Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Nat Genet ; 10(3): 301-6, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7670468

ABSTRACT

Dystonia musculorum (dt) is a hereditary neurodegenerative disease in mice that leads to a sensory ataxia. We describe cloning of a candidate dt gene, dystonin, that is predominantly expressed in the dorsal root ganglia and other sites of neurodegeneration in dt mice. Dystonin encodes an N-terminal actin binding domain and a C-terminal portion comprised of the hemidesmosomal protein, bullous pemphigoid antigen 1 (bpag1). dt and bpag1 are part of the same transcription unit which is partially deleted in a transgenic strain of mice, Tg4, that harbours an insertional mutation at the dt locus, and in mice that carry a spontaneous dt mutation, dtAlb. We also demonstrate abnormal dystonin transcripts in a second dt mutant, dt24J. We conclude that mutations in the dystonin gene are the primary genetic lesion in dt mice.


Subject(s)
Autoantigens/genetics , Carrier Proteins , Collagen , Cytoskeletal Proteins/genetics , Dystonia Musculorum Deformans/genetics , Nerve Tissue Proteins/genetics , Non-Fibrillar Collagens , Pemphigoid, Bullous/immunology , Amino Acid Sequence , Animals , Base Sequence , DNA, Complementary/genetics , Dystonia Musculorum Deformans/immunology , Dystonin , Gene Expression , Humans , In Situ Hybridization , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Pemphigoid, Bullous/genetics , Sequence Homology, Amino Acid , Species Specificity , Transcription, Genetic , Collagen Type XVII
2.
Hum Mol Genet ; 19(7): 1211-20, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20053670

ABSTRACT

The molecular mechanisms regulating expression of utrophin A are of therapeutic interest since upregulating its expression at the sarcolemma can compensate for the lack of dystrophin in animal models of Duchenne Muscular Dystrophy (DMD). The 5'-UTR of utrophin A has been previously shown to drive cap-independent internal ribosome entry site (IRES)-mediated translation in response to muscle regeneration and glucocorticoid treatment. To determine whether the utrophin A IRES displays tissue specific activity, we generated transgenic mice harboring control (CMV/betaGAL/CAT) or utrophin A 5'-UTR (CMV/betaGAL/UtrA/CAT) bicistronic reporter transgenes. Examination of multiple tissues from two CMV/betaGAL/UtrA/CAT lines revealed that the utrophin A 5'-UTR drives cap-independent translation of the reporter gene exclusively in skeletal muscles and no other examined tissues. This expression pattern suggested that skeletal muscle-specific factors are involved in IRES-mediated translation of utrophin A. We performed RNA-affinity chromatography experiments combined with mass spectrometry to identify trans-factors that bind the utrophin A 5'-UTR and identified eukaryotic elongation factor 1A2 (eEF1A2). UV-crosslinking experiments confirmed the specificity of this interaction. Regions of the utrophin A 5'-UTR that bound eEF1A2 also mediated cap-independent translation in C2C12 muscle cells. Cultured cells lacking eEF1A2 had reduced IRES activity compared with cells overexpressing eEF1A2. Together, these results suggest an important role for eEF1A2 in driving cap-independent translation of utrophin A in skeletal muscle. The trans-factors and signaling pathways driving skeletal-muscle specific IRES-mediated translation of utrophin A could provide unique targets for developing pharmacological-based DMD therapies.


Subject(s)
5' Untranslated Regions , Peptide Elongation Factor 1/metabolism , Protein Biosynthesis , Utrophin/genetics , Animals , Binding Sites , Cells, Cultured , Gene Expression Regulation , Genes, Reporter , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Organ Specificity , Ribosomes
3.
Brain Res ; 1226: 33-8, 2008 Aug 21.
Article in English | MEDLINE | ID: mdl-18619576

ABSTRACT

The trigeminal ganglion (TG) and mesencephalic trigeminal tract nucleus (Mes5) were investigated in wild type and dystonia musculorum (dt) mice to study the effect of dystonin deficiency on primary sensory neurons in the trigeminal nervous system. At postnatal day 14, the number of TG neurons was markedly decreased in dt mice when compared to wild type mice (43.1% reduction). In addition, dystonin disruption decreased the number of sensory neurons which bound to isolectin B4, and contained calcitonin gene-related peptide or high-affinity nerve growth factor receptor TrkA. Immunohistochemistry for caspase-3 demonstrated that dystonin deficiency induced excess cell death of TG neurons during the early postnatal period. In contrast, Mes5 neurons were barely affected in dt mice. These data together suggest that dystonin is necessary for survival of nociceptors but not proprioceptors in the trigeminal nervous system.


Subject(s)
Cytoskeletal Proteins/deficiency , Nerve Tissue Proteins/deficiency , Nociceptors/metabolism , Sensory Receptor Cells/metabolism , Trigeminal Ganglion/cytology , Trigeminal Nuclei/cytology , Animals , Carrier Proteins , Caspase 3/metabolism , Dystonin , Gene Expression Regulation/genetics , Lectins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Receptor, trkA/metabolism
4.
Curr Biol ; 9(20): 1203-6, 1999 Oct 21.
Article in English | MEDLINE | ID: mdl-10531040

ABSTRACT

In mammals, growth of the fetal heart is regulated by proliferation of cardiac muscle cells. At later stages of pre-natal life, this proliferation diminishes profoundly [1] [2] and the dramatic expansion in heart size during the transition to adulthood is due exclusively to hypertrophy of individual cardiomyocytes [3] [4] [5]. Cardiomyocyte hypertrophy also contributes to the pathology of most post-natal heart disease [6] [7] [8] [9] [10]. Within this context, numerous signal transduction pathways have been implicated as the link between the effector(s) and altered cardiac gene expression [11] [12] [13] [14] [15] [16]. A common pathway has yet to be discovered, however. Here, we found that the activity of the stress-activated kinase p38 was enhanced in both types of cardiomyocyte hypertrophy. We also found that a target of the activated p38 kinase is the cardiac transcription factor MEF2. Transgenic mice expressing a dominant-negative form of MEF2C displayed attenuated post-natal growth of the myocardium. These results provide the first evidence for a single pathway regulating both normal and pathologic cardiomyocyte hypertrophy.


Subject(s)
Cardiomegaly/genetics , DNA-Binding Proteins/genetics , Heart/growth & development , Transcription Factors/genetics , Animals , Cardiomegaly/metabolism , DNA-Binding Proteins/metabolism , MEF2 Transcription Factors , Mice , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Myocardium/metabolism , Myogenic Regulatory Factors , Transcription Factors/metabolism , Up-Regulation , p38 Mitogen-Activated Protein Kinases
5.
Brain Res ; 1129(1): 142-6, 2007 Jan 19.
Article in English | MEDLINE | ID: mdl-17156752

ABSTRACT

The anterior part of the tongue was examined in wild type and dystonia musculorum mice to assess the effect of dystonin loss on fungiform papillae. In the mutant mouse, the density of fungiform papillae and their taste buds was severely decreased when compared to wild type littermates (papilla, 67% reduction; taste bud, 77% reduction). The mutation also reduced the size of these papillae (17% reduction) and taste buds (29% reduction). In addition, immunohistochemical analysis demonstrated that the dystonin mutation reduced the number of PGP 9.5 and calbindin D28k-containing nerve fibers in fungiform papillae. These data together suggest that dystonin is required for the innervation and development of fungiform papillae and taste buds.


Subject(s)
Carrier Proteins/genetics , Cytoskeletal Proteins/genetics , Nerve Tissue Proteins/genetics , Taste Buds/abnormalities , Taste Buds/metabolism , Taste Disorders/metabolism , Tongue/abnormalities , Tongue/metabolism , Animals , Calbindin 1 , Calbindins , Chorda Tympani Nerve/abnormalities , Chorda Tympani Nerve/metabolism , Chorda Tympani Nerve/physiopathology , Disease Models, Animal , Dystonic Disorders/genetics , Dystonic Disorders/metabolism , Dystonic Disorders/physiopathology , Dystonin , Geniculate Ganglion/abnormalities , Geniculate Ganglion/metabolism , Geniculate Ganglion/physiopathology , Immunohistochemistry , Mice , Mice, Knockout , Mutation/genetics , S100 Calcium Binding Protein G/metabolism , Sensory Receptor Cells/abnormalities , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiopathology , Taste Buds/physiopathology , Taste Disorders/genetics , Taste Disorders/physiopathology , Tongue/physiopathology , Ubiquitin Thiolesterase/metabolism
6.
Mol Cell Biol ; 4(9): 1785-91, 1984 Sep.
Article in English | MEDLINE | ID: mdl-6092938

ABSTRACT

RTG-2 cells, a line of fibroblasts from rainbow trout (Salmo gairdnerii), are induced to synthesize a distinct set of heat-shock polypeptides after exposure to elevated temperature or to low concentrations of sodium arsenite. We isolated and characterized two cDNA sequences, THS70.7 and THS70.14, encoding partial information for two distinct species of 70-kilodalton heat shock polypeptide (hsp70) from these cells. These sequences are identical at 73.3% of the nucleotide positions in their regions of overlap, and their degree of sequence conservation at the polypeptide level is 88.1%. The two derived trout hsp70 polypeptide sequences show extensive homology with derived amino acid sequences for hsp70 polypeptides from Drosophila melanogaster and Saccharomyces cerevisiae. Northern blot analysis of RNA from arsenite-induced RTG-2 cells, with the trout hsp70 cDNAs as probes, revealed the presence of three hsp70 mRNA species. Southern blot analysis of trout testis DNA cleaved with various restriction endonucleases revealed a small number of bands hybridizing to the hsp70 cDNAs, suggesting the existence of a small family of hsp70 genes in this species. Finally, trout hsp70 cDNA sequences cross-hybridized with restriction fragments in genomic DNA from HeLa cells, bovine liver, Caenorhabditis elegans, and D. melanogaster.


Subject(s)
DNA/analysis , Heat-Shock Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , DNA Restriction Enzymes , Drosophila melanogaster/genetics , Fibroblasts/metabolism , Molecular Weight , Nucleic Acid Hybridization , Saccharomyces cerevisiae/genetics , Species Specificity , Trout
7.
Nat Biotechnol ; 18(7): 746-9, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10888842

ABSTRACT

An important issue in developmental biology is the identification of homeoprotein target genes. We have developed a strategy based on the internalization and nuclear addressing of exogenous homeodomains, using an engrailed homeodomain (EnHD) to screen an embryonic stem (ES) cell gene trap library. Eight integrated gene trap loci responded to EnHD. One is within the bullous pemphigoid antigen 1 (BPAG1) locus, in a region that interrupts two neural isoforms. By combining in vivo electroporation with organotypic cultures, we show that an already identified BPAG1 enhancer/promoter is differentially regulated by homeoproteins Hoxc-8 and Engrailed in the embryonic spinal cord and mesencephalon. This strategy can therefore be used for identifying and mutating homeoprotein targets. Because homeodomain third helices can internalize proteins, peptides, phosphopeptides, and antisense oligonucleotides, this strategy should be applicable to other intracellular targets for characterizing genetic networks involved in a large number of physiopathological states.


Subject(s)
Carrier Proteins , Cytoskeletal Proteins , Homeodomain Proteins/genetics , Nerve Tissue Proteins , Non-Fibrillar Collagens , Sequence Analysis, DNA/methods , Transcription Factors , Animals , Autoantigens/biosynthesis , Autoantigens/genetics , Brain/embryology , Brain/metabolism , Cell Nucleus/metabolism , Collagen/biosynthesis , Collagen/genetics , Cytoplasm/metabolism , Dystonin , Electroporation , Embryo, Mammalian/cytology , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Mice , Models, Genetic , Plasmids/metabolism , Promoter Regions, Genetic , Protein Isoforms , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/embryology , Spinal Cord/metabolism , Stem Cells/cytology , Collagen Type XVII
8.
Neuroscience ; 137(2): 531-6, 2006.
Article in English | MEDLINE | ID: mdl-16289886

ABSTRACT

The vagal and glossopharyngeal sensory ganglia and their peripheral tissues were examined in wild type and dystonia musculorum mice to assess the effect of dystonin loss of function on chemoreceptive neurons. In the mutant mouse, the number of vagal and glossopharyngeal sensory neurons was severely decreased (70% reduction) when compared with wild type littermates. The mutation also reduced the size of the circumvallate papilla (45% reduction) and the number of taste buds (89% reduction). In addition, immunohistochemical analysis demonstrated that the dystonin mutation reduced the number of PGP 9.5-, calcitonin gene-related peptide-, P2X3 receptor- and tyrosine hydroxylase-containing neurons. Their peripheral endings also decreased in the taste bud and epithelium of circumvallate papillae. These data together suggest that the survival of vagal and glossopharyngeal sensory neurons is dependent upon dystonin.


Subject(s)
Carrier Proteins/physiology , Cytoskeletal Proteins/physiology , Ganglia, Sensory/abnormalities , Glossopharyngeal Nerve/abnormalities , Nerve Tissue Proteins/physiology , Neurons, Afferent/metabolism , Vagus Nerve/abnormalities , Animals , Animals, Newborn , Calcitonin Gene-Related Peptide/metabolism , Carrier Proteins/genetics , Cell Differentiation/genetics , Cell Survival/genetics , Chemoreceptor Cells/abnormalities , Chemoreceptor Cells/metabolism , Chemoreceptor Cells/pathology , Cytoskeletal Proteins/genetics , Down-Regulation/genetics , Dystonin , Ganglia, Sensory/metabolism , Ganglia, Sensory/pathology , Glossopharyngeal Nerve/metabolism , Glossopharyngeal Nerve/pathology , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons, Afferent/pathology , Nodose Ganglion/abnormalities , Nodose Ganglion/metabolism , Nodose Ganglion/pathology , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2X3 , Sensory Receptor Cells/abnormalities , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/pathology , Taste/genetics , Taste Buds/abnormalities , Taste Buds/pathology , Tyrosine 3-Monooxygenase/metabolism , Ubiquitin Thiolesterase/metabolism , Vagus Nerve/metabolism , Vagus Nerve/pathology
9.
Curr Mol Med ; 16(9): 779-792, 2016.
Article in English | MEDLINE | ID: mdl-27894243

ABSTRACT

Spinal muscular atrophy (SMA) is the most common genetically inherited neurodegenerative disease resulting in infant mortality. SMA is caused by genetic deletion or mutation in the survival of motor neuron 1 (SMN1) gene, which results in reduced levels of the survival of motor neuron (SMN) protein. SMN protein deficiency preferentially affects α- motor neurons, leading to their degeneration and subsequent atrophy of limb and trunk muscles, progressing to death in severe forms of the disease. More recent studies have shown that SMN protein depletion is detrimental to the functioning of other tissues including skeletal muscle, heart, autonomic and enteric nervous systems, metabolic/endocrine (e.g. pancreas), lymphatic, bone and reproductive system. In this review, we summarize studies discussing SMN protein's function in various cell and tissue types and their involvement in the context of SMA disease etiology. Taken together, these studies indicate that SMA is a multi-organ disease, which suggests that truly effective disease intervention may require body-wide correction of SMN protein levels.


Subject(s)
Motor Neurons/pathology , Muscular Atrophy, Spinal/etiology , Muscular Atrophy, Spinal/pathology , Animals , Atrophy/genetics , Atrophy/metabolism , Atrophy/pathology , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy, Spinal/genetics , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism
10.
Mech Dev ; 35(1): 25-31, 1991 Aug.
Article in English | MEDLINE | ID: mdl-1659441

ABSTRACT

Transgenic mice carrying the human cytomegalovirus immediate early gene promoter driving the E. coli lacZ gene displayed an unusual cell specific expression of beta-galactosidase during development. LacZ expression was first detected in cells lining the apex of the neural fold of day 8.5 embryos. By day 10 of gestation, expression was prominent in the spinal ganglia, the ganglia of cranial nerves V, VII, VIII, IX, and X, in a line of cells marking the ventrolateral pathway adjacent to the dermamyotome, and in a column of differentiated cells in the entire ventrolateral neural tube posterior to the mesencephalon. Expression was also found in the myotomes. Neural tube explants from day 8.5 embryos cultured in vitro showed lacZ expression in cells migrating away from the explant. We conclude that the HCMV-IEP-lacZ transgene is expressed in a subpopulation of neural crest cells and its early derivatives.


Subject(s)
Cytomegalovirus/genetics , Gene Expression Regulation, Viral/genetics , Genes, Viral/genetics , Promoter Regions, Genetic/genetics , Animals , Base Sequence , DNA/genetics , Embryo, Mammalian/metabolism , Escherichia coli/genetics , Female , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Neural Crest/cytology , Neural Crest/embryology , Pregnancy , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
11.
Mol Cell Neurosci ; 10(5/6): 243-57, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9618216

ABSTRACT

The mouse neurological mutant dystonia musculorum (dt) suffers from a hereditary sensory neuropathy. We have previously described the cloning and characterization of the dt gene, which we named dystonin (Dst). We had shown that dystonin is a neural isoform of bullous pemphigoid antigen 1 (Bpag1) with an N-terminal actin-binding domain. It has been shown previously that dystonin is a cytoskeletal linker protein, forming a bridge between F-actin and intermediate filaments. Here, we have used two different antibody preparations against dystonin and detected a high-molecular-weight protein in immunoblot analysis of spinal cord extracts. We also show that this high-molecular-weight protein was not detectable in the nervous system of all dt alleles tested. Immunohistochemical analysis revealed that dystonin was present in different compartments of neurons-cell bodies, dendrites, and axons, regions which are rich in the three elements of the cytoskeleton (F-actin, neurofilaments, and microtubules). Ultrastructural analysis of dt dorsal root axons revealed disorganization of the neurofilament network and surprisingly also of the microtubule network. In this context it is of interest that we observed altered levels of the microtubule-associated proteins MAP2 and tau in spinal cord neurons of different dt alleles. Finally, dt dorsal root ganglion neurons formed neurites in culture, but the cytoskeleton was disorganized within these neurites. Our results demonstrate that dystonin is essential for maintaining neuronal cytoskeleton integrity but is not required for establishing neuronal morphology. Copyright 1998 Academic Press.

12.
J Comp Neurol ; 432(2): 155-68, 2001 Apr 02.
Article in English | MEDLINE | ID: mdl-11241383

ABSTRACT

We have investigated the fate of different neurotrophin-responsive subpopulations of dorsal root ganglion neurons in dystonia musculorum (dt) mice. These mice have a null mutation in the cytoskeletal linker protein, dystonin. Dystonin is expressed by all sensory neurons and cross links actin filaments, intermediate filaments, and microtubules. The dt mice undergo massive sensory neurodegeneration postnatally and die at around 4 weeks of age. We assessed the surviving and degenerating neuronal populations by comparing the dorsal root ganglion (DRG) neurons and central and peripheral projections in dt mice and wildtype mice. Large, neurofilament-H-positive neurons, many of which are muscle afferents and are neurotrophin-3 (NT-3)-responsive, were severely decreased in number in dt DRGs. The loss of muscle afferents was correlated with a degeneration of muscle spindles in skeletal muscle. Nerve growth factor (NGF)-responsive populations, which were visualized using calcitonin gene-related peptide and p75, appeared qualitatively normal in the lumbar spinal cord, DRG, and hindlimb skin. In contrast, glial cell line-derived neurotrophic factor (GDNF)-responsive populations, which were visualized using the isolectin B-4 and thiamine monophosphatase, were severely diminished in the lumbar spinal cord, DRG, and hindlimb skin. Analysis of NT-3, NGF, and GDNF mRNA levels using semiquantitative reverse transcriptase-polymerase chain reaction revealed normal trophin synthesis in the peripheral targets of dt mice, arguing against decreased trophic synthesis as a possible cause of neuronal degeneration. Thus, the absence of dystonin results in the selective survival of NGF-responsive neurons and the postnatal degeneration of many NT-3- and GDNF-responsive neurons. Our results reveal that the loss of this ubiquitously expressed cytoskeletal linker has diverse effects on sensory subpopulations. Moreover, we show that dystonin is critical for the maintenance of certain DRG neurons, and its function may be related to neurotrophic support.


Subject(s)
Carrier Proteins , Cytoskeletal Proteins/deficiency , Ganglia, Spinal/metabolism , Muscle Spindles/metabolism , Nerve Growth Factors , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/metabolism , Neurons, Afferent/metabolism , Neurotrophin 3/metabolism , Animals , Axons/metabolism , Axons/pathology , Cell Size/physiology , Dystonia/genetics , Dystonin , Ganglia, Spinal/growth & development , Ganglia, Spinal/pathology , Glial Cell Line-Derived Neurotrophic Factor , Lectins/metabolism , Mice , Mice, Mutant Strains , Muscle Spindles/pathology , Nerve Growth Factor/metabolism , Neurons, Afferent/pathology , Phosphoric Monoester Hydrolases/metabolism
13.
Brain Res Dev Brain Res ; 72(1): 99-105, 1993 Mar 19.
Article in English | MEDLINE | ID: mdl-8453768

ABSTRACT

We have reinvestigated the neural crest defect of Splotch (Sp1H) mutant embryos using the tissue specific expression of lacZ by the HCMV-IEP-lacZ (CMZ) transgene as a marker. The CMZ transgene was backcrossed onto the Sp1H mutant background, which has been shown to carry mutations in the Pax-3 gene. The CMZ transgene has previously been shown to be expressed in some neural crest-derived neural tissues of midgestation embryos. The pattern of CMZ expression in Splotch mutants is not caused by alterations of transgene transcription, but demonstrates morphological deviations of neural crest development. The gradual size reduction of spinal ganglia along a rostrocaudal gradient is shown to occur concomitantly with a size reduction of the sympathetic ganglia. CMZ expression also reveals the total absence of sympathetic ganglion cells in thoracic and lumbar segments of Sp1H homozygotes, which is confirmed in serial sections. Observations in whole mounts of CMZ transgenic homozygotes suggest that cranial nerve ganglia develop normally in these embryos. CMZ is expressed in epithelial cells around the neural tube defect in Splotch mutants at the epidermal/neuroepithelial boundary. It is proposed that this expression represents premigratory neural crest cells that remain within the epithelial layer around the neural tube defect. These observations are discussed with reference to the normal pattern of Pax-3 expression.


Subject(s)
Neural Tube Defects/embryology , Transcription Factors , beta-Galactosidase/genetics , Animals , Cell Adhesion Molecules, Neuronal/analysis , DNA/genetics , DNA/isolation & purification , DNA-Binding Proteins/genetics , Embryo, Mammalian/cytology , Female , Immunohistochemistry , Mice , Mice, Neurologic Mutants , Mice, Transgenic , Neural Tube Defects/pathology , PAX3 Transcription Factor , Paired Box Transcription Factors , Pregnancy , Sympathetic Nervous System/embryology , beta-Galactosidase/analysis
14.
Anat Embryol (Berl) ; 187(2): 153-60, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8238963

ABSTRACT

Homozygosity for the Splotch mutation causes neural tube and neural crest defects in mice. It has been demonstrated that Splotch mutant mice carry mutations in the homeodomain of the Pax-3 gene. Pax-3 is expressed in the neural tube, some neural crest derivatives, the mesenchyme of the limb bud and the somites. We have examined the development of the somite-derived skeletal muscles in homozygotes carrying the Splotch (Sp1H) mutation. Our results suggest that the Splotch mutation affects the development of skeletal muscles in a region-specific way: 1. The expression of the CMZ transgene in homozygotes reveals a disorganisation of the dermomyotome in whole stained embryos. 2. The axial musculature is reduced in size along a rostro-caudal gradient. 3. The muscle anlagen in the limbs develop much more slowly. Muscles of the head and the ventral body wall are normally developed in the mutant on day 13.5 of gestation. Recently, it has been shown that the myogenic precursors of the limbs are derived from the lateral half of the somite. The specific disturbance of muscle development in the limbs of Splotch mutants thus suggests a role for Pax-3 in the organisation of the somite, the production of trophic factors in the limb mesenchyme or an alteration of myogenic and mesenchymal cells.


Subject(s)
Embryonic and Fetal Development , Extremities/embryology , Mice, Neurologic Mutants/embryology , Muscles/embryology , Mutation , Animals , Homozygote , Mice , Mice, Neurologic Mutants/genetics , Neural Tube Defects/genetics
15.
Curr Mol Med ; 13(7): 1160-74, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23514457

ABSTRACT

Spinal muscular atrophy (SMA) is caused by mutations that reduce the level of the survival motor neuron protein (SMN) resulting in death of alpha-motor neurons, yet it is unclear why these cells are preferentially affected by a reduction in this ubiquitously-expressed protein. In mouse models of SMA, one of the earliest events detected is defects at the neuromuscular junction (NMJ). Although NMJs are established at a normal frequency, there are structural as well as functional perturbations and a lack of maturation of the primitive synapse. These early defects are followed by loss of the NMJ, denervation of the muscle and onset of muscle atrophy. In this review, we discuss our current understanding of the contribution of NMJ dysfunction in SMA disease pathogenesis, and also provide an overview of therapies currently under preclinical and clinical development for treatment of SMA.


Subject(s)
Muscular Atrophy, Spinal/genetics , Nerve Degeneration/genetics , Neuromuscular Junction/genetics , Survival of Motor Neuron 1 Protein/genetics , Animals , Disease Models, Animal , Disease Progression , Humans , Mice , Motor Neurons/pathology , Muscular Atrophy, Spinal/pathology , Mutation , Nerve Degeneration/pathology , Neuromuscular Junction/pathology , Synapses/pathology
17.
Dev Genet ; 22(2): 160-8, 1998.
Article in English | MEDLINE | ID: mdl-9581287

ABSTRACT

Dystonia musculorum (dt) is a recessive hereditary neuropathy of the mouse. Affected animals display loss of limb coordination and twisting of the trunk. Sensory nerve fibers of these mice are severely reduced in number, and the remaining fibers present numerous axonal swellings. The gene defective in dt, dystonin (Dst), encodes a cytoskeletal linker protein that forms the bridge between F-actin and intermediate filaments. Dst is expressed during embryogenesis, whereas overt phenotype in dt mice only appears during the second week after birth. Here we show that axonal swellings are present in sensory nerve fibers of dt embryos as early as E15.5, before myelination and radial axonal growth have begun. Thus disease progression is gradual in dt mice, having begun during embryogenesis. In dt embryos, microtubule network disorganization and cytoplasmic organelle accumulation within axonal swellings were consistently observed. In addition, a few of the axonal swellings presented intermediate filament accumulation. These results demonstrate that dystonin is required for cytoskeleton organization during axonogenesis. They also suggest that axonal transport defects, through microtubule network perturbation, may be the primary mechanism of neurodegeneration in dt mice.


Subject(s)
Carrier Proteins , Dystonia Musculorum Deformans/embryology , Dystonia Musculorum Deformans/pathology , Gene Expression Regulation, Developmental , Nervous System/pathology , Animals , Cytoskeletal Proteins/genetics , Cytoskeleton/genetics , Cytoskeleton/pathology , Dystonia Musculorum Deformans/genetics , Dystonin , Mice , Mice, Mutant Strains , Mutation , Nerve Tissue Proteins/genetics , Nervous System/embryology
18.
Can J Biochem ; 60(3): 347-55, 1982 Mar.
Article in English | MEDLINE | ID: mdl-6805927

ABSTRACT

The heat-shock response has been characterized in cultured fibroblasts of the rainbow trout, Salmo gairdnerii. The response has been elicited by two different stress situations; cells were either subjected to higher temperatures than normal (27 to 29 degrees C as opposed to 22 degrees C) or were incubated in medium containing sodium arsenite (15 to 100 microM final concentration). The response of the cells to these conditions is to synthesize a set of new polypeptides, the "heat-shock polypeptides" (hsps), that are not present or present in extremely low amounts in noninduced cells. Furthermore, during prolonged arsenite induction, the synthesis of normal cellular proteins is repressed. In trout fibroblasts, at least six hsps are detectable. These range from 30 000 to 87 000 in molecular weight and are referred to as hsp30, hsp32, hsp42, hsp62, hsp70, and hsp87. The hsp30 and hsp70 components are the most abundant and can be visualized by Coomassie blue staining of gels after prolonged induction. The heat-shock response is a reversible process in trout cells. Results of in vitro translation of mRNA from induced cells indicate that the control of hsp induction may be at the transcriptional level. Hsp70 from trout comigrates with the major hsp from Drosophila melanogaster on sodium dodecyl sulfate - polyacrylamide gels, suggesting that this protein may be highly conserved in evolution.


Subject(s)
Arsenic/pharmacology , Arsenites , Protein Biosynthesis , Animals , Cells, Cultured , Drosophila melanogaster/genetics , Fibroblasts/metabolism , Gene Expression Regulation , Heat-Shock Proteins , Hot Temperature , Time Factors , Transcription, Genetic , Trout/genetics
19.
Exp Cell Res ; 260(2): 304-12, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11035925

ABSTRACT

Whether drug-selectable genes can influence expression of the beta-globin gene linked to its LCR was assessed here. With the tkNeo gene placed in cis and used to select transfected cells, the beta-globin gene was expressed fourfold lower when it was positioned upstream of the LCR rather than downstream. This difference did not occur when the pgkPuro gene replaced tkNeo. Moreover, the beta-globin gene situated upstream of the LCR was transcribed without position effects when it was cotransfected with a pgkPuro-containing plasmid, whereas cotransfection with a tkNeo plasmid gave measurable position effects. Previous results from transfected cells selected via a linked tkNeo gene suggested that the 3' end of the beta-globin gene has no impact on LCR-enhanced expression. Here, removal of the 3' end of the beta-globin gene resulted in lower and much more variable expression in both transgenic mice and cells cotransfected with pgkPuro. Together, the results suggest that tkNeo, but not pgkPuro, can strongly influence expression of the beta-globin gene linked to its LCR. The findings could partly explain why data on beta-globin gene regulation obtained from transfected cells have often not agreed with those obtained using transgenic mice. Hence, one must be careful in choosing a drug-selectable gene for cell transfection studies.


Subject(s)
Gene Expression Regulation , Globins/genetics , Locus Control Region , Neomycin/pharmacology , Phosphoglycerate Kinase/genetics , Plasmids , Puromycin/pharmacology , Thymidine Kinase/genetics , Animals , Mice , Mice, Transgenic , Transfection
20.
Pediatr Res ; 42(3): 327-34, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9284273

ABSTRACT

Sodium reabsorption by the amiloride-sensitive sodium channel of epithelial cells plays a crucial role in the management of ionic composition and fluid volume in the body. In the respiratory system, sodium transport is involved in the clearance of pulmonary edema and of liquid secreted during fetal life at birth. We have cloned a partial cDNA of the alpha subunit of the mouse amiloride-sensitive sodium channel (alpha mENaC). In the region of comparison, the mouse alpha subunit shows 92% identity at the DNA level and 95% identity at the amino acid level with the rat sequence. The kidneys, lungs, and distal colon are major sites of expression of a 3.5-kb alpha mENaC mRNA. During mouse development, alpha mENaC transcripts appear late during gestation (d 17.5) and are expressed continuously thereafter. In the distal colon, a short 1.2-kb mRNA deleted of the 5' part of the transcript is detected during gestation and is replaced gradually by the mature 3.5-kb transcript after birth. Alpha mENaC and alpha1 Na+-K+-ATPase mRNAs have an expression profile that is modulated similarly during development for a given tissue. The expression of alpha mENaC transcripts increases transiently in the lungs at birth (2.5-fold), as for alpha1 Na+-K+-ATPase mRNAs (1.5-fold), suggesting that the expression of several components of the sodium transport system is modulated in the lungs at that time. In the kidney, there is no significant increase of alpha mENaC and alpha1 Na+-K+-ATPase mRNAs in newborns.


Subject(s)
Animals, Newborn/genetics , Fetus/chemistry , Sodium Channels , Sodium Channels/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Colon/chemistry , Colon/physiology , DNA Probes/genetics , Epithelial Sodium Channels , Female , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Gestational Age , Kidney/chemistry , Kidney/physiology , Lung/chemistry , Lung/physiology , Mice , Mice, Inbred Strains , Molecular Sequence Data , Polymerase Chain Reaction/methods , Pregnancy , RNA, Messenger/analysis , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Sodium Channels/chemistry , Sodium Channels/physiology , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/physiology , Transcription, Genetic/genetics , Transcription, Genetic/physiology
SELECTION OF CITATIONS
SEARCH DETAIL