Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Vet Dermatol ; 19(6): 358-67, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19037915

RESUMEN

The mouse In(15)2Rl (hairy ears, Eh) mutation is a paracentric inversion of the distal half of chromosome 15 (Chr 15). Heterozygous Eh/+ mice display misshaped and hairy ears that have more and longer hair than the ears of their wild-type littermates. We mapped, cloned and sequenced both inversion breakpoints. No protein-coding transcript was disrupted by either breakpoint. The proximal breakpoint is located between syntrophin basic 1 (Sntb1) and hyaluronan synthase 2 (Has2), and the distal breakpoint maps between homeobox C4 (Hoxc4) and single-strand selective monofunctional uracil DNA glycosylase (Smug1), near the middle and the telomere ends of Chr 15, respectively. The inversion spans ~47 megabases. Our genetic analysis suggests that the hairy-ear phenotype is caused by the proximal breakpoint of the inversion-bearing Chr 15. Quantitative RNA analysis by real-time polymerase chain reaction for the genes flanking the breakpoint indicated no changes in expression levels except for some homeobox C (Hoxc) genes whose expression was elevated in developing and mature skin of the ears but not of other body regions. The increased hair length on the ears of Eh/+ mice was due to an extension of the anagen stage in the hair cycle, as determined by histological analysis. Our data indicate that the Eh phenotype arises from mis-expression of Hoxc genes.


Asunto(s)
Inversión Cromosómica/genética , Oído/fisiología , Regulación de la Expresión Génica/genética , Cabello/crecimiento & desarrollo , Proteínas de Homeodominio/genética , Animales , Mapeo Cromosómico , Clonación Molecular , Femenino , Genotipo , Cabello/ultraestructura , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Mutación
2.
J Am Soc Nephrol ; 19(10): 1955-64, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18701606

RESUMEN

A screen of recessive mutations generated by the chemical mutagen n-ethyl-n-nitrosourea (ENU) mapped a new mutant locus (5772SB) termed sudden juvenile death syndrome (sjds) to chromosome 7 in mice. These mutant mice, which exhibit severe proximal tubule injury and formation of giant vacuoles in the renal cortex, die from renal failure, a phenotype that resembles aquaporin 11 (Aqp11) knockout mice. In this report, the ENU-induced single-nucleotide variant (sjds mutation) is identified. To determine whether this variant, which causes an amino acid substitution (Cys227Ser) in the predicted E-loop region of aquaporin 11, is responsible for the sjds lethal renal phenotype, Aqp11-/sjds compound heterozygous mice were generated from Aqp11 +/sjds and Aqp11 +/- intercrosses. The compound heterozygous Aqp11 -/sjds offspring exhibited a lethal renal phenotype (renal failure by 2 wk), similar to the Aqp11 sjds/sjds and Aqp11-/- phenotypes. These results demonstrate that the identified mutation causes renal failure in Aqp11 sjds/sjds mutant mice, providing a model for better understanding of the structure and function of aquaporin 11 in renal physiology.


Asunto(s)
Sustitución de Aminoácidos/genética , Acuaporinas/genética , Etilnitrosourea , Mutación Puntual/genética , Insuficiencia Renal/genética , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Mutantes , Polimorfismo de Nucleótido Simple/genética , Síndrome
3.
J Am Soc Nephrol ; 18(1): 103-12, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17151334

RESUMEN

Diabetic nephropathy (DN) is a late diabetic complication that comprises progressively increasing albuminuria, declining GFR, and increased cardiovascular risk. Only a minority of patients with diabetes (25 to 40%) develop nephropathy, and there is evidence that heritable genetic factors predispose these "at-risk" individuals to DN. Comparing variability among inbred mouse strains with respect to a specific phenotype can model interhuman variability, and each strain represents a genetically homogeneous system with a defined risk for nephropathy. C57BL/6 mice, which are relatively resistant to DN, were mutagenized using N-ethyl-N-nitrosourea and screened for mutants that developed excess albuminuria on a sensitizing type 1 diabetic background contributed by the dominant Akita mutation in insulin-2 gene (Ins2(Akita)). Two of 375 diabetic G1 founders were found to exhibit albumin excretion rates persistently 10-fold greater than albumin excretion rates in nonmutagenized Ins2(Akita) controls. This albuminuria trait was heritable and transmitted to approximately 50% of Ins2(Akita) G2 and G3 progeny, consistent with a simple, dominantly inherited trait, but was never observed in nondiabetic offspring. During the course of 1 yr, albuminuric Ins2(Akita) G2 and G3 progeny developed reduced inulin clearance with elevated blood urea nitrogen and plasma creatinine. Glomerular histology revealed mesangial expansion, and glomerular basement membrane thickening as determined by electron microscopy was enhanced in diabetic mutant kidneys. Hereditary albuminuric N-ethyl-N-nitrosourea-induced mutants were redesignated as Nphrp1 (nephropathy1) and Nphrp2 (nephropathy2) mice for two generated lines. These novel mutants provide new, robust mouse models of DN and should help to elucidate the underlying genetic basis of predisposition to DN.


Asunto(s)
Nefropatías Diabéticas/genética , Etilnitrosourea/toxicidad , Mutágenos/toxicidad , Albuminuria/genética , Animales , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/fisiopatología , Modelos Animales de Enfermedad , Femenino , Genes Dominantes , Predisposición Genética a la Enfermedad , Pruebas Genéticas/métodos , Pruebas Genéticas/estadística & datos numéricos , Humanos , Insulina/genética , Riñón/patología , Riñón/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , Fenotipo , Sensibilidad y Especificidad
4.
Hum Mol Genet ; 15(8): 1329-41, 2006 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-16537572

RESUMEN

The mammalian Nell1 gene encodes a protein kinase C-beta1 (PKC-beta1) binding protein that belongs to a new class of cell-signaling molecules controlling cell growth and differentiation. Over-expression of Nell1 in the developing cranial sutures in both human and mouse induces craniosynostosis, the premature fusion of the growing cranial bone fronts. Here, we report the generation, positional cloning and characterization of Nell1(6R), a recessive, neonatal-lethal point mutation in the mouse Nell1 gene, induced by N-ethyl-N-nitrosourea. Nell1(6R) has a T-->A base change that converts a codon for cysteine into a premature stop codon [Cys(502)Ter], resulting in severe truncation of the predicted protein product and marked reduction in steady-state levels of the transcript. In addition to the expected alteration of cranial morphology, Nell1(6R) mutants manifest skeletal defects in the vertebral column and ribcage, revealing a hitherto undefined role for Nell1 in signal transduction in endochondral ossification. Real-time quantitative reverse transcription-PCR assays of 219 genes showed an association between the loss of Nell1 function and reduced expression of genes for extracellular matrix (ECM) proteins critical for chondrogenesis and osteogenesis. Several affected genes are involved in the human cartilage disorder Ehlers-Danlos Syndrome and other disorders associated with spinal curvature anomalies. Nell1(6R) mutant mice are a new tool for elucidating basic mechanisms in osteoblast and chrondrocyte differentiation in the developing skull and vertebral column and understanding how perturbations in the production of ECM proteins can lead to anomalies in these structures.


Asunto(s)
Proteínas de Unión al Calcio/genética , Proteínas de la Matriz Extracelular/metabolismo , Glicoproteínas/genética , Cráneo/anomalías , Columna Vertebral/anomalías , Anomalías Inducidas por Medicamentos/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Calcificación Fisiológica/genética , Proteínas de Unión al Calcio/metabolismo , Moléculas de Adhesión Celular/metabolismo , Condrogénesis , Cisteína/genética , Cisteína/metabolismo , Femenino , Glicoproteínas/metabolismo , Masculino , Ratones , Ratones Endogámicos , Datos de Secuencia Molecular , Osteogénesis , Mutación Puntual , Transducción de Señal , Tirosina/genética , Tirosina/metabolismo
5.
Vis Neurosci ; 22(5): 595-604, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16332270

RESUMEN

The Tennessee Mouse Genome Consortium (TMGC) is in its fifth year of a ethylnitrosourea (ENU)-based mutagenesis screen to detect recessive mutations that affect the eye and brain. Each pedigree is tested by various phenotyping domains including the eye, neurohistology, behavior, aging, ethanol, drug, social behavior, auditory, and epilepsy domains. The utilization of a highly efficient breeding protocol and coordination of various universities across Tennessee makes it possible for mice with ENU-induced mutations to be evaluated by nine distinct phenotyping domains within this large-scale project known as the TMGC. Our goal is to create mutant lines that model human diseases and disease syndromes and to make the mutant mice available to the scientific research community. Within the eye domain, mice are screened for anterior and posterior segment abnormalities using slit-lamp biomicroscopy, indirect ophthalmoscopy, fundus photography, eye weight, histology, and immunohistochemistry. As of January 2005, we have screened 958 pedigrees and 4800 mice, excluding those used in mapping studies. We have thus far identified seven pedigrees with primary ocular abnormalities. Six of the mutant pedigrees have retinal or subretinal aberrations, while the remaining pedigree presents with an abnormal eye size. Continued characterization of these mutant mice should in most cases lead to the identification of the mutated gene, as well as provide insight into the function of each gene. Mice from each of these pedigrees of mutant mice are available for distribution to researchers for independent study.


Asunto(s)
Oftalmopatías/genética , Genoma , Ratones Mutantes/genética , Fenómenos Fisiológicos Oculares , Animales , Cromosomas/genética , Etilnitrosourea/toxicidad , Ojo/patología , Anomalías del Ojo/genética , Anomalías del Ojo/patología , Color del Ojo/genética , Oftalmopatías/patología , Femenino , Genotipo , Inmunohistoquímica , Masculino , Ratones , Ratones Mutantes/clasificación , Mutágenos/toxicidad , Tamaño de los Órganos , Linaje , Fenotipo , Embarazo
6.
BMC Genomics ; 6: 164, 2005 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-16300676

RESUMEN

BACKGROUND: Analysis of an allelic series of point mutations in a gene, generated by N-ethyl-N-nitrosourea (ENU) mutagenesis, is a valuable method for discovering the full scope of its biological function. Here we present an efficient gene-driven approach for identifying ENU-induced point mutations in any gene in C57BL/6J mice. The advantage of such an approach is that it allows one to select any gene of interest in the mouse genome and to go directly from DNA sequence to mutant mice. RESULTS: We produced the Cryopreserved Mutant Mouse Bank (CMMB), which is an archive of DNA, cDNA, tissues, and sperm from 4,000 G1 male offspring of ENU-treated C57BL/6J males mated to untreated C57BL/6J females. Each mouse in the CMMB carries a large number of random heterozygous point mutations throughout the genome. High-throughput Temperature Gradient Capillary Electrophoresis (TGCE) was employed to perform a 32-Mbp sequence-driven screen for mutations in 38 PCR amplicons from 11 genes in DNA and/or cDNA from the CMMB mice. DNA sequence analysis of heteroduplex-forming amplicons identified by TGCE revealed 22 mutations in 10 genes for an overall mutation frequency of 1 in 1.45 Mbp. All 22 mutations are single base pair substitutions, and nine of them (41%) result in nonconservative amino acid substitutions. Intracytoplasmic sperm injection (ICSI) of cryopreserved spermatozoa into B6D2F1 or C57BL/6J ova was used to recover mutant mice for nine of the mutations to date. CONCLUSIONS: The inbred C57BL/6J CMMB, together with TGCE mutation screening and ICSI for the recovery of mutant mice, represents a valuable gene-driven approach for the functional annotation of the mammalian genome and for the generation of mouse models of human genetic diseases. The ability of ENU to induce mutations that cause various types of changes in proteins will provide additional insights into the functions of mammalian proteins that may not be detectable by knockout mutations.


Asunto(s)
Técnicas Genéticas , Mutagénesis , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Animales , Cruzamientos Genéticos , Criopreservación , ADN/metabolismo , Análisis Mutacional de ADN , ADN Complementario/metabolismo , Bases de Datos Genéticas , Etilnitrosourea/farmacología , Femenino , Genotipo , Mutación de Línea Germinal , Homocigoto , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Genéticos , Mutágenos , Mutación , Fenotipo , Mutación Puntual , Reacción en Cadena de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Inyecciones de Esperma Intracitoplasmáticas , Espermatozoides/metabolismo , Distribución Tisular
7.
Mamm Genome ; 16(9): 661-71, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16245023

RESUMEN

Chromosomal deletions have long been used as genetic tools in dissecting the functions of complex genomes, and new methodologies are still being developed to achieve the maximum coverage. In the mouse, where the chromosomal deletion coverage is far less extensive than that in Drosophila, substantial coverage of the genome with deletions is strongly desirable. This article reports the generation of three deletion complexes in the distal part of mouse Chromosome (Chr) 15. Chromosomal deletions were efficiently induced by X rays in embryonic stem (ES) cells around the Otoconin 90 (Oc 90), SRY-box-containing gene 10 (Sox 10), and carnitine palmitoyltransferase 1b (Cpt 1 b) loci. Deletions encompassing the Oc 90 and Sox 10 loci were transmitted to the offspring of the chimeric mice that were generated from deletion-bearing ES cells. Whereas deletion complexes encompassing the Sox 10 and the Cpt 1 b loci overlap each other, no overlap of the Oc 90 complex with the Sox 10 complex was found, possibly indicating the existence of a haploinsufficient gene located between Oc 90 and Sox 10. Deletion frequency and size induced by X rays depend on the selective locus, possibly reflecting the existence of haplolethal genes in the vicinity of these loci that yield fewer and smaller deletions. Deletions induced in ES cells by X rays vary in size and location of breakpoints, which makes them desirable for mapping and for functional genomics studies.


Asunto(s)
Deleción Cromosómica , Cromosomas de los Mamíferos/efectos de la radiación , Ratones/genética , Células Madre/efectos de la radiación , Rayos X , Animales , Proteínas de Unión al Calcio , Cromosomas de los Mamíferos/genética , Proteínas de Unión al ADN/genética , Proteínas de la Matriz Extracelular , Componentes del Gen , Genómica/métodos , Glicoproteínas/genética , Proteínas del Grupo de Alta Movilidad/genética , Hibridación Fluorescente in Situ , Factores de Transcripción SOXE , Factores de Transcripción/genética
8.
Mamm Genome ; 16(8): 555-66, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16180137

RESUMEN

We have used the new high-throughput mutation-scanning technique temperature-gradient capillary electrophoresis (TGCE) for the identification of point mutations induced by N-ethyl-N-nitrosourea (ENU) in the mouse genome. TGCE detects the presence of heteroduplex molecules formed between a wild-type gene segment and the corresponding homologous segment containing an induced mutation or a naturally occurring single nucleotide polymorphism (SNP). Partially denatured heteroduplex molecules are resolved from homoduplexes by virtue of their differential mobilities during capillary electrophoresis conducted in a finely controlled temperature gradient. Simultaneous heteroduplex analysis of 96 amplicons ranging from 150 to 600 bp in size is achieved in approximately 45 min without the need for predetermining the melting profile of each fragment. Initially, we exploited known mouse mutations to develop TGCE protocols for analyzing unpurified PCR samples amplified from crude tail-DNA preparations. TGCE was then applied to the rapid identification of three new ENU-induced mutations recovered from regional mutagenesis screens of a segment of mouse Chromosome 7. Enzyme assays and quantitative reverse transcription-PCR (qRT-PCR) methods validated these new mutations. Our data demonstrate that rapid mutation scanning with TGCE, followed by sequence verification only of detected positives, is an efficient approach to the identification of point mutations in the mouse genome.


Asunto(s)
Cromosomas de los Mamíferos/genética , Análisis Mutacional de ADN , Etilnitrosourea/farmacología , Mutagénesis/efectos de los fármacos , Mutación Puntual/genética , Animales , Secuencia de Bases , Cromatografía Líquida de Alta Presión , Electroforesis Capilar , Análisis Heterodúplex , Isoenzimas/genética , L-Lactato Deshidrogenasa/genética , Ratones , Ratones Endogámicos BALB C , Oxigenasas/genética , Fenotipo , Proteína Amiloide A Sérica/genética
9.
Mol Vis ; 11: 569-81, 2005 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-16088326

RESUMEN

PURPOSE: The 44TNJ mutant mouse was generated by the Tennessee Mouse Genome Consortium (TMGC) using an ENU-based mutagenesis screen to produce recessive mutations that affect the eye and brain. Herein we present its retinal phenotype and genetic basis. METHODS: Fourth generation offspring (G4) and confirmed mutants were examined using slit lamp biomicroscopy, funduscopy, histology, immunohistochemistry, and electroretinography (ERG). 44TNJ mutant mice were crossed to C3BLiA or DBA/2 mice for chromosomal mapping purposes. Linkage analysis by PCR-based microsatellite marker genotyping was used to identify the disease locus. The Rs1h cDNA and its genomic DNA were sequenced directly. RESULTS: The 44TNJ pedigree was the first mutant pedigree identified by the ocular phenotyping domain of the TMGC. Examination of the fundus revealed numerous small and homogeneous intraretinal microflecks in the peripapillary region, which became courser and more irregular in the periphery. Males were typically more affected than females. Histology and immunohistochemistry revealed a disruption of the lamination of the retina, particularly at both margins of the outer nuclear layer, along with reduced calbindin immunostaining. ERG analyses revealed reduced amplitudes of both a-waves and b-waves. Linkage analysis mapped the 44TNJ mutation to the X chromosome close to the marker DXMit117. Sequence analysis of the positional candidate gene Rs1h revealed a T->C exchange at the second base of intron 2 of the Rs1h gene. CONCLUSIONS: We have generated and characterized a mutant mouse line that was produced using ENU-based mutagenesis. The 44TNJ pedigree manifests with photoreceptor dysfunction and concurrent structural and functional aberrations at the post-receptoral level. Genetic analysis revealed a mutation in Rs1h, making this the first murine model of X-linked retinoschisis in which the gene is expressed.


Asunto(s)
Alquilantes/toxicidad , Moléculas de Adhesión Celular/genética , Modelos Animales de Enfermedad , Etilnitrosourea/toxicidad , Proteínas del Ojo/genética , Mutación/efectos de los fármacos , Retina/fisiopatología , Retinosquisis/genética , Animales , Secuencia de Bases , Mapeo Cromosómico , Análisis Mutacional de ADN , Electrorretinografía , Femenino , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Mutantes , Datos de Secuencia Molecular , Mutagénesis , Retinosquisis/fisiopatología
10.
Age (Dordr) ; 27(1): 75-90, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23598606

RESUMEN

With the goal of discovering genes that contribute to late-onset neurological and ocular disorders and also genes that extend the healthy life span in mammals, we are phenotyping mice carrying new mutations induced by the chemical N-ethyl-N-nitrosourea (ENU). The phenotyping plan includes basic behavioral, neurohistological, and vision testing in sibling cohorts of mice aged to 18 months, and then evaluation for markers of growth trajectory and stress response in these same cohorts aged up to 28 months. Statistical outliers are identified by comparison to test results of similar aged cohorts, and potential mutants are recovered for re-aging to confirm heritability of the phenotype.

11.
Genetics ; 167(2): 889-95, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15238537

RESUMEN

Chromosomal inversions are valuable genetic tools for mutagenesis screens, where appropriately marked inversions can be used as balancer chromosomes to recover and maintain mutations in the corresponding chromosomal region. For any inversion to be effective as a balancer, it should exhibit both dominant and recessive visible traits; ideally the recessive trait should be a fully penetrant lethality in which inversion homozygotes die before birth. Unfortunately, most inversions recovered by classical radiation or chemical mutagenesis techniques do not have an overt phenotype in either the heterozygous or the homozygous state. However, they can be modified by relatively simple procedures to make them suitable as an appropriately marked balancer. We have used homologous recombination to modify, in embryonic stem cells, the recessive-lethal In(15)21Rk inversion to endow it with a dominant-visible phenotype. Several ES cell lines were derived from inversion heterozygotes, and a keratin-14 (K14) promoter-driven agouti minigene was introduced onto the inverted chromosome 15 in the ES cells by gene targeting. Mice derived from the targeted ES cells carry the inverted chromosome 15 and, at the same time, exhibit lighter coat color on their ears and tails, making this modified In(15)21Rk useful as a balancer for proximal mouse chromosome 15.


Asunto(s)
Inversión Cromosómica/genética , Mapeo Cromosómico , Animales , Exones/genética , Ingeniería Genética , Heterocigoto , Homocigoto , Ratones , Mutagénesis , Mutagénesis Insercional
12.
Proc Natl Acad Sci U S A ; 100(14): 8360-5, 2003 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-12832620

RESUMEN

Recessive N-ethyl-N-nitrosourea (ENU)-induced mutations recovered at the fitness-1 (fit1) locus in mouse chromosome 7 cause hematopoietic abnormalities, growth retardation, and shortened life span, with varying severity of the defects in different alleles. Abnormal iron distribution and metabolism and frequent scoliosis have also been associated with an allele of intermediate severity (fit14R). We report that fit14R, as well as the most severe fit15R allele, are nonsense point mutations in the mouse ortholog of the human phosphatidylinositol-binding clathrin assembly protein (PICALM) gene, whose product is involved in clathrin-mediated endocytosis. A variety of leukemias and lymphomas have been associated with translocations that fuse human PICALM with the putative transcription factor gene AF10. The Picalmfit1-5R and Picalmfit1-4R mutations are splice-donor alterations resulting in transcripts that are less abundant than normal and missing exons 4 and 17, respectively. These exon deletions introduce premature termination codons predicted to truncate the proteins near the N and C termini, respectively. No mutations in the genes encoding Picalm, clathrin, or components of the adaptor protein complex 2 (AP2) have been previously described in which the suite of disorders present in the Picalmfit1 mutant mice is apparent. These mutants thus provide unique models for exploring how the endocytic function of mouse Picalm and the transport processes mediated by clathrin and the AP2 complex contribute to normal hematopoiesis, iron metabolism, and growth.


Asunto(s)
Hematopoyesis/genética , Hierro/metabolismo , Ratones Mutantes/genética , Proteínas de Ensamble de Clatrina Monoméricas/genética , Sitios de Empalme de ARN/genética , Alelos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Codón sin Sentido , Endocitosis , Exones/genética , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Proteínas de Ensamble de Clatrina Monoméricas/deficiencia , Proteínas de Ensamble de Clatrina Monoméricas/fisiología , Mutación Missense , Mutación Puntual
13.
Proc Natl Acad Sci U S A ; 99(2): 844-9, 2002 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-11792855

RESUMEN

Eleven independent, recessive, N-ethyl-N-nitrosourea-induced mutations that map to a approximately 1- to 2-cM region of mouse chromosome (Chr) 7 homologous to human Chr 11p14-p15 were recovered from a screen of 1,218 gametes. These mutations were initially identified in a hemizygous state opposite a large p-locus deletion and subsequently were mapped to finer genomic intervals by crosses to a panel of smaller p deletions. The 11 mutations also were classified into seven complementation groups by pairwise crosses. Four complementation groups were defined by seven prenatally lethal mutations, including a group (l7R3) comprised of two alleles of obvious differing severity. Two allelic mutations (at the psrt locus) result in a severe seizure and runting syndrome, but one mutation (at the fit2 locus) results in a more benign runting phenotype. This experiment has added seven loci, defined by phenotypes of presumed point mutations, to the genetic map of a small (1-2 cM) region of mouse Chr 7 and will facilitate the task of functional annotation of DNA sequence and transcription maps both in the mouse and the corresponding human 11p14-p15 homology region.


Asunto(s)
Mapeo Cromosómico , Cromosomas Humanos Par 11/genética , ADN/genética , Mutación , Animales , Cruzamientos Genéticos , Análisis Mutacional de ADN , Etilnitrosourea/toxicidad , Femenino , Eliminación de Gen , Genes Letales , Genes Recesivos , Prueba de Complementación Genética , Humanos , Masculino , Ratones , Ratones Mutantes , Mutagénesis , Mutágenos/toxicidad , Fenotipo , Especificidad de la Especie
14.
Oncogene ; 21(2): 299-306, 2002 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-11803473

RESUMEN

The mouse Polycomb-group gene, embryonic ectoderm development (eed), appears to regulate cellular growth and differentiation in a developmental and tissue specific manner. During embryogenesis, eed regulates axial patterning, whereas in the adult eed represses proliferation of myeloid and B cell precursors. The present report demonstrates two novel functional activities of eed: alteration of thymocyte maturation and suppression of thymic lymphoma development. Mice that inherit the viable hypomorphic 17Rn5(1989SB) eed allele sustain a partial developmental block at or before the CD4(-)CD8(-)CD44(-)CD25(+) stage of thymocyte differentiation. Furthermore, mice that are homozygous or heterozygous for the hypomorphic eed allele have an increased incidence and decreased latency of N-methyl-N-nitrosourea-induced thymic lymphoma compared to wild-type littermates. These findings support the notion that Polycomb-group genes exert pleiotropic effects dictated by developmental stage and cellular context.


Asunto(s)
Carcinógenos/toxicidad , Linfoma de Células T/genética , Proteínas Represoras/metabolismo , Linfocitos T/citología , Animales , Diferenciación Celular/genética , Linfoma de Células T/inducido químicamente , Linfoma de Células T/prevención & control , Metilnitrosourea/toxicidad , Ratones , Ratones Mutantes , Mutagénesis , Complejo Represivo Polycomb 2 , Proteínas Represoras/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...