Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Lab Anim ; 40(2): 111-4, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16600070

RESUMEN

In 2003, under the auspices of the main UK funders of biological and biomedical research, a working group was established with a remit to review potential welfare issues for genetically altered (GA) mice, to summarize current practice, and to recommend contemporary best practice for welfare assessments. The working group has produced a report which makes practical recommendations for GA mouse welfare assessment and dissemination of welfare information between establishments using a 'mouse passport'. The report can be found at www.nc3rs.org.uk/GAmice and www.lal.org.uk/gaa and includes templates for the recommended welfare assessment scheme and the mouse passport. An overview is provided below.


Asunto(s)
Crianza de Animales Domésticos/normas , Bienestar del Animal/normas , Animales Modificados Genéticamente , Animales , Animales de Laboratorio , Guías como Asunto , Ratones
2.
Development ; 128(18): 3595-607, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11566863

RESUMEN

During development of the vertebrate hindbrain, Hox genes play multiple roles in the segmental processes that regulate anteroposterior (AP) patterning. Paralogous Hox genes, such as Hoxa3, Hoxb3 and Hoxd3, generally have very similar patterns of expression, and gene targeting experiments have shown that members of paralogy group 3 can functionally compensate for each other. Hence, distinct functions for individual members of this family may primarily depend upon differences in their expression domains. The earliest domains of expression of the Hoxa3 and Hoxb3 genes in hindbrain rhombomeric (r) segments are transiently regulated by kreisler, a conserved Maf b-Zip protein, but the mechanisms that maintain expression in later stages are unknown. In this study, we have compared the segmental expression and regulation of Hoxa3 and Hoxb3 in mouse and chick embryos to investigate how they are controlled after initial activation. We found that the patterns of Hoxa3 and Hoxb3 expression in r5 and r6 in later stages during mouse and chick hindbrain development were differentially regulated. Hoxa3 expression was maintained in r5 and r6, while Hoxb3 was downregulated. Regulatory comparisons of cis-elements from the chick and mouse Hoxa3 locus in both transgenic mouse and chick embryos have identified a conserved enhancer that mediates the late phase of Hoxa3 expression through a conserved auto/cross-regulatory loop. This block of similarity is also present in the human and horn shark loci, and contains two bipartite Hox/Pbx-binding sites that are necessary for its in vivo activity in the hindbrain. These HOX/PBC sites are positioned near a conserved kreisler-binding site (KrA) that is involved in activating early expression in r5 and r6, but their activity is independent of kreisler. This work demonstrates that separate elements are involved in initiating and maintaining Hoxa3 expression during hindbrain segmentation, and that it is regulated in a manner different from Hoxb3 in later stages. Together, these findings add further strength to the emerging importance of positive auto- and cross-regulatory interactions between Hox genes as a general mechanism for maintaining their correct spatial patterns in the vertebrate nervous system.


Asunto(s)
Proteínas Aviares , Tipificación del Cuerpo/genética , Proteínas de Homeodominio/genética , Proteínas Oncogénicas , Rombencéfalo/embriología , Proteínas de Xenopus , Animales , Secuencia de Bases , Sitios de Unión , Evolución Biológica , Embrión de Pollo , Secuencia Conservada , Proteínas de Unión al ADN , Elementos de Facilitación Genéticos , Factores de Unión a la G-Box , Regulación del Desarrollo de la Expresión Génica , Factor de Transcripción MafB , Ratones , Ratones Transgénicos , Modelos Genéticos , Secuencias Reguladoras de Ácidos Nucleicos , Homología de Secuencia de Ácido Nucleico , Especificidad de la Especie , Factores de Transcripción
3.
Dev Biol ; 233(2): 468-81, 2001 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-11336508

RESUMEN

During hindbrain development, segmental regulation of the paralogous Hoxa2 and Hoxb2 genes in rhombomeres (r) 3 and 5 involves Krox20-dependent enhancers that have been conserved during the duplication of the vertebrate Hox clusters from a common ancestor. Examining these evolutionarily related control regions could provide important insight into the degree to which the basic Krox20-dependent mechanisms, cis-regulatory components, and their organization have been conserved. Toward this goal we have performed a detailed functional analysis of a mouse Hoxa2 enhancer capable of directing reporter expression in r3 and r5. The combined activities of five separate cis-regions, in addition to the conserved Krox20 binding sites, are involved in mediating enhancer function. A CTTT (BoxA) motif adjacent to the Krox20 binding sites is important for r3/r5 activity. The BoxA motif is similar to one (Box1) found in the Hoxb2 enhancer and indicates that the close proximity of these Box motifs to Krox20 sites is a common feature of Krox20 targets in vivo. Two other rhombomeric elements (RE1 and RE3) are essential for r3/r5 activity and share common TCT motifs, indicating that they interact with a similar cofactor(s). TCT motifs are also found in the Hoxb2 enhancer, suggesting that they may be another common feature of Krox20-dependent control regions. The two remaining Hoxa2 cis-elements, RE2 and RE4, are not conserved in the Hoxb2 enhancer and define differences in some of components that can contribute to the Krox20-dependent activities of these enhancers. Furthermore, analysis of regulatory activities of these enhancers in a Krox20 mutant background has uncovered differences in their degree of dependence upon Krox20 for segmental expression. Together, this work has revealed a surprising degree of complexity in the number of cis-elements and regulatory components that contribute to segmental expression mediated by Krox20 and sheds light on the diversity and evolution of Krox20 target sites and Hox regulatory elements in vertebrates.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Rombencéfalo/embriología , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Animales , Secuencia de Bases , ADN/genética , Proteína 2 de la Respuesta de Crecimiento Precoz , Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Genes Homeobox , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Datos de Secuencia Molecular , Homología de Secuencia de Ácido Nucleico
4.
EMBO J ; 19(24): 6675-85, 2000 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11118202

RESUMEN

Repair of DNA damage by homologous recombination has only recently been established as an important mechanism in maintaining genetic stability in mammalian cells. The recently cloned Xrcc2 gene is a member of the mammalian Rad51 gene family, thought to be central to homologous recombination repair. To understand its function in mammals, we have disrupted Xrcc2 in mice. No Xrcc2(-/-) animals were found alive, with embryonic lethality occurring from mid-gestation. Xrcc2(-/-) embryos surviving until later stages of embryogenesis commonly showed developmental abnormalities and died at birth. Neonatal lethality, apparently due to respiratory failure, was associated with a high frequency of apoptotic death of post- mitotic neurons in the developing brain, leading to abnormal cortical structure. Embryonic cells showed genetic instability, revealed by a high level of chromosomal aberrations, and were sensitive to gamma-rays. Our findings demonstrate that homologous recombination has an important role in endogenous damage repair in the developing embryo. Xrcc2 disruption identifies a range of defects that arise from malfunction of this repair pathway, and establishes a previously unidentified role for homologous recombination repair in correct neuronal development.


Asunto(s)
Blastocisto/citología , Proteínas de Unión al ADN/metabolismo , Desarrollo Embrionario y Fetal/genética , Sistema Nervioso/embriología , Neuronas/fisiología , Animales , Apoptosis , Blastocisto/fisiología , Quimera , Mapeo Cromosómico , Clonación Molecular , Daño del ADN/genética , Reparación del ADN/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Muerte Fetal , Genotipo , Ratones , Ratones Noqueados , Neuronas/citología , Reacción en Cadena de la Polimerasa , Células Madre
5.
Development ; 127(1): 155-66, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10654609

RESUMEN

Direct auto- and cross-regulatory interactions between Hox genes serve to establish and maintain segmentally restricted patterns in the developing hindbrain. Rhombomere r4-specific expression of both Hoxb1 and Hoxb2 depends upon bipartite cis Hox response elements for the group 1 paralogous proteins, Hoxal and Hoxbl. The DNA-binding ability and selectivity of these proteins depend upon the formation of specific heterodimeric complexes with members of the PBC homeodomain protein family (Pbx genes). The r4 enhancers from Hoxb1 and Hoxb2 have the same activity, but differ with respect to the number and organisation of bipartite Pbx/Hox (PH) sites required, suggesting the intervention of other components/sequences. We report here that another family of homeodomain proteins (TALE, Three-Amino acids-Loop-Extension: Prep1, Meis, HTH), capable of dimerizing with Pbx/EXD, is involved in the mechanisms of r4-restricted expression. We show that: (1) the r4-specific Hoxb1 and Hoxb2 enhancers are complex elements containing separate PH and Prep/Meis (PM) sites; (2) the PM site of the Hoxb2, but not Hoxb1, enhancer is essential in vivo for r4 expression and also influences other sites of expression; (3) both PM and PH sites are required for in vitro binding of Prepl-Pbx and formation and binding of a ternary Hoxbl-Pbxla (or 1b)-Prepl complex. (4) A similar ternary association forms in nuclear extracts from embryonal P19 cells, but only upon retinoic acid induction. This requires synthesis of Hoxbl and also contains Pbx with either Prepl or Meisl. Together these findings highlight the fact that PM sites are found in close proximity to bipartite PH motifs in several Hox responsive elements shown to be important in vivo and that such sites play an essential role in potentiating regulatory activity in combination with the PH motifs.


Asunto(s)
Tipificación del Cuerpo/fisiología , Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Rombencéfalo/embriología , Factores de Transcripción/genética , Animales , Secuencia de Bases , Extractos Celulares , Núcleo Celular , Proteínas de Unión al ADN/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Datos de Secuencia Molecular , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Proteínas Proto-Oncogénicas/genética , Tretinoina/metabolismo
6.
Gene ; 240(2): 389-98, 1999 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-10580159

RESUMEN

We report the cloning of a cDNA for the mouse unconventional myosin Myo9b, the orthologue of the rat myr5 and human MYOIXb genes. A full-length spleen cDNA of 7087bp encoding a protein of 1961 amino acids was isolated. By RT-PCR, we show that Myo9b is expressed in a wide range of tissues, including heart, brain, muscle and inner ear. In addition, we have identified two alternatively spliced exons. Equivalent exons have not been previously reported for either the human or rat homologues. These exons are located in the Myo9b specific actin-binding site insert of the head domain and in the tail region. A third splice form utilizing an alternative reading frame within the 3'UTR is also described. Several polymorphisms within the coding region were identified; of interest is an in-frame 33bp imperfect duplication within the tail region that was observed only in the C57Bl/6 strain. Myo9b has been previously mapped to mouse chromosome 8 and is a candidate for the mouse mutations myodystrophy and quinky.


Asunto(s)
Empalme Alternativo , Miosinas/genética , Regiones no Traducidas 3'/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , ADN Complementario/química , ADN Complementario/genética , ADN Complementario/aislamiento & purificación , Oído Interno/embriología , Oído Interno/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Endogámicos , Datos de Secuencia Molecular , Muridae , Polimorfismo Genético , Isoformas de Proteínas/genética , ARN/genética , ARN/metabolismo , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Distribución Tisular
7.
Neuron ; 22(4): 677-91, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10230789

RESUMEN

Little is known about how the generation of specific neuronal types at stereotypic positions within the hindbrain is linked to Hox gene-mediated patterning. Here, we show that during neurogenesis, Hox paralog group 2 genes control both anteroposterior (A-P) and dorsoventral (D-V) patterning. Hoxa2 and Hoxb2 differentially regulate, in a rhombomere-specific manner, the expression of several genes in broad D-V-restricted domains or narrower longitudinal columns of neuronal progenitors, immature neurons, and differentiating neuronal subtypes. Moreover, Hoxa2 and Hoxb2 can functionally synergize in controlling the development of ventral neuronal subtypes in rhombomere 3 (r3). Thus, in addition to their roles in A-P patterning, Hoxa2 and Hoxb2 have distinct and restricted functions along the D-V axis during neurogenesis, providing insights into how neuronal fates are assigned at stereotypic positions within the hindbrain.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Homeodominio/genética , Neuronas/fisiología , Rombencéfalo/embriología , Factores de Transcripción/genética , Animales , Tipificación del Cuerpo , Diferenciación Celular/fisiología , Nervio Facial/fisiología , Ratones , Ratones Mutantes , Neuronas Motoras/fisiología , Rombencéfalo/citología , Rombencéfalo/metabolismo
8.
Development ; 126(7): 1483-94, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10068641

RESUMEN

Hoxa2 is expressed in cranial neural crest cells that migrate into the second branchial arch and is essential for proper patterning of neural-crest-derived structures in this region. We have used transgenic analysis to begin to address the regulatory mechanisms which underlie neural-crest-specific expression of Hoxa2. By performing a deletion analysis on an enhancer from the Hoxa2 gene that is capable of mediating expression in neural crest cells in a manner similar to the endogenous gene, we demonstrated that multiple cis-acting elements are required for neural-crest-specific activity. One of these elements consists of a sequence that binds to the three transcription factor AP-2 family members. Mutation or deletion of this site in the Hoxa2 enhancer abrogates reporter expression in cranial neural crest cells but not in the hindbrain. In both cell culture co-transfection assays and transgenic embryos AP-2 family members are able to trans-activate reporter expression, showing that this enhancer functions as an AP-2-responsive element in vivo. Reporter expression is not abolished in an AP-2(alpha) null mutant embryos, suggesting redundancy with other AP-2 family members for activation of the Hoxa2 enhancer. Other cis-elements identified in this study critical for neural-crest-specific expression include an element that influences levels of expression and a conserved sequence, which when multimerized directs expression in a broad subset of neural crest cells. These elements work together to co-ordinate and restrict neural crest expression to the second branchial arch and more posterior regions. Our findings have identified the cis-components that allow Hoxa2 to be regulated independently in rhombomeres and cranial neural crest cells.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de Homeodominio/genética , Cresta Neural/embriología , Factores de Transcripción/genética , Animales , Encéfalo/crecimiento & desarrollo , Región Branquial/embriología , Células Cultivadas , Elementos de Facilitación Genéticos/genética , Genes Reporteros , Ratones , Ratones Transgénicos , Mutación , Fosfoproteínas/genética , Eliminación de Secuencia/genética , Factor de Transcripción AP-2 , Activación Transcripcional/genética
9.
Genes Dev ; 11(14): 1885-95, 1997 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-9242495

RESUMEN

Correct regulation of the segment-restricted patterns of Hox gene expression is essential for proper patterning of the vertebrate hindbrain. We have examined the molecular basis of restricted expression of Hoxb2 in rhombomere 4 (r4), by using deletion analysis in transgenic mice to identify an r4 enhancer from the mouse gene. A bipartite Hox/Pbx binding motif is located within this enhancer, and in vitro DNA binding experiments showed that the vertebrate labial-related protein Hoxb1 will cooperatively bind to this site in a Pbx/Exd-dependent manner. The Hoxb2 r4 enhancer can be transactivated in vivo by the ectopic expression of Hoxb1, Hoxa1, and Drosophila labial in transgenic mice. In contrast, ectopic Hoxb2 and Hoxb4 are unable to induce expression, indicating that in vivo this enhancer preferentially responds to labial family members. Mutational analysis demonstrated that the bipartite Hox/Pbx motif is required for r4 enhancer activity and the responses to retinoids and ectopic Hox expression. Furthermore, three copies of the Hoxb2 motif are sufficient to mediate r4 expression in transgenic mouse embryos and a labial pattern in Drosophila embryos. This reporter expression in Drosophila embryos is dependent upon endogenous labial and exd, suggesting that the ability of this Hox/Pbx site to interact with labial-related proteins has been evolutionarily conserved. The endogenous Hoxb2 gene is no longer upregulated in r4 in Hoxb1 homozygous mutant embryos. On the basis of these experiments we conclude that the r4-restricted domain of Hoxb2 in the hindbrain is the result of a direct cross-regulatory interaction by Hoxb1 involving vertebrate Pbx proteins as cofactors. This suggests that part of the functional role of Hoxb1 in maintaining r4 identity may be mediated by the Hoxb2 gene.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Animales , Secuencia de Bases , Drosophila/embriología , Drosophila/genética , Elementos de Facilitación Genéticos , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Datos de Secuencia Molecular , Unión Proteica , Factores de Transcripción/metabolismo , Activación Transcripcional
11.
EMBO J ; 15(19): 5383-96, 1996 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-8895582

RESUMEN

Segmentation is a key feature of the development of the vertebrate hindbrain where it involves the generation of repetitive morphological units termed rhombomeres (r). Hox genes are likely to play an essential role in the specification of segmental identity and we have been investigating their regulation. We show here that the mouse and chicken Hoxb-2 genes are dependent for their expression in r3 and r5 on homologous enhancer elements and on binding to this enhancer of the r3/r5-specific transcriptional activator Krox-20. Among the three Krox-20 binding sites of the mouse Hoxb-2 enhancer, only the high-affinity site is absolutely necessary for activity. In contrast, we have identified an additional cis-acting element, Box1, essential for r3/r5 enhancer activity. It is conserved both in sequence and in position respective to the high-affinity Krox-20 binding site within the mouse and chicken enhancers. Furthermore, a short 44 bp sequence spanning the Box1 and Krox-20 sites can act as an r3/r5 enhancer when oligomerized. Box1 may therefore constitute a recognition sequence for another factor cooperating with Krox-20. Taken together, these data demonstrate the conservation of Hox gene regulation and of Krox-20 function during vertebrate evolution.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Elementos de Facilitación Genéticos/genética , Proteínas de Homeodominio/genética , Rombencéfalo/embriología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional/genética , Animales , Secuencia de Bases , Sitios de Unión , Embrión de Pollo , Clonación Molecular , Secuencia Conservada/genética , ADN/metabolismo , Proteína 2 de la Respuesta de Crecimiento Precoz , Regulación del Desarrollo de la Expresión Génica/genética , Genes Homeobox/genética , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Mutación , Homología de Secuencia de Ácido Nucleico
12.
Proc Natl Acad Sci U S A ; 93(18): 9339-45, 1996 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-8790331

RESUMEN

Transient segmentation in the hindbrain is a fundamental morphogenetic phenomenon in the vertebrate embryo, and the restricted expression of subsets of Hox genes in the developing rhombomeric units and their derivatives is linked with regional specification. Here we show that patterning of the vertebrate hindbrain involves the direct upregulation of the chicken and pufferfish group 2 paralogous genes, Hoxb-2 and Hoxa-2, in rhombomeres 3 and 5 (r3 and r5) by the zinc finger gene Krox-20. We identified evolutionarily conserved r3/r5 enhancers that contain high affinity Krox-20. binding sites capable of mediating transactivation by Krox-20. In addition to conservation of binding sites critical for Krox-20 activity in the chicken Hoxa-2 and pufferfish Hoxb-2 genes, the r3/r5 enhancers are also characterized by the presence of a number of identical motifs likely to be involved in cooperative interactions with Krox-20 during the process of hindbrain patterning in vertebrates.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Regulación del Desarrollo de la Expresión Génica , Genes Homeobox , Proteínas de Homeodominio/genética , Rombencéfalo/embriología , Transactivadores/genética , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Animales , Secuencia de Bases , Sitios de Unión , Pollos , Mapeo Cromosómico , Secuencia Conservada , Proteínas de Unión al ADN/genética , Proteína 2 de la Respuesta de Crecimiento Precoz , Elementos de Facilitación Genéticos , Peces , Humanos , Hibridación in Situ , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Dedos de Zinc/genética
13.
Transgenic Res ; 5(2): 123-30, 1996 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8866894

RESUMEN

Dystrophin has a modular structure and is believed to be critical for muscle cell cytoarchitecture by linking the cytoskeleton to the extracellular matrix. The N-terminus binds to actin and two domains at the C-terminus, the cysteine-rich and C-terminal domains, are associated with the sarcolemma indirectly via the dystroglycan complex. We have generated a mutation in mouse embryonic stem (ES) cells which serves to delete the cysteine-rich and C-terminal domains to address directly their role. We show that these two domains are not necessary for normal costameric organization at the sarcolemma in myotubes derived from the mutant cell line. Furthermore sarcolemmal localization is also apparent in mouse chimaeric muscle in vivo.


Asunto(s)
Distrofina/análisis , Distrofina/química , Sarcolema/química , Alelos , Animales , Diferenciación Celular , Quimera , Cisteína , Distrofina/genética , Exones/genética , Genes Reporteros/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/química , Mutación , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/biosíntesis , Eliminación de Secuencia , Células Madre/citología , beta-Galactosidasa/genética
14.
Development ; 122(2): 543-54, 1996 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8625806

RESUMEN

The hindbrain is a segmented structure divided into repeating metameric units termed rhombomeres (r). The Hox family, vertebrate homologs of the Drosophila HOM-C homeotic selector genes, are expressed in rhombomere-restricted patterns and are believed to participate in regulating segmental identities. Krox-20, a zinc finger gene, has a highly conserved pattern of expression in r3 and r5 and is functionally required for their maintenance in mouse embryos. Krox-20 has been shown to directly regulate the Hoxb-2 gene and we wanted to determine if it was involved in regulating multiple Hox genes as a part of its functional role. Hoxa-2 is the only known paralog of Hoxb-2, and we examined the patterns of expression of the mouse Hoxa-2 gene with particular focus on r3 and r5 in wild type and Krox-20-/- mutant embryos. There was a clear loss of expression in r3, which indicated that Hoxa-2 was downstream of Krox-20. Using transgenic analysis with E. coli lacZ reporter genes we have identified and mapped an r3/r5 enhancer in the 5' flanking region of the Hoxa-2 gene. Deletion analysis narrowed this region to an 809 bp Bg/II fragment, and in vitro binding and competition assays with bacterially expressed Krox-20 protein identified two sites within the enhancer. Mutation of these Krox-20 sites in the regulatory region specifically abolished r3/r5 activity, but did not affect neural crest and mesodermal components. This indicated that the two Krox-20 sites are required in vivo for enhancer function. Furthermore, ectopic expression of Krox-20 in r4 was able to transactivate the Hoxa 2/lacZ reporter in this rhombomere. Together our findings suggest that Krox-20 directly participates in the transcriptional regulation of Hoxa-2 during hindbrain segmentation, and is responsible for the upregulation of the r3 and r5 domains of expression of both vertebrate group 2 Hox paralogs. Therefore, the segmental phenotypes in the Krox-20 mutants are likely to reflect the role of Krox-20 in directly regulating multiple Hox genes.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Genes Homeobox , Proteínas del Tejido Nervioso/biosíntesis , Rombencéfalo/embriología , Factores de Transcripción/biosíntesis , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Secuencia Conservada , Proteínas de Unión al ADN/biosíntesis , Proteína 2 de la Respuesta de Crecimiento Precoz , Elementos de Facilitación Genéticos , Homocigoto , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Sondas de Oligonucleótidos , Fenotipo , Proteínas Recombinantes/análisis , Proteínas Recombinantes/biosíntesis , Mapeo Restrictivo , Rombencéfalo/citología , Rombencéfalo/metabolismo , Vertebrados , Dedos de Zinc , beta-Galactosidasa/análisis , beta-Galactosidasa/biosíntesis
15.
Annu Rev Genet ; 30: 529-56, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-8982464

RESUMEN

The Hox homeobox gene family plays a pivotal role in regulating patterning and axial morphogenesis in vertebrates. Molecular characterization of the four Hox clusters has shown that they are evolutionarily related with respect to sequence, organization, and expression, suggesting they arose by duplication and divergence. Transgenic analysis has clearly demonstrated the functional roles of individual genes in a broad range of embryonic tissues, and in compound mutants has addressed the issues of cooperativity and redundancy. There is an emerging picture of the cis-regulatory elements underlying Hox expression, and for the 3' members of the clusters there is a considerable degree of conservation between paralogous genes with respect to their functional roles and regulatory control.


Asunto(s)
Genes Homeobox , Animales , Análisis Mutacional de ADN , Regulación de la Expresión Génica , Humanos
17.
Mol Gen Genet ; 234(2): 211-6, 1992 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-1354839

RESUMEN

We have isolated a gene from Coprinus cinereus which cross-hybridises to the facA and acu-5 genes of Aspergillus nidulans and Neurospora crassa, respectively. These genes encode acetyl-CoA synthetase, an enzyme which is inducible by acetate and required for growth on acetate as sole carbon source. We have designated the C. cinereus gene acs-1 and have used transformation to demonstrate its functional homology to the ascomycete genes by complementation of an N. crassa acu-5 mutation. The acs-1 gene has never been identified by mutation; mutations leading to loss of acetyl-CoA synthetase function map to another gene, acu-1. Using Northern analyses we have shown that acu-1 has a regulatory function that is required for acetate-induced transcription of acs-1 and of another acetate utilisation gene, acu-7, the isocitrate lyase structural gene.


Asunto(s)
Acetatos/farmacología , Acetil-CoA Carboxilasa/genética , Coprinus/enzimología , Regulación Fúngica de la Expresión Génica/efectos de los fármacos , Genes Reguladores/genética , Ácido Acético , Northern Blotting , Clonación Molecular , Coprinus/genética , Escherichia coli/genética , Biblioteca Genómica , Isocitratoliasa/genética , Plásmidos/genética , Mapeo Restrictivo , Homología de Secuencia de Ácido Nucleico
18.
Mamm Genome ; 2(1): 64-8, 1992.
Artículo en Inglés | MEDLINE | ID: mdl-1543903

RESUMEN

We have isolated two genomic clones from the murine dystrophin locus, containing single exons encoding protein sequence from the putative actin-binding domain of the amino-terminus and the terminal portion of the triple helical domain. Using interspecific backcross progeny mice, both clones were shown to be X-linked. Sequence analysis indicated that the amino-terminal clone contains a 173 bp exon exhibiting 90% nucleotide sequence identity to human dystrophin exon 6, whilst the C-terminal clone contains a 61 bp exon with 93% nucleotide sequence identity to the human cDNA sequence.


Asunto(s)
Distrofina/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , ADN , Exones , Ratones , Datos de Secuencia Molecular , Mapeo Restrictivo , Alineación de Secuencia
19.
Nucleic Acids Res ; 19(12): 3255-60, 1991 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-2062641

RESUMEN

A repeat sequence island, located at the A3 Giemsa dark band on the mouse X chromosome and consisting of 50 copies of a localised long complex repeat unit (LCRU), features an unusually high concentration of L1 LINE repeat sequences juxtaposed and inserted within the LCRU. Sequence analysis of three independent genomic clones containing L1 LINE elements juxtaposed with the LCRU demonstrates a common junction sequence at the L1/LCRU boundary, suggesting that the high concentration of L1 LINE sequences in the repeat sequence island has arisen by association of an L1 element with an LCRU followed by amplification. The LCRU target site at this common junction sequence bears no resemblance to the target site of an L1 element inserted within one LCRU, indicating there is no specific preferential target site for L1 integration. We propose that co-amplification of L1 LINE elements with localised low copy repeat families throughout the genome could have a major effect on the chromosomal distribution of L1 LINE elements.


Asunto(s)
Amplificación de Genes , Secuencias Repetitivas de Ácidos Nucleicos , Cromosoma X , Animales , Secuencia de Bases , Bandeo Cromosómico , Clonación Molecular , ADN , ADN Satélite/química , Ratones , Datos de Secuencia Molecular , Mapeo Restrictivo , Homología de Secuencia de Ácido Nucleico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...