Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Pain ; 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39190341

RESUMEN

ABSTRACT: The thalamus plays an important role in sensory and motor information processing by mediating communication between the periphery and the cerebral cortex. Alterations in thalamic development have profound consequences on sensory and motor function. In this study, we investigated a mouse model in which thalamic nuclei formation is disrupted because of the absence of Sonic hedgehog (Shh) expression from 2 key signaling centers that are required for embryonic forebrain development. The resulting defects observed in distinct thalamic sensory nuclei in Shh mutant embryos persisted into adulthood prompting us to examine their effect on behavioral responses to somatosensory stimulation. Our findings reveal a role for first-order posterior medial thalamic neurons and their projections to layer 4 of the secondary somatosensory cortex in the transmission of nociceptive information. Together, these results establish a connection between a neurodevelopmental lesion in the thalamus and a modality-specific disruption in pain perception.

2.
PLoS Genet ; 19(1): e1010584, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36656851

RESUMEN

Loss or absence of hearing is common at both extremes of human lifespan, in the forms of congenital deafness and age-related hearing loss. While these are often studied separately, there is increasing evidence that their genetic basis is at least partially overlapping. In particular, both common and rare variants in genes associated with monogenic forms of hearing loss also contribute to the more polygenic basis of age-related hearing loss. Here, we directly test this model in the Penn Medicine BioBank-a healthcare system cohort of around 40,000 individuals with linked genetic and electronic health record data. We show that increased burden of predicted deleterious variants in Mendelian hearing loss genes is associated with increased risk and severity of adult-onset hearing loss. As a specific example, we identify one gene-TCOF1, responsible for a syndromic form of congenital hearing loss-in which deleterious variants are also associated with adult-onset hearing loss. We also identify four additional novel candidate genes (COL5A1, HMMR, RAPGEF3, and NNT) in which rare variant burden may be associated with hearing loss. Our results confirm that rare variants in Mendelian hearing loss genes contribute to polygenic risk of hearing loss, and emphasize the utility of healthcare system cohorts to study common complex traits and diseases.


Asunto(s)
Sordera , Pérdida Auditiva Sensorineural , Pérdida Auditiva , Humanos , Adulto , Sordera/genética , Pérdida Auditiva/genética , Pérdida Auditiva Sensorineural/genética , Herencia Multifactorial , Audición , Mutación
3.
Cell Rep ; 41(10): 111768, 2022 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-36476860

RESUMEN

The thalamus is the principal information hub of the vertebrate brain, with essential roles in sensory and motor information processing, attention, and memory. The complex array of thalamic nuclei develops from a restricted pool of neural progenitors. We apply longitudinal single-cell RNA sequencing and regional abrogation of Sonic hedgehog (Shh) to map the developmental trajectories of thalamic progenitors, intermediate progenitors, and post-mitotic neurons as they coalesce into distinct thalamic nuclei. These data reveal that the complex architecture of the thalamus is established early during embryonic brain development through the coordinated action of four cell differentiation lineages derived from Shh-dependent and -independent progenitors. We systematically characterize the gene expression programs that define these thalamic lineages across time and demonstrate how their disruption upon Shh depletion causes pronounced locomotor impairment resembling infantile Parkinson's disease. These results reveal key principles of thalamic development and provide mechanistic insights into neurodevelopmental disorders resulting from thalamic dysfunction.


Asunto(s)
Tálamo , Tálamo/citología
4.
Otolaryngol Head Neck Surg ; 166(3): 537-539, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34058916

RESUMEN

"Cookie-bite" or U-shaped audiograms-specifically, those showing midfrequency sensorineural hearing loss (HL)-are traditionally taught to be associated with genetic HL; however, their utility as a screening tool has not been reported. We aim to determine the performance of a cookie-bite audiogram shape in stratifying patients carrying putative loss-of-function variants in known HL genes from wild-type controls. We merged audiometric and exome sequencing data from adults enrolled in a large biobank at a tertiary care center. Of 321 patients, 50 carried a putative loss-of-function variant in an HL gene. The cookie-bite shape was present in 9 of those patients, resulting in low sensitivity (18%) and positive predictive value (15%) in stratifying genetic carrier status; 84% of patients with a cookie-bite audiogram did not carry a genetic variant. A cookie-bite audiogram should not be used to screen adults for possible genetic testing.


Asunto(s)
Sordera , Pérdida Auditiva Sensorineural , Pérdida Auditiva , Adulto , Audiometría/métodos , Audiometría de Tonos Puros , Pérdida Auditiva/genética , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/genética , Humanos , Secuenciación del Exoma
5.
Otolaryngol Head Neck Surg ; 166(4): 746-752, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34281439

RESUMEN

OBJECTIVE: To investigate the importance of rare variants in adult-onset hearing loss. STUDY DESIGN: Genomic association study. SETTING: Large biobank from tertiary care center. METHODS: We investigated rare variants (minor allele frequency <5%) in 42 autosomal dominant (DFNA) postlingual hearing loss (HL) genes in 16,657 unselected individuals in the Penn Medicine Biobank. We determined the prevalence of known pathogenic and predicted deleterious variants in subjects with audiometric-proven sensorineural hearing loss. We scanned across known postlingual DFNA HL genes to determine those most significantly contributing to the phenotype. We replicated findings in an independent cohort (UK Biobank). RESULTS: While rare individually, when considering the accumulation of variants in all postlingual DFNA genes, more than 90% of participants carried at least 1 rare variant. Rare variants predicted to be deleterious were enriched in adults with audiometric-proven hearing loss (pure-tone average >25 dB; P = .015). Patients with a rare predicted deleterious variant had an odds ratio of 1.27 for HL compared with genotypic controls (P = .029). Gene burden in DIABLO, PTPRQ, TJP2, and POU4F3 were independently associated with sensorineural hearing loss. CONCLUSION: Although prior reports have focused on common variants, we find that rare predicted deleterious variants in DFNA postlingual HL genes are enriched in patients with adult-onset HL in a large health care system population. We show the value of investigating rare variants to uncover hearing loss phenotypes related to implicated genes.


Asunto(s)
Sordera , Pérdida Auditiva Sensorineural , Pérdida Auditiva , Audiometría , Pérdida Auditiva/genética , Pérdida Auditiva Sensorineural/genética , Humanos , Linaje , Fenotipo , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética
6.
Dev Cell ; 56(10): 1526-1540.e7, 2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-33964205

RESUMEN

In mammals, sound is detected by mechanosensory hair cells that are activated in response to vibrations at frequency-dependent positions along the cochlear duct. We demonstrate that inner ear supporting cells provide a structural framework for transmitting sound energy through the cochlear partition. Humans and mice with mutations in GAS2, encoding a cytoskeletal regulatory protein, exhibit hearing loss due to disorganization and destabilization of microtubule bundles in pillar and Deiters' cells, two types of inner ear supporting cells with unique cytoskeletal specializations. Failure to maintain microtubule bundle integrity reduced supporting cell stiffness, which in turn altered cochlear micromechanics in Gas2 mutants. Vibratory responses to sound were measured in cochleae from live mice, revealing defects in the propagation and amplification of the traveling wave in Gas2 mutants. We propose that the microtubule bundling activity of GAS2 imparts supporting cells with mechanical properties for transmitting sound energy through the cochlea.


Asunto(s)
Cóclea/citología , Citoesqueleto/metabolismo , Audición/fisiología , Proteínas de Microfilamentos/metabolismo , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Secuencia de Bases , Citoesqueleto/ultraestructura , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/ultraestructura , Pérdida Auditiva/metabolismo , Pérdida Auditiva/patología , Pérdida Auditiva/fisiopatología , Humanos , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/genética , Microtúbulos/metabolismo , Mutación/genética , Transporte de Proteínas , Sonido , Vibración , Secuenciación del Exoma
7.
Hum Genet ; 140(6): 957-967, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33745059

RESUMEN

While newborns and children with hearing loss are routinely offered genetic testing, adults are rarely clinically tested for a genetic etiology. One clinically actionable result from genetic testing in children is the discovery of variants in syndromic hearing loss genes. EYA4 is a known hearing loss gene which is also involved in important pathways in cardiac tissue. The pleiotropic effects of rare EYA4 variants are poorly understood and their prevalence in a large cohort has not been previously reported. We investigated cardio-auditory phenotypes in 11,451 individuals in a large biobank using a rare variant, genome-first approach to EYA4. We filtered 256 EYA4 variants carried by 6737 participants to 26 rare and predicted deleterious variants carried by 42 heterozygotes. We aggregated predicted deleterious EYA4 gene variants into a combined variable (i.e. "gene burden") and performed association studies across phenotypes compared to wildtype controls. We validated findings with replication in three independent cohorts and human tissue expression data. EYA4 gene burden was significantly associated with audiometric-proven HL (p = [Formula: see text], Mobitz Type II AV block (p = [Formula: see text]) and the syndromic presentation of HL and primary cardiomyopathy (p = 0.0194). Analyses on audiogram, echocardiogram, and electrocardiogram data validated these associations. Prior reports have focused on identifying variants in families with severe or syndromic phenotypes. In contrast, we found, using a genotype-first approach, that gene burden in EYA4 is associated with more subtle cardio-auditory phenotypes in an adult medical biobank population, including cardiac conduction disorders which have not been previously reported. We show the value of using a focused approach to uncover human disease related to pleiotropic gene variants and suggest a role for genetic testing in adults presenting with hearing loss.


Asunto(s)
Cardiomiopatías/genética , Genoma Humano , Pérdida Auditiva/genética , Mutación , Transactivadores/genética , Audiometría , Bancos de Muestras Biológicas , Población Negra , Cardiomiopatías/diagnóstico por imagen , Cardiomiopatías/etnología , Cardiomiopatías/patología , Ecocardiografía , Electrocardiografía , Expresión Génica , Pérdida Auditiva/diagnóstico por imagen , Pérdida Auditiva/etnología , Pérdida Auditiva/patología , Humanos , Masculino , Pennsylvania , Fenotipo , Índice de Severidad de la Enfermedad , Población Blanca , Secuenciación del Exoma
8.
Laryngoscope ; 131(2): 401-409, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32243624

RESUMEN

Literature and clinical practice around adult-onset hearing loss (HL) has traditionally focused on environmental risk factors, including noise exposure, ototoxic drug exposure, and cardiovascular disease. The most common diagnosis in adult-onset HL is presbycusis. However, the age of onset of presbycusis varies, and patients often describe family history of HL as well as individual variation in progression and severity. In recent years, there has been accumulating evidence of gene-environment interactions underlying adult cases of HL. Susceptibility loci for age-related HL have been identified, and genes related to postlingual nonsyndromic HL continue to be discovered through individual reports and genome-wide association studies. This review will outline main concepts in genetics as related to HL, identify implicated genes, and discuss clinical implications. Laryngoscope, 131:401-409, 2021.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Presbiacusia/genética , Edad de Inicio , Anciano , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Persona de Mediana Edad
9.
Protein Cell ; 11(8): 565-583, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32193873

RESUMEN

The anterior pituitary gland drives highly conserved physiologic processes in mammalian species. These hormonally controlled processes are central to somatic growth, pubertal transformation, fertility, lactation, and metabolism. Current cellular models of mammalian anteiror pituitary, largely built on candidate gene based immuno-histochemical and mRNA analyses, suggest that each of the seven hormones synthesized by the pituitary is produced by a specific and exclusive cell lineage. However, emerging evidence suggests more complex relationship between hormone specificity and cell plasticity. Here we have applied massively parallel single-cell RNA sequencing (scRNA-seq), in conjunction with complementary imaging-based single-cell analyses of mRNAs and proteins, to systematically map both cell-type diversity and functional state heterogeneity in adult male and female mouse pituitaries at single-cell resolution and in the context of major physiologic demands. These quantitative single-cell analyses reveal sex-specific cell-type composition under normal pituitary homeostasis, identify an array of cells associated with complex complements of hormone-enrichment, and undercover non-hormone producing interstitial and supporting cell-types. Interestingly, we also identified a Pou1f1-expressing cell population that is characterized by a unique multi-hormone gene expression profile. In response to two well-defined physiologic stresses, dynamic shifts in cellular diversity and transcriptome profiles were observed for major hormone producing and the putative multi-hormone cells. These studies reveal unanticipated cellular complexity and plasticity in adult pituitary, and provide a rich resource for further validating and expanding our molecular understanding of pituitary gene expression programs and hormone production.


Asunto(s)
Plasticidad de la Célula/genética , Hipófisis/citología , Hipófisis/metabolismo , ARN Mensajero/genética , RNA-Seq , Caracteres Sexuales , Análisis de la Célula Individual , Transcriptoma , Animales , Femenino , Homeostasis/genética , Masculino , Ratones , Ratones Transgénicos
10.
Development ; 146(18)2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31488567

RESUMEN

The mammalian cochlea develops from a ventral outgrowth of the otic vesicle in response to Shh signaling. Mouse embryos lacking Shh or its essential signal transduction components display cochlear agenesis; however, a detailed understanding of the transcriptional network mediating this process is unclear. Here, we describe an integrated genomic approach to identify Shh-dependent genes and associated regulatory sequences that promote cochlear duct morphogenesis. A comparative transcriptome analysis of otic vesicles from mouse mutants exhibiting loss (Smoecko ) and gain (Shh-P1) of Shh signaling reveal a set of Shh-responsive genes partitioned into four expression categories in the ventral half of the otic vesicle. This target gene classification scheme provides novel insight into several unanticipated roles for Shh, including priming the cochlear epithelium for subsequent sensory development. We also mapped regions of open chromatin in the inner ear by ATAC-seq that, in combination with Gli2 ChIP-seq, identified inner ear enhancers in the vicinity of Shh-responsive genes. These datasets are useful entry points for deciphering Shh-dependent regulatory mechanisms involved in cochlear duct morphogenesis and establishment of its constituent cell types.


Asunto(s)
Cóclea/embriología , Cóclea/metabolismo , Genoma , Proteínas Hedgehog/metabolismo , Morfogénesis/genética , Animales , Secuencia de Bases , Embrión de Mamíferos/metabolismo , Elementos de Facilitación Genéticos/genética , Regulación del Desarrollo de la Expresión Génica , Ratones Transgénicos , Reproducibilidad de los Resultados
11.
Development ; 145(21)2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30291164

RESUMEN

Sonic hedgehog (Shh) plays well characterized roles in brain and spinal cord development, but its functions in the hypothalamus have been more difficult to elucidate owing to the complex neuroanatomy of this brain area. Here, we use fate mapping and conditional deletion models in mice to define requirements for dynamic Shh activity at distinct developmental stages in the tuberal hypothalamus, a brain region with important homeostatic functions. At early time points, Shh signaling regulates dorsoventral patterning, neurogenesis and the size of the ventral midline. Fate-mapping experiments demonstrate that Shh-expressing and -responsive progenitors contribute to distinct neuronal subtypes, accounting for some of the cellular heterogeneity in tuberal hypothalamic nuclei. Conditional deletion of the hedgehog transducer smoothened (Smo), after dorsoventral patterning has been established, reveals that Shh signaling is necessary to maintain proliferation and progenitor identity during peak periods of hypothalamic neurogenesis. We also find that mosaic disruption of Smo causes a non-cell autonomous gain in Shh signaling activity in neighboring wild-type cells, suggesting a mechanism for the pathogenesis of hypothalamic hamartomas, benign tumors that form during hypothalamic development.


Asunto(s)
Proteínas Hedgehog/metabolismo , Hipotálamo/embriología , Hipotálamo/metabolismo , Transducción de Señal , Animales , Núcleo Arqueado del Hipotálamo/embriología , Núcleo Arqueado del Hipotálamo/metabolismo , Tipificación del Cuerpo , Núcleo Celular/metabolismo , Proliferación Celular , Embrión de Mamíferos/metabolismo , Ratones , Neurogénesis , Neuronas/metabolismo , Receptor Smoothened/metabolismo , Factores de Tiempo , Proteína con Dedos de Zinc GLI1/metabolismo
12.
Dev Cell ; 43(3): 318-331.e5, 2017 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-29107558

RESUMEN

Alternative splicing contributes to gene expression dynamics in many tissues, yet its role in auditory development remains unclear. We performed whole-exome sequencing in individuals with sensorineural hearing loss (SNHL) and identified pathogenic mutations in Epithelial Splicing-Regulatory Protein 1 (ESRP1). Patient-derived induced pluripotent stem cells showed alternative splicing defects that were restored upon repair of an ESRP1 mutant allele. To determine how ESRP1 mutations cause hearing loss, we evaluated Esrp1-/- mouse embryos and uncovered alterations in cochlear morphogenesis, auditory hair cell differentiation, and cell fate specification. Transcriptome analysis revealed impaired expression and splicing of genes with essential roles in cochlea development and auditory function. Aberrant splicing of Fgfr2 blocked stria vascularis formation due to erroneous ligand usage, which was corrected by reducing Fgf9 gene dosage. These findings implicate mutations in ESRP1 as a cause of SNHL and demonstrate the complex interplay between alternative splicing, inner ear development, and auditory function.


Asunto(s)
Empalme Alternativo/genética , Cóclea/embriología , Pérdida Auditiva/genética , Mutación/genética , Proteínas de Unión al ARN/genética , Animales , Diferenciación Celular/genética , Cóclea/metabolismo , Ratones Noqueados
13.
Dis Model Mech ; 10(1): 29-37, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27935818

RESUMEN

Septo-optic dysplasia (SOD) is a congenital disorder characterized by optic nerve, pituitary and midline brain malformations. The clinical presentation of SOD is highly variable with a poorly understood etiology. The majority of SOD cases are sporadic, but in rare instances inherited mutations have been identified in a small number of transcription factors, some of which regulate the expression of Sonic hedgehog (Shh) during mouse forebrain development. SOD is also associated with young maternal age, suggesting that environmental factors, including alcohol consumption at early stages of pregnancy, might increase the risk of developing this condition. Here, we address the hypothesis that SOD is a multifactorial disorder stemming from interactions between mutations in Shh pathway genes and prenatal ethanol exposure. Mouse embryos with mutations in the Shh co-receptor, Cdon, were treated in utero with ethanol or saline at embryonic day 8 (E8.0) and evaluated for optic nerve hypoplasia (ONH), a prominent feature of SOD. We show that both Cdon-/- mutation and prenatal ethanol exposure independently cause ONH through a similar pathogenic mechanism that involves selective inhibition of Shh signaling in retinal progenitor cells, resulting in their premature cell-cycle arrest, precocious differentiation and failure to properly extend axons to the optic nerve. The ONH phenotype was not exacerbated in Cdon-/- embryos treated with ethanol, suggesting that an intact Shh signaling pathway is required for ethanol to exert its teratogenic effects. These results support a model whereby mutations in Cdon and prenatal ethanol exposure increase SOD risk through spatiotemporal perturbations in Shh signaling activity.


Asunto(s)
Etanol/efectos adversos , Proteínas Hedgehog/metabolismo , Mutación/genética , Nervio Óptico/anomalías , Efectos Tardíos de la Exposición Prenatal/genética , Animales , Moléculas de Adhesión Celular/genética , Diferenciación Celular , Proliferación Celular , Embrión de Mamíferos/patología , Femenino , Ratones , Modelos Biológicos , Nervio Óptico/embriología , Nervio Óptico/patología , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Transducción de Señal , Células Madre/metabolismo , Proteína con Dedos de Zinc GLI1/metabolismo
15.
Nat Genet ; 48(5): 575-80, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27064252

RESUMEN

Genomic approaches have predicted hundreds of thousands of tissue-specific cis-regulatory sequences, but the determinants critical to their function and evolutionary history are mostly unknown. Here we systematically decode a set of brain enhancers active in the zona limitans intrathalamica (zli), a signaling center essential for vertebrate forebrain development via the secreted morphogen Sonic hedgehog (Shh). We apply a de novo motif analysis tool to identify six position-independent sequence motifs together with their cognate transcription factors that are essential for zli enhancer activity and Shh expression in the mouse embryo. Using knowledge of this regulatory lexicon, we discover new Shh zli enhancers in mice and a functionally equivalent element in hemichordates, indicating an ancient origin of the Shh zli regulatory network that predates the chordate phylum. These findings support a strategy for delineating functionally conserved enhancers in the absence of overt sequence homologies and over extensive evolutionary distances.


Asunto(s)
Cordados/genética , Elementos de Facilitación Genéticos , Evolución Molecular , Prosencéfalo/embriología , Animales , Cordados/embriología , Cordados/metabolismo , Femenino , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Masculino , Ratones , Ratones Transgénicos , Prosencéfalo/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo
16.
Dev Biol ; 399(1): 177-187, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25592224

RESUMEN

Wnt1 and Wnt3a secreted from the dorsal neural tube were previously shown to regulate a gene expression program in the dorsal otic vesicle that is necessary for vestibular morphogenesis (Riccomagno et al., 2005. Genes Dev. 19, 1612-1623). Unexpectedly, Wnt1(-/-); Wnt3a(-/-) embryos also displayed a pronounced defect in the outgrowth of the ventrally derived cochlear duct. To determine how Wnt signaling in the dorsal otocyst contributes to cochlear development we performed a series of genetic fate mapping experiments using two independent Wnt responsive driver strains (TopCreER and Gbx2(CreER)) that when crossed to inducible responder lines (Rosa(lacZ) or Rosa(zsGreen)) permanently labeled dorsomedial otic progenitors and their derivatives. Tamoxifen time course experiments revealed that most vestibular structures showed some degree of labeling when recombination was induced between E7.75 and E12.5, consistent with continuous Wnt signaling activity in this tissue. Remarkably, a population of Wnt responsive cells in the dorsal otocyst was also found to contribute to the sensory epithelium of the cochlear duct, including auditory hair and support cells. Similar results were observed with both TopCreER and Gbx2(CreER) strains. The ventral displacement of Wnt responsive cells followed a spatiotemporal sequence that initiated in the anterior otic cup at, or immediately prior to, the 17-somite stage (E9) and then spread progressively to the posterior pole of the otic vesicle by the 25-somite stage (E9.5). These lineage-tracing experiments identify the earliest known origin of auditory sensory progenitors within a population of Wnt responsive cells in the dorsomedial otic cup.


Asunto(s)
Cóclea/metabolismo , Oído Interno/metabolismo , Epitelio/metabolismo , Vía de Señalización Wnt/genética , Animales , Linaje de la Célula/genética , Movimiento Celular/genética , Proliferación Celular/genética , Cóclea/citología , Cóclea/embriología , Oído Interno/citología , Oído Interno/embriología , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Epitelio/embriología , Antagonistas de Estrógenos/farmacología , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Hibridación in Situ , Masculino , Ratones Transgénicos , Microscopía Confocal , Morfogénesis/efectos de los fármacos , Morfogénesis/genética , Tamoxifeno/farmacología , Factores de Tiempo
17.
Development ; 140(11): 2299-309, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23674600

RESUMEN

Tbx2 and Tbx3 are two highly related members of the T-box transcription factor gene family that regulate patterning and differentiation of a number of tissue rudiments in the mouse. Both genes are partially co-expressed in the ventral diencephalon and the infundibulum; however, a functional requirement in murine pituitary development has not been reported. Here, we show by genetic lineage tracing that Tbx2(+) cells constitute the precursor population of the neurohypophysis. However, Tbx2 is dispensable for neurohypophysis development as revealed by normal formation of this organ in Tbx2-deficient mice. By contrast, loss of Tbx3 from the ventral diencephalon results in a failure to establish the Tbx2(+) domain in this region, and a lack of evagination of the infundibulum and formation of the neurohypophysis. Rathke's pouch is severely hypoplastic, exhibits defects in dorsoventral patterning, and degenerates after E12.5. In Tbx3-deficient embryos, the ventral diencephalon is hyperproliferative and displays an abnormal cellular architecture, probably resulting from a failure to repress transcription of Shh. We further show that Tbx3 and Tbx2 repress Shh by sequestering the SRY box-containing transcription factor Sox2 away from a Shh forebrain enhancer (SBE2), thus preventing its activation. These data suggest that Tbx3 is required in the ventral diencephalon to establish a Shh(-) domain to allow formation of the infundibulum.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Neurohipófisis/metabolismo , Factores de Transcripción SOXB1/metabolismo , Proteínas de Dominio T Box/metabolismo , Animales , Encéfalo/embriología , Células COS , Proliferación Celular , Chlorocebus aethiops , Diencéfalo/embriología , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Hipófisis/embriología , Factores de Tiempo
18.
Development ; 140(8): 1730-9, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23487315

RESUMEN

The morphogenetic program that shapes the three semicircular canals (SSCs) must be executed with extreme precision to satisfy their complex vestibular function. The SSCs emerge from epithelial outgrowths of the dorsal otocyst, the central regions of which fuse and resorb to leave three fluid-filled canals. The Wnt/ß-catenin signaling pathway is active at multiple stages of otic development, including during vestibular morphogenesis. How Wnt/ß-catenin functionally integrates with other signaling pathways to sculpt the SSCs and their sensory patches is unknown. We used a genetic strategy to spatiotemporally modulate canonical Wnt signaling activity during SSC development in mice. Our findings demonstrate that Wnt/ß-catenin signaling functions in a multifaceted manner during SSC formation. In the early phase, Wnt/ß-catenin signaling is required to preserve the epithelial integrity of the vertical canal pouch perimeter (presumptive anterior and posterior SSCs) by establishing a sensory-dependent signaling relay that maintains expression of Dlx5 and opposes expression of the fusion plate marker netrin 1. Without this Wnt signaling activity the sensory to non-sensory signaling cascade fails to be activated, resulting in loss of vestibular hair and support cells and the anterior and posterior SSCs. In the later phase, Wnt/ß-catenin signaling becomes restricted to the fusion plate where it facilitates the timely resorption of this tissue. Mosaic recombination of ß-catenin in small clusters of canal pouch cells prevents their resorption, causing instead the formation of ectopic SSCs. Together, these disparate functions of the Wnt/ß-catenin pathway in epithelial maintenance and resorption help regulate the size, shape and number of SSCs.


Asunto(s)
Epitelio/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Morfogénesis/fisiología , Canales Semicirculares/embriología , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo , Animales , Bromodesoxiuridina , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Hibridación in Situ , Ratones , Factores de Crecimiento Nervioso/metabolismo , Netrina-1 , Proteínas Supresoras de Tumor/metabolismo , beta-Galactosidasa
19.
Dev Biol ; 366(2): 393-403, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22561201

RESUMEN

Six6, a sine oculis homeobox protein, plays a crucial and conserved role in the development of the forebrain and eye. To understand how the expression of Six6 is regulated during embryogenesis, we screened ~250 kb of genomic DNA encompassing the Six6 locus for cis-regulatory elements capable of directing reporter gene expression to sites of Six6 transcription in transgenic mouse embryos. Here, we describe two novel enhancer elements, that are highly conserved in vertebrate species and whose activities recapitulate Six6 expression in the ventral forebrain and eye, respectively. Cross-species comparisons of the Six6 forebrain enhancer sequences revealed highly conserved binding sites matching the consensus for homeodomain and SoxB1 transcription factors. Deletion of either of the binding sites resulted in loss of the forebrain enhancer activity in the ventral forebrain. Moreover, our studies show that members of the SoxB1 family, including Sox2 and Sox3, are expressed in the overlapping region of the ventral forebrain with Six6 and can bind to the Six6 forebrain enhancer. Loss of function of SoxB1 genes in vivo further emphasizes their role in regulating Six6 forebrain enhancer activity. Thus, our data strongly suggest that SoxB1 transcription factors are direct activators of Six6 expression in the ventral forebrain.


Asunto(s)
Elementos de Facilitación Genéticos , Proteínas de Homeodominio/genética , Prosencéfalo/fisiología , Factores de Transcripción SOXB1/genética , Transactivadores/genética , Animales , Ojo/embriología , Proteínas del Ojo/genética , Regulación del Desarrollo de la Expresión Génica , Ratones , Prosencéfalo/embriología , Unión Proteica , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción SOXB1/metabolismo
20.
Front Neurosci ; 6: 57, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22529771

RESUMEN

The thalamus is strategically positioned within the caudal diencephalic area of the forebrain, between the mesencephalon and telencephalon. This location is important for unique aspects of thalamic function, to process and relay sensory and motor information to and from the cerebral cortex. How the thalamus comes to reside within this region of the central nervous system has been the subject of much investigation. Extracellular signals secreted from key locations both extrinsic and intrinsic to the thalamic primordium have recently been identified and shown to play important roles in the growth, regionalization, and specification of thalamic progenitors. One factor in particular, the secreted morphogen Sonic hedgehog (Shh), has been implicated in spatiotemporal and threshold models of thalamic development that differ from other areas of the CNS due, in large part, to its expression within two signaling centers, the basal plate and the zona limitans intrathalamica, a dorsally projecting spike that separates the thalamus from the subthalamic region. Shh signaling from these dual sources exhibit unique and overlapping functions in the control of thalamic progenitor identity and nuclei specification. This review will highlight recent advances in our understanding of Shh function during thalamic development, revealing similarities, and differences that exist between species.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA