Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Molecules ; 29(10)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38792190

RESUMEN

As a conformationally restricted amino acid, hydroxy-l-proline is a versatile scaffold for the synthesis of diverse multi-functionalized pyrrolidines for probing the ligand binding sites of biological targets. With the goal to develop new inhibitors of the widely expressed amino acid transporters SLC1A4 and SLC1A5 (also known as ASCT1 and ASCT2), we synthesized and functionally screened synthetic hydroxy-l-proline derivatives using electrophysiological and radiolabeled uptake methods against amino acid transporters from the SLC1, SLC7, and SLC38 solute carrier families. We have discovered a novel class of alkoxy hydroxy-pyrrolidine carboxylic acids (AHPCs) that act as selective high-affinity inhibitors of the SLC1 family neutral amino acid transporters SLC1A4 and SLC1A5. AHPCs were computationally docked into a homology model and assessed with respect to predicted molecular orientation and functional activity. The series of hydroxyproline analogs identified here represent promising new agents to pharmacologically modulate SLC1A4 and SLC1A5 amino acid exchangers which are implicated in numerous pathophysiological processes such as cancer and neurological diseases.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC , Antígenos de Histocompatibilidad Menor , Sistema de Transporte de Aminoácidos ASC/antagonistas & inhibidores , Sistema de Transporte de Aminoácidos ASC/metabolismo , Sistema de Transporte de Aminoácidos ASC/química , Antígenos de Histocompatibilidad Menor/metabolismo , Antígenos de Histocompatibilidad Menor/química , Humanos , Prolina/química , Prolina/análogos & derivados , Animales , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad , Células HEK293 , Pirrolidinas/química , Pirrolidinas/farmacología , Pirrolidinas/síntesis química , Descubrimiento de Drogas , Sistemas de Transporte de Aminoácidos Neutros/antagonistas & inhibidores , Sistemas de Transporte de Aminoácidos Neutros/química , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/genética
2.
Mamm Genome ; 34(4): 572-585, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37642681

RESUMEN

Solute carrier family 1 member 4 (SLC1A4), also referred to as Alanine/Serine/Cysteine/Threonine-preferring Transporter 1 (ASCT1), is a sodium-dependent neutral amino acid transporter. It is expressed in many tissues, including the brain, where it is expressed primarily on astrocytes and plays key roles in neuronal differentiation and development, maintaining neurotransmitter homeostasis, and N-methyl-D-aspartate neurotransmission, through regulation of L- and D-serine. Mutations in SLC1A4 are associated with the rare autosomal recessive neurodevelopmental disorder spastic tetraplegia, thin corpus callosum, and progressive microcephaly (SPATCCM, OMIM 616657). Psychomotor development and speech are significantly impaired in these patients, and many develop seizures. We generated and characterized a knock-in mouse model for the most common mutant allele, which results in a single amino acid change (p.Glu256Lys, or E256K). Homozygous mutants had increased D-serine uptake in the brain, microcephaly, and thin corpus callosum and cortex layer 1. While p.E256K homozygotes showed some significant differences in exploratory behavior relative to wildtype mice, their performance in assays for motor coordination, endurance, learning, and memory was normal, and they showed no significant differences in long-term potentiation. Taken together, these results indicate that the impact of the p.E256K mutation on cognition and motor function is minimal in mice, but other aspects of SLC1A4 function in the brain are conserved. Mice homozygous for p.E256K may be a good model for understanding the developmental basis of the corpus callosum and microcephaly phenotypes observed in SPATCCM patients and assessing whether they are rescued by serine supplementation.


Asunto(s)
Microcefalia , Humanos , Ratones , Animales , Microcefalia/genética , Microcefalia/complicaciones , Cuerpo Calloso/metabolismo , Encéfalo/metabolismo , Cuadriplejía/complicaciones , Serina
3.
Res Sq ; 2023 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-37162879

RESUMEN

SLC1A4 (solute carrier family 1 member 4, also referred to as ASCT1, Alanine/Serine/Cysteine/Threonine-preferring Transporter 1) is a sodium-dependent neutral amino acid transporter. It is highly expressed in many tissues, including the brain, where it is expressed primarily on astrocytes and plays key roles in neuronal differentiation and development, maintaining neurotransmitter homeostasis, and N-methyl-D-aspartate (NMDA) neurotransmission, through regulation of L- and D-serine. Mutations in SLC1A4 are associated with the rare autosomal recessive neurodevelopmental disorder spastic tetraplegia, thin corpus callosum, and progressive microcephaly (SPATCCM, OMIM 616657). Psychomotor development and speech are significantly impaired in these patients, and many develop seizures. We generated and characterized a knock-in mouse model for the most common mutant allele, which results in a single amino acid change (p.Glu256Lys, or E256K). Homozygous mutants had increased D-serine uptake in the brain, microcephaly, and thin corpus callosum and cortex layer 1. While p.E256K homozygotes showed some significant differences in exploratory behavior relative to wildtype mice, their performance in assays for motor coordination, endurance, learning, and memory was normal, and they showed no significant differences in long-term potentiation. Taken together, these results indicate that some aspects of SLC1A4 function in brain development are conserved between mice and humans, but the impact of the p.E256K mutation on cognition and motor function is minimal in mice.

4.
J Virol ; 97(2): e0167222, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36651748

RESUMEN

Phenotypic screening has yielded small-molecule inhibitors of prion replication that are effective in vivo against certain prion strains but not others. Here, we sought to test the small molecule anle138b in multiple mouse models of prion disease. In mice inoculated with the RML strain of prions, anle138b doubled survival and durably suppressed astrogliosis measured by live-animal bioluminescence imaging. In knock-in mouse models of the D178N and E200K mutations that cause genetic prion disease, however, we were unable to identify a clear, quantifiable disease endpoint against which to measure therapeutic efficacy. Among untreated animals, the mutations did not impact overall survival, and bioluminescence remained low out to >20 months of age. Vacuolization and PrP deposition were observed in some brain regions in a subset of mutant animals but appeared to be unable to carry the weight of a primary endpoint in a therapeutic study. We conclude that not all animal models of prion disease are suited to well-powered therapeutic efficacy studies, and care should be taken in choosing the models that will support drug development programs. IMPORTANCE There is an urgent need to develop drugs for prion disease, a currently untreatable neurodegenerative disease. In this effort, there is a debate over which animal models can best support a drug development program. While the study of prion disease benefits from excellent animal models because prions naturally afflict many different mammals, different models have different capabilities and limitations. Here, we conducted a therapeutic efficacy study of the drug candidate anle138b in mouse models with two of the most common mutations that cause genetic prion disease. In a more typical model where prions are injected directly into the brain, we found anle138b to be effective. In the genetic models, however, the animals never reached a clear, measurable point of disease onset. We conclude that not all prion disease animal models are ideally suited to drug efficacy studies, and well-defined, quantitative disease metrics should be a priority.


Asunto(s)
Enfermedades por Prión , Pirazoles , Animales , Ratones , Modelos Animales de Enfermedad , Ratones Transgénicos , Enfermedades por Prión/tratamiento farmacológico , Enfermedades por Prión/genética , Priones/genética , Pirazoles/uso terapéutico
5.
Mamm Genome ; 30(5-6): 151-165, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31089807

RESUMEN

Spongiform encephalopathy is an intriguing yet poorly understood neuropathology characterized by vacuoles, demyelination, and gliosis. It is observed in patients with prion disease, primary mitochondrial disease, HIV-1 infection of the brain, and some inherited disorders, but the underlying mechanism of disease remains unclear. The brains of mice lacking the MGRN1 E3 ubiquitin ligase develop vacuoles by 9 months of age. MGRN1-dependent ubiquitination has been reported to regulate mitofusin 1 and GP78, suggesting MGRN1 may have a direct effect on mitochondrial homeostasis. Here, we demonstrate that some MGRN1 localizes to mitochondria, most likely due to N-myristoylation, and mitochondria in cells from Mgrn1 null mutant mice display fragmentation and depolarization without recruitment of the parkin E3 ubiquitin ligase. The late onset of pathology in the brains of Mgrn1 null mutant mice suggests that a further, age-dependent effect on mitochondrial homeostasis may be required to trigger vacuolation. Parkin protein and mRNA levels showed a significant decline in the brains of Mgrn1 null mutant mice by 12 months of age. To test whether loss of parkin triggers vacuolation through a synergistic effect, we generated Mgrn1; parkin double mutant mice. By 1 month of age, their brains demonstrated more severe mitochondrial dysfunction than Mgrn1 null mutants, but there was no effect on the age-of-onset of spongiform neurodegeneration. Expression of the ATF4 transcription factor, a key regulator of the mitochondrial stress response, also declined in the brains of aged Mgrn1 null mutant mice. Together, the data presented here indicate that loss of MGRN1 has early, direct effects on mitochondrial homeostasis and late, indirect effects on the ability of cells to respond to mitochondrial stress.


Asunto(s)
Envejecimiento/genética , Mitocondrias/fisiología , Enfermedades Neurodegenerativas/genética , Ubiquitina-Proteína Ligasas/genética , Envejecimiento/patología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Expresión Génica , Homeostasis , Humanos , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/patología , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/metabolismo , Vacuolas/patología
6.
Mamm Genome ; 26(1-2): 80-93, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25399070

RESUMEN

Mice homozygous for the gray tremor (gt) mutation have a pleiotropic phenotype that includes pigmentation defects, megacolon, whole body tremors, sporadic seizures, hypo- and dys-myelination of the central nervous system (CNS) and peripheral nervous system, vacuolation of the CNS, and early death. Vacuolation similar to that caused by prions was originally reported to be transmissible, but subsequent studies showed the inherited disease was not infectious. The gt mutation mapped to distal mouse chromosome 15, to the same region as Sox10, which encodes a transcription factor with essential roles in neural crest survival and differentiation. As dominant mutations in mouse or human SOX10 cause white spotting and intestinal aganglionosis, we screened the Sox10 coding region for mutations in gt/gt DNA. An adenosine to guanine transversion was identified in exon 2 that changes a highly conserved glutamic acid residue in the SOX10 DNA binding domain to glycine. This mutant allele was not seen in wildtype mice, including the related GT/Le strain, and failed to complement a Sox10 null allele. Gene expression analysis revealed significant down-regulation of genes involved in myelin lipid biosynthesis pathways in gt/gt brains. Knockout mice for some of these genes develop CNS vacuolation and/or myelination defects, suggesting that their down-regulation may contribute to these phenotypes in gt mutants and could underlie the neurological phenotypes associated with peripheral demyelinating neuropathy-central dysmyelinating leukodystrophy-Waardenburg syndrome-Hirschsprung disease, caused by mutations in human SOX10.


Asunto(s)
Regulación de la Expresión Génica/genética , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/fisiopatología , Factores de Transcripción SOXE/metabolismo , Animales , Vías Biosintéticas/genética , Análisis Mutacional de ADN , Cartilla de ADN/genética , Galactósidos , Perfilación de la Expresión Génica , Humanos , Indoles , Ratones , Ratones Noqueados , Ratones Mutantes , Repeticiones de Microsatélite/genética , Mutación Missense/genética , Vaina de Mielina/metabolismo , Factores de Transcripción SOXE/genética
7.
PLoS One ; 8(1): e55575, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23383230

RESUMEN

Prion diseases are rare but invariably fatal neurodegenerative disorders. They are associated with spongiform encephalopathy, a histopathology characterized by the presence of large, membrane-bound vacuolar structures in the neuropil of the brain. While the primary cause is recognized as conversion of the normal form of prion protein (PrP(C)) to a conformationally distinct, pathogenic form (PrP(Sc)), the cellular pathways and mechanisms that lead to spongiform change, neuronal dysfunction and death are not known. Mice lacking the Mahogunin Ring Finger 1 (MGRN1) E3 ubiquitin ligase develop spongiform encephalopathy by 9 months of age but do not become ill. In cell culture, PrP aberrantly present in the cytosol was reported to interact with and sequester MGRN1. This caused endo-lysosomal trafficking defects similar to those observed when Mgrn1 expression is knocked down, implicating disrupted MGRN1-dependent trafficking in the pathogenesis of prion disease. As these defects were rescued by over-expression of MGRN1, we investigated whether reduced or elevated Mgrn1 expression influences the onset, progression or pathology of disease in mice inoculated with PrP(Sc). No differences were observed, indicating that disruption of MGRN1-dependent pathways does not play a significant role in the pathogenesis of transmissible spongiform encephalopathy.


Asunto(s)
Enfermedades por Prión/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Enfermedades por Prión/genética , Enfermedades por Prión/mortalidad , Priones/metabolismo , Ubiquitina-Proteína Ligasas/genética
8.
Pigment Cell Melanoma Res ; 26(2): 263-8, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23253940

RESUMEN

Mice lacking the E3 ubiquitin ligase mahogunin ring finger-1 (MGRN1) have a pleiotropic phenotype that includes spongiform neurodegeneration, embryonic patterning defects, and dark fur due to a defect in pigment-type switching. The only MGRN1 ubiquitination target identified to date is tumor susceptibility gene 101 (TSG101), a component of the endosomal trafficking machinery. Here, we show that MGRN1 also interacts with but does not ubiquitinate NEDD4, a HECT-domain ubiquitin ligase involved in endosomal trafficking. Using transgenesis in mice, we demonstrate that pigment-type switching likely requires MGRN1's ubiquitin ligase activity but not its ability to bind TSG101 or NEDD4. This indicates that MGRN1-dependent ubiquitination of an as-yet unidentified target protein is required for agouti-mediated melanocortin signaling.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Pigmentación , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Animales , Células HEK293 , Humanos , Ratones , Ratones Transgénicos , Ubiquitina-Proteína Ligasas Nedd4 , Unión Proteica , Pigmentación de la Piel , Transgenes/genética
9.
Birth Defects Res A Clin Mol Teratol ; 91(4): 204-17, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21472842

RESUMEN

BACKGROUND: Hypertrophic cardiomyopathy, characterized by thickened ventricular walls and reduced ventricular chamber volume, is a common cause of sudden cardiac death in young people. Most inherited forms result from mutations in genes encoding sarcomeric proteins. METHODS: Histologic analysis identified embryonic cardiac hypertrophy in dark-like mutant mice. BrdU analysis was performed to measure proliferation and cardiomyocytes were isolated to measure cell size. The dark-like mutation was identified by positional cloning. RESULTS: The dark-like mutation causes cardiomyocyte hypertrophy due to loss-of-function of peptidase d (Pepd), which encodes prolidase, a cytosolic enzyme that recycles proline for collagen re-synthesis. Prolidase deficiency is a rare autosomal recessive disease in humans with a broad phenotypic spectrum not reported to include heart defects, but a conserved role for prolidase in heart development was confirmed by morpholino knockdown in zebrafish. We tested the hypothesis that loss of prolidase function disrupts collagen-mediated integrin signaling and determined that the levels of several key integrin transducers were reduced in the hearts of dark-like mutant embryos. CONCLUSIONS: This work identifies dark-like mice as a model of prolidase deficiency that will be valuable for studying the role of proline metabolism in normal physiology and disease processes, and suggests that integrin signaling may regulate the onset of hypertrophic cardiac growth.


Asunto(s)
Cardiomegalia/genética , Cardiomegalia/fisiopatología , Mutación , Deficiencia de Prolidasa/genética , Animales , Cardiomegalia/embriología , Tamaño de la Célula , Clonación Molecular , Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Corazón/embriología , Corazón/fisiopatología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos CBA , Miocitos Cardíacos/patología , Fenotipo , Prolina/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo
11.
Nucleic Acids Res ; 37(8): 2699-711, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19273531

RESUMEN

Protein kinase CK2 is a serine/threonine kinase known to phosphorylate numerous substrates. CK2 is implicated in several physiologic and pathologic processes, particularly in cancer biology. CK2 is comprised of several subunits, including CK2alpha, CK2alpha' and CK2beta. Inactivation of CK2alpha' leads to chromatin degeneration of germ cells, resulting in male sterility. To identify additional targets of CK2alpha' in testes and to determine the role of CK2alpha' in germ cell nuclear integrity, GST pull-down and protein-protein interaction assays were conducted. A novel testis-specific gene, CKT2 (CK2 Target protein 2), was found whose product interacts with and is phosphorylated by CK2 in vitro and in vivo. CKT2 is a 30.2 kDa protein with one coiled-coil domain and six putative phosphorylation sites. High expression of CKT2 correlated with chromatin condensation of spermatids in murine testes. Findings reported herein demonstrate that CKT2 is a target protein of native CK2alpha' in testes and suggest that CKT2 plays a role in chromatin regulation of male germ cells.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Fosfoproteínas/metabolismo , Testículo/metabolismo , Animales , Línea Celular , Humanos , Masculino , Ratones , Proteínas Nucleares/metabolismo , Fosfoproteínas/química , Fosfoproteínas/genética , Fosforilación , Subunidades de Proteína/metabolismo , Espermátides/metabolismo , Testículo/enzimología , Técnicas del Sistema de Dos Híbridos
12.
Am J Hum Genet ; 80(4): 800-4, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17357085

RESUMEN

Branchio-oto-renal syndrome (BOR) is an autosomal dominant developmental disorder characterized by the association of branchial arch defects, hearing loss, and renal anomalies. Mutations in EYA1 are known to cause BOR. More recently, mutations in SIX1, which interacts with EYA1, were identified as an additional cause of BOR. A second member of the SIX family of proteins, unc-39 (SIX5), has also been reported to directly interact with eya-1 in Caenorhabditis elegans. We hypothesized that this interaction would be conserved in humans and that interactors of EYA1 represent good candidate genes for BOR. We therefore screened a cohort of 95 patients with BOR for mutations in SIX5. Four different heterozygous missense mutations were identified in five individuals. Functional analyses of these mutations demonstrated that two mutations affect EYA1-SIX5 binding and the ability of SIX5 or the EYA1-SIX5 complex to activate gene transcription. We thereby identified heterozygous mutations in SIX5 as a novel cause of BOR.


Asunto(s)
Síndrome Branquio Oto Renal/genética , Predisposición Genética a la Enfermedad , Proteínas de Homeodominio/genética , Mutación Missense/genética , Factores de Transcripción/genética , Secuencia de Bases , Pruebas Genéticas , Proteínas de Homeodominio/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Luciferasas , Datos de Secuencia Molecular , Proteínas Nucleares/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Factores de Transcripción/metabolismo
13.
Dev Biol ; 298(2): 430-41, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-16916509

RESUMEN

Members of the Eyes absent (Eya) gene family are important for auditory system development. While mutations in human EYA4 cause late-onset deafness at the DFNA10 locus, mutations in human EYA1 cause branchio-oto-renal (BOR) syndrome. Inactivation of Eya1 in mice causes an early arrest of the inner ear development at the otocyst stage. To better understand the role of Eya1 in inner ear development, we analyzed the cellular and molecular basis of the early defect observed in the Eya1 mutant embryos. We report here that Eya1-/- otic epithelium shows reduced cell proliferation from E8.5 and increased cell apoptosis from E9.0, thus providing insights into the cellular basis of inner ear defect which occurred in the absence of Eya1. Previous studies have suggested that Pax, Eya and Six genes function in a parallel or independent pathway during inner ear development. However, it remains unknown whether Pax genes interact with Eya1 or Six1 during inner ear morphogenesis. To further evaluate whether Pax genes function in the Eya1-Six1 pathway or whether they interact with Eya1 or Six1 during inner ear morphogenesis, we have analyzed the expression pattern of Eya1, Pax2 and Pax8 on adjacent sections of otic epithelium from E8.5 to 9.5 by in situ hybridization and the inner ear gross structures of Pax2, Eya1 and Six1 compound mutants at E17.5 by latex paintfilling. Our data strongly suggest that Pax2 interacts with Eya1 during inner ear morphogenesis, and this interaction is critical for the development of all sensory areas in the inner ear. Furthermore, otic marker analysis in both Eya1-/- and Pax2-/- embryos indicates that Eya1 but not Pax2 regulates the establishment of regional specification of the otic vesicle. Together, these results show that, while Eya1 exerts an early function essential for normal growth and patterning of the otic epithelium, it also functionally synergizes with Pax2 during the morphogenesis of all sensory areas of mammalian inner ear.


Asunto(s)
Oído Interno/embriología , Oído/embriología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas Nucleares/fisiología , Factor de Transcripción PAX2/metabolismo , Proteínas Tirosina Fosfatasas/fisiología , Animales , Tipificación del Cuerpo , Muerte Celular , Proliferación Celular , Oído/fisiología , Oído Interno/metabolismo , Oído Interno/fisiología , Conducto Endolinfático/anomalías , Epitelio/embriología , Epitelio/metabolismo , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Transgénicos , Morfogénesis , Proteínas Nucleares/genética , Factor de Transcripción PAX8 , Factores de Transcripción Paired Box/metabolismo , Proteínas Tirosina Fosfatasas/genética
14.
Dev Biol ; 293(2): 499-512, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16530750

RESUMEN

Previous studies have suggested a role of the homeodomain Six family proteins in patterning the developing vertebrate head that involves appropriate segmentation of three tissue layers, the endoderm, the paraxial mesoderm and the neural crest cells; however, the developmental programs and mechanisms by which the Six genes act in the pharyngeal endoderm remain largely unknown. Here, we examined their roles in pharyngeal pouch development. Six1-/- mice lack thymus and parathyroid and analysis of Six1-/- third pouch endoderm demonstrated that the patterning of the third pouch into thymus/parathyroid primordia is initiated. However, the endodermal cells of the thymus/parathyroid rudiments fail to maintain the expression of the parathyroid-specific gene Gcm2 and the thymus-specific gene Foxn1 and subsequently undergo abnormal apoptosis, leading to a complete disappearance of organ primordia by E12.5. This thus defines the thymus/parathyroid defects present in the Six1 mutant. Analyses of the thymus/parathyroid development in Six1-/-;Six4-/- double mutant show that both Six1 and Six4 act synergistically to control morphogenetic movements of early thymus/parathyroid tissues, and the threshold of Six1/Six4 appears to be crucial for the regulation of the organ primordia-specific gene expression. Previous studies in flies and mice suggested that Eya and Six genes may function downstream of Pax genes. Our data clearly show that Eya1 and Six1 expression in the pouches does not require Pax1/Pax9 function, suggesting that they may function independently from Pax1/Pax9. In contrast, Pax1 expression in all pharyngeal pouches requires both Eya1 and Six1 function. Moreover, we show that the expression of Tbx1, Fgf8 and Wnt5b in the pouch endoderm was normal in Six1-/- embryos and slightly reduced in Six1-/-;Six4-/- double mutant, but was largely reduced in Eya1-/- embryos. These results indicate that Eya1 appears to be upstream of very early events in the initiation of thymus/parathyroid organogenesis, while Six genes appear to act in an early differentiation step during thymus/parathyroid morphogenesis. Together, these analyses establish an essential role for Eya1 and Six genes in patterning the third pouch into organ-specific primordia.


Asunto(s)
Región Branquial/embriología , Proteínas de Homeodominio/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Nucleares/genética , Glándulas Paratiroides/embriología , Proteínas Tirosina Fosfatasas/genética , Timo/embriología , Transactivadores/genética , Animales , Tipificación del Cuerpo/genética , Región Branquial/metabolismo , Factor 8 de Crecimiento de Fibroblastos/genética , Factores de Transcripción Forkhead/genética , Edad Gestacional , Glicoproteínas/genética , Proteínas de Homeodominio/metabolismo , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Noqueados , Ratones Mutantes , Modelos Biológicos , Proteínas Nucleares/deficiencia , Proteínas Nucleares/metabolismo , Factores de Transcripción Paired Box/genética , Glándulas Paratiroides/metabolismo , Proteínas Tirosina Fosfatasas/deficiencia , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas de Dominio T Box/genética , Timo/metabolismo , Transactivadores/deficiencia , Transactivadores/metabolismo , Factores de Transcripción/genética , Proteínas Wnt/genética
15.
Dev Biol ; 284(2): 323-36, 2005 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16018995

RESUMEN

Although it is well established that the Gdnf-Ret signal transduction pathway initiates metanephric induction, no single regulator has yet been identified to specify the metanephric mesenchyme or blastema within the intermediate mesoderm, the earliest step of metanephric kidney development and the molecular mechanisms controlling Gdnf expression are essentially unknown. Previous studies have shown that a loss of Eya 1 function leads to renal agenesis that is a likely result of failure of metanephric induction. The studies presented here demonstrate that Eya 1 specifies the metanephric blastema within the intermediate mesoderm at the caudal end of the nephrogenic cord. In contrast to its specific roles in metanephric development, Eya 1 appears dispensable for the formation of nephric duct and mesonephric tubules. Using a combination of null and hypomorphic Eya 1 mutants, we now demonstrated that approximately 20% of normal Eya 1 protein level is sufficient for establishing the metanephric blastema and inducing the ureteric bud formation but not for its normal branching. Using Eya 1, Gdnf, Six 1 and Pax 2 mutant mice, we show that Eya 1 probably functions at the top of the genetic hierarchy controlling kidney organogenesis and it acts in combination with Six 1 and Pax 2 to regulate Gdnf expression during UB outgrowth and branching. These findings uncover an essential function for Eya 1 as a critical determination factor in acquiring metanephric fate within the intermediate mesoderm and as a key regulator of Gdnf expression during ureteric induction and branching morphogenesis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Riñón/embriología , Mesodermo/metabolismo , Transactivadores/metabolismo , Alelos , Animales , Cruzamientos Genéticos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Inducción Embrionaria , Factor Neurotrófico Derivado de la Línea Celular Glial , Heterocigoto , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Hibridación in Situ , Péptidos y Proteínas de Señalización Intracelular , Riñón/citología , Mesodermo/citología , Ratones , Ratones Noqueados , Mutación , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Proteínas Nucleares , Técnicas de Cultivo de Órganos , Factor de Transcripción PAX2 , Proteínas Tirosina Fosfatasas , Transactivadores/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Uréter/citología , Uréter/embriología
16.
Development ; 131(22): 5561-72, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15496442

RESUMEN

Eya1 encodes a transcriptional co-activator and is expressed in cranial sensory placodes. It interacts with and functions upstream of the homeobox gene Six1 during otic placodal development. Here, we have examined their role in cranial sensory neurogenesis. Our data show that the initial cell fate determination for the vestibuloacoustic neurons and their delamination appeared to be unaffected in the absence of Eya1 or Six1 as judged by the expression of the basic helix-loop-helix genes, Neurog1 that specifies the neuroblast cell lineage, and Neurod that controls neuronal differentiation and survival. However, both genes are necessary for normal maintenance of neurogenesis. During the development of epibranchial placode-derived distal cranial sensory ganglia, while the phenotype appears less severe in Six1 than in Eya1 mutants, an early arrest of neurogenesis was observed in the mutants. The mutant epibranchial progenitor cells fail to express Neurog2 that is required for the determination of neuronal precursors, and other basic helix-loop-helix as well as the paired homeobox Phox2 genes that are essential for neural differentiation and maintenance. Failure to activate their normal differentiation program resulted in abnormal apoptosis of the progenitor cells. Furthermore, we show that disruption of viable ganglion formation leads to pathfinding errors of branchial motoneurons. Finally, our results suggest that the Eya-Six regulatory hierarchy also operates in the epibranchial placodal development. These findings uncover an essential function for Eya1 and Six1 as critical determination factors in acquiring both neuronal fate and neuronal subtype identity from epibranchial placodal progenitors. These analyses define a specific role for both genes in early differentiation and survival of the placodally derived cranial sensory neurons.


Asunto(s)
Oído Interno/embriología , Ectodermo/metabolismo , Cabeza/embriología , Cabeza/inervación , Proteínas de Homeodominio/metabolismo , Transactivadores/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Región Branquial/anomalías , Región Branquial/embriología , Región Branquial/metabolismo , Muerte Celular/genética , Oído Interno/anomalías , Oído Interno/patología , Ectodermo/citología , Endodermo/citología , Endodermo/metabolismo , Ganglios/citología , Ganglios/metabolismo , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Noqueados , Mutación/genética , Proteínas del Tejido Nervioso , Neuronas/citología , Neuronas/metabolismo , Proteínas Nucleares , Faringe/embriología , Faringe/metabolismo , Fenotipo , Proteínas Tirosina Fosfatasas , Células Madre/citología , Células Madre/metabolismo , Factores de Tiempo , Transactivadores/deficiencia , Transactivadores/genética , Factores de Transcripción/metabolismo
17.
Proc Natl Acad Sci U S A ; 101(21): 8090-5, 2004 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-15141091

RESUMEN

Urinary tract malformations constitute the most frequent cause of chronic renal failure in the first two decades of life. Branchio-otic (BO) syndrome is an autosomal dominant developmental disorder characterized by hearing loss. In branchio-oto-renal (BOR) syndrome, malformations of the kidney or urinary tract are associated. Haploinsufficiency for the human gene EYA1, a homologue of the Drosophila gene eyes absent (eya), causes BOR and BO syndromes. We recently mapped a locus for BOR/BO syndrome (BOS3) to human chromosome 14q23.1. Within the 33-megabase critical genetic interval, we located the SIX1, SIX4, and SIX6 genes, which act within a genetic network of EYA and PAX genes to regulate organogenesis. These genes, therefore, represented excellent candidate genes for BOS3. By direct sequencing of exons, we identified three different SIX1 mutations in four BOR/BO kindreds, thus identifying SIX1 as a gene causing BOR and BO syndromes. To elucidate how these mutations cause disease, we analyzed the functional role of these SIX1 mutations with respect to protein-protein and protein-DNA interactions. We demonstrate that all three mutations are crucial for Eya1-Six1 interaction, and the two mutations within the homeodomain region are essential for specific Six1-DNA binding. Identification of SIX1 mutations as causing BOR/BO offers insights into the molecular basis of otic and renal developmental diseases in humans.


Asunto(s)
Síndrome Branquio Oto Renal/genética , ADN/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Mutación/genética , Transactivadores/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Línea Celular , ADN/genética , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros/genética , Proteínas de Homeodominio/química , Humanos , Péptidos y Proteínas de Señalización Intracelular , Sustancias Macromoleculares , Datos de Secuencia Molecular , Proteínas Nucleares , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Tirosina Fosfatasas
18.
Mol Reprod Dev ; 66(2): 190-201, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12950107

RESUMEN

Csnk2a2 encodes the CK2alpha'catalytic subunit of CK2 that is predominantly expressed in testis. Male mice in which Csnk2a2 has been disrupted were infertile and displayed oligozoospermia with an abnormal shape of the spermatid nucleus. In this study, Csnk2a2 null testes revealed extensive germ cell degenerative processes at all stages of spermatogenesis, including the first spermatogenesis wave. Nuclear envelope (NE) protrusions with loss of nuclear pores, swelling of the outer membrane, and disruption of the inner membrane were observed in cells ranging from spermatogonia to early spermatids. Most early round spermatids were depleted, and DNA-specific fluorescent dyes showed a large chromatin-free nuclear domain near the chromocenter. Spermatids that were not eliminated retained NE defects that could explain the acrosomal and nuclear abnormalities of Csnk2a2 null spermatozoa. Data suggest that CK2alpha' deficiency could impair the phosphorylation of nuclear proteins of male germ cells leading to a particular cell-death pathway characterized by NE protrusions and an unusual pattern of chromatin modifications in spermatids.


Asunto(s)
Núcleo Celular/ultraestructura , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Espermátides/fisiología , Espermátides/ultraestructura , Espermatogénesis , Animales , Quinasa de la Caseína II , Muerte Celular , Supervivencia Celular , ADN/aislamiento & purificación , Sondas de ADN , Epidídimo/citología , Epidídimo/ultraestructura , Técnica del Anticuerpo Fluorescente Directa , Genotipo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica , Reacción en Cadena de la Polimerasa , Proteínas Serina-Treonina Quinasas/deficiencia , Testículo/citología , Testículo/ultraestructura
19.
Development ; 130(17): 3989-4000, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12874121

RESUMEN

The homeobox Six genes, homologues to Drosophila sine oculis (so) gene, are expressed in multiple organs during mammalian development. However, their roles during auditory system development have not been studied. We report that Six1 is required for mouse auditory system development. During inner ear development, Six1 expression was first detected in the ventral region of the otic pit and later is restricted to the middle and ventral otic vesicle within which, respectively, the vestibular and auditory epithelia form. By contrast, Six1 expression is excluded from the dorsal otic vesicle within which the semicircular canals form. Six1 is also expressed in the vestibuloacoustic ganglion. At E15.5, Six1 is expressed in all sensory epithelia of the inner ear. Using recently generated Six1 mutant mice, we found that all Six1(+/-) mice showed some degree of hearing loss because of a failure of sound transmission in the middle ear. By contrast, Six1(-/-) mice displayed malformations of the auditory system involving the outer, middle and inner ears. The inner ear development in Six1(-/-) embryos arrested at the otic vesicle stage and all components of the inner ear failed to form due to increased cell death and reduced cell proliferation in the otic epithelium. Because we previously reported that Six1 expression in the otic vesicle is Eya1 dependent, we first clarified that Eya1 expression was unaffected in Six1(-/-) otic vesicle, further demonstrating that the Drosophila Eya-Six regulatory cassette is evolutionarily conserved during mammalian inner ear development. We also analyzed several other otic markers and found that the expression of Pax2 and Pax8 was unaffected in Six1(-/-) otic vesicle. By contrast, Six1 is required for the activation of Fgf3 expression and the maintenance of Fgf10 and Bmp4 expression in the otic vesicle. Furthermore, loss of Six1 function alters the expression pattern of Nkx5.1 and Gata3, indicating that Six1 is required for regional specification of the otic vesicle. Finally, our data suggest that the interaction between Eya1 and Six1 is crucial for the morphogenesis of the cochlea and the posterior ampulla during inner ear development. These analyses establish a role for Six1 in early growth and patterning of the otic vesicle.


Asunto(s)
Oído Interno/embriología , Proteínas de Homeodominio/metabolismo , Proteínas Nucleares , Animales , Proteína Morfogenética Ósea 4 , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas de Unión al ADN/metabolismo , Factor 10 de Crecimiento de Fibroblastos , Factor 3 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , Factor de Transcripción GATA3 , Pérdida Auditiva/embriología , Pérdida Auditiva/genética , Péptidos y Proteínas de Señalización Intracelular , Ratones , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción PAX2 , Factor de Transcripción PAX8 , Factores de Transcripción Paired Box , Proteínas Tirosina Fosfatasas , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo
20.
Development ; 130(14): 3085-94, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12783782

RESUMEN

The murine Six gene family, homologous to Drosophila sine oculis (so) which encodes a homeodomain transcription factor, is composed of six members (Six1-6). Among the six members, only the Six2 gene has been previously shown to be expressed early in kidney development, but its function is unknown. We have recently found that the Six1 gene is also expressed in the kidney. In the developing kidney, Six1 is expressed in the uninduced metanephric mesenchyme at E10.5 and in the induced mesenchyme around the ureteric bud at E11.5. At E17.5 to P0, Six1 expression became restricted to a subpopulation of collecting tubule epithelial cells. To study its in vivo function, we have recently generated Six1 mutant mice. Loss of Six1 leads to a failure of ureteric bud invasion into the mesenchyme and subsequent apoptosis of the mesenchyme. These results indicate that Six1 plays an essential role in early kidney development. In Six1(-/-) kidney development, we have found that Pax2, Six2 and Sall1 expression was markedly reduced in the metanephric mesenchyme at E10.5, indicating that Six1 is required for the expression of these genes in the metanephric mesenchyme. In contrast, Eya1 expression was unaffected in Six1(-/-) metanephric mesenchyme at E10.5, indicating that Eya1 may function upstream of Six1. Moreover, our results show that both Eya1 and Six1 expression in the metanephric mesenchyme is preserved in Pax2(-/-) embryos at E10.5, further indicating that Pax2 functions downstream of Eya1 and Six1 in the metanephric mesenchyme. Thus, the epistatic relationship between Pax, Eya and Six genes in the metanephric mesenchyme during early kidney development is distinct from a genetic pathway elucidated in the Drosophila eye imaginal disc. Finally, our results show that Eya1 and Six1 genetically interact during mammalian kidney development, because most compound heterozygous embryos show hypoplastic kidneys. These analyses establish a role for Six1 in the initial inductive step for metanephric development.


Asunto(s)
Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Riñón/embriología , Animales , Apoptosis , Proteínas de Unión al ADN/metabolismo , Genotipo , Heterocigoto , Proteínas de Homeodominio/metabolismo , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Péptidos y Proteínas de Señalización Intracelular , Túbulos Renales/embriología , Mesodermo/metabolismo , Ratones , Mutación , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares , Técnicas de Cultivo de Órganos , Factor de Transcripción PAX2 , Fenotipo , Proteínas Tirosina Fosfatasas , Factores de Tiempo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...