Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Oncogene ; 35(25): 3335-41, 2016 06 23.
Article in English | MEDLINE | ID: mdl-26477318

ABSTRACT

Cadherin subtype switching from E-cadherin to N-cadherin is associated with the epithelial-to-mesenchymal transition (EMT), a process required for invasion and dissemination of carcinoma cells. We found that N-cadherin is expressed in human and mouse pancreatic intraepithelial neoplasia (PanIN), suggesting that N-cadherin may also have a role in early-stage pancreatic cancer. To investigate the role of N-cadherin in mouse PanIN (mPanIN), we simultaneously activated oncogenic K-ras(G12D) and deleted the N-cadherin (Cdh2) gene in the murine pancreas. Genetic ablation of N-cadherin (N-cad KO) caused hyperproliferation, accelerated mPanIN progression, and early tumor development in K-ras(G12D) mice. Decreased E-cadherin and redistribution of ß-catenin accompanied the loss of N-cadherin in pancreatic ductal epithelial cells (PDEC). Nuclear accumulation of ß-catenin and its transcription co-activator Tcf4 led to activation of Wnt/ß-catenin target genes. Unexpectedly, loss of N-cadherin in the K-ras(G12D) model resulted in increased mPanIN progression and tumor incidence. These in vivo results demonstrate for the first time that N-cadherin functions as a growth suppressor in the context of oncogenic K-ras.


Subject(s)
Cadherins/genetics , Cell Proliferation/genetics , Pancreatic Neoplasms/genetics , Precancerous Conditions/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Cadherins/metabolism , Cell Line , Disease Models, Animal , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Humans , Immunoblotting , Immunohistochemistry , Mice, Knockout , Mice, Transgenic , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/genetics , beta Catenin/metabolism
2.
Oncogene ; 31(41): 4484-9, 2012 Oct 11.
Article in English | MEDLINE | ID: mdl-22158044

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is often detected at a late stage, hence the identification of new therapies that have potential to block tumor progression is critical for this lethal disease. N-cadherin upregulation has been observed in many cancers including PDA, however, a causal role for this cell adhesion receptor in disease progression has yet to be defined. The concomitant expression of oncogenic Kras(G12D) and mutant p53 (Trp53(R172H)) in the murine pancreas results in metastatic PDA that recapitulates the cognate features of human pancreatic cancer providing an excellent animal model to identify genes required for tumor progression. Here we determine the consequences of genetically manipulating N-cadherin expression in a mouse model of PDA. Remarkably, mice with reduced N-cadherin expression (that is, Ncad(-/+)) survived 25% longer (177 vs 142 days, P<0.05) than animals expressing two wild-type N-cadherin (Cdh2) alleles. The survival benefit is likely due to a cumulative effect of N-cadherin's role in different aspects of tumorigenesis including tumor-cell survival, growth, migration and invasion. Interestingly, reduced hedgehog signaling may contribute to the better prognosis for the Ncad(-/+) mice. Moreover, the matrix metalloproteinase MMP-7, associated with poor prognosis in PDA, was reduced in Ncad(-/+) tumors. Finally, Ncad(-/+) tumor cells exhibited decreased FGF-stimulated ERK1/2 activation consistent with N-cadherin's ability to promote FGFR signaling. These data support a critical role for N-cadherin in PDA and its potential prognostic value. Additionally, this study provides in vivo genetic evidence that the cell-surface protein N-cadherin represents a promising therapeutic target for the treatment of pancreatic cancer.


Subject(s)
Cadherins/genetics , Carcinoma, Pancreatic Ductal/genetics , Haploinsufficiency , Pancreatic Neoplasms/genetics , Animals , Apoptosis , Cadherins/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Caspase 3/metabolism , Cell Proliferation , Cell Survival , Fibroblast Growth Factors/physiology , Humans , Kaplan-Meier Estimate , Matrix Metalloproteinase 7/metabolism , Mice , Mice, Transgenic , Mutation, Missense , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
3.
J Cell Biol ; 154(1): 217-30, 2001 Jul 09.
Article in English | MEDLINE | ID: mdl-11449002

ABSTRACT

Connexin 43 (Cx43alpha1) gap junction has been shown to have an essential role in mediating functional coupling of neural crest cells and in modulating neural crest cell migration. Here, we showed that N-cadherin and wnt1 are required for efficient dye coupling but not for the expression of Cx43alpha1 gap junctions in neural crest cells. Cell motility was found to be altered in the N-cadherin-deficient neural crest cells, but the alterations were different from that elicited by Cx43alpha1 deficiency. In contrast, wnt1-deficient neural crest cells showed no discernible change in cell motility. These observations suggest that dye coupling may not be a good measure of gap junction communication relevant to motility. Alternatively, Cx43alpha1 may serve a novel function in motility. We observed that p120 catenin (p120ctn), an Armadillo protein known to modulate cell motility, is colocalized not only with N-cadherin but also with Cx43alpha1. Moreover, the subcellular distribution of p120ctn was altered with N-cadherin or Cx43alpha1 deficiency. Based on these findings, we propose a model in which Cx43alpha1 and N-cadherin may modulate neural crest cell motility by engaging in a dynamic cross-talk with the cell's locomotory apparatus through p120ctn signaling.


Subject(s)
Cadherins/metabolism , Connexin 43/metabolism , Gap Junctions , Neural Crest/cytology , Zebrafish Proteins , Animals , Bromodeoxyuridine/metabolism , Catenins , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/physiology , Cell Communication , Cell Division , Cell Movement , Cells, Cultured , Genotype , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Video , Phosphoproteins/metabolism , Phosphoproteins/physiology , Protein Binding , Proto-Oncogene Proteins/metabolism , Time Factors , Wnt Proteins , Wnt1 Protein , Delta Catenin
4.
Dev Biol ; 234(1): 72-9, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11356020

ABSTRACT

Cadherin-mediated cell-cell interactions are thought to be critical in controlling cell sorting during embryogenesis. Here, we report that chimeric embryos generated with N-cadherin-deficient (N-cadherin(-/-)) embryonic stem cells develop further than embryos completely lacking N-cadherin only when the myocardium consists of N-cadherin-positive cells. Initially, the N-cadherin-negative and -positive cells mix together to form chimeric tissues; however, by embryonic day 9.5, the N-cadherin(-/-) cells segregate from the wild-type cells forming distinct aggregates. The chimeric embryos have large aggregates of N-cadherin(-/-) myocardial cells in the heart lumen, indicating that the cells are unable to maintain cell-cell contacts with N-cadherin-positive myocytes. This sorting-out phenomenon also is apparent in somites, neural tube, and developing brain where N-cadherin(-/-) cells form distinct lumenal structures. These studies demonstrate that N-cadherin-mediated adhesion is critical for maintaining cell-cell interactions in tissues undergoing active cellular rearrangements and increased mechanical stress associated with morphogenesis.


Subject(s)
Cadherins/metabolism , Morphogenesis , Animals , Cadherins/genetics , Cell Adhesion , Chimera , Embryonic and Fetal Development , Heart/embryology , Mice , Myocardium/cytology , Stem Cells
5.
Cell Commun Adhes ; 8(4-6): 321-4, 2001.
Article in English | MEDLINE | ID: mdl-12064611

ABSTRACT

Our previous studies showed an essential role for connexin 43 or alpha1 connexin (Cx43alpha1) gap junctions in the modulation of neural crest cell motility. Cx43alpha1 gap junctions and N-cadherin containing adherens junctions are expressed in migrating cardiac neural crest cells. Analysis of the N-cadherin knockout (KO) mouse model revealed that N-cadherin is essential for gap junction mediated dye coupling but not for expression of Cx43alpha1 gap junctions in neural crest cells. Time lapse videomicroscopy and motion analysis showed that the motility of N-cadherin KO neural crest cells were altered, but the motility changes differed compared to Cx43alpha1 KO neural crest cells. These observations suggest that the role of N-cadherin in cell motility is not simply mediated via the modulation of Cx43alpha1 mediated cell-cell communication. This was confirmed by a parallel analysis of wnt-1 deficient neural crest cells, which also showed a reduction in dye coupling, and yet no change in cell motility. Analysis of p120 catenin (p120ctn), an Amardillo family protein known to play a role in cell motility, showed that it is colocalized with N-cadherin and Cx43alpha1 in migrating neural crest cells. This subcellular distribution was altered in the N-cadherin and Cx43alpha1 KO neural crest cells. Given these results, we propose that N-cadherin and Cx43alpha1 may modulate neural crest cell motility by engaging in a dynamic cross-talk with the cell's locomotory apparatus through p120ctn signaling.


Subject(s)
Cadherins/metabolism , Cell Movement/physiology , Connexin 43/metabolism , Neural Crest/physiology , Zebrafish Proteins , Animals , Catenins , Cell Adhesion Molecules/metabolism , Cell Communication/physiology , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Gap Junctions/metabolism , Heart/embryology , Mice , Microscopy, Video , Neural Crest/cytology , Phosphoproteins/metabolism , Proto-Oncogene Proteins/metabolism , Time Factors , Wnt Proteins , Wnt1 Protein , Delta Catenin
6.
Dev Biol ; 238(1): 202-12, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11784004

ABSTRACT

Members of the cadherin family of cell adhesion molecules are thought to be crucial regulators of tissue patterning and organogenesis. During pancreatic ontogeny N-cadherin is initially expressed in the pancreatic mesenchyme and later in pancreatic endoderm. Analysis of N-cadherin-deficient mice revealed that these mice suffer from selective agenesis of the dorsal pancreas. Further analysis demonstrated that the mechanism for the lack of a dorsal pancreas involves an essential function of N-cadherin as a survival factor in the dorsal pancreatic mesenchyme.


Subject(s)
Cadherins/genetics , Cadherins/physiology , Pancreas/metabolism , Animals , Apoptosis , Cell Adhesion , Cell Differentiation , Endoderm/metabolism , Genotype , Heterozygote , Immunohistochemistry , In Situ Hybridization , In Situ Nick-End Labeling , Mesoderm/metabolism , Mice , Mice, Transgenic , Microscopy, Electron , Mutation , Pancreas/embryology , Time Factors , Tissue Distribution
7.
J Invest Dermatol ; 114(5): 948-52, 2000 May.
Article in English | MEDLINE | ID: mdl-10771476

ABSTRACT

To investigate the cooperativity of different cell adhesion molecules in maintaining the structural integrity of the epidermis, we have generated mice deficient for both a classical cadherin, P-cadherin, and a desmosomal cadherin, desmoglein 3. In epithelial cells, P-cadherin is localized to the adherens junction, whereas desmoglein 3 is found in desmosomes. Previous studies have shown that these two junctional complexes are important for keratinocyte cell-cell adhesion. Both P-cadherin and desmoglein 3 expression are restricted to the basal and most immediate suprabasal cells of the epidermis, whereas both proteins are found throughout the oral mucosal epithelium. Although P-cadherin mutant mice have no apparent defect in epithelial cell adhesion, the desmoglein 3 mutant phenotype resembles that of patients with the autoimmune disease pemphigus vulgaris, in that the mice develop spontaneous mucous membrane blisters and trauma-induced skin blisters. The oral lesions in DSG3-/- mice reduce their food intake, resulting in a runted phenotype; however, most animals recover and live past weaning age. In contrast, animals mutant for both P-cadherin and desmoglein 3 die before weaning. The majority of the double mutant animals die around 1 wk after birth, apparently due to malnutrition. These studies suggest that loss of P-cadherin leads to a more severe desmoglein 3 mutant phenotype in the double knockout mice. This is the first in vivo evidence of possible synergism between a classical and desmosomal cadherin.


Subject(s)
Cadherins/physiology , Epidermis/physiology , Homeostasis , Animals , Animals, Newborn , Cadherins/analysis , Cadherins/genetics , Desmoglein 3 , Female , Lactation , Mice , Mice, Knockout , Mouth Mucosa/chemistry
8.
Mamm Genome ; 10(10): 959, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10501960
9.
Am J Pathol ; 154(5): 1391-406, 1999 May.
Article in English | MEDLINE | ID: mdl-10329592

ABSTRACT

Studies suggest that cell-cell interactions may regulate apoptosis, and in particular, the calcium-dependent cell adhesion molecule N-cadherin has been shown to be capable of modulating this process. Rat granulosa cells (GCs) are known to express N-cadherin whereas cAMP is known to induce apoptosis in human and rat GCs. Based on these observations, we hypothesized that N-cadherin regulates human GC apoptosis via a cAMP-dependent mechanism. N-cadherin expression was evaluated in ovarian follicles and corpora lutea utilizing immunohistochemical techniques and in luteinized GCs in culture using immunoblotting, flow cytometric analysis, immunohistochemistry, and indirect immunofluorescence techniques utilizing anti-N-cadherin antibodies directed against both the extracellular and cytoplasmic domains of the molecule. Apoptosis was assessed by TUNEL and DNA fragmentation analysis and confirmed by flow cytometric cell cycle analysis and electron microscopy. The rate of GC apoptosis was found to be two- to three-fold lower among aggregated cells, as compared with single cells. N-cadherin was found to be expressed by aggregating GCs in vitro and GCs cultured in the presence of either N-cadherin function disrupting antibodies or peptides exhibiting enhanced rates of apoptosis. GCs in situ stained intensely for N-cadherin in preantral and normal growing preovulatory follicles as well as early corpora lutea. N-cadherin was weak in atretic follicles and regressing corpora lutea. Exposure of GCs to cAMP increased apoptosis while decreasing N-cadherin protein expression in a dose-dependent manner. Cell culture under serum-free conditions increased apoptosis and decreased N-cadherin expression, in part through cleavage of the extracellular domain of the molecule. The metalloproteinase inhibitor 1-10-phenanthroline inhibited the cleavage of the extracellular domain of N-cadherin and concomitantly inhibited the serum-deprivation-induced apoptosis of aggregated GCs. Collectively, these observations suggest that down-regulation of N-cadherin or the absence of a functional extracellular domain of the molecule prevents cell aggregation and is associated with GC apoptosis. In addition, cAMP induces apoptosis in a dose-dependent manner, and this process is dependent, at least in part, on regulation of the N-cadherin molecule at the surface of the cells. We conclude that N-cadherin-mediated GC signaling plays a central role in follicular and luteal cell survival.


Subject(s)
Apoptosis/physiology , Cadherins/physiology , Follicular Atresia/physiology , Granulosa Cells/pathology , Luteolytic Agents/metabolism , Adult , Cell Adhesion/physiology , Cell Aggregation/physiology , Cells, Cultured , Cyclic AMP/physiology , Down-Regulation , Female , Humans , In Situ Nick-End Labeling , Protein Structure, Tertiary
10.
Int J Dev Biol ; 43(8): 831-4, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10707907

ABSTRACT

Many mutations of the murine genome are recessive embryonic lethals precluding phenotype analysis at subsequent stages of development. This is true for embryos genetically lacking either N-cadherin or N- and P-cadherin. To circumvent this, we have generated pluripotent embryonal stem (ES) cells of the same genotype in vitro and differentiated them in vivo in the form of teratomas. All of the ES cells isolated in this study had a normal ES cell morphology in vitro and were able to generate teratomas. Histological analysis revealed that some differentiation and histogenesis had occurred within the teratomas. Epithelial formation was, for example, unaffected in all cadherin null cells. Surprisingly, however, the differentiation of cells lacking both N- and P-cadherin was, in general, even more pronounced both quantitatively and qualitatively. Tumours lacking either N- cadherin or N- and P-cadherin contained more striated muscle (apparently cardiac muscle) than heterozygote controls, and this was most strikingly conspicuous in teratomas from N- and P-cadherin null cells. This more pronounced differentiation was not seen for all tissues, however, as structures with a simple neural tube-like morphology were never found in teratomas lacking both N- and P-cadherin and organoid-like structures were rare in Ncad-/-tissue.


Subject(s)
Cadherins/genetics , Cadherins/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Differentiation , Cells, Cultured , Genotype , Male , Mice , Mice, Knockout , Teratoma/genetics , Teratoma/metabolism , Teratoma/pathology
11.
Dev Biol ; 202(1): 85-102, 1998 Oct 01.
Article in English | MEDLINE | ID: mdl-9758705

ABSTRACT

Somitogenesis during early stages in the chick and mouse embryo was examined in relation to N-cadherin-mediated adhesion. Previous studies indicated that N-cadherin localizes to the somite regions during their formation. Those observations were extended to include a spatiotemporal immunohistochemical analyses of beta-catenin and alpha-catenin, as well as a more detailed study of N-cadherin, during segmentation, compaction, and compartmentalization of the somite. N-cadherin and the catenins appear early within the segmental plate and are expressed as small patch-like foci throughout this tissue. The small foci of immunostaining coalesce into larger clusters of N-cadherin/catenin-expressing regions. The clusters subsequently coalesce into a region of centrally localized cells that express N-cadherin/catenins at their apical surfaces. The multiple clusters are spaced wide apart in the anterior segmental plates that form the first 6 somite pairs, as contrasted to segmental plates that form somites 7 and beyond. To examine the functional significance of N-cadherin, segmental plates were exposed to antibodies that perturb N-cadherin-mediated adhesion in the chick embryo. The multiple, anomalous somites that result in these experiments indicate that each N-cadherin/catenin-expressing cluster can give rise to a somitic structure. beta-Catenin involvement in somitogenesis suggests a role for Wnt-mediated signaling. Embryos treated with LiCl also show induction of similar anomalous somites indicating further the possibility that Wnt-mediated signaling may be involved in the clustering event. It is suggested that beta-catenin serves to initiate the adhesion process which is spread then by N-cadherin. Later during compartmentalization, N-cadherin/catenins remain expressed by the myotome compartment. Taken together, these results suggest that the Ca2+-dependent cell adhesion molecule N-cadherin and the intracellular catenins are important in segmentation and formation of the somite and myotome compartment. It is proposed that the N-cadherin-mediated adhesion process may serve as a common, evolutionarily conserved, link in the differentiation pathways of skeletal and cardiac muscle.


Subject(s)
Cadherins/physiology , Cytoskeletal Proteins/physiology , Somites/cytology , Trans-Activators , Animals , Antibodies, Monoclonal , Cadherins/analysis , Cadherins/genetics , Chick Embryo , Cytoskeletal Proteins/analysis , Lithium Chloride/pharmacology , Mice , Mutation , Somites/chemistry , alpha Catenin , beta Catenin
12.
J Cell Biol ; 139(4): 1025-32, 1997 Nov 17.
Article in English | MEDLINE | ID: mdl-9362520

ABSTRACT

To investigate the functions of P-cadherin in vivo, we have mutated the gene encoding this cell adhesion receptor in mice. In contrast to E- and N-cadherin- deficient mice, mice homozygous for the P-cadherin mutation are viable. Although P-cadherin is expressed at high levels in the placenta, P-cadherin-null females are fertile. P-cadherin expression is localized to the myoepithelial cells surrounding the lumenal epithelial cells of the mammary gland. The role of the myoepithelium as a contractile tissue necessary for milk secretion is clear, but its function in the nonpregnant animal is unknown. The ability of the P-cadherin mutant female to nurse and maintain her litter indicates that the contractile function of the myoepithelium is not dependent on the cell adhesion molecule P-cadherin. The virgin P-cadherin-null females display precocious differentiation of the mammary gland. The alveolar-like buds in virgins resemble the glands of an early pregnant animal morphologically and biochemically (i.e., milk protein synthesis). The P-cadherin mutant mice develop hyperplasia and dysplasia of the mammary epithelium with age. In addition, abnormal lymphocyte infiltration was observed in the mammary glands of the mutant animals. These results indicate that P-cadherin-mediated adhesion and/or signals derived from cell-cell interactions are important determinants in negative growth control in the mammary gland. Furthermore, the loss of P-cadherin from the myoepithelium has uncovered a novel function for this tissue in maintaining the undifferentiated state of the underlying secretory epithelium.


Subject(s)
Cadherins/physiology , Mammary Glands, Animal/growth & development , Animals , Cadherins/genetics , Cell Adhesion , Cell Differentiation , Embryonic Induction , Epithelial Cells/cytology , Female , Hyperplasia , Lactation , Mammary Glands, Animal/cytology , Mice , Mice, Knockout , Pregnancy
13.
J Cell Biol ; 138(2): 331-6, 1997 Jul 28.
Article in English | MEDLINE | ID: mdl-9230075

ABSTRACT

Myoblast fusion is essential to muscle tissue development yet remains poorly understood. N-cadherin, like other cell surface adhesion molecules, has been implicated by others in muscle formation based on its pattern of expression and on inhibition of myoblast aggregation and fusion by antibodies or peptide mimics. Mice rendered homozygous null for N-cadherin revealed the general importance of the molecule in early development, but did not test a role in skeletal myogenesis, since the embryos died before muscle formation. To test genetically the proposed role of N-cadherin in myoblast fusion, we successfully obtained N-cadherin null primary myoblasts in culture. Fusion of myoblasts expressing or lacking N-cadherin was found to be equivalent, both in vitro by intracistronic complementation of lacZ and in vivo by injection into the muscles of adult mice. An essential role for N-cadherin in mediating the effects of basic fibroblast growth factor was also excluded. These methods for obtaining genetically homozygous null somatic cells from adult tissues should have broad applications. Here, they demonstrate clearly that the putative fusion molecule, N-cadherin, is not essential for myoblast fusion.


Subject(s)
Cadherins/physiology , Cell Fusion/physiology , Muscle, Skeletal/cytology , Animals , Cadherins/genetics , Cells, Cultured , Clone Cells , Fibroblast Growth Factor 2/physiology , Homozygote , Mice , Mice, Nude , Muscle Fibers, Skeletal/cytology , Mutation
14.
Dev Biol ; 181(1): 64-78, 1997 Jan 01.
Article in English | MEDLINE | ID: mdl-9015265

ABSTRACT

To investigate the functions of N-cadherin in vivo, we have mutated the gene encoding this adhesion protein in mice. Although N-cadherin is expressed at the time of gastrulation and neurulation, both neurulation and somitogenesis initiate apparently normally in homozygous mutant embryos. However, the resulting structures are often malformed. The somites of the mutant embryos are small, irregularly shaped, and less cohesive compared with those of their wild-type littermates, and the epithelial organization of the somites is partially disrupted. Undulation of the neural tube is also observed in the mutant embryos. Homozygous mutant embryos die by Day 10 of gestation. The mesodermal and endodermal cell layers of the yolk sac are separated in the mutants. The most dramatic cell adhesion defect is observed in the primitive heart; although myocardial tissue forms initially, the myocytes subsequently dissociate and the heart tube fails to develop normally. In vitro studies of cardiac myocytes derived from N-cadherin mutant embryos show that the cells can loosely aggregate and beat synchronously, demonstrating that electrical coupling can occur between N-cadherin-deficient cardiac myocytes. These results show that N-cadherin plays a critical role in early heart development as well as in other morphogenetic processes.


Subject(s)
Cadherins/physiology , Embryo, Mammalian/abnormalities , Embryonic and Fetal Development/physiology , Animals , Cadherins/genetics , Cell Adhesion , Cell Differentiation , Embryo, Mammalian/pathology , Endoderm/pathology , Fetal Death/genetics , Fetal Death/pathology , Fetal Heart/abnormalities , Fetal Heart/pathology , Gene Expression Regulation, Developmental , Mesoderm/pathology , Mice , Mice, Knockout , Morphogenesis/genetics , Muscle, Skeletal/embryology , Yolk Sac/pathology
15.
Mamm Genome ; 7(6): 417-9, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8662222

ABSTRACT

The location of three mutations on proximal Chromosome (Chr) 18 was determined by analysis of the offspring of several backcrosses. The results demonstrate that ataxia and the insertional mutation TgN9257Mm are separated by less than 1 cM and are located approximately 3 cM from the centromere, while the balding locus is 7 cM more distal. Previous data demonstrated that the twirler locus also maps within 1 cM of ataxia. The corrected locations will contribute to identification of appropriate candidate genes for these mutations. Two polymorphic microsatellite markers for proximal Chr 18 are described, D18Umi1 and D18Umi2. The Lama3 locus encoding the alpha 3 subunit of nicein was mapped distal to ataxia and did not recombine with Tg9257.


Subject(s)
Ataxia/genetics , Chromosome Mapping , Mutation , Animals , Base Sequence , Genetic Linkage/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Molecular Sequence Data , Transgenes/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...