Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Ecotoxicol Environ Saf ; 260: 115080, 2023 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-37262967

RESUMEN

BACKGROUND: Glyphosate (GLY) is the most widely used herbicide in the world. Due to its mode of action as an inhibitor of the 5-enolpyruvylshikimate-3-phosphate synthase, an important step in the shikimate pathway, specifically in plants, GLY is considered to be of low toxicity to non-target organisms. However, various studies have shown the negative effects of GLY on the mortality and development of different non-target organisms, including insects, rodents, fish and amphibians. To better understand the various effects of GLY in more detail, we studied the effects of GLY without co-formulants during the embryogenesis of the aquatic model organism Xenopus laevis. RESULTS: A treatment with GLY affected various morphological endpoints in X. laevis tadpoles (body length, head width and area, eye area). Additionally, GLY interfered with the mobility as well as the neural and cardiac development of the embryos at stage 44/45. We were able to detect detailed structural changes in the cranial nerves and the heart and gained insights into the negative effects of GLY on cardiomyocyte differentiation. CONCLUSION: The application of GLY without co-formulants resulted in negative effects on several endpoints in the early embryonic development of X. laevis at concentrations that are environmentally relevant and concentrations that reflect the worst-case scenarios. This indicates that GLY could have a strong negative impact on the survival and lives of amphibians in natural waters. As a result, future GLY approvals should consider its impact on the environment.


Asunto(s)
Desarrollo Embrionario , Animales , Xenopus laevis/fisiología , Sudáfrica , Glifosato
2.
Hum Mutat ; 43(12): 1866-1871, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36116039

RESUMEN

Galloway-Mowat syndrome (GAMOS) is a very rare condition characterized by early-onset nephrotic syndrome and microcephaly with variable neurologic features. While considerable genetic heterogeneity of GAMOS has been identified, the majority of cases are caused by pathogenic variants in genes encoding the four components of the Kinase, endopeptidase, and other proteins of small size (KEOPS) complex, one of which is TP53RK. Here we describe a 3-year-old male with progressive microcephaly, neurodevelopmental deficits, and glomerular proteinuria. He was found to carry a novel homozygous TP53RK missense variant, c.163C>G (p.Arg55Gly), which was considered as potentially disease-causing. We generated a morpholino tp53rk knockdown model in Xenopus laevis showing that the depletion of endogenous Tp53rk caused abnormal eye and head development. This phenotype could be rescued by the expression of human wildtype TP53RK but not by the c.163C>G mutant nor by another previously described GAMOS-associated mutant c.125G>A (p.Gly42Asp). These findings support the pathogenic role of the novel TP53RK variant.


Asunto(s)
Hernia Hiatal , Microcefalia , Nefrosis , Síndrome Nefrótico , Masculino , Humanos , Preescolar , Microcefalia/genética , Mutación , Nefrosis/genética , Hernia Hiatal/genética , Síndrome Nefrótico/genética
3.
Am J Hum Genet ; 104(2): 246-259, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30661772

RESUMEN

SOX4, together with SOX11 and SOX12, forms group C of SRY-related (SOX) transcription factors. They play key roles, often in redundancy, in multiple developmental pathways, including neurogenesis and skeletogenesis. De novo SOX11 heterozygous mutations have been shown to cause intellectual disability, growth deficiency, and dysmorphic features compatible with mild Coffin-Siris syndrome. Using trio-based exome sequencing, we here identify de novo SOX4 heterozygous missense variants in four children who share developmental delay, intellectual disability, and mild facial and digital morphological abnormalities. SOX4 is highly expressed in areas of active neurogenesis in human fetuses, and sox4 knockdown in Xenopus embryos diminishes brain and whole-body size. The SOX4 variants cluster in the highly conserved, SOX family-specific HMG domain, but each alters a different residue. In silico tools predict that each variant affects a distinct structural feature of this DNA-binding domain, and functional assays demonstrate that these SOX4 proteins carrying these variants are unable to bind DNA in vitro and transactivate SOX reporter genes in cultured cells. These variants are not found in the gnomAD database of individuals with presumably normal development, but 12 other SOX4 HMG-domain missense variants are recorded and all demonstrate partial to full activity in the reporter assay. Taken together, these findings point to specific SOX4 HMG-domain missense variants as the cause of a characteristic human neurodevelopmental disorder associated with mild facial and digital dysmorphism.


Asunto(s)
Anomalías Múltiples/genética , Mutación Missense/genética , Trastornos del Neurodesarrollo/genética , Factores de Transcripción SOXC/genética , Secuencia de Aminoácidos , Animales , Niño , Preescolar , Síndrome de Coffin-Lowry/genética , Estudios de Cohortes , Secuencia Conservada , ADN/genética , ADN/metabolismo , Femenino , Dominios HMG-Box/genética , Heterocigoto , Humanos , Masculino , Factores de Transcripción SOX/química , Factores de Transcripción SOX/genética , Factores de Transcripción SOXC/química , Factores de Transcripción SOXC/metabolismo , Activación Transcripcional , Xenopus/anatomía & histología , Xenopus/embriología , Xenopus/genética , Proteínas de Xenopus/química , Proteínas de Xenopus/genética
4.
J Am Soc Nephrol ; 32(3): 580-596, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33593823

RESUMEN

BACKGROUND: Galloway-Mowat syndrome (GAMOS) is characterized by neurodevelopmental defects and a progressive nephropathy, which typically manifests as steroid-resistant nephrotic syndrome. The prognosis of GAMOS is poor, and the majority of children progress to renal failure. The discovery of monogenic causes of GAMOS has uncovered molecular pathways involved in the pathogenesis of disease. METHODS: Homozygosity mapping, whole-exome sequencing, and linkage analysis were used to identify mutations in four families with a GAMOS-like phenotype, and high-throughput PCR technology was applied to 91 individuals with GAMOS and 816 individuals with isolated nephrotic syndrome. In vitro and in vivo studies determined the functional significance of the mutations identified. RESULTS: Three biallelic variants of the transcriptional regulator PRDM15 were detected in six families with proteinuric kidney disease. Four families with a variant in the protein's zinc-finger (ZNF) domain have additional GAMOS-like features, including brain anomalies, cardiac defects, and skeletal defects. All variants destabilize the PRDM15 protein, and the ZNF variant additionally interferes with transcriptional activation. Morpholino oligonucleotide-mediated knockdown of Prdm15 in Xenopus embryos disrupted pronephric development. Human wild-type PRDM15 RNA rescued the disruption, but the three PRDM15 variants did not. Finally, CRISPR-mediated knockout of PRDM15 in human podocytes led to dysregulation of several renal developmental genes. CONCLUSIONS: Variants in PRDM15 can cause either isolated nephrotic syndrome or a GAMOS-type syndrome on an allelic basis. PRDM15 regulates multiple developmental kidney genes, and is likely to play an essential role in renal development in humans.


Asunto(s)
Proteínas de Unión al ADN/genética , Hernia Hiatal/genética , Microcefalia/genética , Mutación Missense , Nefrosis/genética , Factores de Transcripción/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Línea Celular , Preescolar , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/deficiencia , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Recién Nacido , Masculino , Modelos Moleculares , Síndrome Nefrótico/genética , Podocitos/metabolismo , Polimorfismo de Nucleótido Simple , Pronefro/embriología , Pronefro/metabolismo , Estabilidad Proteica , Factores de Transcripción/química , Factores de Transcripción/deficiencia , Xenopus laevis/embriología , Xenopus laevis/genética , Dedos de Zinc/genética
5.
Dev Dyn ; 250(8): 1096-1112, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33570783

RESUMEN

BACKGROUND: Retinol binding protein 1 (Rbp1) acts as an intracellular regulator of vitamin A metabolism and retinoid transport. In mice, Rbp1 deficiency decreases the capacity of hepatic stellate cells to take up all-trans retinol and sustain retinyl ester stores. Furthermore, Rbp1 is crucial for visual capacity. Although the function of Rbp1 has been studied in the mature eye, its role during early anterior neural development has not yet been investigated in detail. RESULTS: We showed that rbp1 is expressed in the eye, anterior neural crest cells (NCCs) and prosencephalon of the South African clawed frog Xenopus laevis. Rbp1 knockdown led to defects in eye formation, including microphthalmia and disorganized retinal lamination, and to disturbed induction and differentiation of the eye field, as shown by decreased rax and pax6 expression. Furthermore, it resulted in reduced rax expression in the prosencephalon and affected cranial cartilage. Rbp1 inhibition also interfered with neural crest induction and migration, as shown by twist and slug. Moreover, it led to a significant reduction of the all-trans retinoic acid target gene pitx2 in NCC-derived periocular mesenchyme. The Rbp1 knockdown phenotypes were rescued by pitx2 RNA co-injection. CONCLUSION: Rbp1 is crucial for the development of the anterior neural tissue.


Asunto(s)
Desarrollo Embrionario/fisiología , Cresta Neural/metabolismo , Prosencéfalo/metabolismo , Proteínas Celulares de Unión al Retinol/genética , Transducción de Señal/fisiología , Tretinoina/metabolismo , Animales , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Factor de Transcripción PAX6/genética , Factor de Transcripción PAX6/metabolismo , Proteínas Celulares de Unión al Retinol/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis
6.
Dev Biol ; 449(1): 1-13, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30797757

RESUMEN

Wnt proteins can activate different intracellular signaling pathways. These pathways need to be tightly regulated for proper cardiogenesis. The canonical Wnt/ß-catenin inhibitor Dkk1 has been shown to be sufficient to trigger cardiogenesis in gain-of-function experiments performed in multiple model systems. Loss-of-function studies however did not reveal any fundamental function for Dkk1 during cardiogenesis. Using Xenopus laevis as a model we here show for the first time that Dkk1 is required for proper differentiation of cardiomyocytes, whereas specification of cardiomyocytes remains unaffected in absence of Dkk1. This effect is at least in part mediated through regulation of non-canonical Wnt signaling via Wnt11. In line with these observations we also found that Isl1, a critical regulator for specification of the common cardiac progenitor cell (CPC) population, acts upstream of Dkk1.


Asunto(s)
Diferenciación Celular , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Miocardio/citología , Vía de Señalización Wnt , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Xenopus laevis/metabolismo , Animales , Biomarcadores/metabolismo , Tipificación del Cuerpo , Sistema Digestivo/embriología , Sistema Digestivo/metabolismo , Regulación hacia Abajo/genética , Embrión no Mamífero/metabolismo , Endodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas con Homeodominio LIM/metabolismo , Mesodermo/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Organogénesis/genética , Factores de Transcripción/metabolismo , Proteínas Wnt/metabolismo
7.
Development ; 144(2): 321-333, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27993984

RESUMEN

The signal-induced proliferation-associated family of proteins comprises four members, SIPA1 and SIPA1L1-3. Mutations of the human SIPA1L3 gene result in congenital cataracts. In Xenopus, loss of Sipa1l3 function led to a severe eye phenotype that was distinguished by smaller eyes and lenses including lens fiber cell maturation defects. We found a direct interaction between Sipa1l3 and Epha4, building a functional platform for proper ocular development. Epha4 deficiency phenocopied loss of Sipa1l3 and rescue experiments demonstrated that Epha4 acts upstream of Sipa1l3 during eye development, with both Sipa1l3 and Epha4 required for early eye specification. The ocular phenotype, upon loss of either Epha4 or Sipa1l3, was partially mediated by rax We demonstrate that canonical Wnt signaling is inhibited downstream of Epha4 and Sipa1l3 during normal eye development. Depletion of either Sipa1l3 or Epha4 resulted in an upregulation of axin2 expression, a direct Wnt/ß-catenin target gene. In line with this, Sipa1l3 or Epha4 depletion could be rescued by blocking Wnt/ß-catenin or activating non-canonical Wnt signaling. We therefore conclude that this pathomechanism prevents proper eye development and maturation of lens fiber cells, resulting in congenital cataracts.


Asunto(s)
Ojo/embriología , Proteínas Activadoras de GTPasa/fisiología , Cristalino/embriología , Cristalino/crecimiento & desarrollo , Receptor EphA4/fisiología , Vía de Señalización Wnt/fisiología , Animales , Animales Modificados Genéticamente , Catarata/genética , Diferenciación Celular/genética , Embrión no Mamífero , Ojo/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Cristalino/metabolismo , Organogénesis/genética , Unión Proteica , Receptor EphA4/metabolismo , Xenopus/embriología , Xenopus/genética
8.
Med Teach ; 41(11): 1245-1251, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31282246

RESUMEN

Purpose: Basic research about test-enhanced learning points towards its effectiveness to improve students' learning and is still underutilized in educational practice. Therefore, we developed an evidence-based instructional design to investigate the usefulness of test-enhanced learning within a flipped/inverted classroom approach. Materials and Methods: We developed two modes of learning material for the self-study phase of a flipped classroom for 139 students: in addition to educational films, one group of students received a reader and another group received multiple-choice questions that corresponded to the reader in content and length. An assessment of the content of the learning material was conducted at the subsequent on-site phase. Also, ratings about students' perceptions of the additional learning material were gathered. Results: At the assessment, students that prepared with films and multiple-choice questions outperformed students that prepared with films and the reader. Furthermore, students perceived the multiple-choice questions as more helpful, more motivating and felt better prepared for the assessment than students that used the reader. Conclusions: This study shows that test-enhanced learning can be utilized to promote students' learning within the self-study phase of a flipped classroom. Not only assessment scores are positively affected but also the motivation to learn and preparedness towards an assessment.


Asunto(s)
Bioquímica/educación , Educación Médica/métodos , Evaluación Educacional/métodos , Modelos Educacionales , Aprendizaje Basado en Problemas/métodos , Curriculum , Evaluación Educacional/normas , Humanos , Estudiantes de Medicina , Adulto Joven
9.
BMC Med Educ ; 19(1): 64, 2019 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-30819178

RESUMEN

BACKGROUND: The inverted classroom approach is characterized by a primary self-study phase for students followed by an on-site, face-to-face teaching phase that is used to deepen the prior acquired knowledge. Obviously, this teaching approach relies on the students preparing before the on-site phase, which in turn requires optimized preparatory material as well as defined working instructions. The major aim of this study, therefore, was to investigate the effect of different preparatory materials and working instructions for the self-study phase of an e-learning-based inverted classroom on the knowledge gained by medical students in biochemistry. Furthermore, we analyzed whether collaborative dyadic learning during the self-study phase is more effective than individual learning with respect to knowledge gain. METHODS: The study was performed in a biochemistry seminar for second semester medical students at Ulm University in Germany. This seminar was held using an e-learning-based inverted classroom. A total of 196 students were divided into three homogeneous study groups that differed in terms of the working material and instructions provided for the self-study phase. Knowledge gain was measured by formative tests at the beginning of the on-site phases. Questionnaires were also handed out asking about motivation, interest and learning time in the self-study phases. RESULTS: Students who were told to prepare in collaborating dyads during the self-study phase performed better in formative tests taken at the beginning of on-site phases than learners who were told to prepare individually. The study material that was provided was of minor importance for the differences in formative testing since almost all students prepared for the on-site phases. With the dyadic learning approach, both students benefited from this collaboration, characterized by a higher motivation and interest in the topic, as well as a longer time spent on task. CONCLUSION: Our study provides strong evidence that the study material, but more importantly the instructions provided for the self-study phase, affect students` knowledge gain in an e-learning-based inverted classroom. The instructed collaboratively working group was the most successful.


Asunto(s)
Bioquímica/educación , Instrucción por Computador/métodos , Prácticas Interdisciplinarias/métodos , Estudiantes de Medicina , Actitud del Personal de Salud , Conducta Cooperativa , Curriculum , Alemania , Humanos , Modelos Teóricos , Encuestas y Cuestionarios
10.
Dev Dyn ; 247(9): 1070-1082, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30055071

RESUMEN

BACKGROUND: The nitric oxide synthase interacting protein (Nosip) has been associated with diverse human diseases including psychological disorders. In line, early neurogenesis of mouse and Xenopus is impaired upon Nosip deficiency. Nosip knockout mice show craniofacial defects and the down-regulation of Nosip in the mouse and Xenopus leads to microcephaly. Until now, the exact underlying molecular mechanisms of these malformations were still unknown. RESULTS: Here, we show that nosip is expressed in the developing ocular system as well as the anterior neural crest cells of Xenopus laevis. Furthermore, Nosip inhibition causes severe defects in eye formation in the mouse and Xenopus. Retinal lamination as well as dorso-ventral patterning of the retina were affected in Nosip-depleted Xenopus embryos. Marker gene analysis using rax, pax6 and otx2 reveals an interference with the eye field induction and differentiation. A closer look on Nosip-deficient Xenopus embryos furthermore reveals disrupted cranial cartilage structures and an inhibition of anterior neural crest cell induction and migration shown by twist, snai2, and egr2. Moreover, foxc1 as downstream factor of retinoic acid signalling is affected upon Nosip deficiency. CONCLUSIONS: Nosip is a crucial factor for the development of anterior neural tissue such the eyes and neural crest cells. Developmental Dynamics 247:1070-1082, 2018. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Ojo/crecimiento & desarrollo , Cresta Neural/crecimiento & desarrollo , Ubiquitina-Proteína Ligasas/genética , Proteínas de Xenopus/genética , Xenopus laevis/crecimiento & desarrollo , Animales , Cartílago/embriología , Cartílago/crecimiento & desarrollo , Embrión no Mamífero , Desarrollo Embrionario , Ojo/embriología , Técnicas de Silenciamiento del Gen , Ratones , Cresta Neural/embriología , Neurogénesis , Cráneo , Xenopus laevis/embriología
11.
Dev Biol ; 423(1): 66-76, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28104388

RESUMEN

The Fezzin family member Nedd4-binding protein 3 (N4BP3) is known to regulate axonal and dendritic branching. Here, we show that n4bp3 is expressed in the neural tissue of the early Xenopus laevis embryo including the eye, the brain and neural crest cells. Knockdown of N4bp3 in the Xenopus anterior neural tissue results in severe developmental impairment of the eye, the brain and neural crest derived cranial cartilage structures. Moreover, we demonstrate that N4bp3 depletion leads to a significant reduction of both eye and brain specific marker genes and reduced neural crest cell migration. Finally, we demonstrate an impact of N4bp3 deficiency on cell apoptosis and proliferation. Our studies indicate that N4bp3 is required for early anterior neural development of vertebrates. This is in line with a study implicating that genetic disruption of N4BP3 in humans might be related to neurodevelopmental disease.


Asunto(s)
Proteínas Portadoras/metabolismo , Sistema Nervioso/embriología , Sistema Nervioso/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Xenopus laevis/metabolismo , Animales , Apoptosis , Biomarcadores/metabolismo , Encéfalo/embriología , Encéfalo/metabolismo , Proteínas Portadoras/genética , Cartílago/embriología , Cartílago/metabolismo , Movimiento Celular/genética , Proliferación Celular , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Desarrollo Embrionario/genética , Ojo/embriología , Ojo/metabolismo , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Etiquetado Corte-Fin in Situ , Cresta Neural/citología , Coloración y Etiquetado , Proteínas de Xenopus/genética
12.
Dev Biol ; 424(1): 28-39, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28237811

RESUMEN

Wnt proteins are critical for embryonic cardiogenesis and cardiomyogenesis by regulating different intracellular signalling pathways. Whereas canonical Wnt/ß-catenin signalling is required for mesoderm induction and proliferation of cardiac progenitor cells, ß-catenin independent, non-canonical Wnt signalling regulates cardiac specification and terminal differentiation. Although the diverse cardiac malformations associated with the loss of non-canonical Wnt11 in mice such as outflow tract (OFT) defects, reduced ventricular trabeculation, myofibrillar disorganization and reduced cardiac marker gene expression are well described, the underlying molecular mechanisms are still not completely understood. Here we aimed to further characterize Wnt11 mediated signal transduction during vertebrate cardiogenesis. Using Xenopus as a model system, we show by loss of function and corresponding rescue experiments that the non-canonical Wnt signalling mediator Rcsd1 is required downstream of Wnt11 for ventricular trabeculation, terminal differentiation of cardiomyocytes and cardiac morphogenesis. We here place Rcsd1 downstream of Wnt11 during cardiac development thereby providing a novel mechanism for how non-canonical Wnt signalling regulates vertebrate cardiogenesis.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Miocardio/metabolismo , Organogénesis , Proteínas Wnt/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Secuencias de Aminoácidos , Animales , Diferenciación Celular , Citoplasma/metabolismo , Desarrollo Embrionario , Eliminación de Gen , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Ratones , Miocardio/patología , Células 3T3 NIH , Fenotipo , Unión Proteica , Transducción de Señal , Proteínas de Xenopus/química
13.
Dev Biol ; 426(1): 69-83, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28427856

RESUMEN

Formation of a functional eye during vertebrate embryogenesis requires different processes such as cell differentiation, cell migration, cell-cell interactions as well as intracellular signalling processes. It was previously shown that the non-canonical Wnt receptor Frizzled 3 (Fzd3) is required for proper eye formation, however, the underlying mechanism is poorly understood. Here we demonstrate that loss of Fzd3 induces severe malformations of the developing eye and that this defect is phenocopied by loss of the activated leukocyte cell adhesion molecule (Alcam). Promoter analysis revealed the presence of a Fzd3 responsive element within the alcam promoter, which is responsible for alcam expression during anterior neural development. In-depth analysis identified the jun N-terminal protein kinase 1 (JNK1) and the transcription factor paired box 2 (Pax2) to be important for the activation of alcam expression. Altogether our study reveals that alcam is activated through non-canonical Wnt signalling during embryonic eye development in Xenopus laevis and shows that this pathway plays a similar role in different tissues.


Asunto(s)
Molécula de Adhesión Celular del Leucocito Activado/genética , Ojo/embriología , Receptores Frizzled/genética , Proteínas de Xenopus/genética , Xenopus laevis/embriología , Molécula de Adhesión Celular del Leucocito Activado/metabolismo , Animales , Adhesión Celular/fisiología , Comunicación Celular/fisiología , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Ojo/ultraestructura , Receptores Frizzled/metabolismo , Técnicas de Inactivación de Genes , Microscopía Electrónica de Transmisión , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Morfolinos/genética , Neurogénesis/genética , Neurogénesis/fisiología , Factor de Transcripción PAX2/metabolismo , Regiones Promotoras Genéticas/genética , Vía de Señalización Wnt , Proteínas de Xenopus/metabolismo
14.
Dev Biol ; 429(1): 200-212, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28663132

RESUMEN

BACKGROUND: Genetic deletion of Nosip in mice causes holoprosencephaly, however, the function of Nosip in neurogenesis is currently unknown. RESULTS: We combined two vertebrate model organisms, the mouse and the South African clawed frog, Xenopus laevis, to study the function of Nosip in neurogenesis. We found, that size and cortical thickness of the developing brain of Nosip knockout mice were reduced. Accordingly, the formation of postmitotic neurons was greatly diminished, concomitant with a reduced number of apical and basal neural progenitor cells in vivo. Neurospheres derived from Nosip knockout embryos exhibited reduced growth and the differentiation capability into neurons in vitro was almost completely abolished. Mass spectrometry analysis of the neurospheres proteome revealed a reduced expression of Rbp1, a regulator of retinoic acid synthesis, when Nosip was absent. We identified the homologous nosip gene to be expressed in differentiated neurons in the developing brain of Xenopus embryos. Knockdown of Nosip in Xenopus resulted in a reduction of brain size that could be rescued by reintroducing human NOSIP mRNA. Furthermore, the expression of pro-neurogenic transcription factors was reduced and the differentiation of neuronal cells was impaired upon Nosip knockdown. In Xenopus as well as in mouse we identified reduced proliferation and increased apoptosis as underlying cause of microcephaly upon Nosip depletion. In Xenopus Nosip and Rbp1 are similarly expressed and knockdown of Nosip resulted in down regulation of Rbp1. Knockdown of Rbp1 caused a similar microcephaly phenotype as the depletion of Nosip and synergy experiments indicated that both proteins act in the same signalling pathway. CONCLUSIONS: Nosip is a novel factor critical for neural stem cell/progenitor self-renewal and neurogenesis during mouse and Xenopus development and functions upstream of Rbp1 during early neurogenesis.


Asunto(s)
Neurogénesis , Ubiquitina-Proteína Ligasas/deficiencia , Proteínas de Xenopus/deficiencia , Xenopus laevis/embriología , Xenopus laevis/metabolismo , Animales , Apoptosis , Proliferación Celular , Separación Celular , Supervivencia Celular , Corteza Cerebral/embriología , Corteza Cerebral/patología , Regulación hacia Abajo , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ratones Noqueados , Microcefalia/patología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Proteoma/metabolismo , Proteínas Celulares de Unión al Retinol/metabolismo , Esferoides Celulares/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
15.
Nucleic Acids Res ; 44(10): 4703-20, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-26912830

RESUMEN

The transcriptional shift from repression to activation of target genes is crucial for the fidelity of Notch responses through incompletely understood mechanisms that likely involve chromatin-based control. To activate silenced genes, repressive chromatin marks are removed and active marks must be acquired. Histone H3 lysine-4 (H3K4) demethylases are key chromatin modifiers that establish the repressive chromatin state at Notch target genes. However, the counteracting histone methyltransferase required for the active chromatin state remained elusive. Here, we show that the RBP-J interacting factor SHARP is not only able to interact with the NCoR corepressor complex, but also with the H3K4 methyltransferase KMT2D coactivator complex. KMT2D and NCoR compete for the C-terminal SPOC-domain of SHARP. We reveal that the SPOC-domain exclusively binds to phosphorylated NCoR. The balance between NCoR and KMT2D binding is shifted upon mutating the phosphorylation sites of NCoR or upon inhibition of the NCoR kinase CK2ß. Furthermore, we show that the homologs of SHARP and KMT2D in Drosophila also physically interact and control Notch-mediated functions in vivo Together, our findings reveal how signaling can fine-tune a committed chromatin state by phosphorylation of a pivotal chromatin-modifier.


Asunto(s)
Cromatina/metabolismo , Proteínas Co-Represoras/metabolismo , Regulación de la Expresión Génica , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas Nucleares/metabolismo , Receptores Notch/metabolismo , Transcripción Genética , Animales , Quinasa de la Caseína II/metabolismo , Línea Celular , Línea Celular Tumoral , Proteínas de Unión al ADN , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Código de Histonas , N-Metiltransferasa de Histona-Lisina , Proteínas de Homeodominio/química , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Proteínas Nucleares/química , Fosforilación , Dominios y Motivos de Interacción de Proteínas , Proteínas de Unión al ARN , Xenopus laevis
17.
Development ; 141(10): 2064-74, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24764076

RESUMEN

Proper development of nephrons is essential for kidney function. ß-Catenin-independent Wnt signaling through Fzd8, Inversin, Daam1, RhoA and Myosin is required for nephric tubule morphogenesis. Here, we provide a novel mechanism through which non-canonical Wnt signaling contributes to tubular development. Using Xenopus laevis as a model system, we found that the cell-adhesion molecule Alcam is required for proper nephrogenesis and functions downstream of Fzd3 during embryonic kidney development. We found alcam expression to be independent of Fzd8 or Inversin, but to be transcriptionally regulated by the ß-Catenin-independent Wnt/JNK pathway involving ATF2 and Pax2 in a direct manner. These novel findings indicate that several branches of Wnt signaling are independently required for proximal tubule development. Moreover, our data indicate that regulation of morphogenesis by non-canonical Wnt ligands also involves direct transcriptional responses in addition to the effects on a post-translational level.


Asunto(s)
Molécula de Adhesión Celular del Leucocito Activado/fisiología , Desarrollo Embrionario/genética , Riñón/embriología , Sistema de Señalización de MAP Quinasas/genética , Vía de Señalización Wnt/genética , Molécula de Adhesión Celular del Leucocito Activado/genética , Animales , Embrión no Mamífero , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Regulación del Desarrollo de la Expresión Génica , Riñón/metabolismo , Factor de Transcripción PAX2/fisiología , Pronefro/embriología , Pronefro/metabolismo , Elementos de Respuesta/genética , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Xenopus laevis/genética
18.
J Med Genet ; 53(3): 152-62, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26543203

RESUMEN

BACKGROUND: SOX11 is a transcription factor proposed to play a role in brain development. The relevance of SOX11 to human developmental disorders was suggested by a recent report of SOX11 mutations in two patients with Coffin-Siris syndrome. Here we further investigate the role of SOX11 variants in neurodevelopmental disorders. METHODS: We used array based comparative genomic hybridisation and trio exome sequencing to identify children with intellectual disability who have deletions or de novo point mutations disrupting SOX11. The pathogenicity of the SOX11 mutations was assessed using an in vitro gene expression reporter system. Loss-of-function experiments were performed in xenopus by knockdown of Sox11 expression. RESULTS: We identified seven individuals with chromosome 2p25 deletions involving SOX11. Trio exome sequencing identified three de novo SOX11 variants, two missense (p.K50N; p.P120H) and one nonsense (p.C29*). The biological consequences of the missense mutations were assessed using an in vitro gene expression system. These individuals had microcephaly, developmental delay and shared dysmorphic features compatible with mild Coffin-Siris syndrome. To further investigate the function of SOX11, we knocked down the orthologous gene in xenopus. Morphants had significant reduction in head size compared with controls. This suggests that SOX11 loss of function can be associated with microcephaly. CONCLUSIONS: We thus propose that SOX11 deletion or mutation can present with a Coffin-Siris phenotype.


Asunto(s)
Anomalías Múltiples/genética , Cara/anomalías , Deformidades Congénitas de la Mano/genética , Discapacidad Intelectual/genética , Micrognatismo/genética , Cuello/anomalías , Trastornos del Neurodesarrollo/genética , Factores de Transcripción SOXC/genética , Eliminación de Secuencia , Anomalías Múltiples/fisiopatología , Adolescente , Adulto , Animales , Niño , Preescolar , Cara/fisiopatología , Femenino , Técnicas de Silenciamiento del Gen , Deformidades Congénitas de la Mano/fisiopatología , Humanos , Discapacidad Intelectual/fisiopatología , Masculino , Microcefalia , Micrognatismo/fisiopatología , Cuello/fisiopatología , Trastornos del Neurodesarrollo/fisiopatología , Xenopus
19.
J Neurochem ; 136(1): 28-35, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26364583

RESUMEN

Rap GTPase-activating proteins (RapGAPs) are essential for synaptic function as they tightly regulate synaptic Rap signaling. Among the most abundant synaptic RapGAPs in brain are the Spine-associated RapGAPs (SPARs) Sipa1l1/SPAR and Sipa1l2/SPAR2, whereas nothing has been reported on Sipa1l3/SPAR3. In this study, we show that Sipa1l3/SPAR3 is conserved across species, has a distinct expression pattern in the developing rat brain and is localized at excitatory postsynapses. We further demonstrate that the Sipa1l3/SPAR3 C-terminus is required for postsynaptic targeting and represents an interaction module for Fezzins such as ProSAPiP1/Lzts3, a binding partner of the postsynaptic scaffold protein Shank3. Taken together, our data imply that Sipa1l3/SPAR3 is a hitherto unknown synaptic RapGAP, which is targeted to postsynaptic specializations and interacts with Fezzins. Spine-associated RapGAPs (SPARs) are essential modulators of synaptic signaling. Our study is the first to characterize the SPAR family member Sipa1l3/SPAR3 in neuronal tissue. We show that Sipa1l3/SPAR3 is conserved across species, has a distinct expression pattern in brain and is localized to excitatory postsynapses via its C-terminus, which represents an interaction module for other postsynaptic proteins including the Fezzin ProSAPiP1/Lzts3.


Asunto(s)
Proteínas Portadoras/biosíntesis , Proteínas Activadoras de GTPasa/biosíntesis , Proteínas de la Membrana/biosíntesis , Sinapsis/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Animales , Encéfalo/metabolismo , Células COS , Células Cultivadas , Chlorocebus aethiops , Perros , Femenino , Humanos , Masculino , Ratones , Pan troglodytes , Unión Proteica/fisiología , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie
20.
Dev Genes Evol ; 226(5): 369-82, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27384056

RESUMEN

The signal-induced proliferation-associated (SIPA) protein family belongs to the RapGAP protein superfamily. Previous studies mainly focused on the expression and function of SIPA genes in vertebrate neuronal tissue. Only limited data about the embryonic expression pattern of the genes are currently available. Our study provides the first expression analysis of sipa1, sipa1l1, sipa1l2, and sipa1l3 during early development of the vertebrate organism Xenopus laevis. In silico, analysis revealed that all genes are highly conserved across species. Semi-quantitative RT-PCR experiments demonstrated that the RNA of all genes was maternally supplied. By whole mount in situ hybridization approaches, we showed that sipa1 is mainly expressed in various sensory organs, the respiratory and blood system, heart, neural tube, and eye. In contrast, sipa1l1 showed a broad expression during development in particular within the brain, somites, eye, and heart. Sipa1l2 was detected in the branchial arches, glomerulus, and the developing eye. In contrast, sipa1l3 revealed a tissue specific expression within the olfactory and otic vesicles, the cranial placodes and ganglia, neural tube, pronephros, retina, and lens. In summary, all sipa gene family members are expressed throughout the whole developing Xenopus organism and might play an important role during vertebrate early embryogenesis.


Asunto(s)
Proteínas Nucleares/genética , Xenopus laevis/crecimiento & desarrollo , Xenopus laevis/genética , Animales , Embrión no Mamífero/metabolismo , Desarrollo Embrionario , Dominios Proteicos , Proteínas de Xenopus/genética , Xenopus laevis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA