Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Toxicol Appl Pharmacol ; 298: 56-65, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26905748

ABSTRACT

Fumonisin B1 (FB1) is a mycotoxin produced by a common fungal contaminant of corn. Administration of FB1 to pregnant LM/Bc mice induces exencephaly in embryos, and ingestion of FB1-contaminated food during early pregnancy is associated with increased risk for neural tube defects (NTDs) in humans. FB1 inhibits ceramide synthase enzymes in sphingolipid biosynthesis, causing sphinganine (Sa) and bioactive sphinganine-1-phosphate (Sa1P) accumulation in blood, cells, and tissues. Sphingosine kinases (Sphk) phosphorylate Sa to form Sa1P. Upon activation, Sphk1 associates primarily with the plasma membrane, while Sphk2 is found predominantly in the nucleus. In cells over-expressing Sphk2, accumulation of Sa1P in the nuclear compartment inhibits histone deacetylase (HDAC) activity, causing increased acetylation of histone lysine residues. In this study, FB1 treatment in LM/Bc mouse embryonic fibroblasts (MEFs) resulted in significant accumulation of Sa1P in nuclear extracts relative to cytoplasmic extracts. Elevated nuclear Sa1P corresponded to decreased histone deacetylase (HDAC) activity and increased histone acetylation at H2BK12, H3K9, H3K18, and H3K23. Treatment of LM/Bc MEFs with a selective Sphk1 inhibitor, PF-543, or with ABC294640, a selective Sphk2 inhibitor, significantly reduced nuclear Sa1P accumulation after FB1, although Sa1P levels remained significantly increased relative to basal levels. Concurrent treatment with both PF-543 and ABC294640 prevented nuclear accumulation of Sa1P in response to FB1. Other HDAC inhibitors are known to cause NTDs, so these results suggest that FB1-induced disruption of sphingolipid metabolism leading to nuclear Sa1P accumulation, HDAC inhibition, and histone hyperacetylation is a potential mechanism for FB1-induced NTDs.


Subject(s)
Cell Nucleus/drug effects , Environmental Pollutants/toxicity , Fibroblasts/drug effects , Fumonisins/toxicity , Histone Deacetylases/metabolism , Neural Tube Defects/metabolism , Sphingosine/analogs & derivatives , Animals , Blotting, Western , Cell Line , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cytoplasm/drug effects , Cytoplasm/metabolism , Embryo, Mammalian , Fibroblasts/metabolism , Fibroblasts/pathology , Mice , Neural Tube Defects/embryology , Neural Tube Defects/pathology , Primary Cell Culture , Spectrometry, Mass, Electrospray Ionization , Sphingosine/metabolism , Tandem Mass Spectrometry
2.
Toxicol Sci ; 150(1): 161-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26719367

ABSTRACT

FTY720 (fingolimod) is a U.S. Food and Drug Administration-approved drug to treat relapsing remitting multiple sclerosis. FTY720 treatment in pregnant inbred LM/Bc mice results in approximately 60% of embryos having a neural tube defect (NTD). Sphingosine kinases (Sphk1, Sphk2) phosphorylate FTY720 in vivo to form the bioactive metabolite FTY720-1-phosphate (FTY720-P). Cytoplasmic FTY720-P is an agonist for 4 of the 5 sphingosine-1-phosphate (S1P) receptors (S1P1, 3-5) and can also act as a functional antagonist of S1P1, whereas FTY720-P generated in the nucleus inhibits histone deacetylases (HDACs), leading to increased histone acetylation. This study demonstrates that treatment of LM/Bc mouse embryonic fibroblasts (MEFs) with FTY720 results in a significant accumulation of FTY720-P in both the cytoplasmic and nuclear compartments. Elevated nuclear FTY720-P is associated with decreased HDAC activity and increased histone acetylation at H3K18 and H3K23 in LM/Bc MEFs. Treatment of LM/Bc MEFs with FTY720 and a selective Sphk2 inhibitor, ABC294640, significantly reduces the amount of FTY720-P that accumulates in the nucleus. The data provide insight into the relative amounts of FTY720-P generated in the nuclear versus cytoplasmic subcellular compartments after FTY720 treatment and the specific Sphk isoforms involved. The results of this study suggest that FTY720-induced NTDs may involve multiple mechanisms, including: (1) sustained and/or altered S1P receptor activation and signaling by FTY720-P produced in the cytoplasm and (2) HDAC inhibition and histone hyperacetylation by FTY720-P generated in the nucleus that could lead to epigenetic changes in gene regulation.


Subject(s)
Cell Nucleus/drug effects , Cytoplasm/drug effects , Fibroblasts/drug effects , Neural Tube Defects/chemically induced , Organophosphates/toxicity , Sphingosine/analogs & derivatives , Animals , Cell Line , Cell Nucleus/metabolism , Cell Survival/drug effects , Cytoplasm/metabolism , Fibroblasts/metabolism , Histone Deacetylases/metabolism , Histones/drug effects , Mice, Inbred Strains , Neural Tube Defects/embryology , Neural Tube Defects/metabolism , Organophosphates/metabolism , Protein Processing, Post-Translational/drug effects , Sphingosine/metabolism , Sphingosine/toxicity
3.
Neurogenetics ; 16(4): 277-85, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26051944

ABSTRACT

The progressive and permanent loss of cerebellar Purkinje cells (PC) is a hallmark of many inherited ataxias. Mutations in several genes involved in the regulation of Ca(2+) release from intracellular stores by the second messenger IP3 have been associated with PC dysfunction or death. While much is known about the defects in production and response to IP3, less is known about the defects in breakdown of the IP3 second messenger. A mutation in Inpp4a of the pathway is associated with a severe, early-onset PC degeneration in the mouse model weeble. The step preceding the removal of the 4-phosphate is the removal of the 5-phosphate by Inpp5a. Gene expression analysis was performed on an Inpp5a (Gt(OST50073)Lex) mouse generated by gene trap insertion using quantitative real-time PCR (qRT-PCR), immunohistochemistry, and Western blot. Phenotypic analyses were performed using rotarod, ß-galactosidase staining, and phosphatase activity assay. Statistical significance was calculated. The deletion of Inpp5a causes an early-onset yet slowly progressive PC degeneration and ataxia. Homozygous mutants (90%) exhibit perinatal lethality; surviving homozygotes show locomotor instability at P16. A consistent pattern of PC loss in the cerebellum is initially detectable by weaning and widespread by P60. Phosphatase activity toward phosphoinositol substrates is reduced in the mutant relative to littermates. The ataxic phenotype and characteristics neurodegeneration of the Inpp5a (Gt(OST50073)Lex) mouse indicate a crucial role for Inpp5a in PC survival. The identification of the molecular basis of the selective PC survival will be important in defining a neuroprotective gene applicable to establishing a disease mechanism.


Subject(s)
Cerebellar Ataxia/genetics , Gene Deletion , Phosphoric Monoester Hydrolases/genetics , Animals , Cerebellar Ataxia/embryology , Cerebellar Ataxia/enzymology , Cerebellar Ataxia/pathology , Disease Models, Animal , Inositol Polyphosphate 5-Phosphatases , Mice , Motor Activity/genetics , Phosphoric Monoester Hydrolases/metabolism , Phosphoric Monoester Hydrolases/physiology , Purkinje Cells/pathology , Rotarod Performance Test
4.
Birth Defects Res A Clin Mol Teratol ; 94(10): 790-803, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22991331

ABSTRACT

BACKGROUND: Fumonisin B(1) (FB(1)) is a mycotoxin produced by a common fungal contaminant of corn. Ingestion of FB(1)-contaminated food is associated with increased risk for neural tube defects (NTDs). FB(1) induces NTDs in inbred LM/Bc mice. FB(1) inhibits ceramide synthase in de novo sphingolipid biosynthesis, resulting in accumulation of sphinganine and sphinganine-1-phosphate (Sa1P). Sa1P functions as a ligand for a family of G protein-coupled S1P receptors. METHODS: Pregnant SWV and LM/Bc mice were treated with FB(1) (20 mg/kg/day intraperitoneally on embryonic day (ED) 7.5-8.5) or the known S1P receptor agonist FTY720 (10 mg/kg/day oral gavage on ED 6.5-8.5). LC/MS was used to detect sphingoid base-1-phosphates in maternal blood spots, plasma, and embryonic tissue. Strain-specific SWV and LM/Bc mouse embryonic fibroblasts (MEFs) and serum free mouse embryo (SFME) neural progenitor cells were treated with FB(1) (40 µM for 24 hr) and LC/MS was used to detect sphingoid base-1-phosphates. RESULTS: FTY720 induced NTDs in both the SWV and the LM/Bc strains of mice. Sphinganine-1-P (Sa1P) and FTY720-P were elevated in the blood spots and plasma of mice treated with FB(1) or FTY720, respectively. FTY720-P was elevated in ED 9.5 exencephalic embryos. Sa1P was elevated in SFME and MEF cells treated with FB(1), and Sa1P was higher in MEFs generated from the FB(1)-NTD-susceptible LM/Bc strain. CONCLUSIONS: Elevated sphingoid base-1-P after FB(1) or FTY720 suggest a potential role for these bioactive lipid ligands and activation of S1P receptor signaling pathways in the failure of neural tube closure after FB(1) or FTY720. Sa1P may represent a biomarker for FB(1)-NTD risk assessment.


Subject(s)
Fumonisins/adverse effects , Neural Tube Defects/chemically induced , Propylene Glycols/adverse effects , Sphingosine/analogs & derivatives , Animals , Cells, Cultured , Embryo, Mammalian , Female , Fingolimod Hydrochloride , Fumonisins/pharmacology , Mice , Mice, Inbred Strains , Models, Biological , Neural Tube Defects/pathology , Pregnancy , Prenatal Exposure Delayed Effects/blood , Prenatal Exposure Delayed Effects/chemically induced , Propylene Glycols/pharmacology , Sphingosine/adverse effects , Sphingosine/blood , Sphingosine/pharmacology , Up-Regulation/drug effects
5.
PLoS One ; 6(7): e21858, 2011.
Article in English | MEDLINE | ID: mdl-21779340

ABSTRACT

Variation in genetic background can significantly influence the phenotypic outcome of both disease and non-disease associated traits. Additionally, differences in temporal and strain specific gene expression can also contribute to phenotypes in the mammalian retina. This is the first report of microarray based cross-strain analysis of gene expression in the retina investigating genetic background effects. Microarray analyses were performed on retinas from the following mouse strains: C57BL6/J, AKR/J, CAST/EiJ, and NOD.NON-H2(-nb1) at embryonic day 18.5 (E18.5) and postnatal day 30.5 (P30.5). Over 3000 differentially expressed genes were identified between strains and developmental stages. Differential gene expression was confirmed by qRT-PCR, Western blot, and immunohistochemistry. Three major gene networks were identified that function to regulate retinal or photoreceptor development, visual perception, cellular transport, and signal transduction. Many of the genes in these networks are implicated in retinal diseases such as bradyopsia, night-blindness, and cone-rod dystrophy. Our analysis revealed strain specific variations in cone photoreceptor cell patterning and retinal function. This study highlights the substantial impact of genetic background on both development and function of the retina and the level of gene expression differences tolerated for normal retinal function. These strain specific genetic variations may also be present in other tissues. In addition, this study will provide valuable insight for the development of more accurate models for human retinal diseases.


Subject(s)
Genetic Variation/genetics , Retina/metabolism , Animals , Blotting, Western , Immunohistochemistry , Mice , Oligonucleotide Array Sequence Analysis , Phenotype , Retina/cytology , Retinal Cone Photoreceptor Cells/cytology , Retinal Cone Photoreceptor Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction
6.
PLoS One ; 6(3): e17494, 2011 Mar 08.
Article in English | MEDLINE | ID: mdl-21408158

ABSTRACT

The majority of diseases in the retina are caused by genetic mutations affecting the development and function of photoreceptor cells. The transcriptional networks directing these processes are regulated by genes such as nuclear hormone receptors. The nuclear hormone receptor gene Rev-erb alpha/Nr1d1 has been widely studied for its role in the circadian cycle and cell metabolism, however its role in the retina is unknown. In order to understand the role of Rev-erb alpha/Nr1d1 in the retina, we evaluated the effects of loss of Nr1d1 to the developing retina and its co-regulation with the photoreceptor-specific nuclear receptor gene Nr2e3 in the developing and mature retina. Knock-down of Nr1d1 expression in the developing retina results in pan-retinal spotting and reduced retinal function by electroretinogram. Our studies show that NR1D1 protein is co-expressed with NR2E3 in the outer neuroblastic layer of the developing mouse retina. In the adult retina, NR1D1 is expressed in the ganglion cell layer and is co-expressed with NR2E3 in the outer nuclear layer, within rods and cones. Several genes co-targeted by NR2E3 and NR1D1 were identified that include: Nr2c1, Recoverin, Rgr, Rarres2, Pde8a, and Nupr1. We examined the cyclic expression of Nr1d1 and Nr2e3 over a twenty-four hour period and observed that both nuclear receptors cycle in a similar manner. Taken together, these studies reveal a novel role for Nr1d1, in conjunction with its cofactor Nr2e3, in regulating transcriptional networks critical for photoreceptor development and function.


Subject(s)
Gene Regulatory Networks/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Orphan Nuclear Receptors/metabolism , Retina/metabolism , Aging/metabolism , Aging/radiation effects , Animals , Cell Count , Enhancer Elements, Genetic/genetics , Injections , Light , Light Signal Transduction/genetics , Light Signal Transduction/radiation effects , Mice , Organ Specificity/radiation effects , Protein Transport/radiation effects , RNA, Small Interfering/metabolism , Retina/cytology , Retina/growth & development , Retina/radiation effects , Staining and Labeling , Transcriptional Activation/genetics , Transcriptional Activation/radiation effects
7.
Invest Ophthalmol Vis Sci ; 51(6): 3115-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20042664

ABSTRACT

PURPOSE: Intraocular injection of linezolid, a synthetic oxazolidinone antibiotic, was performed in rabbits to assess its safety as a possible treatment for endophthalmitis. METHODS: Linezolid, 300 microg/0.1 mL, 200 microg/0.1 mL, or 100 microg/0.1 mL, was injected into the vitreous of the right eye of 12 rabbits. Balanced saline solution was injection into the left eye of each rabbit as a control. A standard electroretinogram (ERG) was obtained before injection and repeated 2 days and 1 and 4 weeks after injection. Intraocular pressure (IOP) was also measured after injection. After the experiment, the rabbits were euthanatized and the retinas were examined by light and electron microscopy. Differences between the two eyes in the ERGs, IOP, and histopathology were recorded. RESULTS: There were no statistically significant differences in the electroretinograms obtained between the linezolid-injected eyes and the control eyes. Histopathology showed no changes in the study eyes compared with the control eyes. CONCLUSIONS: Preservative-free linezolid is nontoxic to the retinas of rabbits when injected intravitreally, and this route can therefore be considered for the administration of linezolid in the treatment of endophthalmitis.


Subject(s)
Acetamides/toxicity , Anti-Infective Agents/toxicity , Oxazolidinones/toxicity , Retina/drug effects , Animals , Electroretinography/drug effects , Injections , Intraocular Pressure , Linezolid , Microscopy, Electron , Rabbits , Vitreous Body
8.
PLoS One ; 4(12): e8270, 2009 Dec 14.
Article in English | MEDLINE | ID: mdl-20011524

ABSTRACT

The weeble mutant mouse has a frame shift mutation in inositol polyphosphate 4-phosphatase type I (Inpp4a). The phenotype is characterized by an early onset cerebellar ataxia and neurodegeneration, especially apparent in the Purkinje cells. Purkinje cell loss is a common pathological finding in many human and mouse ataxic disorders. Here we show that in the Inpp4a(wbl) mutant, Purkinje cells are lost in a specific temporal and spatial pattern. Loss occurs early in postnatal development; however, prior to the appearance of climbing fibers in the developing molecular layer, the mutant has a normal complement of Purkinje cells and they are properly positioned. Degeneration and reactive gliosis are present at postnatal day 5 and progress rapidly in a defined pattern of patches; however, Inpp4a is expressed uniformly across Purkinje cells. In late stage mutants, patches of surviving Purkinje cells appear remarkably normal with the exception that the climbing fibers have been excessively eliminated. Surviving Purkinje cells express Eaat4, a glutamate transporter that is differentially expressed in subsets of Purkinje cells during development and into adult stages. Prior to Purkinje cell loss, reactive gliosis and dendritic atrophy can be seen in Eaat4 negative stripes. Our data suggest that Purkinje cell loss in the Inpp4a(wbl) mutant is due to glutamate excitotoxicity initiated by the climbing fiber, and that Eaat4 may exert a protective effect.


Subject(s)
Cerebellum/enzymology , Cerebellum/pathology , Cytoprotection , Excitatory Amino Acid Transporter 4/metabolism , Neurons/enzymology , Neurons/pathology , Phosphoric Monoester Hydrolases/metabolism , Animals , Mice , Mice, Neurologic Mutants , Phenotype , Purkinje Cells/enzymology , Purkinje Cells/pathology , Survival Analysis , Time Factors , Wasting Syndrome/enzymology , Wasting Syndrome/pathology , Weaning
9.
Exp Eye Res ; 89(3): 365-72, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19379737

ABSTRACT

The retinal transcription factor Nr2e3 plays a key role in photoreceptor development and function. In this study we examine gene expression in the retina of Nr2e3(rd7/rd7) mutants with respect to wild-type control mice, to identify genes that are misregulated and hence potentially function in the Nr2e3 transcriptional network. Quantitative candidate gene real time PCR and subtractive hybridization approaches were used to identify transcripts that were misregulated in Nr2e3(rd7/rd7) mice. Chromatin immunoprecipitation assays were then used to determine which of the misregulated transcripts were direct targets of NR2E3. We identified 24 potential targets of NR2E3. In the developing retina, NR2E3 targets transcription factors such as Ror1, Rorg, and the nuclear hormone receptors Nr1d1 and Nr2c1. In the mature retina NR2E3 targets several genes including the rod specific gene Gnb1 and cone specific genes blue opsin, and two of the cone transducin subunits, Gnat2 and Gnb3. In addition, we identified 5 novel transcripts that are targeted by NR2E3. While mislocalization of proteins between rods and cones was not observed, we did observe diminished concentration of GNB1 protein in adult Nr2e3(rd7/rd7) retinas. These studies identified novel transcriptional pathways that are potentially targeted by Nr2e3 in the retina and specifically demonstrate a novel role for NR2E3 in regulating genes involved in phototransduction.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Light Signal Transduction/genetics , Photoreceptor Cells, Vertebrate/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Retinal Degeneration/genetics , Animals , Disease Models, Animal , Eye Proteins/metabolism , Mice , Mice, Mutant Strains , Orphan Nuclear Receptors , Retina/growth & development , Retina/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/physiopathology , Transcription Factors/physiology
10.
Mamm Genome ; 19(9): 623-33, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18763016

ABSTRACT

The nmf193 mutant was generated by a large-scale ENU mutagenesis screen and originally described as having a dominantly inherited phenotype characterized by fundus abnormalities. We determined that nmf193 mice exhibit outer-segment defects and progressive retinal degeneration. Clinical examination revealed retinal spotting apparent at 6 weeks of age. Histologic analysis of homozygous mutant mice at 6 weeks indicated an absence of outer segments (OS) and a 50% reduction of photoreceptor cells which progressed to complete loss of photoreceptors by 10 months. Mice heterozygous for the nmf193 mutation had a less severe phenotype of shortened outer segments at 2 months with progressive loss of photoreceptor cells to 50% by 10 months. A positional cloning approach using a DNA pooling strategy was performed to identify the causative mutation in nmf193 mice. The nmf193 mutation was linked to chromosome 17 and fine mapped to an interval containing the peripherin/rds (Prph2) gene. Mutation analysis identified a single base change in Prph2 that causes aberrant splicing between exons 1 and 2. Interestingly, a comparative histologic analysis demonstrated that Prph2 ( nmf193/+ ) mutants have similar photoreceptor degeneration to that of Nr2e3 ( rd7/rd7 ). We show that Prph2 mRNA and protein levels are reduced in the Nr2e3 ( rd7/rd7 ) mutant compared to control littermates. Chromatin immunoprecipitation analysis shows that Prph2 is a direct target of NR2E3. In addition, the downregulation of Prph2 gene expression is similar in both the Nr2e3 ( rd7/rd7 ) and Prph2 ( nmf193/+ ) mutants, suggesting that the reduction of Prph2 may contribute to the degenerative pathology seen in Nr2e3 ( rd7/rd7 ).


Subject(s)
Intermediate Filament Proteins/genetics , Membrane Glycoproteins/genetics , Mutation , Nerve Tissue Proteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Retinal Degeneration/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , DNA Primers/genetics , Ethylnitrosourea/toxicity , Female , Heterozygote , Homozygote , Intermediate Filament Proteins/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Orphan Nuclear Receptors , Peripherins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinal Degeneration/pathology , Retinal Degeneration/physiopathology , Rod Cell Outer Segment/pathology , Rod Cell Outer Segment/physiology
11.
Mamm Genome ; 18(11): 749-56, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17934773

ABSTRACT

The identification of novel mutant alleles is important for understanding critical functional domains of a protein and establishing genotype:phenotype correlations. The recoil wobbler (rcw) allelic series of spontaneous ataxic mutants and the ENU-induced mutant nmf373 genetically mapped to a shared region of chromosome 10. Their mutant phenotypes are strikingly similar; all have an ataxic phenotype that is recessive, early-onset, and is not associated with neurodegeneration. In this study we used complementation tests to show that these series of mutants are allelic to a knockout mutant of Grm1. Subsequently, a duplication of exon 4 and three missense mutations were identified in Grm1: I160T, E292D, and G337E. All mutations occurred within the ligand-binding region and changed conserved amino acids. In the rcw mutant, the Grm1 gene is expressed and the protein product is properly localized to the molecular layer of the cerebellar cortex. Grm1 is responsible for the generation of inositol 1,4,5-trisphosphate (IP(3)). The inositol second messenger system is the central mechanism for calcium release from intracellular stores in cerebellar Purkinje cells. Several of the genes involved in this pathway are mutated in mouse ataxic disorders. The novel rcw mutants represent a resource that will have utility for further studies of inositol second-messenger-system defects in neurogenetic disorders.


Subject(s)
Mutation , Receptors, Metabotropic Glutamate/genetics , Alleles , Amino Acid Sequence , Animals , Ataxia/genetics , Ataxia/metabolism , Base Sequence , Binding Sites/genetics , Calcium Signaling , Cerebellar Cortex/metabolism , Chromosome Mapping , DNA/genetics , Exons , Female , Genetic Complementation Test , Inositol 1,4,5-Trisphosphate/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation, Missense , Phenotype , Purkinje Cells/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Metabotropic Glutamate/metabolism , Second Messenger Systems , Sequence Homology, Amino Acid
12.
Neurogenetics ; 8(1): 1-10, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17102983

ABSTRACT

The soluble N-ethylmaleimide sensitive factor attachment receptors are a large family of membrane-associated proteins that are critical for Ca(2+)-mediated synaptic vesicle release. This family includes the VAMP, synaptosomal-associated protein, and syntaxin proteins. In this report, we describe a mutation in vesicle-associated membrane protein 1(VAMP1)/synaptobrevin in the mouse neurological mutant lethal-wasting (lew). The lethal-wasting mutant phenotype is characterized by a general lack of movement and wasting, eventually leading to death before weaning. Mutants are visibly immobile and lay on their side by postnatal day 10 (P10). Before this stage, mutants can be identified by a failure to attempt to right themselves. Affected mice die on average at P15. We used a positional cloning strategy to identify the mutation associated with this neurological phenotype. Lethal wasting had previously been linked to chromosome 6. We further narrowed the genetic disease interval and selected a small number of candidate genes for mutation screening. Genes were evaluated by quantitative reverse transcription-polymerase chain reaction (RT-PCR) to detect differences in their expression levels between control and mutant brain ribonucleic acid (RNA) samples. VAMP1 mRNA was found to be significantly downregulated in the lethal-wasting brain compared to wild-type littermates. Subsequently, a nonsense mutation was identified in the coding region of the gene. This mutation is predicted to truncate approximately half of the protein; however, Western blot analysis showed that no protein is detectable in the mutant. VAMP1 is selectively expressed in the retina and in discrete areas of the brain including the zona incerta and rostral periolivary region, although no gross histological abnormalities were observed in these tissues. Taken together, these data indicate that VAMP1 has a vital role in a subset of central nervous system tissues.


Subject(s)
Brain/pathology , Codon, Nonsense , Nervous System Diseases/genetics , R-SNARE Proteins/genetics , Vesicle-Associated Membrane Protein 1/genetics , Wasting Syndrome/genetics , Amino Acid Sequence , Animals , Chromosome Mapping , Disease Models, Animal , Genotype , Mice , Mice, Neurologic Mutants , Molecular Sequence Data , Nervous System Diseases/pathology , Reverse Transcriptase Polymerase Chain Reaction , Wasting Syndrome/mortality , Wasting Syndrome/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...