Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Dis Model Mech ; 12(6)2019 06 17.
Article in English | MEDLINE | ID: mdl-31101663

ABSTRACT

In a screen for organogenesis defects in N-ethyl-N-nitrosourea (ENU)-induced mutant mice, we discovered a line carrying a mutation in Colgalt1 [collagen beta(1-O)galactosyltransferase type 1], which is required for proper galactosylation of hydroxylysine residues in a number of collagens. Colgalt1 mutant embryos have not been previously characterized; here, we show that they exhibit skeletal and muscular defects. Analysis of mutant-derived embryonic fibroblasts reveals that COLGALT1 acts on collagen IV and VI, and, while collagen VI appears stable and its secretion is not affected, collagen IV accumulates inside of cells and within the extracellular matrix, possibly due to instability and increased degradation. We also generated mutant zebrafish that do not express the duplicated orthologs of mammalian Colgalt1 The double-homozygote mutants have muscle defects; they are viable through the larvae stage but do not survive to 10 days post-fertilization. We hypothesize that the Colgalt1 mutant could serve as a model of a human connective tissue disorder and/or congenital muscular dystrophy or myopathy.


Subject(s)
Collagen/metabolism , Galactosyltransferases/deficiency , Loss of Function Mutation/genetics , Musculoskeletal System/pathology , Protein Processing, Post-Translational , Zebrafish Proteins/metabolism , Alleles , Animals , Embryo, Mammalian/pathology , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Galactosyltransferases/metabolism , Glycosylation , Mice , Molecular Weight , Muscles/metabolism , Muscles/pathology , Mutation, Missense/genetics , Phenotype , Skin/metabolism , Skin/pathology , Zebrafish
2.
G3 (Bethesda) ; 8(2): 401-409, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29208648

ABSTRACT

Positional cloning of ENU-induced mutations has traditionally relied on analysis of polymorphic variation between two strains. In contrast, the application of whole-genome sequencing (WGS) has enabled gene discovery in mutant lines maintained on an inbred genetic background. This approach utilizes genetic variation derived from ENU-induced variants for mapping and reduces the likelihood of phenotypic variation, making it an ideal method for genetic modifier screening. Here, we describe the results of such a screen, wherein we determined the minimal number of mutant genomic DNA samples to include in our analyses and improved the sensitivity of our screen by individually barcoding each genomic DNA library. We present several unique cases to illustrate this approach's efficacy, including the discovery of two distinct mutations that generate essentially identical mutant phenotypes, the ascertainment of a non-ENU-induced candidate variant through homozygosity mapping, and an approach for the identification of putative dominant genetic modifiers.


Subject(s)
Chromosome Mapping/methods , Genes, Dominant/genetics , Genomics/methods , Mutation , Alkylating Agents/toxicity , Animals , Embryo, Mammalian/drug effects , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Ethylnitrosourea/toxicity , Female , Genotype , Male , Mice, Inbred C57BL , Mutagenesis/drug effects , Phenotype
3.
BMC Genomics ; 16: 913, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26552429

ABSTRACT

BACKGROUND: The development of powerful new methods for DNA sequencing enable the discovery of sequence variants, their utilization for the mapping of mutant loci, and the identification of causal variants in a single step. We have applied this approach for the analysis of ENU-mutagenized mice maintained on an inbred background. RESULTS: We ascertained ENU-induced variants in four different phenotypically mutant lines. These were then used as informative markers for positional cloning of the mutated genes. We tested both whole genome (WGS) and whole exome (WES) datasets. CONCLUSION: Both approaches were successful as a means to localize a region of homozygosity, as well as identifying mutations of candidate genes, which could be individually assessed. As expected, the WGS strategy was more reliable, since many more ENU-induced variants were ascertained.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Animals , Exome/genetics , Female , Male , Mice , Mutation/genetics , Pedigree
4.
PLoS Genet ; 11(10): e1005569, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26496357

ABSTRACT

Skeletal dysplasias are a common, genetically heterogeneous cause of short stature that can result from disruptions in many cellular processes. We report the identification of the lesion responsible for skeletal dysplasia and male infertility in the spontaneous, recessive mouse mutant chagun. We determined that Poc1a, encoding protein of the centriole 1a, is disrupted by the insertion of a processed Cenpw cDNA, which is flanked by target site duplications, suggestive of a LINE-1 retrotransposon-mediated event. Mutant fibroblasts have impaired cilia formation and multipolar spindles. Male infertility is caused by defective spermatogenesis early in meiosis and progressive germ cell loss. Spermatogonial stem cell transplantation studies revealed that Poc1a is essential for normal function of both Sertoli cells and germ cells. The proliferative zone of the growth plate is small and disorganized because chondrocytes fail to re-align after cell division and undergo increased apoptosis. Poc1a and several other genes associated with centrosome function can affect the skeleton and lead to skeletal dysplasias and primordial dwarfisms. This mouse mutant reveals how centrosome dysfunction contributes to defects in skeletal growth and male infertility.


Subject(s)
Cytoskeletal Proteins/genetics , Dwarfism/genetics , Infertility, Male/genetics , Long Interspersed Nucleotide Elements/genetics , Spermatogenesis/genetics , Animals , Cell Cycle Proteins , Centrioles/genetics , Centrosome/metabolism , Chromosomal Proteins, Non-Histone/genetics , Dwarfism/pathology , Humans , Infertility, Male/pathology , Male , Meiosis/genetics , Mice , Proteins/genetics , Proteins/metabolism , Sertoli Cells/metabolism , Spermatogonia/metabolism
5.
Biol Reprod ; 93(5): 121, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26333994

ABSTRACT

Chromatin remodeling influences gene expression in developing and adult organisms. Active and repressive marks of histone methylation dictate the embryonic expression boundaries of developmentally regulated genes, including the Hox gene cluster. Drosophila ash1 (absent, small or homeotic discs 1) gene encodes a histone methyltransferase essential for regulation of Hox gene expression that interacts genetically with other members of the trithorax group (TrxG). While mammalian members of the mixed lineage leukemia (Mll) family of TrxG genes have roles in regulation of Hox gene expression, little is known about the expression and function of the mammalian ortholog of the Drosophila ash1 gene, Ash1-like (Ash1l). Here we report the expression of mouse Ash1l gene in specific structures within various organs and provide evidence that reduced Ash1l expression has tissue-specific effects on mammalian development and adult homeostasis. Mutants exhibit partially penetrant postnatal lethality and failure to thrive. Surviving mutants have growth insufficiency, skeletal transformations, and infertility associated with developmental defects in both male and female reproductive organs. Specifically, expression of Hoxa11 and Hoxd10 are altered in the epididymis of Ash1l mutant males and Hoxa10 is reduced in the uterus of Ash1l mutant females. In summary, we show that the histone methyltransferase Ash1l is important for the development and function of several tissues and for proper expression of homeotic genes in mammals.


Subject(s)
DNA-Binding Proteins/deficiency , Epididymis/abnormalities , Fertility , Transcription Factors/deficiency , Uterus/abnormalities , Alleles , Animals , DNA-Binding Proteins/genetics , Epididymis/metabolism , Female , Genes, Homeobox , Histone-Lysine N-Methyltransferase , Homeodomain Proteins/metabolism , Male , Mice, Inbred C57BL , Transcription Factors/genetics
6.
Annu Rev Genomics Hum Genet ; 16: 199-227, 2015.
Article in English | MEDLINE | ID: mdl-25939055

ABSTRACT

Skeletal dysplasias result from disruptions in normal skeletal growth and development and are a major contributor to severe short stature. They occur in approximately 1/5,000 births, and some are lethal. Since the most recent publication of the Nosology and Classification of Genetic Skeletal Disorders, genetic causes of 56 skeletal disorders have been uncovered. This remarkable rate of discovery is largely due to the expanded use of high-throughput genomic technologies. In this review, we discuss these recent discoveries and our understanding of the molecular mechanisms behind these skeletal dysplasia phenotypes. We also cover potential therapies, unusual genetic mechanisms, and novel skeletal syndromes both with and without known genetic causes. The acceleration of skeletal dysplasia genetics is truly spectacular, and these advances hold great promise for diagnostics, risk prediction, and therapeutic design.


Subject(s)
Bone Diseases, Developmental/genetics , Mutation , Animals , Body Height/genetics , Disease Models, Animal , Dwarfism/genetics , Epigenesis, Genetic , High-Throughput Nucleotide Sequencing/methods , Histone Acetyltransferases/genetics , Humans , Mice , MicroRNAs , Osteochondrodysplasias/genetics , Proteus Syndrome/genetics
7.
Hum Mol Genet ; 22(2): 345-57, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23065701

ABSTRACT

We discovered a new spontaneous mutant allele of Npr2 named peewee (pwe) that exhibits severe disproportionate dwarfism and female infertility. The pwe phenotype is caused by a four base-pair deletion in exon 3 that generates a premature stop codon at codon 313 (L313X). The Npr2(pwe/pwe) mouse is a model for the human skeletal dysplasia acromesomelic dysplasia, Maroteaux type (AMDM). We conducted a thorough analysis of the female reproductive tract and report that the primary cause of Npr2(pwe/pwe) female infertility is premature oocyte meiotic resumption, while the pituitary and uterus appear to be normal. Npr2 is expressed in chondrocytes and osteoblasts. We determined that the loss of Npr2 causes a reduction in the hypertrophic and proliferative zones of the growth plate, but mineralization of skeletal elements is normal. Mutant tibiae have increased levels of the activated form of ERK1/2, consistent with the idea that natriuretic peptide receptor type 2 (NPR2) signaling inhibits the activation of the MEK/ERK mitogen activated protein kinase pathway. Treatment of fetal tibiae explants with mitogen activated protein kinase 1 and 2 inhibitors U0126 and PD325901 rescues the Npr2(pwe/pwe) growth defect, providing a promising foundation for skeletal dysplasia therapeutics.


Subject(s)
Bone Diseases, Developmental/genetics , Mutation , Receptors, Atrial Natriuretic Factor/genetics , Reproduction/genetics , Animals , Base Sequence , Bone Density/genetics , Bone Diseases, Developmental/drug therapy , Bone and Bones/metabolism , Dwarfism/genetics , Female , Genotype , Humans , Infertility, Female/genetics , MAP Kinase Signaling System/drug effects , Male , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phenotype , Phosphorylation/drug effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...