Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Hum Mol Genet ; 22(22): 4562-78, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23804750

ABSTRACT

Duchenne muscular dystrophy (DMD) is a neuromuscular disease caused by mutations in the dystrophin gene. The subcellular mechanisms of DMD remain poorly understood and there is currently no curative treatment available. Using a Caenorhabditis elegans model for DMD as a pharmacologic and genetic tool, we found that cyclosporine A (CsA) reduces muscle degeneration at low dose and acts, at least in part, through a mitochondrial cyclophilin D, CYN-1. We thus hypothesized that CsA acts on mitochondrial permeability modulation through cyclophilin D inhibition. Mitochondrial patterns and dynamics were analyzed, which revealed dramatic mitochondrial fragmentation not only in dystrophic nematodes, but also in a zebrafish model for DMD. This abnormal mitochondrial fragmentation occurs before any obvious sign of degeneration can be detected. Moreover, we demonstrate that blocking/delaying mitochondrial fragmentation by knocking down the fission-promoting gene drp-1 reduces muscle degeneration and improves locomotion abilities of dystrophic nematodes. Further experiments revealed that cytochrome c is involved in muscle degeneration in C. elegans and seems to act, at least in part, through an interaction with the inositol trisphosphate receptor calcium channel, ITR-1. Altogether, our findings reveal that mitochondria play a key role in the early process of muscle degeneration and may be a target of choice for the design of novel therapeutics for DMD. In addition, our results provide the first indication in the nematode that (i) mitochondrial permeability transition can occur and (ii) cytochrome c can act in cell death.


Subject(s)
Cyclophilins/metabolism , Cyclosporine/pharmacology , Cytochromes c/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mitochondrial Dynamics , Muscular Dystrophy, Animal/drug therapy , Muscular Dystrophy, Duchenne/metabolism , Animals , Animals, Genetically Modified , Binding Sites , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Death , Cyclophilins/antagonists & inhibitors , Cytochromes c/genetics , Gene Knockdown Techniques , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Methazolamide/pharmacology , Mitochondrial Dynamics/drug effects , Mitochondrial Dynamics/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Phylogeny , Sequence Homology , Zebrafish/embryology , Zebrafish/genetics
2.
Mol Biol Cell ; 24(8): 1232-49, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23427270

ABSTRACT

In vertebrates, zyxin is a LIM-domain protein belonging to a family composed of seven members. We show that the nematode Caenorhabditis elegans has a unique zyxin-like protein, ZYX-1, which is the orthologue of the vertebrate zyxin subfamily composed of zyxin, migfilin, TRIP6, and LPP. The ZYX-1 protein is expressed in the striated body-wall muscles and localizes at dense bodies/Z-discs and M-lines, as well as in the nucleus. In yeast two-hybrid assays ZYX-1 interacts with several known dense body and M-line proteins, including DEB-1 (vinculin) and ATN-1 (α-actinin). ZYX-1 is mainly localized in the middle region of the dense body/Z-disk, overlapping the apical and basal regions containing, respectively, ATN-1 and DEB-1. The localization and dynamics of ZYX-1 at dense bodies depend on the presence of ATN-1. Fluorescence recovery after photobleaching experiments revealed a high mobility of the ZYX-1 protein within muscle cells, in particular at dense bodies and M-lines, indicating a peripheral and dynamic association of ZYX-1 at these muscle adhesion structures. A portion of the ZYX-1 protein shuttles from the cytoplasm into the nucleus, suggesting a role for ZYX-1 in signal transduction. We provide evidence that the zyx-1 gene encodes two different isoforms, ZYX-1a and ZYX-1b, which exhibit different roles in dystrophin-dependent muscle degeneration occurring in a C. elegans model of Duchenne muscular dystrophy.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/metabolism , Dystrophin/metabolism , Muscles/metabolism , Zyxin/physiology , Actinin/metabolism , Amino Acid Sequence , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans Proteins/chemistry , Gene Expression , Molecular Sequence Data , Muscles/cytology , Organ Specificity , Phylogeny , Protein Isoforms/chemistry , Protein Isoforms/physiology , Protein Transport , Sequence Homology, Amino Acid , Zyxin/chemistry
3.
PLoS One ; 7(2): e30482, 2012.
Article in English | MEDLINE | ID: mdl-22347378

ABSTRACT

Methods that use homologous recombination to engineer the genome of C. elegans commonly use strains carrying specific insertions of the heterologous transposon Mos1. A large collection of known Mos1 insertion alleles would therefore be of general interest to the C. elegans research community. We describe here the optimization of a semi-automated methodology for the construction of a substantial collection of Mos1 insertion mutant strains. At peak production, more than 5,000 strains were generated per month. These strains were then subject to molecular analysis, and more than 13,300 Mos1 insertions characterized. In addition to targeting directly more than 4,700 genes, these alleles represent the potential starting point for the engineered deletion of essentially all C. elegans genes and the modification of more than 40% of them. This collection of mutants, generated under the auspices of the European NEMAGENETAG consortium, is publicly available and represents an important research resource.


Subject(s)
Caenorhabditis elegans/genetics , DNA Transposable Elements , DNA-Binding Proteins , Genetic Engineering/methods , Genome/genetics , Recombination, Genetic , Transposases , Animals , Animals, Genetically Modified , Homologous Recombination , Mutagenesis, Insertional , Research
4.
Methods ; 56(1): 103-13, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22041718

ABSTRACT

We previously reported the use of the cheap and fast-growing nematode Caenorhabditis elegans to search for molecules, which reduce muscle degeneration in a model for Duchenne Muscular Dystrophy (DMD). We showed that Prednisone, a steroid that is generally prescribed as a palliative treatment to DMD patients, also reduced muscle degeneration in the C. elegans DMD model. We further showed that this strategy could lead to the discovery of new and unsuspected small molecules, which have been further validated in a mammalian model of DMD, i.e. the mdx mouse model. These proof-of-principles demonstrate that C. elegans can serve as a screening tool to search for drugs against neuromuscular disorders. Here, we report and discuss two methodologies used to screen chemical libraries for drugs against muscle disorders in C. elegans. We first describe a manual method used to find drugs against DMD. We further present a semi-automated method, which is currently in use for the search of drugs against the Schwartz-Jampel Syndrome (SJS). Both assays are simple to implement and can be readily transposed and/or adapted to screens against other muscle/neuromuscular diseases, which can be modeled in the worm. Finally we discuss, with respect to our experience and knowledge, the different parameters that have to be taken into account before choosing one or the other method.


Subject(s)
Caenorhabditis elegans/drug effects , Drug Evaluation, Preclinical/methods , Small Molecule Libraries/pharmacology , Animals , Automation , Disease Models, Animal , Humans , Muscular Dystrophy, Animal , Osteochondrodysplasias
5.
PLoS One ; 6(5): e19937, 2011.
Article in English | MEDLINE | ID: mdl-21611156

ABSTRACT

Determining the sub-cellular localization of a protein within a cell is often an essential step towards understanding its function. In Caenorhabditis elegans, the relatively large size of the body wall muscle cells and the exquisite organization of their sarcomeres offer an opportunity to identify the precise position of proteins within cell substructures. Our goal in this study is to generate a comprehensive "localizome" for C. elegans body wall muscle by GFP-tagging proteins expressed in muscle and determining their location within the cell. For this project, we focused on proteins that we know are expressed in muscle and are orthologs or at least homologs of human proteins. To date we have analyzed the expression of about 227 GFP-tagged proteins that show localized expression in the body wall muscle of this nematode (e.g. dense bodies, M-lines, myofilaments, mitochondria, cell membrane, nucleus or nucleolus). For most proteins analyzed in this study no prior data on sub-cellular localization was available. In addition to discrete sub-cellular localization we observe overlapping patterns of localization including the presence of a protein in the dense body and the nucleus, or the dense body and the M-lines. In total we discern more than 14 sub-cellular localization patterns within nematode body wall muscle. The localization of this large set of proteins within a muscle cell will serve as an invaluable resource in our investigation of muscle sarcomere assembly and function.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Muscles/metabolism , Animals , Caenorhabditis elegans/cytology , Cloning, Molecular , Genome/genetics , Green Fluorescent Proteins/metabolism , Humans , Mitochondrial Proteins/metabolism , Muscles/cytology , Open Reading Frames/genetics , Phenotype , Protein Transport , RNA Interference , Recombinant Fusion Proteins/metabolism , Subcellular Fractions/metabolism
6.
Neuromuscul Disord ; 21(5): 313-27, 2011 May.
Article in English | MEDLINE | ID: mdl-21392993

ABSTRACT

Duchenne muscular dystrophy, a genetic disease caused by the absence of functional dystrophin, remains without adequate treatment. Although great hopes are attached to gene and cell therapies, identification of active small molecules remains a valid option for new treatments. We have studied the effect of 20 approved pharmaceutical compounds on the muscles of dystrophin-deficient mdx5Cv mice. These compounds were selected as the result of a prior screen of 800 approved molecules on a dystrophin mutant of the invertebrate animal model Cænorhabditis elegans. Drugs were administered to the mice through maternal feeding since 2weeks of life and mixed in their food after the 3rd week of life. The effects of the drugs on mice were evaluated both at 6weeks and 16weeks. Each drug was tested at two concentrations. Prednisone was added to the molecule list as a positive control. To investigate treatment efficiency, more than 30 histological, biochemical and functional parameters were recorded. This extensive study reveals that tricyclics (Imipramine and Amitriptyline) are beneficial to the fast muscles of mdx mice. It also highlights a great variability of responses according to time, muscles and assays.


Subject(s)
Drug Approval/methods , Drug Evaluation, Preclinical/methods , Muscular Dystrophy, Duchenne/drug therapy , Animals , Creatine Kinase/blood , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Compounding , Dystrophin/genetics , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Contraction/drug effects , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/physiology , Muscles/drug effects , Muscles/pathology , Muscular Dystrophy, Duchenne/blood , Muscular Dystrophy, Duchenne/pathology
7.
Aging Cell ; 10(1): 39-54, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21040400

ABSTRACT

Dietary restriction (DR) is the most universal intervention known to extend animal lifespan. DR also prevents tumor development in mammals, and this effect requires the tumor suppressor PTEN. However, the metabolic and cellular processes that underly the beneficial effects of DR are poorly understood. We identified slcf-1 in an RNAi screen for genes that extend Caenorhabditis elegans lifespan in a PTEN/daf-18-dependent manner. We showed that slcf-1 mutation, which increases average lifespan by 40%, mimics DR in worms fed ad libitum. An NMR-based metabolomic characterization of slcf-1 mutants revealed lower lipid levels compared to wild-type animals, as expected for dietary-restricted animals, but also higher pyruvate content. Epistasis experiments and metabolic measurements support a model in which the long lifespan of slcf-1 mutants relies on increased mitochondrial pyruvate metabolism coupled to an adaptive response to oxidative stress. This response requires DAF-18/PTEN and the previously identified DR effectors PHA-4/FOXA, HSF-1/HSF1, SIR-2.1/SIRT-1, and AMPK/AAK-2. Overall, our data show that pyruvate homeostasis plays a central role in lifespan control in C. elegans and that the beneficial effects of DR results from a hormetic mechanism involving the mitochondria. Analysis of the SLCF-1 protein sequence predicts that slcf-1 encodes a plasma membrane transporter belonging to the conserved monocarboxylate transporter family. These findings suggest that inhibition of this transporter homolog in mammals might also promote a DR response.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Longevity/genetics , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Monocarboxylic Acid Transporters/genetics , Mutation/physiology , Pyruvic Acid , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/physiology , Caloric Restriction , Epistasis, Genetic/physiology , High-Throughput Screening Assays , Metabolism/genetics , Mitochondria/genetics , Mitochondria/metabolism , Monocarboxylic Acid Transporters/metabolism , Oxidative Stress , PTEN Phosphohydrolase/physiology , Pyruvate Dehydrogenase Complex/metabolism , Pyruvic Acid/metabolism , RNA Interference , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Br J Pharmacol ; 160(2): 204-16, 2010 May.
Article in English | MEDLINE | ID: mdl-20423335

ABSTRACT

Current high-throughput screening methods for drug discovery rely on the existence of targets. Moreover, most of the hits generated during screenings turn out to be invalid after further testing in animal models. To by-pass these limitations, efforts are now being made to screen chemical libraries on whole animals. One of the most commonly used animal model in biology is the murine model Mus musculus. However, its cost limit its use in large-scale therapeutic screening. In contrast, the nematode Caenorhabditis elegans, the fruit fly Drosophila melanogaster, and the fish Danio rerio are gaining momentum as screening tools. These organisms combine genetic amenability, low cost and culture conditions that are compatible with large-scale screens. Their main advantage is to allow high-throughput screening in a whole-animal context. Moreover, their use is not dependent on the prior identification of a target and permits the selection of compounds with an improved safety profile. This review surveys the versatility of these animal models for drug discovery and discuss the options available at this day.


Subject(s)
Drug Delivery Systems , Drug Design , High-Throughput Screening Assays/methods , Animals , Drug Discovery , Humans , Models, Animal , Small Molecule Libraries
10.
Hum Mol Genet ; 18(21): 4089-101, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19648295

ABSTRACT

Duchenne Muscular Dystrophy is an inherited muscle degeneration disease for which there is still no efficient treatment. However, compounds active on the disease may already exist among approved drugs but are difficult to identify in the absence of cellular models. We used the Caenorhabditis elegans animal model to screen a collection of 1000 already approved compounds. Two of the most active hits obtained were methazolamide and dichlorphenamide, carbonic anhydrase inhibitors widely used in human therapy. In C. elegans, these drugs were shown to interact with CAH-4, a putative carbonic anhydrase. The therapeutic efficacy of these compounds was further validated in long-term experiments on mdx mice, the mouse model of Duchenne Muscular Dystrophy. Mice were treated for 120 days with food containing methazolamide or dichlorphenamide at two doses each. Musculus tibialis anterior and diaphragm muscles were histologically analyzed and isometric muscle force was measured in M. extensor digitorum longus. Both substances increased the tetanic muscle force in the treated M. extensor digitorum longus muscle group, dichlorphenamide increased the force significantly by 30%, but both drugs failed to increase resistance of muscle fibres to eccentric contractions. Histological analysis revealed a reduction of centrally nucleated fibers in M. tibialis anterior and diaphragm in the treated groups. These studies further demonstrated that a C. elegans-based screen coupled with a mouse model validation strategy can lead to the identification of potential pharmacological agents for rare diseases.


Subject(s)
Carbonic Anhydrase Inhibitors/pharmacology , Disease Models, Animal , Dystrophin/deficiency , Muscular Dystrophy, Animal/prevention & control , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Carbonic Anhydrase Inhibitors/metabolism , Carbonic Anhydrases/genetics , Carbonic Anhydrases/metabolism , Dichlorphenamide/pharmacology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Dystrophin/genetics , Humans , Methazolamide/pharmacology , Mice , Mice, Inbred mdx , Motor Activity , Muscle Contraction/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/physiopathology , RNA Interference , Time Factors
11.
J Proteome Res ; 8(5): 2542-50, 2009 May.
Article in English | MEDLINE | ID: mdl-19267476

ABSTRACT

In this study, we present a methodology for metabotyping of C. elegans using 1H high resolution magic angle spinning (HRMAS) whole-organism nuclear magnetic resonance (NMR). We demonstrate and characterize the robustness of our metabolic phenotyping method, discriminating wild-type N2 from mutant sod-1(tm776) animals, with the latter being an otherwise silent mutation, and we identify and quantify several confounding effects to establish guidelines to ensure optimal quality of the raw data across time and space. We monitor the sample stability under experimental conditions and examine variations arising from effects that can potentially confuse the biological interpretation or prevent the automation of the protocol, including sample culture (breeding of the worms by two biologists), sample preparation (freezing), NMR acquisition (acquisition by different spectroscopists, acquisition in different facilities), and the effect of the age of the animals. When working with intact model organisms, some of these exogenous effects are shown to be significant and therefore require control through experimental design and sample randomization.


Subject(s)
Caenorhabditis elegans/metabolism , Magnetic Resonance Spectroscopy/methods , Metabolome , Metabolomics/methods , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Models, Biological , Mutation
12.
Mol Biol Cell ; 19(3): 785-96, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18094057

ABSTRACT

In Caenorhabditis elegans, mutations of the dystrophin homologue, dys-1, produce a peculiar behavioral phenotype (hyperactivity and a tendency to hypercontract). In a sensitized genetic background, dys-1 mutations also lead to muscle necrosis. The dyc-1 gene was previously identified in a genetic screen because its mutation leads to the same phenotype as dys-1, suggesting that the two genes are functionally linked. Here, we report the detailed characterization of the dyc-1 gene. dyc-1 encodes two isoforms, which are expressed in neurons and muscles. Isoform-specific RNAi experiments show that the absence of the muscle isoform, and not that of the neuronal isoform, is responsible for the dyc-1 mutant phenotype. In the sarcomere, the DYC-1 protein is localized at the edges of the dense body, the nematode muscle adhesion structure where actin filaments are anchored and linked to the sarcolemma. In yeast two-hybrid assays, DYC-1 interacts with ZYX-1, the homologue of the vertebrate focal adhesion LIM domain protein zyxin. ZYX-1 localizes at dense bodies and M-lines as well as in the nucleus of C. elegans striated muscles. The DYC-1 protein possesses a highly conserved 19 amino acid sequence, which is involved in the interaction with ZYX-1 and which is sufficient for addressing DYC-1 to the dense body. Altogether our findings indicate that DYC-1 may be involved in dense body function and stability. This, taken together with the functional link between the C. elegans DYC-1 and DYS-1 proteins, furthermore suggests a requirement of dystrophin function at this structure. As the dense body shares functional similarity with both the vertebrate Z-disk and the costamere, we therefore postulate that disruption of muscle cell adhesion structures might be the primary event of muscle degeneration occurring in the absence of dystrophin, in C. elegans as well as vertebrates.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Carrier Proteins/metabolism , Cytoplasmic Structures/metabolism , Cytoskeletal Proteins/metabolism , Dystrophin/metabolism , Muscle Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Caenorhabditis elegans/ultrastructure , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Carrier Proteins/chemistry , Carrier Proteins/genetics , Conserved Sequence , Cytoplasmic Structures/ultrastructure , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Gene Expression Regulation , LIM Domain Proteins , Microscopy, Immunoelectron , Models, Biological , Molecular Sequence Data , Muscles/cytology , Muscles/metabolism , Mutation/genetics , Neurons/cytology , Neurons/metabolism , Phenotype , Protein Binding , Protein Isoforms/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Subcellular Fractions/metabolism , Zyxin
13.
Adv Exp Med Biol ; 642: 192-206, 2008.
Article in English | MEDLINE | ID: mdl-19181102

ABSTRACT

Model organisms are vital to our understanding of human muscle biology and disease. The potential of the nematode Caenorhabditis elegans, the fruitfly, Drosophila melanogaster and the zebrafish, Danio rerio, as model genetic organisms for the study of human muscle disease is discussed by examining their muscle biology, muscle genetics and development. The powerful genetic tools available with each organism are outlined. It is concluded that these organisms have already demonstrated potential in facilitating the study of muscle disease and in screening for therapeutic agents.


Subject(s)
Muscular Diseases/metabolism , Sarcomeres/metabolism , Animals , Cell Differentiation , Disease Models, Animal , Genome/genetics , Humans , Muscular Diseases/genetics , Muscular Diseases/pathology
14.
Proc Natl Acad Sci U S A ; 104(50): 19808-12, 2007 Dec 11.
Article in English | MEDLINE | ID: mdl-18077412

ABSTRACT

Assigning functions to every gene in a living organism is the next challenge for functional genomics. In fact, 85-90% of the 19,000 genes of the nematode Caenorhabditis elegans genome do not produce any visible phenotype when inactivated, which hampers determining their function, especially when they do not belong to previously characterized gene families. We used (1)H high-resolution magic angle spinning NMR spectroscopy ((1)H HRMAS-NMR) to reveal the latent phenotype associated to superoxide dismutase (sod-1) and catalase (ctl-1) C. elegans mutations, both involved in the elimination of radical oxidative species. These two silent mutations are significantly discriminated from the wild-type strain and from each other. We identify a metabotype significantly associated with these mutations involving a general reduction of fatty acyl resonances from triglycerides, unsaturated lipids being known targets of free radicals. This work opens up perspectives for the use of (1)H HRMAS-NMR as a molecular phenotyping device for model organisms. Because it is amenable to high throughput and is shown to be highly informative, this approach may rapidly lead to a functional and integrated metabonomic mapping of the C. elegans genome at the systems biology level.


Subject(s)
Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Phenotype , Animals , Caenorhabditis elegans/classification , Genome, Helminth , Genomics , Magnetic Resonance Spectroscopy , Mutation , Oxidative Stress/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
15.
J Muscle Res Cell Motil ; 28(1): 79-87, 2007.
Article in English | MEDLINE | ID: mdl-17492481

ABSTRACT

During evolution, both the architecture and the cellular physiology of muscles have been remarkably maintained. Striated muscles of invertebrates, although less complex, strongly resemble vertebrate skeletal muscles. In particular, the basic contractile unit called the sarcomere is almost identical between vertebrates and invertebrates. In vertebrate muscles, sarcomeric actin filaments are anchored to attachment points called Z-disks, which are linked to the extra-cellular matrix (ECM) by a muscle specific focal adhesion site called the costamere. In this review, we focus on the dense body of the animal model Caenorhabditis elegans. The C. elegans dense body is a structure that performs two in one roles at the same time, that of the Z-disk and of the costamere. The dense body is anchored in the muscle membrane and provides rigidity to the muscle by mechanically linking actin filaments to the ECM. In the last few years, it has become increasingly evident that, in addition to its structural role, the dense body also performs a signaling function in muscle cells. In this paper, we review recent advances in the understanding of the C. elegans dense body composition and function.


Subject(s)
Caenorhabditis elegans/ultrastructure , Cytoplasmic Structures/metabolism , Muscle Proteins/metabolism , Muscles/metabolism , Muscles/ultrastructure , Actin Cytoskeleton/metabolism , Animals , Caenorhabditis elegans/metabolism , Cytoplasmic Structures/ultrastructure , Sarcolemma/ultrastructure , Sarcomeres/metabolism , Sarcomeres/ultrastructure , Signal Transduction
16.
ACS Chem Biol ; 2(4): 231-6, 2007 Apr 24.
Article in English | MEDLINE | ID: mdl-17455900

ABSTRACT

Invertebrate animal models (mainly the nematode Caenorhabditis elegans and the fruit fly Drosophila melanogaster) are gaining momentum as screening tools in drug discovery. These organisms combine genetic amenability, low cost, and culture conditions compatible with large-scale screens. Their main advantage is to allow high-throughput screening in a physiological context. On the down side, protein divergence between invertebrates and humans causes a high rate of false negatives. Despite important limitations, invertebrate models are an imperfect yet much needed tool to bridge the gap between traditional in vitro and preclinical animal assays.


Subject(s)
Models, Animal , Pharmaceutical Preparations/isolation & purification , Animals , Caenorhabditis elegans , Disease Models, Animal , Drosophila melanogaster , Drug Design , Drug Evaluation, Preclinical , Muscular Diseases/drug therapy , Neurodegenerative Diseases/drug therapy
17.
Neuromuscul Disord ; 17(1): 56-60, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17134897

ABSTRACT

In mammals, the lack of dystrophin leads to a degeneration of skeletal muscles. It has been known for many years that this pathology can be blocked by denervation or immobilization of muscles. It is not yet clear, however, whether this suppressing effect is due to the absence of fiber contraction per se, or to other mechanisms which may be induced by such treatments. We took advantage of the genetic tools available in the animal model Caenorhabditis elegans to address this question. Using RNA interference and existing mutants, we genetically impaired the excitation-contraction cascade at specific points in a dystrophin-deficient C. elegans strain which normally undergoes extensive muscle degeneration. Our data show that reducing sarcomere contraction by slightly impairing the contraction machinery is sufficient to dramatically suppress muscle degeneration. Thus, it is the physical tension exerted on the muscle fibers which is the key deleterious event in the absence of dystrophin.


Subject(s)
Dystrophin/physiology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/physiopathology , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/physiology , Dystrophin/deficiency , Dystrophin/genetics , Models, Biological , Muscle Contraction/drug effects , Muscle Contraction/genetics , Muscle Denervation/methods , Muscle Proteins , Muscular Dystrophy, Animal/drug therapy , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/physiology , Nuclear Proteins , RNA, Double-Stranded/therapeutic use , Transcription Factors
18.
ACS Chem Biol ; 1(5): 277-8, 2006 Jun 20.
Article in English | MEDLINE | ID: mdl-17163755

ABSTRACT

Identification of bioactive molecules and their targets impedes the process of drug development. In a recent paper, a genetically tractable organism, the Caenorhabditis elegans worm, is shown to be a viable screening system in which the drug target and the pathway it activates can be readily identified.


Subject(s)
Caenorhabditis elegans/drug effects , Caenorhabditis elegans/metabolism , Calcium Channel Blockers/pharmacology , Dihydropyridines/pharmacology , Animals , Caenorhabditis elegans/genetics , Calcium Channels, L-Type/metabolism , Drug Delivery Systems/methods , Drug Evaluation, Preclinical/methods
19.
Invert Neurosci ; 6(4): 189-205, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17082916

ABSTRACT

Serotonin (5-HT) regulates a wide range of behaviors in Caenorhabditis elegans, including egg laying, male mating, locomotion and pharyngeal pumping. So far, four serotonin receptors have been described in the nematode C. elegans, three of which are G protein-coupled receptors (GPCR), (SER-1, SER-4 and SER-7), and one is an ion channel (MOD-1). By searching the C. elegans genome for additional 5-HT GPCR genes, we identified five further genes which encode putative 5-HT receptors, based on sequence similarities to 5-HT receptors from other species. Using loss-of-function mutants and RNAi, we performed a systematic study of the role of the eight GPCR genes in serotonin-modulated behaviors of C. elegans (F59C12.2, Y22D7AR.13, K02F2.6, C09B7.1, M03F4.3, F16D3.7, T02E9.3, C24A8.1). We also examined their expression patterns. Finally, we tested whether the most likely candidate receptors were able to modulate adenylate cyclase activity in transfected cells in a 5-HT-dependent manner. This paper is the first comprehensive study of G protein-coupled serotonin receptors of C. elegans. It provides a direct comparison of the expression patterns and functional roles for 5-HT receptors in C. elegans.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Gene Expression , Receptors, G-Protein-Coupled/genetics , Receptors, Serotonin/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , COS Cells , Caenorhabditis elegans Proteins/metabolism , Chlorocebus aethiops , Female , Gene Expression Profiling , Humans , Male , Molecular Sequence Data , Polymerase Chain Reaction , RNA Interference , Receptors, G-Protein-Coupled/metabolism , Receptors, Serotonin/metabolism , Reproductive Behavior , Sequence Homology, Amino Acid
20.
Genomics ; 88(5): 642-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16962739

ABSTRACT

The Caenorhabditis elegans genome contains a single dystrophin/utrophin orthologue, dys-1. Point mutations in this gene, dys-1(cx35) and dys-1(cx18), result in truncated proteins. Such mutants offer potentially valuable worm models of human Duchenne muscular dystrophy. We have used microarrays to examine genes expressed differentially between wild-type C. elegans and dys-1 mutants. We found 106 genes (115 probe sets) to be differentially expressed when the two mutants are compared to wild-type worms, 49 of which have been assigned to six functional categories. The main categories of regulated genes in C. elegans are genes encoding intracellular signalling, cell-cell communication, cell-surface, and extracellular matrix proteins; genes in these same categories have been shown by others to be differentially expressed in muscle biopsies of muscular dystrophy patients. The C. elegans model may serve as a convenient vehicle for future genetic and chemical screens to search for new drug targets.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Dystrophin/genetics , Genes, Helminth , Animals , Base Sequence , DNA Primers/genetics , DNA, Helminth/genetics , Gene Expression Profiling , Humans , Muscular Dystrophy, Duchenne/genetics , Mutation , Oligonucleotide Array Sequence Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...