Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
iScience ; 25(12): 105464, 2022 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-36404917

RESUMEN

D/E repeats are stretches of aspartic and/or glutamic acid residues found in over 150 human proteins. We examined genomic stability of D/E repeats and functional characteristics of D/E repeat-containing proteins vis-à-vis the proteins with poly-Q or poly-A repeats, which are known to undergo pathologic expansions. Mining of tumor sequencing data revealed that D/E repeat-coding regions are similar to those coding poly-Qs and poly-As in increased incidence of trinucleotide insertions/deletions but differ in types and incidence of substitutions. D/E repeat-containing proteins preferentially function in chromatin metabolism and are the more likely to be nuclear and interact with core histones, the longer their repeats are. One of the longest D/E repeats of unknown function is in ATAD2, a bromodomain family ATPase frequently overexpressed in tumors. We demonstrate that D/E repeat deletion in ATAD2 suppresses its binding to nascent and mature chromatin and to the constitutive pericentromeric heterochromatin, where ATAD2 represses satellite transcription.

2.
Front Mol Biosci ; 9: 1048726, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36710880

RESUMEN

Background: The cGAS/STING pathway, part of the innate immune response to foreign DNA, can be activated by cell's own DNA arising from the processing of the genome, including the degradation of nascent DNA at arrested replication forks, which can be upregulated in cancer cells. Recent evidence raises a possibility that the cGAS/STING pathway may also modulate the very processes that trigger it, e.g., DNA damage repair or processing of stalled forks. Methods: We manipulated STING levels in human cells by depleting or re-expressing it, and assessed the effects of STING on replication using microfluidics-assisted replication track analysis, or maRTA, a DNA fiber assay, as well as immuno-precipitation of nascent DNA, or iPOND. We also assessed STING subcellular distribution and its ability to activate. Results: Depletion of STING suppressed and its re-expression in STING-deficient cancer cells upregulated the degradation of nascent DNA at arrested replication forks. Replication fork arrest was accompanied by the STING pathway activation, and a STING mutant that does not activate the pathway failed to upregulate nascent DNA degradation. cGAS was required for STING's effect on degradation, but this requirement could be bypassed by treating cells with a STING agonist. Cells expressing inactive STING had a reduced level of RPA on parental and nascent DNA of arrested forks and a reduced CHK1 activation compared to cells with the wild type STING. STING also affected unperturbed fork progression in a subset of cell lines. STING fractionated to the nuclear fractions enriched for structural components of chromatin and nuclear envelope, and furthermore, it associated with the chromatin of arrested replication forks as well as post-replicative chromatin. Conclusion: Our data highlight STING as a determinant of stalled replication fork integrity, thus revealing a novel connection between the replication stress and innate immune responses.

3.
Oncogene ; 40(3): 578-591, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33199827

RESUMEN

Rhabdomyosarcoma (RMS) is a devastating pediatric sarcoma. The survival outcomes remain poor for patients with relapsed or metastatic disease. Effective targeted therapy is lacking due to our limited knowledge of the underlying cellular and molecular mechanisms leading to disease progression. In this study, we used functional assays in vitro and in vivo (zebrafish and xenograft mouse models) to demonstrate the crucial role of HDAC6, a cytoplasmic histone deacetylase, in driving RMS tumor growth, self-renewal, and migration/invasion. Treatment with HDAC6-selective inhibitors recapitulates the HDAC6 loss-of-function phenotypes. HDAC6 regulates cytoskeletal dynamics to promote tumor cell migration and invasion. RAC1, a Rho family GTPase, is an essential mediator of HDAC6 function, and is necessary and sufficient for RMS cell migration and invasion. High expression of RAC1 correlates with poor clinical prognosis in RMS patients. Targeting the HDAC6-RAC1 axis represents a promising therapeutic option for improving survival outcomes of RMS patients.


Asunto(s)
Movimiento Celular , Proliferación Celular , Histona Desacetilasa 6/metabolismo , Proteínas de Neoplasias/metabolismo , Rabdomiosarcoma/metabolismo , Línea Celular Tumoral , Histona Desacetilasa 6/genética , Humanos , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Rabdomiosarcoma/genética , Rabdomiosarcoma/patología
4.
Mol Cell Biol ; 40(9)2020 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-32015101

RESUMEN

Newly synthesized histone H4 that is incorporated into chromatin during DNA replication is acetylated on lysines 5 and 12. Histone deacetylase 1 (HDAC1) and HDAC2 are responsible for reducing H4 acetylation as chromatin matures. Using CRISPR-Cas9-generated hdac1- or hdac2-null fibroblasts, we determined that HDAC1 and HDAC2 do not fully compensate for each other in removing de novo acetyls on H4 in vivo Proteomics of nascent chromatin and proximity ligation assays with newly replicated DNA revealed the binding of ATAD2, a bromodomain-containing posttranslational modification (PTM) reader that recognizes acetylated H4. ATAD2 is a transcription facilitator overexpressed in several cancers and in the simian virus 40 (SV40)-transformed human fibroblast model cell line used in this study. The recruitment of ATAD2 to nascent chromatin was increased in hdac2 cells over the wild type, and ATAD2 depletion reduced the levels of nascent chromatin-associated, acetylated H4 in wild-type and hdac2 cells. We propose that overexpressed ATAD2 shifts the balance of H4 acetylation by protecting this mark from removal and that HDAC2 but not HDAC1 can effectively compete with ATAD2 for the target acetyls. ATAD2 depletion also reduced global RNA synthesis and nascent DNA-associated RNA. A moderate dependence on ATAD2 for replication fork progression was noted only for hdac2 cells overexpressing the protein.


Asunto(s)
Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Histonas/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Acetilación , Línea Celular , Cromatina/metabolismo , ADN/metabolismo , Replicación del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Humanos , Lisina/metabolismo , Procesamiento Proteico-Postraduccional
5.
BMC Genet ; 16: 143, 2015 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-26678520

RESUMEN

BACKGROUND: Etl4(lacZ) (Enhancer trap locus 4) and Skt(Gt) (Sickle tail) are lacZ reporter gene integrations into the same locus on mouse chromosome 2 targeting a gene that is expressed in the notochord of early embryos and in multiple epithelia during later development. Both insertions caused recessive mutations that resulted exclusively in mild defects in the caudal vertebral column. Since notochord-derived signals are essential for formation of the vertebral column the phenotypes suggested that the lacZ insertions interfered with some notochord-dependent aspect of vertebral development. As both insertions occurred in introns it was unclear whether they represent hypomorphic alleles or abolish gene function. Here, we have generated a definitive null allele of the Skt/Etl4 gene and analysed homozygous mutants. RESULTS: We have introduced loxP sites into three positions of the gene based on additional upstream exons that we identified, and deleted approximately 870 kb of the locus by a combination of inter- and intra-chromosomal Cre-mediated recombinations in the female germ line of mice. This deletion removes about 90 % of the coding region and results in the loss of the SKT/ETL4 protein. Similar to the Etl4(lacZ) and Skt(Gt) alleles our deletion mutants are viable and fertile and show only mild defects in caudal vertebrae due to abnormal intervertebral disc development, although with higher penetrance. No other tissue with Skt/Etl4 expression that we analysed showed obvious defects. CONCLUSION: The complete loss of Skt/Etl4 function affects only development of caudal notochord derivatives and is compensated for in its other expression domains.


Asunto(s)
Marcación de Gen/métodos , Proteínas/genética , Animales , Cromosomas de los Mamíferos/metabolismo , Embrión de Mamíferos/metabolismo , Eliminación de Gen , Genes Reporteros , Disco Intervertebral/metabolismo , Operón Lac , Ratones , Proteínas/metabolismo , Cola (estructura animal)/embriología
6.
J Neuroinflammation ; 12: 154, 2015 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-26329692

RESUMEN

BACKGROUND: Neuroinflammation is an important secondary mechanism that is a key mediator of the long-term consequences of neuronal injury that occur in traumatic brain injury (TBI). Microglia are highly plastic cells with dual roles in neuronal injury and recovery. Recent studies suggest that the chemokine fractalkine (CX3CL1, FKN) mediates neural/microglial interactions via its sole receptor CX3CR1. CX3CL1/CX3CR1 signaling modulates microglia activation, and depending upon the type and time of injury, either protects or exacerbates neurological diseases. METHODS: In this study, mice deficient in CX3CR1 were subjected to mild controlled cortical impact injury (CCI), a model of TBI. We evaluated the effects of genetic deletion of CX3CR1 on histopathology, cell death/survival, microglia activation, and cognitive function for 30 days post-injury. RESULTS: During the acute post-injury period (24 h-15 days), motor deficits, cell death, and neuronal cell loss were more profound in injured wild-type than in CX3CR1(-/-) mice. In contrast, during the chronic period of 30 days post-TBI, injured CX3CR1(-/-) mice exhibited greater cognitive dysfunction and increased neuronal death than wild-type mice. The protective and deleterious effects of CX3CR1 were associated with changes in microglia phenotypes; during the acute phase CX3CR1(-/-) mice showed a predominant anti-inflammatory M2 microglial response, with increased expression of Ym1, CD206, and TGFß. In contrast, increased M1 phenotypic microglia markers, Marco, and CD68 were predominant at 30 days post-TBI. CONCLUSION: Collectively, these novel data demonstrate a time-dependent role for CX3CL1/CX3CR1 signaling after TBI and suggest that the acute and chronic responses to mild TBI are modulated in part by distinct microglia phenotypes.


Asunto(s)
Lesiones Encefálicas , Encéfalo/patología , Receptores de Quimiocina/metabolismo , Análisis de Varianza , Animales , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Receptor 1 de Quimiocinas CX3C , Modelos Animales de Enfermedad , Conducta Exploratoria/fisiología , Citometría de Flujo , Fluoresceínas/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Leucocitos Mononucleares/patología , Activación de Macrófagos/genética , Activación de Macrófagos/fisiología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Neuronas/patología , Trastornos Psicomotores/etiología , Receptores de Quimiocina/genética , Prueba de Desempeño de Rotación con Aceleración Constante , Factores de Tiempo
7.
Brain Behav Immun ; 48: 244-57, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25849975

RESUMEN

Interactions between sleep and immune function are bidirectional. Although the mechanisms that govern these interactions are not fully elucidated, the pro-inflammatory cytokine, interleukin-1ß (IL-1), is a known regulator of sleep and mediator of immune responses. To further clarify the underlying substrates of sleep and immune interactions, we engineered two transgenic mouse lines that express interleukin-1 receptor 1 (IL1R1) only in the central nervous system (CNS) and selectively on neurons (NSE-IL1R1) or astrocytes (GFAP-IL1R1). During spontaneous sleep, compared to wild type (WT) animals, NSE-IL1R1 and GFAP-IL1R1 mice have more rapid eye movement sleep (REMS) that is characterized by reduced theta power in the electroencephalogram (EEG) spectra. The non-REM sleep (NREMS) EEG of each of the IL1R1 transgenic mouse strains also is characterized by enhanced power in the delta frequency band. In response to 6h of sleep deprivation, sleep of both IL1R1 transgenic mouse strains is more consolidated than that of WT animals. Additionally, the NREMS EEG of NSE-IL1R1 mice contains less delta power after sleep deprivation, suggesting astroglial IL1R1 activity may modulate sleep homeostasis. Intracerebroventricular injection of IL-1 fails to alter sleep or brain temperature of NSE-IL1R1 or GFAP-IL1R1 mice. These data suggest that selective IL1R1 expression on neurons or on astrocytes is not sufficient for centrally-administered IL-1 to induce sleep or fever. Lack of sleep and febrile responses to IL-1 in these IL1R1 transgenic mouse strains may be due to their inability to produce IL-6 in brain. Overall, these studies demonstrate, through the use of novel transgenic mice, that IL1R1 on neurons and astrocytes differentially mediates aspects of sleep under physiological conditions and in response to central IL-1 administration.


Asunto(s)
Astrocitos/fisiología , Neuronas/fisiología , Receptores de Interleucina-1/metabolismo , Sueño/fisiología , Animales , Astrocitos/efectos de los fármacos , Temperatura Corporal/efectos de los fármacos , Temperatura Corporal/fisiología , Electroencefalografía , Interleucina-1/farmacología , Masculino , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Receptores de Interleucina-1/genética , Sueño/efectos de los fármacos
8.
PLoS Genet ; 4(5): e1000069, 2008 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-18464897

RESUMEN

Inducible and reversible regulation of gene expression is a powerful approach for uncovering gene function. We have established a general method to efficiently produce reversible and inducible gene knockout and rescue in mice. In this system, which we named iKO, the target gene can be turned on and off at will by treating the mice with doxycycline. This method combines two genetically modified mouse lines: a) a KO line with a tetracycline-dependent transactivator replacing the endogenous target gene, and b) a line with a tetracycline-inducible cDNA of the target gene inserted into a tightly regulated (TIGRE) genomic locus, which provides for low basal expression and high inducibility. Such a locus occurs infrequently in the genome and we have developed a method to easily introduce genes into the TIGRE site of mouse embryonic stem (ES) cells by recombinase-mediated insertion. Both KO and TIGRE lines have been engineered for high-throughput, large-scale and cost-effective production of iKO mice. As a proof of concept, we have created iKO mice in the apolipoprotein E (ApoE) gene, which allows for sensitive and quantitative phenotypic analyses. The results demonstrated reversible switching of ApoE transcription, plasma cholesterol levels, and atherosclerosis progression and regression. The iKO system shows stringent regulation and is a versatile genetic system that can easily incorporate other techniques and adapt to a wide range of applications.


Asunto(s)
Expresión Génica , Marcación de Gen , Transgenes , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/patología , Colesterol/sangre , Doxiciclina/administración & dosificación , Doxiciclina/metabolismo , Células Madre Embrionarias/fisiología , Expresión Génica/efectos de los fármacos , Vectores Genéticos/genética , Elementos Aisladores , Ratones , Ratones Transgénicos , Mutagénesis Insercional , Retroviridae/genética , Transactivadores/genética , Transactivadores/metabolismo
9.
Mol Endocrinol ; 21(11): 2795-804, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17666589

RESUMEN

TRH is a neuropeptide with a variety of hormonal and neurotransmitter/neuromodulator functions. In particular, TRH has pronounced acute antidepressant effects in both humans and animals and has been implicated in the mediation of the effects of other antidepressive therapies. Two G protein-coupled receptors, TRH receptor 1 (TRH-R1) and TRH-R2, have been identified. Here we report the generation and phenotypic characterization of mice deficient in TRH-R1. The TRH-R1 knockout mice have central hypothyroidism and mild hyperglycemia but exhibit normal growth and development and normal body weight and food intake. Behaviorally, the TRH-R1 knockout mice display increased anxiety and depression levels while behaving normally in a number of other aspects examined. These results provide the first direct evidence that the endogenous TRH system is involved in mood regulation, and this function is carried out through TRH-R1-mediated neural pathways.


Asunto(s)
Ansiedad/metabolismo , Depresión/metabolismo , Receptores de Hormona Liberadora de Tirotropina/genética , Receptores de Hormona Liberadora de Tirotropina/fisiología , Afecto , Animales , Conducta Animal , Peso Corporal , Hiperglucemia/genética , Hipotiroidismo , Masculino , Ratones , Ratones Noqueados , Modelos Genéticos , Movimiento , Fenotipo
10.
J Neurosci ; 27(33): 8826-35, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17699664

RESUMEN

GPR54 is a G-protein-coupled receptor, which binds kisspeptins and is widely expressed throughout the brain. Kisspeptin-GPR54 signaling has been implicated in the regulation of pubertal and adulthood gonadotropin-releasing hormone (GnRH) secretion, and mutations or deletions of GPR54 cause hypogonadotropic hypogonadism in humans and mice. Other reproductive roles for kisspeptin-GPR54 signaling, including the regulation of developmental GnRH secretion or sexual behavior in adults, have not yet been explored. Using adult wild-type (WT) and GPR54 knock-out (KO) mice, we first tested whether kisspeptin-GPR54 signaling is necessary for male and female sexual behaviors. We found that hormone-replaced gonadectomized GPR54 KO males and females displayed appropriate gender-specific adult sexual behaviors. Next, we examined whether GPR54 signaling is required for proper display of olfactory-mediated partner preference behavior. Testosterone-treated WT males preferred stimulus females rather than males, whereas similarly treated WT females and GPR54 KO males showed no preference for either sex. Because olfactory preference is sexually dimorphic and organized during development by androgens, we assessed whether GPR54 signaling is essential for sexual differentiation of other sexually dimorphic traits. Interestingly, adult testosterone-treated GPR54 KO males displayed "female-like" numbers of tyrosine hydroxylase-immunoreactive and Kiss1 mRNA-containing neurons in the anteroventral periventricular nucleus and likewise possessed fewer motoneurons in the spino-bulbocavernosus nucleus than did WT males. Our findings indicate that kisspeptin-GPR54 signaling is not required for male or female copulatory behavior, provided there is appropriate adulthood hormone replacement. However, GPR54 is necessary for proper male-like development of several sexually dimorphic traits, likely by regulating GnRH-mediated androgen secretion during "critical windows" in perinatal development.


Asunto(s)
Encéfalo/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Diferenciación Sexual/fisiología , Conducta Sexual Animal/fisiología , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Conducta Animal/fisiología , Encéfalo/citología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Kisspeptinas , Hormona Luteinizante/sangre , Masculino , Ratones , Ratones Noqueados , Neuronas/clasificación , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteínas/genética , Proteínas/metabolismo , Receptores Acoplados a Proteínas G/deficiencia , Receptores de Kisspeptina-1 , Caracteres Sexuales , Diferenciación Sexual/efectos de los fármacos , Diferenciación Sexual/genética , Conducta Sexual Animal/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Testosterona/farmacología , Proteínas Supresoras de Tumor/farmacología , Tirosina 3-Monooxigenasa/metabolismo
11.
Pharmacol Biochem Behav ; 86(1): 8-20, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17257664

RESUMEN

The neuropeptide galanin has been implicated in anxiety-related behaviors, cognition, analgesia, and feeding in rodents. Neuromodulatory actions of galanin are mediated by three G-protein coupled receptors, GalR1, GalR2, and GalR3. The present study investigates the role of the GalR2 receptor by evaluating behavioral phenotypes of mice with a targeted mutation in the GalR2 gene. A three-tiered behavioral phenotyping approach first examined control measures of general health, body weight, neurological reflexes, sensory abilities and motor function. Mice were then assessed on several tests for cognitive and anxiety-like behaviors. GalR2 null mutants and heterozygotes were not significantly different from wildtype littermates on two cognitive tests previously shown to be sensitive to galanin manipulation: acquisition of the Morris water maze spatial task, and trace cued and contextual fear conditioning, an emotional learning and memory task. Two independent cohorts of GalR2 null mutant mice demonstrated an anxiogenic-like phenotype in the elevated plus-maze. No genotype differences were detected on several other measures of anxiety-like behavior. The discovery of an anxiogenic phenotype specific to the elevated plus-maze, similar to findings in GalR1 null mutants, highlights the potential therapeutic efficacy of targeting GalR1 and GalR2 receptors in treating anxiety disorders.


Asunto(s)
Ansiedad/genética , Ansiedad/psicología , Receptor de Galanina Tipo 2/genética , Receptor de Galanina Tipo 2/fisiología , Animales , Peso Corporal/fisiología , Condicionamiento Psicológico/fisiología , Conducta Exploratoria/fisiología , Miedo/fisiología , Miedo/psicología , Femenino , Genotipo , Salud , Heterocigoto , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Noqueados , Movimiento/fisiología , Dolor/genética , Dolor/psicología , Dimensión del Dolor , Fenotipo , Equilibrio Postural/fisiología , Reflejo/fisiología , Sensación/fisiología , Estrés Psicológico/genética , Estrés Psicológico/psicología
12.
Mol Cell Biol ; 26(24): 9352-63, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17030627

RESUMEN

Neuromedin U (NMU) is a highly conserved neuropeptide with a variety of physiological functions mediated by two receptors, peripheral NMUR1 and central nervous system NMUR2. Here we report the generation and phenotypic characterization of mice deficient in the central nervous system receptor NMUR2. We show that behavioral effects, such as suppression of food intake, enhanced pain response, and excessive grooming induced by intracerebroventricular NMU administration were abolished in the NMUR2 knockout (KO) mice, establishing a causal role for NMUR2 in mediating NMU's central effects on these behaviors. In contrast to the NMU peptide-deficient mice, NMUR2 KO mice appeared normal with regard to stress, anxiety, body weight regulation, and food consumption. However, the NMUR2 KO mice showed reduced pain sensitivity in both the hot plate and formalin tests. Furthermore, facilitated excitatory synaptic transmission in spinal dorsal horn neurons, a mechanism by which NMU stimulates pain, did not occur in NMUR2 KO mice. These results provide significant insights into a functional dissection of the differential contribution of peripherally or centrally acting NMU system. They suggest that NMUR2 plays a more significant role in central pain processing than other brain functions including stress/anxiety and regulation of feeding.


Asunto(s)
Conducta Alimentaria/fisiología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Dolor/genética , Percepción/fisiología , Receptores de Neurotransmisores/deficiencia , Receptores de Neurotransmisores/genética , Estrés Fisiológico/genética , Animales , Ansiedad/genética , Femenino , Masculino , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Dolor/fisiopatología , Receptores de Neurotransmisores/biosíntesis
13.
Genes Dev ; 18(14): 1725-36, 2004 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15231714

RESUMEN

The floating head (flh) gene in zebrafish encodes a homeodomain protein, which is essential for notochord formation along the entire body axis. flh orthologs, termed Not genes, have been isolated from chick and Xenopus, but no mammalian ortholog has yet been identified. Truncate (tc) is an autosomal recessive mutation in mouse that specifically disrupts the development of the caudal notochord. Here, we demonstrate that truncate arose by a mutation in the mouse Not gene. The truncate allele (Nottc) contains a point mutation in the homeobox of Not that changes a conserved Phenylalanine residue in helix 1 to a Cysteine (F20C), and significantly destabilizes the homeodomain. Reversion of F20C in one allele of homozygous tc embryonic stem (ES) cells is sufficient to restore normal notochord formation in completely ES cell-derived embryos. We have generated a targeted mutation of Not by replacing most of the Not coding sequence, including the homeobox with the eGFP gene. The phenotype of NoteGFP/eGFP, NoteGFP/tc, and Nottc/tc embryos is very similar but slightly more severe in NoteGFP/eGFP than in Nottc/tc embryos. This confirms allelism of truncate and Not, and indicates that tc is not a complete null allele. Not expression is abolished in Foxa2 and T mutant embryos, suggesting that Not acts downstream of both genes during notochord development. This is in contrast to zebrafish embryos, in which flh interacts with ntl (zebrafish T) in a regulatory loop and is essential for development of the entire notochord, and suggests that different genetic control circuits act in different vertebrate species during notochord formation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Genes Homeobox/genética , Proteínas de Homeodominio/genética , Ratones/embriología , Ratones/genética , Notocorda/embriología , Cola (estructura animal)/embriología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Paseo de Cromosoma , Cromosomas Artificiales Bacterianos , Clonación Molecular , Cartilla de ADN , Componentes del Gen , Proteínas Fluorescentes Verdes , Proteínas de Homeodominio/metabolismo , Hibridación in Situ , Proteínas Luminiscentes , Ratones Mutantes , Datos de Secuencia Molecular , Mutación/genética , Filogenia , Análisis de Secuencia de ADN , Análisis Espectral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...