripción/metabolismo
13.
Front Mol Neurosci ; 10: 369, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29187810

RESUMEN

Hoxa5 is a member of the Hox gene family that plays critical roles in successive steps of the central nervous system formation during embryonic and fetal development. In the mouse, Hoxa5 was recently shown to be expressed in the medulla oblongata and the pons from fetal stages to adulthood. In these territories, Hoxa5 transcripts are enriched in many precerebellar neurons and several nuclei involved in autonomic functions, while the HOXA5 protein is detected mainly in glutamatergic and GABAergic neurons. However, whether HOXA5 is functionally required in these neurons after birth remains unknown. As a first approach to tackle this question, we aimed at determining the molecular programs downstream of the HOXA5 transcription factor in the context of the postnatal brainstem. A comparative transcriptomic analysis was performed in combination with gene expression localization, using a conditional postnatal Hoxa5 loss-of-function mouse model. After inactivation of Hoxa5 at postnatal days (P)1-P4, we established the transcriptome of the brainstem from P21 Hoxa5 conditional mutants using RNA-Seq analysis. One major finding was the downregulation of several genes associated with synaptic function in Hoxa5 mutant specimens including different actors involved in glutamatergic synapse, calcium signaling pathway, and GABAergic synapse. Data were confirmed and extended by reverse transcription quantitative polymerase chain reaction analysis, and the expression of several HOXA5 candidate targets was shown to co-localize with Hoxa5 transcripts in precerebellar nuclei. Together, these new results revealed that HOXA5, through the regulation of key actors of the glutamatergic/GABAergic synapses and calcium signaling, might be involved in synaptogenesis, synaptic transmission, and synaptic plasticity of the cortico-ponto-cerebellar circuitry in the postnatal brainstem.

14.
J Exp Biol ; 220(Pt 24): 4571-4577, 2017 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-29074702

RESUMEN

Fetal development of the respiratory tract and diaphragm requires strict coordination between genetically controlled signals and mechanical forces produced by the neural network that generates breathing. HOXA5, which is expressed in the mesenchyme of the trachea, lung and diaphragm, and in phrenic motor neurons, is a key transcription factor regulating lung development and function. Consequently, most Hoxa5-/- mutants die at birth from respiratory failure. However, the extensive effect of the null mutation makes it difficult to identify the origins of respiratory dysfunction in newborns. To address the physiological impact of Hoxa5 tissue-specific roles, we used conditional gene targeting with the Dermo1Cre and Olig2Cre mouse lines to produce specific Hoxa5 deletions in the mesenchyme and motor neurons, respectively. Hoxa5 expression in the mesenchyme is critical for trachea development, whereas its expression in phrenic motor neurons is essential for diaphragm formation. Breathing measurements in adult mice with whole-body plethysmography demonstrated that, at rest, only the motor neuron deletion affects respiration, resulting in higher breathing frequency and decreased tidal volume. But subsequent exposure to a moderate hypoxic challenge (FiO2 =0.12; 10 min) revealed that both mutant mice hyperventilate more than controls. Hoxa5flox/flox;Dermo1+/Cre mice showed augmented tidal volume while Hoxa5flox/flox;Olig2+/Cre mice had the largest increase in breathing frequency. No significant differences were observed between medulla-spinal cord preparations from E18.5 control and Hoxa5flox/flox;Olig2+/Cre mouse embryos that could support a role for Hoxa5 in fetal inspiratory motor command. According to our data, Hoxa5 expression in the mesenchyme and phrenic motor neurons controls distinct aspects of respiratory development.


Asunto(s)
Proteínas de Homeodominio/genética , Pulmón/embriología , Fosfoproteínas/genética , Insuficiencia Respiratoria/genética , Animales , Diafragma/fisiopatología , Eliminación de Gen , Marcación de Gen , Proteínas de Homeodominio/fisiología , Técnicas In Vitro , Pulmón/metabolismo , Pulmón/fisiopatología , Ratones , Ratones Noqueados , Mutación , Fosfoproteínas/fisiología , Pletismografía Total , Insuficiencia Respiratoria/fisiopatología , Tráquea/fisiopatología , Factores de Transcripción
15.
Development ; 144(19): 3547-3561, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28827394

RESUMEN

Hoxa5 is essential for development of several organs and tissues. In the respiratory system, loss of Hoxa5 function causes neonatal death due to respiratory distress. Expression of HOXA5 protein in mesenchyme of the respiratory tract and in phrenic motor neurons of the central nervous system led us to address the individual contribution of these Hoxa5 expression domains using a conditional gene targeting approach. Hoxa5 does not play a cell-autonomous role in lung epithelium, consistent with lack of HOXA5 expression in this cell layer. In contrast, ablation of Hoxa5 in mesenchyme perturbed trachea development, lung epithelial cell differentiation and lung growth. Further, deletion of Hoxa5 in motor neurons resulted in abnormal diaphragm innervation and musculature, and lung hypoplasia. It also reproduced the neonatal lethality observed in null mutants, indicating that the defective diaphragm is the main cause of impaired survival at birth. Thus, Hoxa5 possesses tissue-specific functions that differentially contribute to the morphogenesis of the respiratory tract.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Fosfoproteínas/metabolismo , Sistema Respiratorio/embriología , Sistema Respiratorio/metabolismo , Animales , Animales Recién Nacidos , Tipificación del Cuerpo/genética , Cartílago/embriología , Cartílago/metabolismo , Diferenciación Celular/genética , Cruzamientos Genéticos , Diafragma/inervación , Diafragma/metabolismo , Diafragma/ultraestructura , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Genotipo , Proteínas de Homeodominio/genética , Masculino , Mesodermo/embriología , Mesodermo/metabolismo , Modelos Biológicos , Neuronas Motoras/metabolismo , Desarrollo de Músculos , Fibras Musculares Esqueléticas/metabolismo , Especificidad de Órganos/genética , Fosfoproteínas/genética , Mucosa Respiratoria/metabolismo , Factor de Transcripción SOX9/metabolismo , Transducción de Señal/genética , Análisis de Supervivencia , Tráquea/embriología , Tráquea/metabolismo , Factores de Transcripción
16.
Dev Dyn ; 246(1): 72-82, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27748998

RESUMEN

BACKGROUND: Reciprocal epithelial-mesenchymal communications are critical throughout lung development, dictating branching morphogenesis and cell specification. Numerous signaling molecules are involved in these interactions, but the way epithelial-mesenchymal crosstalk is coordinated remains unclear. The ERK/MAPK pathway transduces several important signals in lung formation. Epithelial inactivation of both Mek genes, encoding ERK/MAPK kinases, causes lung agenesis and death. Conversely, Mek mutation in mesenchyme results in lung hypoplasia, trachea cartilage malformations, kyphosis, omphalocele, and death. Considering the negative impact of kyphosis and omphalocele on intrathoracic space and, consequently, on lung growth, the exact role of ERK/MAPK pathway in lung mesenchyme remains unresolved. RESULTS: To address the role of the ERK/MAPK pathway in lung mesenchyme in absence of kyphosis and omphalocele, we used the Tbx4Cre deleter mouse line, which acts specifically in lung mesenchyme. These Mek mutants did not develop kyphosis and omphalocele but they presented lung hypoplasia, tracheal defects, and neonatal death. Tracheal cartilage anomalies suggested a role for the ERK/MAPK pathway in the control of chondrocyte hypertrophy. Moreover, expression data indicated potential interactions between the ERK/MAPK and canonical Wnt pathways during lung formation. CONCLUSIONS: Lung development necessitates a functional ERK/MAPK pathway in the lung mesenchymal layer in order to coordinate efficient epithelial-mesenchymal interactions. Developmental Dynamics 246:72-82, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Comunicación Celular , Pulmón/crecimiento & desarrollo , Sistema de Señalización de MAP Quinasas/fisiología , Mesodermo/metabolismo , Organogénesis , Animales , Condrocitos/patología , Epitelio/embriología , Epitelio/fisiología , Pulmón/embriología , Sistema de Señalización de MAP Quinasas/genética , Mesodermo/embriología , Mesodermo/fisiología , Ratones , Mutación , Tráquea/anomalías , Vía de Señalización Wnt
17.
Paediatr Respir Rev ; 19: 62-8, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26907828

RESUMEN

It is well established that a number of birth defects are associated with improper formation of the respiratory tract. Important progress has been made in the identification of components of the regulatory networks controlling lung morphogenesis. They comprise a variety of soluble factors, receptors, transcription factors, and miRNAs. However, the underlying molecular mechanisms remain unsolved and fundamental questions, such as those related to lung branching are still unanswered. Congenital cystic lung diseases consist of a heterogeneous group of rare lung diseases mainly detected prenatally and characterized by airway dilatation. Despite their apparent phenotypic heterogeneity, these malformations are proposed to be related to a common malformation sequence occurring during lung branching morphogenesis.


Asunto(s)
Malformación Adenomatoide Quística Congénita del Pulmón/genética , Malformación Adenomatoide Quística Congénita del Pulmón/patología , Blastoma Pulmonar/genética , Blastoma Pulmonar/patología , Animales , Modelos Animales de Enfermedad , Humanos
18.
J Dev Biol ; 4(2)2016 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-29615582

RESUMEN

A critical position in the developmental hierarchy is occupied by the Hox genes, which encode transcription factors. Hox genes are crucial in specifying regional identity along the embryonic axes and in regulating morphogenesis. In mouse, targeted mutations of Hox genes cause skeletal transformations and organ defects that can impair viability. Here, we present the current knowledge about the Hoxa5 gene, a paradigm for the function and the regulation of Hox genes. The phenotypic survey of Hoxa5-/- mice has unveiled its critical role in the regional specification of the skeleton and in organogenesis. Most Hoxa5-/- mice die at birth from respiratory distress due to tracheal and lung dysmorphogenesis and impaired diaphragm innervation. The severity of the phenotype establishes that Hoxa5 plays a predominant role in lung organogenesis versus other Hox genes. Hoxa5 also governs digestive tract morphogenesis, thyroid and mammary glands development, and ovary homeostasis. Deregulated Hoxa5 expression is reported in cancers, indicating Hoxa5 involvement in tumor predisposition and progression. The dynamic Hoxa5 expression profile is under the transcriptional control of multiple cis-acting sequences and trans-acting regulators. It is also modulated by epigenetic mechanisms, implicating chromatin modifications and microRNAs. Finally, lncRNAs originating from alternative splicing and distal promoters encompass the Hoxa5 locus.

19.
Development ; 142(21): 3801, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26534987

RESUMEN

There was an error published in Development 141, 3197-3211. In the key for Fig. 3C, the grey bars were labelled with the incorrect genotype name. The correct genotype is Mek1+/flox;Mek2−/−; Dermo1+/cre. This error does not affect the conclusions of the paper. The authors apologise to readers for this mistake.

20.
Transgenic Res ; 24(6): 1065-77, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26395370

RESUMEN

Temporal control of site-specific recombination is commonly achieved by using a tamoxifen-inducible form of Cre or Flp recombinases. Although powerful protocols of induction have been developed for gene inactivation at adult stages or during embryonic development, induction of recombination at late gestational or early postnatal stages is still difficult to achieve. In this context, using the ubiquitous CMV-CreER(T2) transgenic mice, we have tested and validated two procedures to achieve recombination just before and just after birth. The efficiency of recombination was evaluated in the brain, which is known to be more problematic to target. For the late gestation treatment with tamoxifen, different protocols of complementary administration of progesterone and estrogen were tested. However, delayed delivery and/or mortality of pups due to difficult delivery were always observed. To circumvent this problem, pups were collected from tamoxifen-treated pregnant dams by caesarian section at E18.5 and given to foster mothers. For postnatal treatment, different dosages of tamoxifen were administered by intragastric injection to the pups during 3 or 4 days after birth. The efficiency of these treatments was analyzed at P7 using a transgenic reporter line. They were also validated with the Hoxa5 conditional allele. In conclusion, we have developed efficient procedures that allow achieving efficient recombination of floxed alleles at perinatal stages. These protocols will allow investigating the late/adult functions of many developmental genes, whose characterization has been so far restricted to embryonic development.


Asunto(s)
Proteínas de Homeodominio/genética , Integrasas/metabolismo , Fosfoproteínas/genética , Receptores de Estrógenos/genética , Recombinación Genética , Tamoxifeno/farmacología , Animales , Animales Recién Nacidos , Antagonistas de Estrógenos/farmacología , Femenino , Integrasas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Atención Perinatal , Embarazo , Factores de Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...