Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
mBio ; 14(5): e0212323, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37800917

ABSTRACT

IMPORTANCE: Cryptococcosis studies often utilize the common C57BL/6J mouse model. Unfortunately, infection in these mice fails to replicate the basic course of human disease, particularly hampering immunological studies. This work demonstrates that SJL/J mice can recapitulate human infection better than other mouse strains. The immunological response to Cryptococcus infection in SJL/J mice was markedly different from C57BL/6J and much more productive in combating this infection. Characterization of infected mice demonstrated strain-specific genetic linkage and differential regulation of multiple important immune-relevant genes in response to Cryptococcus infection. While our results validate many of the previously identified immunological features of cryptococcosis, we also demonstrate limitations from previous mouse models as they may be less translatable to human disease. We concluded that SJL/J mice more faithfully recapitulate human cryptococcosis serving as an exciting new animal model for immunological and genetic studies.


Subject(s)
Cryptococcosis , Cryptococcus neoformans , Humans , Mice , Animals , Cryptococcus neoformans/genetics , Mice, Inbred C57BL , Disease Models, Animal
2.
Dev Biol ; 237(2): 245-57, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11543611

ABSTRACT

SOX10 is a high-mobility-group transcription factor that plays a critical role in the development of neural crest-derived melanocytes. At E11.5, mouse embryos homozygous for the Sox10(Dom) mutation entirely lack neural crest-derived cells expressing the lineage marker KIT, MITF, or DCT. Moreover, neural crest cell cultures derived from homozygous embryos do not give rise to pigmented cells. In contrast, in Sox10(Dom) heterozygous embryos, melanoblasts expressing KIT and MITF do occur, albeit in reduced numbers, and pigmented cells eventually develop in nearly normal numbers both in culture and in vivo. Intriguingly, however, Sox10(Dom)/+ melanoblasts transiently lack Dct expression both in culture and in vivo, suggesting that during a critical developmental period SOX10 may serve as a transcriptional activator of Dct. Indeed, we found that SOX10 and DCT colocalized in early melanoblasts and that SOX10 is capable of transactivating the Dct promoter in vitro. Our data suggest that during early melanoblast development SOX10 acts as a critical transactivator of Dct, that MITF, on its own, is insufficient to stimulate Dct expression, and that delayed onset of Dct expression is not deleterious to the melanocyte lineage.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , High Mobility Group Proteins/physiology , Intramolecular Oxidoreductases/metabolism , Melanocytes/metabolism , Neural Crest/embryology , Transcription Factors , Transcription, Genetic , Animals , Animals, Genetically Modified , Cell Lineage , Cells, Cultured , DNA-Binding Proteins/metabolism , Galactosides/metabolism , Genotype , Heterozygote , Homozygote , Immunohistochemistry , In Situ Hybridization , Indoles/metabolism , Luciferases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microphthalmia-Associated Transcription Factor , Mutation , Pigmentation/genetics , Plasmids/metabolism , SOXE Transcription Factors , Time Factors , Transfection
3.
Genesis ; 30(2): 70-6, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11416866

ABSTRACT

This study describes an in utero approach for overexpressing genes in a cell-type directed manner. It uses an avian leukosis retroviral expression system coupled with a transgenic mouse line expressing the viral receptor tv-a from a tissue-specific promoter (RCAS-TVA system) (Federspiel et al., 1994, and reviewed in Fisher et al., 1999). A transgenic mouse line was generated expressing tv-a from the Dopachrome tautomerase promoter (DCT-tv-a) in embryonic melanocyte precursors (melanoblasts). RCAS virus encoding beta-galactosidase (RCAS-LacZ) or tyrosinase (RCAS-Tyr) was injected in utero into embryonic day 12.5 albino (tyrosinase inactive) mouse embryos. Animals were analyzed for beta-galactosidase activity or tyrosinase activity (hair pigmentation). RCAS gene expression was detected in 44% and 25% of the transgenic mice, respectively. We demonstrate the RCAS-TVA system coupled with the DCT-tv-a line of mice can be used for in utero infection.


Subject(s)
Gene Transfer Techniques , Melanocytes/metabolism , Neural Crest/metabolism , Zebrafish Proteins , Animals , Avian Leukosis Virus/genetics , Avian Leukosis Virus/isolation & purification , Avian Proteins , Cell Differentiation , Embryo, Mammalian/cytology , Genetic Complementation Test , Melanocytes/cytology , Mice , Mice, Transgenic , Neural Crest/cytology , Proto-Oncogene Proteins/genetics , Receptors, Virus/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Transfection , Wnt Proteins
4.
DNA Res ; 8(5): 221-6, 2001 Oct 31.
Article in English | MEDLINE | ID: mdl-11759842

ABSTRACT

Avian leukosis type A virus-derived retroviral vectors have been used to introduce genes into cells expressing the corresponding avian receptor tv-a. This includes the use of Replication-Competent Avian sarcoma-leukosis virus (ASLV) long terminal repeat (LTR) with Splice acceptor (RCAS) vectors in the analysis of avian development, human and murine cell cultures, murine cell lineage studies and cancer biology. Previously, cloning of genes into this virus was difficult due to the large size of the vector and sparse cloning sites. To overcome some of the disadvantages of traditional cloning using the RCASBP-Y vector, we have modified the RCASBP-Y to incorporate "Gateway" site-specific recombination cloning of genes into the construct, either with or without HA epitope tags. We have found the repetitive "att" sequences, which are the targets for site-specific recombination, do not impair the production of infectious viral particles or the expression of the gene of interest. This is the first instance of site-specific recombination being used to generate retroviral gene constructs. These viral constructs will allow for the efficient transfer and expression of cDNAs needed for functional genomic analyses.


Subject(s)
Avian Sarcoma Viruses/genetics , Cloning, Molecular/methods , Genetic Vectors/genetics , Amino Acid Sequence , Animals , Base Sequence , Gene Expression Regulation , Green Fluorescent Proteins , Humans , Luminescent Proteins , Mice , Molecular Sequence Data
5.
Hum Genet ; 107(1): 1-6, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10982026

ABSTRACT

Waardenburg syndrome (WS) is associated with neural crest-derived melanocyte deficiency caused by mutations in either one of three transcription factors: MITF, PAX3, and SOX10. However, the hierarchical relationship of these transcription factors is largely unknown. We show that SOX10 is capable of transactivating the MITF promoter 100-fold, and that this transactivation is further stimulated by PAX3. Promoter deletion and mutational analyses indicate that SOX10 can activate MITF expression through binding to a region that is evolutionarily conserved between the mouse and human MITF promoters. A SOX10 mutant that models C-terminal truncations in WS can reduce wild-type SOX10 induction of MITF, suggesting these mutations may act in a dominant-negative fashion. Our data support a model in which the hypopigmentation in WS, of which these factors have been implicated, results from a disruption in function of the central melanocyte transcription factor MITF.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation , High Mobility Group Proteins/genetics , Transcription Factors/genetics , Waardenburg Syndrome/genetics , Animals , Base Sequence , Conserved Sequence , DNA-Binding Proteins/biosynthesis , Evolution, Molecular , Gene Deletion , Genes, Dominant , Genotype , HeLa Cells , High Mobility Group Proteins/biosynthesis , Humans , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Mutant Strains , Microphthalmia-Associated Transcription Factor , Molecular Sequence Data , Mutagenesis , Mutation , PAX3 Transcription Factor , Paired Box Transcription Factors , Phenotype , Promoter Regions, Genetic , SOXE Transcription Factors , Sequence Homology, Nucleic Acid , Time Factors , Transfection
6.
Proc Natl Acad Sci U S A ; 97(18): 10050-5, 2000 Aug 29.
Article in English | MEDLINE | ID: mdl-10963668

ABSTRACT

Wnt1 signaling has been implicated as one factor involved in neural crest-derived melanocyte (NC-M) development. Mice deficient for both Wnt1 and Wnt3a have a marked deficiency in trunk neural crest derivatives including NC-Ms. We have used cell lineage-directed gene targeting of Wnt signaling genes to examine the effects of Wnt signaling in mouse neural crest development. Gene expression was directed to cell lineages by infection with subgroup A avian leukosis virus vectors in lines of transgenic mice that express the retrovirus receptor tv-a. Transgenic mice with tva in either nestin-expressing neural precursor cells (line Ntva) or dopachrome tautomerase (DCT)-expressing melanoblasts (line DCTtva) were analyzed. We overstimulated Wnt signaling in two ways: directed gene transfer of Wnt1 to Ntva(+) cells and transfer of beta-catenin to DCTtva(+) NC-M precursor cells. In both methods, NC-M expansion and differentiation were effected. Significant increases were observed in the number of NC-Ms [melanin(+) and tyrosinase-related protein 1 (TYRP1)(+) cells], the differentiation of melanin(-) TYRP1(+) cells to melanin(+) TYRP1(+) NC-Ms, and the intensity of pigmentation per NC-M. These data are consistent with Wnt1 signaling being involved in both expansion and differentiation of migrating NC-Ms in the developing mouse embryo. The use of lineage-directed gene targeting will allow the dissection of signaling molecules involved in NC development and is adaptable to other mammalian developmental systems.


Subject(s)
Embryonic and Fetal Development , Gene Transfer Techniques , Melanocytes/cytology , Melanocytes/physiology , Nerve Tissue Proteins , Neural Crest/physiology , Proto-Oncogene Proteins/physiology , Trans-Activators , Zebrafish Proteins , Animals , Cytoskeletal Proteins/physiology , Genetic Vectors , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/physiology , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Membrane Glycoproteins , Mice , Mice, Transgenic , Nestin , Organ Culture Techniques , Oxidoreductases , Proto-Oncogene Proteins/genetics , Recombinant Proteins/metabolism , Transfection , Wnt Proteins , Wnt1 Protein , beta Catenin
7.
Pigment Cell Res ; 13(3): 141-6, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10885671

ABSTRACT

Regulation of gene expression is a fundamental process by which cells respond to both intracellular and extracellular signals. For a pigment cell, alterations in gene expression regulate the processes of cell migration, lineage restriction, differentiation, type of pigment produced, and progression from a normal pigment cell to that of melanoma. To date, the identification of genes involved in normal pigment cell development has been accomplished by the cloning of individual mutant alleles, a single gene at a time. Current advances in technology have now made it possible to use expression profile analysis to investigate, on a genomic scale, the process of pigment cell development and function. This review compares and contrasts the methods of subtractive suppressive polymerase chain reaction (PCR) and differential display with that of cDNA microarray analysis.


Subject(s)
Gene Expression Profiling/methods , Melanocytes/physiology , Cell Differentiation/genetics , DNA, Complementary/analysis , Gene Expression Regulation , Humans , Melanoma/genetics , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis/methods , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
8.
Genome Res ; 10(1): 17-29, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10645946

ABSTRACT

We have used the varied expressivity of white spotting (hypopigmentation) observed in intrasubspecific crosses of Ednrb(s) mice (Mayer Ednrb(s)/Ednrb(s) and C3HeB/FeJ Ednrb(s)/Ednrb(s)) to analyze the effects of modifier loci on the patterning of hypopigmentation. We have confirmed that an Ednrb(s) modifier locus is present on mouse Chromosome 10. This locus is now termed k10, using the nomenclature established by Dunn in 1920. The k10(Mayer) allele is a recessive modifier that accounts for almost all of the genetic variance of dorsal hypopigmentation. Using intercross analyses we identified a second allele of this locus or a closely linked gene termed k10(C3H). The k10(C3H) allele is semidominant and is associated with the penetrance and expressivity of a white forelock phenotype similar to that seen in Waardenburg syndrome. Molecular linkage analysis was used to determine that the k10 critical interval was flanked by D10Mit10 and D10Mit162/D10Mit122 and cosegregates with mast cell growth factor (Mgf). Complementation crosses with a Mgf(Sl) allele (a 3-5-cM deletion) confirm the semidominant white forelock feature of the k10(C3H) allele and the dorsal spotting feature of K10(Mayer) allele. MgF was assessed as a candidate gene for k10(Mayer) and k10(C3H) by sequence and genomic analyses. No molecular differences were observed between the Mayer and C57BL/6J alleles of MgF; however, extensive genomic differences were observed between the C3HeB/FeJ and C57BL/6J alleles. This suggests that alteration of MgF expression in C3H mice may account for the k10(C3H) action on white forelock hypopigmentation. Crosses of Ednrb(s) with Kit(WJ-2) (the receptor for MGF)-deficient mice confirmed the hypothesis that synergistic interaction between the Endothelin and MGF signaling pathways regulates proper neural crest-derived melanocyte development in vivo.


Subject(s)
Chromosome Mapping , Hypopigmentation/genetics , Receptors, Endothelin/deficiency , Receptors, Endothelin/genetics , Alleles , Animals , Crosses, Genetic , Female , Gene Expression Regulation, Developmental , Genetic Complementation Test , Genetic Linkage , Genetic Markers , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Receptor, Endothelin B , Receptors, Endothelin/chemistry
9.
Proc Natl Acad Sci U S A ; 96(23): 13264-9, 1999 Nov 09.
Article in English | MEDLINE | ID: mdl-10557309

ABSTRACT

Alveolar rhabdomyosarcoma is an aggressive pediatric cancer of striated muscle characterized in 60% of cases by a t(2;13)(q35;q14). This results in the fusion of PAX3, a developmental transcription factor required for limb myogenesis, with FKHR, a member of the forkhead family of transcription factors. The resultant PAX3-FKHR gene possesses transforming properties; however, the effects of this chimeric oncogene on gene expression are largely unknown. To investigate the actions of these transcription factors, both Pax3 and PAX3-FKHR were introduced into NIH 3T3 cells, and the resultant gene expression changes were analyzed with a murine cDNA microarray containing 2,225 elements. We found that PAX3-FKHR but not PAX3 activated a myogenic transcription program including the induction of transcription factors MyoD, Myogenin, Six1, and Slug as well as a battery of genes involved in several aspects of muscle function. Notable among this group were the growth factor gene Igf2 and its binding protein Igfbp5. Relevance of this model was suggested by verification that three of these genes (IGFBP5, HSIX1, and Slug) were also expressed in alveolar rhabdomyosarcoma cell lines. This study utilizes cDNA microarrays to elucidate the pattern of gene expression induced by an oncogenic transcription factor and demonstrates the profound myogenic properties of PAX3-FKHR in NIH 3T3 cells.


Subject(s)
Artificial Gene Fusion , DNA-Binding Proteins/genetics , MyoD Protein/genetics , Myogenin/genetics , Transcription Factors/genetics , Transcription, Genetic , 3T3 Cells , Animals , DNA, Complementary , Forkhead Box Protein O1 , Forkhead Transcription Factors , Mice , PAX3 Transcription Factor , Paired Box Transcription Factors , Rhabdomyosarcoma, Alveolar/genetics , Transduction, Genetic , Tumor Cells, Cultured
11.
Mamm Genome ; 10(6): 592-6, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10341092

ABSTRACT

The mouse homolog of the human MEN1 gene, which is defective in a dominant familial cancer syndrome, multiple endocrine neoplasia type 1 (MEN1), has been identified and characterized. The mouse Men1 transcript contains an open reading frame encoding a protein of 611 amino acids which has 97% identity and 98% similarity to human menin. Sequence of the entire Men1 gene (9.3 kb) was assembled, revealing 10 exons, with exon 1 being non-coding; a polymorphic tetranucleotide repeat was located in the 5'- flanking region. The exon-intron organization and the size of the coding exons 2-9 were well conserved between the human and mouse genes. Fluorescence in situ hybridization localized the Men1 gene to mouse Chromosome (Chr) 19, a region known to be syntenic to human Chr 11q13, the locus for the MEN1 gene. Northern analysis indicated two messages-2.7 kb and 3.1 kb-expressed in all stages of the embryo analyzed and in all eight adult tissues tested. The larger transcript differs from the smaller by the inclusion of an unspliced intron 1. Whole-mount in situ hybridization of 10.5-day and 11.5-day embryos showed ubiquitous expression of Men1 RNA. Western analysis with antibodies raised against a conserved C-terminal peptide identified an approximately 67-kDa protein in the lysates of adult mouse brain, kidney, liver, pancreas, and spleen tissues, consistent with the size of human menin. The levels of mouse menin do not appear to fluctuate during the cell cycle.


Subject(s)
Chromosome Mapping , Neoplasm Proteins/genetics , Proto-Oncogene Proteins , Amino Acid Sequence , Animals , Blotting, Northern , Cell Cycle/genetics , Cloning, Molecular , DNA, Complementary , Deoxyribonucleases, Type II Site-Specific/genetics , Embryo, Mammalian/physiology , Exons , Gene Expression Regulation, Developmental , Humans , In Situ Hybridization/methods , In Situ Hybridization, Fluorescence , Introns , Mice , Molecular Sequence Data , Neoplasm Proteins/metabolism , Sequence Analysis, DNA , Sequence Homology, Amino Acid
12.
Genome Res ; 9(3): 215-25, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10077527

ABSTRACT

Hirschsprung disease (HSCR) is a multigenic neurocristopathy clinically recognized by aganglionosis of the distal gastrointestinal tract. Patients presenting with aganglionosis in association with hypopigmentation are classified as Waardenburg syndrome type 4 (Waardenburg-Shah, WS4). Variability in the disease phenotype of WS4 patients with equivalent mutations suggests the influence of genetic modifier loci in this disorder. Sox10(Dom)/+ mice exhibit variability of aganglionosis and hypopigmentation influenced by genetic background similar to that observed in WS4 patients. We have constructed Sox10(Dom)/+ congenic lines to segregate loci that modify the neural crest defects in these mice. Consistent with previous studies, increased lethality of Sox10(Dom)/+ animals resulted from a C57BL/6J locus(i). However, we also observed an increase in hypopigmentation in conjunction with a C3HeB/FeJLe-a/a locus(i). Linkage analysis localized a hypopigmentation modifier of the Dom phenotype to mouse chromosome 10 in close proximity to a previously reported modifier of hypopigmentation for the endothelin receptor B mouse model of WS4. To evaluate further the role of SOX10 in development and disease, we have performed comparative genomic analyses. An essential role for this gene in neural crest development is supported by zoo blot hybridizations that reveal extensive conservation throughout vertebrate evolution and by similar Northern blot expression profiles between mouse and man. Comparative sequence analysis of the mouse and human SOX10 gene have defined the exon-intron boundaries of SOX10 and facilitated mutation analysis leading to the identification of two new SOX10 mutations in individuals with WS4. Structural analysis of the HMG DNA-binding domain was performed to evaluate the effect of human mutations in this region.


Subject(s)
DNA-Binding Proteins/genetics , Genes, Dominant/genetics , Genetic Variation/genetics , High Mobility Group Proteins/genetics , Hirschsprung Disease/genetics , Hypopigmentation/genetics , Amino Acid Sequence , Animals , Base Sequence , Crosses, Genetic , DNA-Binding Proteins/biosynthesis , Disease Models, Animal , Female , High Mobility Group Proteins/biosynthesis , Humans , Male , Mice , Mice, Congenic , Mice, Inbred C3H , Mice, Inbred C57BL , Molecular Sequence Data , Rats , SOXE Transcription Factors , Syndrome , Transcription Factors
13.
Mamm Genome ; 9(9): 715-20, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9716656

ABSTRACT

We report the isolation of the murine ortholog of AIM1, a human gene whose expression is associated with the reversal of tumorigenicity in an experimental model of melanoma. Mouse and human AIM1 are more than 90% identical in amino acid sequence in the betagamma-crystallin repeats and the C-terminal domain, and more than 75% identical in the extended N-terminal domain. Consistent with the isolated cDNA representing the authentic AIM1 ortholog, linkage analysis localized mouse Aim1 to proximal mouse Chromosome (Chr) 10 in a conserved linkage group with genes localized to human Chr band 6q21. Searches of EST databases identified a second AIM1-like gene in both mouse and human, suggesting the existence of a gene family. Northern analysis demonstrates Aim1 is expressed most abundantly in adult skin, lung, heart, liver, and kidney and is temporally regulated during embryogenesis. Aim1 is expressed highly in the shaft region of the hair follicles and the presumptive ectoderm, but not at detectable levels in melanocytes or melanocyte precursor cells.


Subject(s)
Crystallins/genetics , Membrane Proteins , Multigene Family/genetics , Proteins/genetics , Sequence Homology, Amino Acid , Amino Acid Sequence , Animals , Cell Line, Transformed , Chromosome Mapping , Chromosomes, Human, Pair 6/genetics , Cloning, Molecular , Embryo, Mammalian/chemistry , Embryo, Mammalian/embryology , Gene Expression Regulation, Developmental , Humans , Melanocytes/chemistry , Melanocytes/cytology , Mice , Models, Molecular , Molecular Sequence Data , Organ Specificity/genetics , Sequence Analysis, DNA , Skin/chemistry , Skin/cytology , Symporters , gamma-Crystallins
15.
Nat Genet ; 18(1): 60-4, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9425902

ABSTRACT

Hirschsprung disease (HSCR, MIM #142623) is a multigenic neurocristopathy (neural crest disorder) characterized by absence of enteric ganglia in a variable portion of the distal colon. Subsets of HSCR individuals also present with neural crest-derived melanocyte deficiencies (Hirschsprung-Waardenburg, HSCR-WS, MIM #277580). Murine models have been instrumental in the identification and analysis of HSCR disease genes. These include mice with deficiencies of endothelin B receptor (Ednrb(s-l); refs 1,2) endothelin 3 (Edn3(ls): refs 1,3) the tyrosine kinase receptor cRet and glial-derived neurotrophic factor. Another mouse model of HSCR disease, Dom, arose spontaneously at the Jackson Laboratory. While Dom/+ heterozygous mice display regional deficiencies of neural crest-derived enteric ganglia in the distal colon, Dom/Dom homozygous animals are embryonic lethal. We have determined that premature termination of Sox10, a member of the SRY-like HMG box family of transcription factors, is responsible for absence of the neural crest derivatives in Dom mice. We demonstrate expression of Sox10 in normal neural crest cells, disrupted expression of both Sox10 and the HSCR disease gene Ednrb in Dom mutant embryos, and loss of neural crest derivatives due to apoptosis. Our studies suggest that Sox10 is essential for proper peripheral nervous system development. We propose SOX10 as a candidate disease gene for individuals with HSCR whose disease does not have an identified genetic origin.


Subject(s)
DNA-Binding Proteins/genetics , High Mobility Group Proteins/genetics , Hirschsprung Disease/genetics , Neural Crest/embryology , Amino Acid Sequence , Animals , Base Sequence , DNA, Complementary , DNA-Binding Proteins/biosynthesis , Disease Models, Animal , Gene Expression Regulation, Developmental , High Mobility Group Proteins/biosynthesis , Hirschsprung Disease/embryology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Sequence Data , Mutation , Neural Crest/growth & development , RNA, Messenger , SOXE Transcription Factors , Transcription Factors
16.
Biochem Cell Biol ; 76(6): 1093-9, 1998.
Article in English | MEDLINE | ID: mdl-10392719

ABSTRACT

In both mice and humans, mutations in the genes encoding the endothelin B receptor and its ligand endothelin 3 lead to deficiencies in neural crest-derived melanocytes and enteric neurons. The discrete steps at which endothelins exert their functions in melanocyte development were examined in mouse neural crest cell cultures. Such cultures, kept in the presence of fetal calf serum, gave rise to cells expressing the early melanoblast marker Dct even in the absence of the phorbol ester tetradecanoyl phorbol acetate (TPA) or endothelins. However, these early Dct+ cells did not proliferate and pigmented cells never formed unless TPA or endothelins were added. In fact, endothelin 2 was as potent as TPA in promoting the generation of both Dct+ melanoblasts and pigmented cells, and endothelin 1 or endothelin 3 stimulated the generation of melanoblasts and of pigmented cells to an even greater extent. The inhibition of this stimulation by the selective endothelin B receptor antagonist BQ-788 (N-cis-2,6-dimethylpiperidinocarbonyl-L-alpha-methylleucyl-D -1-methoxycarbonyltryptophanyl-D-norleucine) suggested that the three endothelins all signal through the endothelin B receptor. This receptor was indeed expressed in Dct+ melanoblasts, in addition to cells lacking Dct expression. The results demonstrate that endothelins are potent stimulators of melanoblast proliferation and differentiation.


Subject(s)
Endothelins/physiology , Melanocytes/physiology , Neural Crest/embryology , Signal Transduction , Animals , Bromodeoxyuridine/metabolism , Cell Division , Cell Survival , Cells, Cultured , Dose-Response Relationship, Drug , Endothelin-1/metabolism , Endothelin-2/metabolism , Endothelin-3/metabolism , In Situ Hybridization , Mice , Mice, Inbred C57BL , Receptor, Endothelin B , Receptors, Endothelin/metabolism , Tetradecanoylphorbol Acetate/metabolism , Time Factors
17.
Oncogene ; 15(19): 2361-8, 1997 Nov 06.
Article in English | MEDLINE | ID: mdl-9393880

ABSTRACT

As part of a cloning strategy to identify genes involved in early mouse liver development we have isolated Praja1, a gene with similar sequences to the Drosophila melanogaster gene goliath (gl) which is involved in the fate of mesodermal cells ultimately forming gut musculatures, fat body, and the heart. Praja1 is a 2.1 kb gene encoding a putative 396 amino acid ORF and includes a COOH-terminal RING-H2 domain. Using the Jackson Laboratory BSS panel, we have localized Praja1 on chromosome X at 36 cM, which may be a candidate gene for mouse sla (sex linked sideroblastic anemia), near the X inactivation center gene, Xist. Northern blot analysis demonstrated three transcripts (3.1, 2.6 and 2.1 kb) in mRNA from adult mouse tissues brain, liver, and kidney as well as in mRNA from developing mouse embryos (days 7, 11, 15 and 17 post coitus, p.c.). In vitro transcription/translation yielded a product with an Mr of 59 kD. Immunohistochemical staining of in vitro liver explant cultures using a heterologous antibody against praja1 demonstrated cytoplasmic staining of cuboidal cells that have hepatocyte morphology and organization. The presence of the RING-H2 domain, a proline-rich region at the COOH-end, and regions rich in acidic amino acids, leads to the hypothesis that the Praja1 product is possibly involved in mediating protein-protein interactions, possibly as part of a protein sorting or transport pathway. This is strengthened by the similarity of Praja1 to rat Neurodap1, whose product has been shown to localize to the endoplasmic reticulum and golgi in brain.


Subject(s)
Proteins/genetics , X Chromosome , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Liver/metabolism , Mice , Molecular Sequence Data , Proteins/chemistry , Sequence Alignment , Ubiquitin-Protein Ligases
18.
Science ; 277(5323): 232-5, 1997 Jul 11.
Article in English | MEDLINE | ID: mdl-9211850

ABSTRACT

An integrated human-mouse positional candidate approach was used to identify the gene responsible for the phenotypes observed in a mouse model of Niemann-Pick type C (NP-C) disease. The predicted murine NPC1 protein has sequence homology to the putative transmembrane domains of the Hedgehog signaling molecule Patched, to the cholesterol-sensing regions of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase and SREBP cleavage-activating protein (SCAP), and to the NPC1 orthologs identified in human, the nematode Caenorhabditis elegans, and the yeast Saccharomyces cerevisiae. The mouse model may provide an important resource for studying the role of NPC1 in cholesterol homeostasis and neurodegeneration and for assessing the efficacy of new drugs for NP-C disease.


Subject(s)
Cholesterol/metabolism , Disease Models, Animal , Niemann-Pick Diseases/genetics , Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Homeostasis , Humans , Hydroxymethylglutaryl CoA Reductases/chemistry , Intracellular Signaling Peptides and Proteins , Lysosomes/metabolism , Membrane Proteins/chemistry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Sequence Data , Mutation , Niemann-Pick C1 Protein , Niemann-Pick Diseases/metabolism , Phenotype , Protein Sorting Signals/chemistry , Proteins/chemistry , Proteins/physiology , Sequence Homology, Amino Acid
19.
Science ; 277(5323): 228-31, 1997 Jul 11.
Article in English | MEDLINE | ID: mdl-9211849

ABSTRACT

Niemann-Pick type C (NP-C) disease, a fatal neurovisceral disorder, is characterized by lysosomal accumulation of low density lipoprotein (LDL)-derived cholesterol. By positional cloning methods, a gene (NPC1) with insertion, deletion, and missense mutations has been identified in NP-C patients. Transfection of NP-C fibroblasts with wild-type NPC1 cDNA resulted in correction of their excessive lysosomal storage of LDL cholesterol, thereby defining the critical role of NPC1 in regulation of intracellular cholesterol trafficking. The 1278-amino acid NPC1 protein has sequence similarity to the morphogen receptor PATCHED and the putative sterol-sensing regions of SREBP cleavage-activating protein (SCAP) and 3-hydroxy-3-methyl-glutaryl coenzyme A (HMG-CoA) reductase.


Subject(s)
Carrier Proteins , Cholesterol/metabolism , Drosophila Proteins , Membrane Glycoproteins , Niemann-Pick Diseases/genetics , Proteins/genetics , Amino Acid Sequence , Cholesterol, LDL/metabolism , Chromosome Mapping , Chromosomes, Human, Pair 18 , Cloning, Molecular , Homeostasis , Humans , Hydroxymethylglutaryl CoA Reductases/chemistry , Insect Proteins/chemistry , Intracellular Signaling Peptides and Proteins , Lysosomes/metabolism , Membrane Proteins/chemistry , Molecular Sequence Data , Mutation , Niemann-Pick C1 Protein , Niemann-Pick Diseases/metabolism , Polymorphism, Single-Stranded Conformational , Proteins/chemistry , Proteins/physiology , Receptors, Cell Surface/chemistry , Sequence Homology, Amino Acid , Transfection
20.
Proc Natl Acad Sci U S A ; 94(14): 7378-83, 1997 Jul 08.
Article in English | MEDLINE | ID: mdl-9207099

ABSTRACT

Niemann-Pick disease type C (NP-C) is an autosomal recessive lipidosis linked to chromosome 18q11-12, characterized by lysosomal accumulation of unesterified cholesterol and delayed induction of cholesterol-mediated homeostatic responses. This cellular phenotype is identifiable cytologically by filipin staining and biochemically by measurement of low-density lipoprotein-derived cholesterol esterification. The mutant Chinese hamster ovary cell line (CT60), which displays the NP-C cellular phenotype, was used as the recipient for a complementation assay after somatic cell fusions with normal and NP-C murine cells suggested that this Chinese hamster ovary cell line carries an alteration(s) in the hamster homolog(s) of NP-C. To narrow rapidly the candidate interval for NP-C, three overlapping yeast artificial chromosomes (YACs) spanning the 1 centimorgan human NP-C interval were introduced stably into CT60 cells and analyzed for correction of the cellular phenotype. Only YAC 911D5 complemented the NP-C phenotype, as evidenced by cytological and biochemical analyses, whereas no complementation was obtained from the other two YACs within the interval or from a YAC derived from chromosome 7. Fluorescent in situ hybridization indicated that YAC 911D5 was integrated at a single site per CT60 genome. These data substantially narrow the NP-C critical interval and should greatly simplify the identification of the gene responsible in mouse and man. This is the first demonstration of YAC complementation as a valuable adjunct strategy for positional cloning of a human gene.


Subject(s)
Chromosomes, Artificial, Yeast/genetics , Chromosomes, Human, Pair 18 , Chromosomes, Human, Pair 7 , Niemann-Pick Diseases/genetics , Animals , CHO Cells , Chromosome Mapping , Cloning, Molecular , Cricetinae , DNA, Complementary , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...