Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters











Publication year range
1.
Genes Immun ; 7(2): 122-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16395389

ABSTRACT

Chlamydia trachomatis is a bacterial pathogen that is a major cause of blindness and infertility in diverse populations across the world. In an effort to model genetic complexities that are observed in human populations and to identify novel genes involved in susceptibility to C. trachomatis, we have adapted a murine model of systemic infection for use in genetic analysis. In this model, chlamydial colonization and replication is measured in the spleens of mice shortly after intravenous delivery of C. trachomatis L2. Here, we show that C57BL/6J and C3H/HeJ inbred mice are differentially susceptible to this systemic infection. Additionally, fibroblasts cultured from C57BL/6J and C3H/HeJ embryos are differentially permissive for chlamydial replication. We have taken advantage of this natural variation to map quantitative trait loci on Chromosomes 2, 3, and 11 that segregate with the bacterial load in F2 cross progeny during the acute phase of C. trachomatis infection in vivo. To validate our mapping results, we also generated mice that are congenic for a portion of Chromosome 11 from the susceptible parent. This congenic interval confers increased susceptibility to C. trachomatis, both in vivo and in vitro, suggesting that our screen identified at least one gene that is involved in cellular resistance to C. trachomatis replication.


Subject(s)
Chlamydia Infections/genetics , Chlamydia trachomatis/pathogenicity , Animals , Cells, Cultured , Chlamydia Infections/immunology , Chlamydia trachomatis/classification , Chlamydia trachomatis/physiology , Chromosome Mapping , Chromosomes , DNA, Bacterial/analysis , Disease Susceptibility , Female , Fibroblasts/cytology , Fibroblasts/immunology , Fibroblasts/microbiology , Genetic Linkage , Genetic Markers , Kinetics , Mice , Mice, Congenic , Mice, Inbred C3H , Mice, Inbred C57BL , Pregnancy , Quantitative Trait Loci , Serotyping , Species Specificity , Spleen/microbiology
2.
J Exp Med ; 194(8): 1081-96, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11602638

ABSTRACT

The products of the Legionella pneumophila dot/icm genes enable the bacterium to replicate within a macrophage vacuole. This study demonstrates that the Dot/Icm machinery promotes macropinocytotic uptake of L. pneumophila into mouse macrophages. In mouse strains harboring a permissive Lgn1 allele, L. pneumophila promoted formation of vacuoles that were morphologically similar to macropinosomes and dependent on the presence of an intact Dot/Icm system. Macropinosome formation appeared to occur during, rather than after, the closure of the plasma membrane about the bacterium, since a fluid-phase marker preloaded into the macrophage endocytic path failed to label the bacterium-laden macropinosome. The resulting macropinosomes were rich in GM1 gangliosides and glycosylphosphatidylinositol-linked proteins. The Lgn1 allele restrictive for L. pneumophila intracellular replication prevented dot/icm-dependent macropinocytosis, with the result that phagosomes bearing the microorganism were targeted into the endocytic network. Analysis of macrophages from recombinant inbred mouse strains support the model that macropinocytotic uptake is controlled by the Lgn1 locus. These results indicate that the products of the dot/icm genes and Lgn1 are involved in controlling an internalization route initiated at the time of bacterial contact with the plasma membrane.


Subject(s)
Genes, Bacterial/physiology , Legionella pneumophila/immunology , Macrophages/immunology , Pinocytosis/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cell Membrane/microbiology , Cell Membrane/physiology , Cells, Cultured , Female , Macrophages/cytology , Macrophages/microbiology , Mice , Mice, Inbred A , Mice, Inbred C57BL
3.
Curr Biol ; 11(19): 1503-11, 2001 Oct 02.
Article in English | MEDLINE | ID: mdl-11591317

ABSTRACT

BACKGROUND: Inbred mouse strains exhibit striking differences in the susceptibility of their macrophages to the effects of anthrax lethal toxin (LeTx). Previous data has shown that this difference in susceptibility lies downstream of toxin entry into macrophages. A locus controlling this phenotype, called Ltxs1, has been mapped to chromosome 11, but the responsible gene has not been identified. RESULTS: Here, we report the identification of the Ltxs1 gene as Kif1C, which encodes a kinesin-like motor protein of the UNC104 subfamily. Kif1C is the only gene in the Ltxs1 interval exhibiting polymorphisms between susceptible and resistant strains. Multiple alleles of Kif1C determine the susceptibility or resistance of cultured mouse macrophages to LeTx. Treatment of resistant macrophages with brefeldin-A (which alters the cellular localization of Kif1C) induces susceptibility to LeTx, while ectopic expression of a resistance allele of Kif1C in susceptible macrophages causes a 4-fold increase in the number of cells surviving LeTx treatment. We also show that cleavage of map kinase kinase 3, a target of LeTx proteolysis, occurs in resistant cells. CONCLUSIONS: We conclude that mutations in Kif1C are responsible for the differences in the susceptibility of inbred mouse macrophages to LeTx and that proper Kif1C function is required for LeTx resistance. Since the LeTx-mediated proteolysis of map kinase kinase 3 occurs even in resistant cells, Kif1C does not affect cellular entry or processing of LeTx and likely influences events occurring later in the intoxication pathway.


Subject(s)
Antigens, Bacterial , Bacillus anthracis , Bacterial Toxins/pharmacology , Kinesins/physiology , Macrophages/drug effects , Alleles , Animals , Brefeldin A/pharmacology , Kinesins/classification , Kinesins/genetics , Macrophages/cytology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mutagenesis
4.
Genomics ; 73(2): 223-31, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11318612

ABSTRACT

Lethal factor (LF) is a toxin secreted by Bacillus anthracis that plays an important role in the pathogenesis of anthrax. Intoxication with LF results in a macrophage-specific cytolysis that is not well understood. Interestingly, inbred mouse strains exhibit dramatic differences in the susceptibility of their cultured macrophages to killing by LF, and a gene that influences this phenotype, called Ltxs1, has been mapped to mouse chromosome 11. Here we report a high-resolution genetic map that confines the Ltxs1 region to a 0.51-cM interval between D11Mit90 and D11Die37/D11Die38. We have also constructed a complete physical map of YAC and BAC clones covering the Ltxs1 region. In conjunction with synteny homology searching, BLAST searches of sequences obtained from the clones in the physical map have revealed 14 known genes and five ESTs that reside in the critical interval. Additionally, a region of 100 kb or more is deleted in the Ltxs1 interval of some strains. Our genetic, physical, and transcript map provides an important resource for the molecular cloning of Ltxs1.


Subject(s)
Anthrax/genetics , Antigens, Bacterial , Bacillus anthracis , Bacterial Toxins/toxicity , Chromosome Mapping , Animals , Chromosomes, Artificial, Yeast , Contig Mapping , Disease Models, Animal , Expressed Sequence Tags , Genetic Markers , Genetic Predisposition to Disease , Mice , Mice, Inbred Strains , Sequence Tagged Sites , Transcription, Genetic
5.
Nat Genet ; 27(3): 259-60, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11242105

ABSTRACT

We have used a novel quantitative trait locus model to study the genetics of survival of F2 progeny of susceptible BALB/cByJ and resistant C57BL/6ByJ mice that have been infected with Listeria monocytogenes. This allowed us to map modifiers of L. monocytogenes susceptibility to chromosomes 5 and 13.


Subject(s)
Listeriosis/genetics , Listeriosis/immunology , Animals , Chromosome Mapping , Crosses, Genetic , Female , Listeriosis/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Quantitative Trait, Heritable
8.
Genome Res ; 10(8): 1095-102, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10958627

ABSTRACT

A mouse locus called Lgn1 determines differences in macrophage permissiveness for the intracellular replication of Legionella pneumophila. The only regional candidate genes for this phenotype difference lie within a cluster of closely linked paralogs of the Neuronal Apoptosis Inhibitory Protein (Naip) gene. Previous genetic and physical mapping of the Lgn1 phenotype narrowed it to an interval containing only Naip2 and Naip5, suggesting that there is not complete functional overlap among the mouse Naip loci. In order to gather more information about polymorphisms among the Naip genes of the 129 mouse haplotype, we have determined the genomic sequence of a substantial portion of the 129 Naip gene array. We have constructed an evolutionary model for the expansion of the Naip gene array from a single progenitor Naip gene. This model predicts the presence of two distinct families of Naip paralogs: Naip1/2/3 and Naip4/5/6/7. Unlike the divergences among all the other Naip paralogs, the splits among Naip4, Naip5, Naip6, and Naip7 occurred relatively recently. The high degree of sequence conservation within the Naip4/5/6/7 family increases the likelihood of functional overlap among these genes.


Subject(s)
Apoptosis/genetics , Genome , Multigene Family , Nerve Tissue Proteins/genetics , Animals , Genetic Markers , Humans , Mice , Molecular Sequence Data , Neuronal Apoptosis-Inhibitory Protein , Sequence Alignment , Sequence Analysis, DNA
9.
Genome Res ; 10(8): 1158-71, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10958634

ABSTRACT

Prior genetic and physical mapping has shown that the Naip gene cluster on mouse chromosome 13D1-D3 contains a gene, Lgn1, that is responsible for determining the permissivity of ex vivo macrophages to Legionella pneumophila replication. We have identified differences in the structure of the Naip array among commonly used inbred mouse strains, although these gross structural differences do not correlate with differences in L. pneumophila permissiveness. A physical map of the region employing clones of the C57BL/6J haplotype confirms that there are fewer copies of Naip in this strain than are in the physical map of the 129 haplotype. We have also refined the genetic location of Lgn1, leaving only Naip2 and Naip5 as candidates for Lgn1. Our genetic map suggests the presence of two hotspots of recombination within the Naip array, indicating that the 3' portion of Naip may be involved in the genomic instability at this locus.


Subject(s)
Apoptosis/genetics , Legionella pneumophila/genetics , Legionella pneumophila/pathogenicity , Legionnaires' Disease/genetics , Nerve Tissue Proteins/genetics , Physical Chromosome Mapping/methods , Animals , Cell Line , Genetic Predisposition to Disease/genetics , Legionnaires' Disease/etiology , Mice , Mice, Inbred A , Mice, Inbred C57BL , Molecular Sequence Data , Multigene Family/genetics , Neuronal Apoptosis-Inhibitory Protein
10.
Proc Natl Acad Sci U S A ; 97(15): 8560-5, 2000 Jul 18.
Article in English | MEDLINE | ID: mdl-10890913

ABSTRACT

Over 2 billion people are estimated to be infected with virulent Mycobacterium tuberculosis, yet fewer than 10% progress to clinical tuberculosis within their lifetime. Twin studies and variations in the outcome of tuberculosis infection after exposure to similar environmental risks suggest genetic heterogeneity among individuals in their susceptibility to disease. In a mouse model of tuberculosis, we have established that resistance and susceptibility to virulent M. tuberculosis is a complex genetic trait. A new locus with a major effect on tuberculosis susceptibility, designated sst1 (susceptibility to tuberculosis 1), was mapped to a 9-centimorgan (cM) interval on mouse chromosome 1. It is located 10-19 cM distal to a previously identified gene, Nramp1, that controls the innate resistance of mice to the attenuated bacillus Calmette-Guérin vaccine strain. The phenotypic expression of the newly identified locus is distinct from that of Nramp1 in that sst1 controls progression of tuberculosis infection in a lung-specific manner. Mice segregating at the sst1 locus exhibit marked differences in the growth rates of virulent tubercle bacilli in the lungs. Lung lesions in congenic sst1-susceptible mice are characterized by extensive necrosis and unrestricted extracellular multiplication of virulent mycobacteria, whereas sst1-resistant mice develop interstitial granulomas and effectively control multiplication of the bacilli. The resistant allele of sst1, although powerful in controlling infection, is not sufficient to confer full protection against virulent M. tuberculosis, indicating that other genes located outside of the sst1 locus are likely also to be important for controlling tuberculosis infection.


Subject(s)
Mycobacterium tuberculosis/pathogenicity , Tuberculosis/genetics , Animals , Chromosome Mapping , Crosses, Genetic , Disease Models, Animal , Genetic Predisposition to Disease/genetics , Immunity, Innate/genetics , Lung/immunology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Pneumonia/immunology , Tuberculosis/microbiology , Virulence
11.
Genomics ; 64(1): 62-81, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10708519

ABSTRACT

The orthologous genomic segments on mouse chromosome 13D1-D3 and human chromosome 5q11.2-q13.3 have been extensively studied because of their involvement in two distinct disease phenotypes, spinal muscular atrophy (SMA) in human and susceptibility to Legionella pneumophila (determined by Lgn1) in mice. The overlapping intervals in both species contain genomic amplifications of distinct structure, indicating an independent origin. We have endeavored to construct a comprehensive comparative gene map of the mouse and human Lgn1/SMA intervals in the hopes that the origins and maintenance of the genomic amplifications may become clear. Our comparative gene map demonstrates that the only regional gene in common between the amplified segments in mouse and human is the Lgn1 candidate Naip/NAIP. We have also determined that mice of the 129 haplotype harbor seven intact and three partial Naip transcription units arranged in a closely linked direct repeat on chromosome 13. Several, but not all, of these Naip loci are contained within the Lgn1 critical interval. We present a model for the origins of the mouse and human repetitive arrays from a common ancestral haplotype.


Subject(s)
Evolution, Molecular , Legionnaires' Disease/genetics , Muscular Atrophy, Spinal/genetics , Nerve Tissue Proteins/genetics , Repetitive Sequences, Nucleic Acid , 5' Untranslated Regions , Animals , Blotting, Southern , Cell Line , Chromosome Mapping , Cyclic AMP Response Element-Binding Protein , Exons , Humans , Legionella pneumophila , Mice , Molecular Sequence Data , Nerve Tissue Proteins/classification , Neuronal Apoptosis-Inhibitory Protein , Physical Chromosome Mapping , RNA-Binding Proteins , SMN Complex Proteins
13.
Genomics ; 60(2): 137-51, 1999 Sep 01.
Article in English | MEDLINE | ID: mdl-10486205

ABSTRACT

Human chromosome 5q11.2-q13.3 and its ortholog on mouse chromosome 13 contain candidate genes for an inherited human neurodegenerative disorder called spinal muscular atrophy (SMA) and for an inherited mouse susceptibility to infection with Legionella pneumophila (Lgn1). These homologous genomic regions also have unusual repetitive organizations that create practical difficulties in mapping and raise interesting issues about the evolutionary origin of the repeats. In an attempt to analyze this region in detail, and as a way to identify additional candidate genes for these diseases, we have determined the sequence of 179 kb of the mouse Lgn1/SMA interval. We have analyzed this sequence using BLAST searches and various exon prediction programs to identify potential genes. Since these methods can generate false-positive exon declarations, our alignments of the mouse sequence with available human orthologous sequence allowed us to discriminate rapidly among this collection of potential coding regions by indicating which regions were well conserved and were more likely to represent actual coding sequence. As a result of our analysis, we accurately mapped two additional genes in the SMA interval that can be tested for involvement in the pathogenesis of SMA. While no new Lgn1 candidates emerged, we have identified new genetic markers that exclude Smn as an Lgn1 candidate. In addition to providing important resources for studying SMA and Lgn1, our data provide further evidence of the value of sequencing the mouse genome as a means to help with the annotation of the human genomic sequence and vice versa.


Subject(s)
Chromosomes, Human, Pair 5/genetics , Legionnaires' Disease/genetics , Muscular Atrophy, Spinal/genetics , Nerve Tissue Proteins/genetics , Animals , Base Sequence , Chromosome Mapping , Cyclic AMP Response Element-Binding Protein , DNA/genetics , DNA Primers/genetics , Exons , Genetic Markers , Humans , Mice , Molecular Sequence Data , Neuronal Apoptosis-Inhibitory Protein , Polymorphism, Genetic , RNA-Binding Proteins , SMN Complex Proteins , Sequence Homology, Nucleic Acid , Species Specificity
14.
Hum Genet ; 105(1-2): 17-27, 1999.
Article in English | MEDLINE | ID: mdl-10480350

ABSTRACT

Recently, the human orthologue to the cell cycle checkpoint genes rad17 (Schizosaccharomyces pombe) and RAD24 (Saccharomyces cerevisiae), called HRAD17, has been isolated and localized to chromosome 4. Independently, we have isolated the HRAD17 transcript and mapped it to chromosome 5q13 between the CCNB1 and BTF2p44cen genes. Furthermore, we have identified the complete exon-intron structure of HRAD17. The gene is organized into 14 exons, the translation initiation site lies within exon 2, and the stop codon within exon 14. Two further HRAD17 pseudogenes, HRAD17P1 and HRAD17P2, were identified on chromosomes 7p21 and 13q14.3, respectively, encompassing exons 3-14 and bearing 84% and 93% homology, respectively. Additionally, we have isolated the coding region of the mouse orthologue, Mrad17, and mapped it on chromosome 13 between Ccnb1 and Btf2p44, the same two genes between which it maps in human. The predicted Mrad17 polypeptide encompasses 687 amino acids and shows 89% similarity to HRAD17. Both genes are most highly expressed in testis compared to all other tissues, as shown by Northern blot hybridization. Histological studies, based on in situ hybridization with radioactively labeled antisense HRAD17 riboprobes, showed a strong expression within the germinal epithelium of the seminiferous tubuli in normal testis whereas in testicular tumors (seminomas) only weak, diffuse signals were seen. In light of the known function of the yeast orthologue at meiotic and mitotic checkpoints, as well as the strong expression in testis and weak expression in seminomas, we suggest a putative involvement of HRAD 17 in testicular tumorigenesis.


Subject(s)
Cell Cycle Proteins/genetics , Seminoma/genetics , Seminoma/metabolism , Testicular Neoplasms/genetics , Testicular Neoplasms/metabolism , Testis/metabolism , Animals , Base Sequence , Blotting, Northern , Blotting, Southern , Chromosome Mapping , Chromosomes, Human, Pair 5 , DNA, Complementary/analysis , DNA-Binding Proteins , Exons , Gene Library , Humans , In Situ Hybridization , In Situ Hybridization, Fluorescence , Intracellular Signaling Peptides and Proteins , Introns , Male , Mice , Models, Genetic , Molecular Sequence Data , Nuclear Proteins , Pseudogenes , Saccharomyces cerevisiae Proteins , Schizosaccharomyces pombe Proteins , Testis/anatomy & histology , Tissue Distribution
15.
Nat Genet ; 22(4): 388-93, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10431246

ABSTRACT

A physical map of the mouse genome is an essential tool for both positional cloning and genomic sequencing in this key model system for biomedical research. Indeed, the construction of a mouse physical map with markers spaced at an average interval of 300 kb is one of the stated goals of the Human Genome Project. Here we report the results of a project at the Whitehead Institute/MIT Center for Genome Research to construct such a physical map of the mouse. We built the map by screening sequenced-tagged sites (STSs) against a large-insert yeast artificial chromosome (YAC) library and then integrating the STS-content information with a dense genetic map. The integrated map shows the location of 9,787 loci, providing landmarks with an average spacing of approximately 300 kb and affording YAC coverage of approximately 92% of the mouse genome. We also report the results of a project at the MRC UK Mouse Genome Centre targeted at chromosome X. The project produced a YAC-based map containing 619 loci (with 121 loci in common with the Whitehead map and 498 additional loci), providing especially dense coverage of this sex chromosome. The YAC-based physical map directly facilitates positional cloning of mouse mutations by providing ready access to most of the genome. More generally, use of this map in addition to a newly constructed radiation hybrid (RH) map provides a comprehensive framework for mouse genomic studies.


Subject(s)
Chromosomes, Artificial, Yeast , Genome , Mice/genetics , Physical Chromosome Mapping , Animals , Chromosome Mapping , Contig Mapping , Genetic Markers , Models, Genetic
16.
J Immunol ; 162(3): 1339-47, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-9973388

ABSTRACT

Th phenotype development is controlled not only by cytokines but also by other parameters including genetic background. One site of genetic variation between murine strains that has direct impact on Th development is the expression of the IL-12 receptor. T cells from B10.D2 and BALB/c mice show distinct control of IL-12 receptor expression. When activated by Ag, B10.D2 T cells express functional IL-12 receptors and maintain IL-12 responsiveness. In contrast, under the same conditions, BALB/c T cells fail to express IL-12 receptors and become unresponsive to IL-12, precluding any Th1-inducing effects if subsequently exposed to IL-12. Previously, we identified a locus, which we termed T cell phenotype modifier 1 (Tpm1), on murine chromosome 11 that controls this differential maintenance of IL-12 responsiveness. In this study, we have produced a higher resolution map around Tpm1. We produced and analyzed a series of recombinants from a first-generation backcross that significantly narrows the genetic boundaries of Tpm1. This allowed us to exclude from consideration certain previous candidates for Tpm1, including IFN-regulatory factor-1. Also, cellular analysis of F1(B10.D2 x BALB/c) T cells demonstrates that Tpm1 exerts its effect on IL-12 receptor expression in a cell-autonomous manner, rather than through influencing the extracellular milieu. This result strongly implies that despite the proximity of our locus to the IL-13/IL-4 gene cluster, these cytokines are not candidates for Tpm1.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interleukin-12/pharmacology , Alleles , Animals , Base Sequence , Chromosome Mapping , Crosses, Genetic , DNA Primers/genetics , Female , Genetic Linkage , Genetic Markers , Genotype , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Phenotype , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Interleukin/genetics , Receptors, Interleukin-12 , Recombination, Genetic , Sequence Tagged Sites
17.
Radiat Meas ; 30(3): 345-59, 1999 Jun.
Article in English | MEDLINE | ID: mdl-11543141

ABSTRACT

In more than 25 years of almost continuous observations, the University of Chicago's Cosmic Ray Telescope (CRT) on IMP-8 has amassed a unique database on high-energy solar heavy ions of potential relevance to manned spaceflight. In the very largest particle events, IMP-8/CRT has even observed solar Fe ions above the Galactic cosmic ray background up to approximately 800 MeV/nucleon, an energy sufficiently high to penetrate nearly 25 g/cm2 of shielding. IMP-8/CRT observations show that high-energy heavy-ion spectra are often surprisingly hard power laws, without the exponential roll-offs suggested by stochastic acceleration fits to lower energy measurements alone. Also, in many solar particle events the Fe/O ratio grows with increasing energy, contrary to the notion that ions with higher mass-to-charge ratios should be less abundant at higher energies. Previous studies of radiation hazards for manned spaceflight have often assumed heavy-ion composition and steeply-falling energy spectra inconsistent with these observations. Conclusions based on such studies should therefore be re-assessed. The significant event-to-event variability observed in the high-energy solar heavy ions also has important implications for strategies in building probabilistic models of solar particle radiation hazards.


Subject(s)
Heavy Ions , Models, Theoretical , Radiation Monitoring/instrumentation , Solar Activity , Space Flight/instrumentation , Astronomy/instrumentation , Cosmic Radiation , Humans , Iron , Oxygen , Radiation Protection , Spacecraft/instrumentation
19.
Nat Genet ; 20(1): 83-6, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9731538

ABSTRACT

Spinal muscular atrophy (SMA) is a common recessive disorder characterized by the loss of lower motor neurons in the spinal cord. The disease has been classified into three types based on age of onset and severity. SMA I-III all map to chromosome 5q13 (refs 2,3), and nearly all patients display deletions or gene conversions of the survival motor neuron (SMN1) gene. Some correlation has been established between SMN protein levels and disease course; nevertheless, the genetic basis for SMA phenotypic variability remains unclear, and it has been postulated that the loss of an additional modifying factor contributes to the severity of type I SMA. Using comparative genomics to screen for such a factor among evolutionarily conserved sequences between mouse and human, we have identified a novel transcript, H4F5, which lies closer to SMN1 than any previously identified gene in the region. A multi-copy microsatellite marker that is deleted in more than 90% of type I SMA chromosomes is embedded in an intron of this gene, indicating that H4F5 is also highly deleted in type I SMA chromosomes, and thus is a candidate phenotypic modifier for SMA.


Subject(s)
Muscular Atrophy, Spinal/genetics , Nerve Tissue Proteins/genetics , Amino Acid Sequence , Animals , Chromosome Mapping , Chromosomes, Human, Pair 5 , Cloning, Molecular , Cyclic AMP Response Element-Binding Protein , Gene Deletion , Genetic Markers , Homozygote , Humans , Mice , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins , SMN Complex Proteins , Sequence Homology, Amino Acid , Survival of Motor Neuron 1 Protein
20.
Mol Microbiol ; 29(2): 581-91, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9720874

ABSTRACT

The lethal factor (LF) toxin that is produced by Bacillus anthracis plays an important role in the pathogenesis of anthrax. LF has mononuclear phagocyte-specific intoxicating effects that are not well understood. We have identified genetic differences in inbred mouse strains that determine whether their cultured macrophages are susceptible to the cytolytic effect of LF intoxication. Our identification of resistant and susceptible mouse strains enabled us to analyse crosses between these strains and to map a single responsible gene (called Ltx1) to chromosome 11. Ltx1 probably influences intoxication events that occur after the delivery of LF to the cytosol, as all mouse macrophages are killed by polypeptides containing the catalytic domain of Diphtheria toxin fused to the domain of LF required for cytosolic transport. Furthermore, the susceptibility phenotype is dominant to resistance, suggesting that resistance is caused by an absence of or polymorphism in a molecule that acts jointly with, or downstream of, the activity of LF. Our mapping of Ltx1 is a crucial first step in its positional cloning, which will provide more information about the mechanism of LF intoxication.


Subject(s)
Antigens, Bacterial , Bacillus anthracis/pathogenicity , Bacterial Toxins/toxicity , Chromosome Mapping , Macrophages/microbiology , Proteins/genetics , Animals , Female , Macrophages/metabolism , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred Strains , Polymorphism, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL